Abstract
Background
Notwithstanding that the past decade has witnessed unprecedented medical progress, gastric cancer (GC) remains a leading cause of cancer death, highlighting the need for effective prognostic markers. The Memorial Sloan Kettering Prognostic Score (MPS) has been validated as a valuable prognostic tool for patients with metastatic pancreatic adenocarcinoma (mPDAC). This study aimed to assess the prognostic value of the MPS in advanced GC.
Methods
Data from 367 patients were analyzed in the present study. The MPS for each patient was calculated based on the sum of scores based on the neutrophil‐to‐lymphocyte ratio and serum albumin levels. Multivariate analyses were performed to identify the independent clinicopathological parameters associated with overall survival (OS). Further subgroup analyses based on clinicopathological features were conducted.
Results
Patients with MPS 0 (n = 161), MPS 1 (n = 158), and MPS 2 (n = 48) exhibited significantly different OS, with a median survival duration of 20.7 (95%CI: 12.2–29.2), 14.9 (95%CI: 12.5–17.3), and 12.7 (95%CI: 9.3–16.0) months, respectively (p < 0.001). Significant differences in survival were observed among different groups of patients receiving chemotherapy (18.5 months vs. 14.7 months vs. 11.0 months, p = 0.03) or the subgroup receiving chemotherapy plus immunotherapy as first‐line treatment (32.6 months vs. 17.7 months vs. 12.7 months, p = 0.02). The MPS was identified as an independent prognostic factor in multivariate analysis. During subgroup analyses, MPS‐low (MPS 0) was consistently associated with a better prognosis than MPS‐high (MPS 1 or 2).
Conclusions
MPS is a practical, simple, and useful prognostic tool for patients with advanced GC. Further studies are warranted to validate its prognostic value in advanced GC.
Keywords: gastric cancer, memorial Sloan Kettering prognostic score, prognosis
1. INTRODUCTION
Gastric cancer (GC) is the fourth leading cause of cancer death worldwide, with over 769,000 documented deaths in 2020. 1 Although the overall prognosis has improved over the past decade, the 5‐year survival rate of GC remains poor. Given the poor specificity of early symptoms, most GC patients are diagnosed at advanced stages, requiring systemic treatment. The emergency of immune checkpoint inhibitor therapy has rapidly changed the landscape of advanced GC treatment. 2 However, the prognosis of some patients remains dismal. It is now understood that traditional prognostic markers, including CEA, 3 AFP, 4 and LDH, 5 exhibit low specificity, making them suboptimal in the modern era of immunotherapy. Although prognostic factors, including PD‐L1 expression, 6 tumor mutation burden (TMB), 7 microsatellite instability (MSI), 8 microRNA (miR/miRNA), 9 and epigenetic alterations 10 have been developed to predict prognosis in GC, biochemical markers that can effectively predict patient outcomes remain largely unknown. In addition, the widespread application of certain molecular biomarkers in clinical practice is hindered by their high cost, time‐consuming nature, and limited availability. Therefore, a cheap and easily accessible prognostic model for advanced GC patients that can predict the survival prognosis of patients is urgently required.
Various malignant tumors have been observed to originate from sites characterized by infection, chronic irritation, and inflammation. The tumor microenvironment (TME), consisting of inflammatory cells, plays a significant role in facilitating the proliferation and progression of malignant tumors. 11 , 12 Inflammatory cells, along with their secreted chemokines and cytokines, play a pivotal role in shaping the TME. These elements exert significant influences on the growth, migration, and differentiation of various cell populations within the TME, influencing the effectiveness of adaptive immunity and anti‐tumor therapies. 13 , 14 Serum albumin level serves as a vital parameter in clinical practice for evaluating the nutritional status of patients afflicted with malignant tumors. Additionally, it exhibits a close association with disease status, inflammation, and liver function. 15 Prior studies have consistently shown a strong correlation between low serum albumin levels and poor prognostic outcomes in individuals with malignant tumors. 16 , 17 , 18 From these observations, the combination of nutritional parameter and biomarker associated with inflammation may be good predictors of the survival prognosis of patients with malignant tumor.
In recent years, Lebenthal et al. introduced the Memorial Sloan Kettering Prognostic Score (MPS) as a novel scoring system to assess the prognosis of patients diagnosed with metastatic pancreatic adenocarcinoma (mPDAC), effectively stratifying patients according to their prognosis. 19 , 20 This prognostic scoring system assigns a score of either 0 or 1 to two variables: the serum albumin level (score 0 for an albumin level ≥4 g/dL, score 1 for an albumin level <4 g/dL) and the neutrophil‐to‐lymphocyte ratio (NLR; score 0 for an NLR ≤4, score 1 for an NLR >4). Three different groups of patients were identified, with a median survival of 12.9 (95%CI, 10.9–14.3), 9.0 (95%CI, 7.4–10.3), and 5.4 (95%CI, 4.3–6.6) months for MPS of 0, 1, and 2, respectively. Increasing evidence showed that the MPS may be effective for other tumor types. Wang et al. demonstrated that the NLR and serum albumin could be combined to provide additional risk stratification for ovarian cancer, 21 it has also been reported that the combination of serum albumin and NLR was significantly associated with OS and disease‐free survival (DFS) of GC patients submitted to curative surgery. 22 , 23 However, whether MPS is a predictor of prognosis of advanced GC has not yet been investigated. Therefore, we performed a retrospective study to examine the prognostic significance of the MPS on the survival outcomes of patients with advanced GC. Our aim was to develop a simple and cost‐effective model that could assist in identifying patients who may benefit from treatment.
2. METHODS
2.1. Study design and patients
This retrospective study enrolled patients diagnosed with advanced or metastatic gastric or gastro‐esophageal junction cancer at the Medical Oncology Department of the First Affiliated Hospital of Zhejiang University between January 2010 and June 2021. The inclusion criteria were as follows: (1) histologically confirmed, locally advanced, inoperable, recurrent, or metastatic gastric adenocarcinoma; (2) received no previous systemic treatment or had disease progression (PD) more than 6 months after adjuvant therapy or postoperative adjuvant chemotherapy; (3) at least one treatment efficacy assessment. The exclusion criteria were as follows: (1) pretreatment complete blood count values not obtained 2 weeks before treatment initiation; (2) double primary cancers; (3) recent operation; (4) infectious diseases; (5) incomplete medical records and missing complete blood count data. All patients received chemotherapy with or without immune checkpoint inhibitors as first‐line treatment. The chemotherapy regimens were as follows: oxaliplatin plus capecitabine (XELOX), S‐1 plus paclitaxel (SPA), oxaliplatin plus fluorouracil and leucovorin (FOLFOX), and S‐1 plus oxaliplatin (SOX). The dosage was determined according to the actual condition and preference of the patients. The protocol was approved by the Ethics Committee of Zhejiang University (reference number: 2022–871).
2.2. Data collection
Pathological and clinical features of the included patients were collected. Peripheral blood was collected in the early morning within 2 weeks before the start of treatment. Data were abstracted from an electronic medical record system. Computed tomography (CT) and/or magnetic resonance imaging (MRI) were performed at baseline and every 2 or 3 cycles. Overall survival (OS) was defined as the time from initial treatment until death. The NLR was defined as the ratio between the neutrophil and lymphocyte counts. In the MPS, two variables, the serum albumin level and NLR, were assigned a prognostic score of either 0 or 1. A score of 0 was given for an albumin level ≥4 g/dL, and a score of 1 was given for an albumin level <4 g/dL. Similarly, a score of 0 was assigned for an NLR ≤4, while a score of 1 was assigned for an NLR >4. The MPS was calculated by summing the scores of these two variables.
2.3. Statistical analysis
The Kaplan–Meier method was used to analyze OS, and the log‐rank test was used to compare differences in OS. The median follow‐up was calculated using the reverse Kaplan–Meier method. Multivariate analysis was performed using the Cox proportional hazards model (Forward LR method), with the following variables included: sex (male vs. female), age (<60 vs. ≥60), differentiation (poor vs. moderate‐well), intra‐abdominal metastasis (no vs. yes), location (GEJ vs. Non‐GEJ), history of GC surgery (no vs. yes), type of therapy (chemotherapy vs. combination therapy), history of smoke (no vs. yes), history of alcohol (no vs. yes), hypertension (no vs. yes), diabetes (no vs. yes), and the MPS. Receiver operating characteristics (ROC) curve analysis was performed, and the area under the curve (AUC) was calculated. Subsequently, calibration plots for the MPS were generated to predict the probability of 1‐year and 2‐year survival. Statistical analyses were performed on R language (R Core Team), Stata software 17.0 (Stata Corp.), SPSS 26.0 (SPSS, Inc.), and GraphPad Prism 9.0 (GraphPad Software).
3. RESULTS
3.1. Characteristics of patients
Figure 1 shows the flowchart outlining the patient selection process. Patients were excluded due to missing follow‐up information (n = 116), unavailability of pretreatment complete blood count within 2 weeks (n = 15), incomplete records (n = 83), recent operation (n = 2), and double primary cancers (n = 4). Pathological and clinical details of patients are summarized in Table 1. The median duration of treatment was 7 cycles. The median follow‐up time was 25.4(95% CI: 21.9–28.8) months, while the median OS was 17.1(95% CI: 15.1–19.2) months. Among 367 patients, the majority of patients were male (n = 267, 72.8%), had a history of GC surgery (n = 136, 37.1%) and received chemotherapy as monotherapy (267, 72.8%). Most patients had an MPS of 0 (n = 161, 43.9%), followed by MPS 1 (n = 158, 43.1%) and MPS 2 (n = 48, 13.1%). There were 45(12.3%) cases of gastroesophageal junction (GEJ) cancer and 170 (46.3%) with poor differentiation. The most common chemotherapy regimens used were SOX (36.5%) and SPA (33.8%). 98 (26.7%) patients were treated with a combination of anti–PD‐1 antibodies, and 2 (0.5%) patients received anti‐PD‐L1 antibodies (Table 1).
FIGURE 1.
Flowchart visualizing the inclusion and exclusion of study subjects.
TABLE 1.
Pathological and clinical characteristics of the patients.
Characteristic | N = 367 |
---|---|
Age | |
<60 | 169(46.0%) |
≥60 | 198(54.0%) |
Sex | |
Male | 267(72.8%) |
Female | 100(27.2%) |
Differentiation | |
Poor | 170(46.3%) |
Moderate‐well | 182(49.6%) |
Unknown | 15(4.1%) |
Location | |
GEJ | 45(12.3%) |
Non‐GEJ | 309(84.2%) |
Unknown | 13(3.5%) |
Stage | |
III | 12(3.3%) |
IV | 355(96.7%) |
Intra‐abdominal metastasis | |
Yes | 224(61.0%) |
No | 143(39.0%) |
History of GC surgery | |
Yes | 136(37.1%) |
No | 231(62.9%) |
History of smoke | |
Yes | 132(36.0%) |
No | 234(63.8%) |
Unknown | 1(0.3%) |
History of alcohol | |
Yes | 104(28.3%) |
No | 262(71.4%) |
Unknown | 1(0.3%) |
Hypertension | |
Yes | 70(19.1%) |
No | 296(80.7%) |
Unknown | 1(0.3%) |
Diabetes | |
Yes | 27(7.4%) |
No | 339(92.4%) |
Unknown | 1(0.3%) |
BMI | |
≥18.5 kg/m2 | 253(68.9%) |
<18.5 kg/m2 | 56(15.3%) |
Unknown | 58(15.8%) |
NLR | |
<4 | 106(28.9%) |
≤4 | 261(71.1%) |
Albumin | |
≥4 | 219(59.7%) |
<4 | 148(40.3%) |
MPS | |
0 | 161(43.9%) |
1 | 158(43.1%) |
2 | 48(13.1%) |
Type of first‐line therapy | |
Chemotherapy | 267(72.8%) |
Immunotherapy combined with chemotherapy | 100(27.2%) |
Chemotherapy | |
SOX | 134(36.5%) |
SPA | 124(33.8%) |
FOLFOX | 44(12.0%) |
XELOX | 64(17.4%) |
Others | 1(0.3%) |
Immunotherapy | |
Anti–PD‐L1 antibody | 2(0.5%) |
Anti–PD‐1 antibody | 98(26.7%) |
Abbreviations: FOLFOX, oxaliplatin plus fluorouracil and leucovorin; GEJ, gastroesophageal junction; GC, gastric cancer; NLR, neutrophil‐to‐lymphocyte ratio; MPS, Memorial Sloan Kettering Prognostic Score; SOX, S‐1 plus oxaliplatin, SPA, S‐1 plus paclitaxel; XELOX, oxaliplatin plus capecitabine.
3.2. Prognostic significance of the MPS in advanced GC
Patients with elevated NLR levels (>4) and reduced albumin levels (<4 g/dL) were allocated a score of 2, and those with only one of these biochemical abnormalities were allocated a score of 1. Patients with neither of these abnormalities were allocated a score of 0 (Figure 2A). Figure 2B shows the OS curve of the total 367 patients with advanced GC, according to their pretreatment MPS. The median OS was 20.7 (95%CI: 12.2–29.2), 14.9 (95%CI: 12.5–17.3), and 12.7 (95%CI: 9.3–16.0) months for MPS 0 (n = 161), MPS 1 (n = 158), and MPS 2 (n = 48) (p < 0.001). The OS rates for patients with MPS 0, 1, and 2 were 75.8%, 65.6%, and 52.0% at 1 year, while the corresponding OS rates at 2 years were 47.4%, 31.8%, and 24.1%, respectively.
FIGURE 2.
(A) Correlation analysis between the neutrophil‐to‐lymphocyte ratio (NLR) and serum albumin; (B) Kaplan–Meier curves for OS in all patients. MPS, Memorial Sloan Kettering Prognostic Score; OS, overall survival.
We then divided patients with advanced GC into two subgroups based on whether immune checkpoint inhibitors treatment was used as first‐line treatment. Similar trends were noted in subgroup analyses among different treatments. In the chemotherapy‐only subgroup, patients with MPS 0 (n = 117), 1 (n = 119) or 2 (n = 31) had significantly different prognoses, with median OS of 18.5(95%CI: 14.1–22.9), 14.7(95%CI: 13.5–15.8), and 11.0 (95%CI: 7.2–14.8) months, respectively (p = 0.03; Figure 3A). Among patients exposed to immune checkpoint inhibitors combined with chemotherapy, the median OS was 32.6 (95%CI: 15.2–50.0) months for MPS 0 (n = 44), 17.7 (95%CI: 13.1–22.3) months for MPS 1 (n = 39), and 12.7 (95%CI: 8.2–17.1) months for MPS 2 (n = 17) (p = 0.02; Figure 3B). In addition, patients were stratified into two groups based on baseline body mass index (BMI). In the BMI ≥18.5 kg/m2 subgroup, high MPS was associated with poor prognosis (n = 253; 28.2 months vs. 18.7 months vs. 12.7 months; p = 0.003; Figure 3C). In the low BMI (<18.5 kg/m2) subgroup, patients with MPS 0 had longer median OS than those with MPS 1 or 2, although no significant difference was observed (n = 56; 35.0 months vs. 14.4 months vs. 16.2 months; p = 0.05; Figure 3D). Additionally, we collected the PFS of these patients (n = 363) and observed a significantly longer PFS in patients with an MPS score of 0 compared to those with an MPS score of 1 or 2 (Figure S1). The median PFS was 8.7 (95%CI: 7.1–10.4), 7.1 (95%CI: 6.5–7.6), and 6.0 (95%CI: 4.8–7.3) months for MPS 0 (n = 158), MPS 1 (n = 157), and MPS 2 (n = 48) (p = 0.01). We also observed a similar trend in the subgroup of patients who received chemotherapy or combined therapy, although the results did not reach statistical significance (Figure S2).
FIGURE 3.
Subgroup survival analysis according to the MPS in patients with advanced GC. (A) Kaplan–Meier curves for OS in 267 patients who received chemotherapy alone; (B) Kaplan–Meier curves for OS in 100 patients who received immunotherapy plus chemotherapy; (C) Kaplan–Meier curves for OS in 253 patients with BMI ≥18.5 kg/m2; (D) Kaplan–Meier curves for OS in 56 patients with BMI <18.5 kg/m2. BMI, body mass index; MPS, Memorial Sloan Kettering Prognostic Score; OS, overall survival.
3.3. Score validation
The results of univariate and multivariate proportional hazard analyses conducted on all 367 patients between baseline clinicopathological characteristics and survival are shown in Table 2. Univariate predictors of OS were diabetes (HR: 1.64, 95% CI: 1.02–2.64; p = 0.004), history of GC surgery (HR: 0.62, 95% CI: 0.46–0.83; p = 0.001), and the MPS (p = 0.001). Multivariate analysis revealed that history of GC surgery (HR: 0.66, 95% CI: 0.49–0.89; p = 0.007) and MPS (p = 0.004) were independent prognostic factors for OS. Compared with patients with MPS 0, an elevated HR was observed for patients with MPS 1 (HR: 1.46, 95% CI: 1.07–1.99) and MPS 2 (HR: 2.01, 95% CI 1.30–3.11) during multivariate analysis.
TABLE 2.
Univariate and multivariate analysis of clinicopathologic variables in relation to OS in advanced GC patients.
Univariate analysis | Multivariate analysis | ||||||
---|---|---|---|---|---|---|---|
N | HR | 95%CI | p‐value | HR | 95%CI | p‐value | |
Sex | 367 | 0.138 | |||||
Male | Ref | ||||||
Female | 1.26 | 0.93–1.70 | |||||
Age | 367 | 0.495 | |||||
<60 | Ref | ||||||
≥60 | 0.91 | 0.69–1.20 | |||||
Differentiation | 352 | 0.46 | |||||
Poor | Ref | ||||||
Moderation‐Well | 0.9 | 0.68–1.19 | |||||
Intra‐abdominal metastasis | 367 | 0.848 | |||||
No | Ref | ||||||
Yes | 1.03 | 0.77–1.37 | |||||
History of smoke | 366 | 0.33 | |||||
No | Ref | ||||||
Yes | 1.15 | 0.87–1.54 | |||||
History of alcohol | 366 | 0.409 | |||||
No | Ref | ||||||
Yes | 1.14 | 0.84–1.54 | |||||
Hypertension | 366 | 0.092 | 0.391 | ||||
No | Ref | Ref | |||||
Yes | 1.35 | 0.95–1.91 | 1.17 | 0.81–1.69 | |||
Diabetes | 366 | 0.004 | 0.290 | ||||
No | Ref | Ref | |||||
Yes | 1.64 | 1.02–2.64 | 1.31 | 0.80–2.15 | |||
Location | 354 | 0.91 | |||||
GEJ | Ref | ||||||
Non‐GEJ | 1.03 | 0.67–1.57 | |||||
History of GC surgery | 367 | 0.001 | 0.007 | ||||
0 | Ref | Ref | |||||
1 | 0.62 | 0.46–0.83 | 0.66 | 0.49–0.89 | |||
MPS | 367 | 0.001 | 0.004 | ||||
0 | Ref | Ref | |||||
1 | 1.50 | 1.10–2.03 | 1.46 | 1.07–1.99 | |||
2 | 2.14 | 1.39–3.29 | 2.01 | 1.30–3.11 |
Abbreviations: GC, gastric cancer; GEJ, gastroesophageal junction; MPS, Memorial Sloan Kettering Prognostic Score.
The AUC of the MPS for predicting 1‐year, 2‐year, and 3‐year OS was 0.592, 0.607, and 0.638, respectively (Figure S3). Calibration plots depicting the probability for 1‐year (A) and 2‐year (B) survival are presented in Figure S2, whereby the diagonal 45° dashed line represents ideal prediction performance, the solid line indicates the performance of the MPS and vertical bars indicate 95% confidence intervals (Figure S4).
3.4. The MPS predicts outcomes in different subgroups
We further categorized the patients into two subgroups: the MPS‐low group (MPS 0) and the MPS‐high group (MPS 1 or 2). Figure 4 shows the forest plot for subgroups in all patients; significant differences in survival were observed in patients with age <65 and ≥65 years, non‐GEJ, poor and moderate‐well differentiation, no hypertension, no diabetes, no smoking, no drinking, and use of chemotherapy alone. Patients in the MPS‐low group experienced superior OS compared with the MPS‐high group across almost all subgroups.
FIGURE 4.
Hazard ratios and median OS comparing MPS categories (0 vs. 1 point and 0 vs. 2 points) in subgroups analysis. GC, gastric cancer; GEJ, gastroesophageal junction.
4. DISCUSSION
The primary objective of this study was to assess the prognostic value of the MPS in advanced GC. Consistent with the findings in mPDAC reported by Lebenthal et al., 20 our study confirmed that the MPS serves as an independent prognostic factor for patients with advanced GC, the median OS was 20.7, 14.9, and 12.7 months for MPS 0, MPS 1, and MPS 2(p < 0.001). The 2‐year OS rates were 47.4%, 31.8%, and 24.1% for patients with MPS 0, 1, and 2, respectively. Importantly, our research included patients who underwent a combination of immunotherapy and chemotherapy, and the MPS demonstrated comparable predictive value within this subgroup. Remarkably, in subgroup analyses, patients who did not meet the criteria of an NLR of 4 or higher and albumin levels of 4 g/dL or lower (MPS = 0) exhibited an excellent prognosis, in contrast to patients who met either or both of these criteria.
There is a growing body of evidence indicating that the combination of preoperative albumin concentration and the NLR holds predictive value for DFS and OS in patients with Stage I–II GC. 22 , 23 Additionally, neutrophil count and albumin level 1 month after curative surgery reflect long‐term prognosis. 24 Previous study has also shown a correlation between higher NLR and lower serum albumin levels and more advanced clinical staging in GC. 25 These data suggested that the MPS may be effective in advanced GC. Our study contributes novel evidence supporting the potential utility of the combined NLR and serum albumin as a prognostic biomarker in advanced GC, shedding light on a previously unexplored aspect. Furthermore, our study suggested that the evaluated scoring system holds prognostic significance for patients receiving first‐line immunotherapy as well. Particularly, patients with MPS 0 exhibited significantly better prognosis in this subgroup. These findings indicated the promising potential of the MPS in the era of immunotherapy and highlighted its potential clinical utility.
The cut‐off value of the NLR utilized in our scoring system is strongly supported by previous studies, including a meta‐analysis involving more than 5000 individuals, 26 as well as a large‐scale study conducted by Shimada et al. 27 The albumin cut‐off value employed in our study corresponded to the lower limit of the normal range commonly used in clinical practice. Notably, the MPS is straightforward to calculate and relies solely on readily available clinical data. Furthermore, our choice of first‐line chemotherapy regimens aligns with the recommendations outlined in the current National Comprehensive Cancer Network (NCCN) guidelines for GC, 28 enhancing the practicality of implementing this scoring system in both clinical and research settings. Given the demonstrated prognostic advantage of the MPS in mPDAC, it holds the potential to serve as a universal prognostic tool for various solid tumors, particularly in advanced stages.
The observed associations between the MPS and prognosis may be explained by the influence of heightened inflammation and compromised nutritional status. In this respect, in 1863, Virchow established that the production of inflammatory cytokines is an important step in tumor development and progression. 29 Furthermore, these cytokines stimulate tumor‐induced immune suppression, 30 permitting tumor cells to escape host immune resistance. 31 Neutrophils have been recognized as an important component of the inflammatory infiltrate in the TME. 32 Current evidence suggests neutrophils mediate angiogenesis through mitogenic and proangiogenic molecules, including elastase, prokineticin 2, and metallopeptidase 9 (MMP9). 33 On the other hand, tumor‐associated neutrophil has been proven to be induced by the immunosuppressive cytokine transforming growth factor‐beta (TGF‐β) in the cancer setting to acquire a protumorigenic (N2) phenotype, playing a role in tumor growth, invasion, angiogenesis, and immunosuppression. 34 , 35 Lymphopenia may affect all or some T or B lymphocyte subpopulations, 36 limiting the critical mechanisms of immune function against cancer. 37 T lymphocytes represent the core cells that mediate anti‐tumor immunity and are the targets of immune checkpoint therapy. 38 Various studies have shown that lymphopenia is correlated with shorter survival in different types of cancers. 39 , 40 In advanced cancers, serum albumin levels are frequently used as a laboratory parameter to indicate symptoms related to inflammatory response or cancer cachexia. 41 There is a rich literature available substantiating that a low albumin level may be associated with poor prognosis in various cancers, including GC. 17 , 42 , 43 The MPS, which combines an inflammatory marker and serum albumin, offers a comprehensive assessment of both nutritional status and inflammation, exhibiting a good performance as an independent prognostic index in mPDAC and in advanced GC. For individual patients, this scoring system can function as an initial screening tool and physicians can conveniently calculate the MPS value by extracting commonly used clinical indicators, empowering them to make more informed decisions regarding patient management promptly. While the AUC of the MPS may be comparatively low, it still possesses significant potential in clinical applications, serving as an additional supplement to the TNM staging in the clinical setting. Furthermore, the MPS deserves further investigation, as it provides a foundation for the development of more clinically valuable models.
However, the limitations of this research should be acknowledged. First of all, our findings were based on patients from a single center. Conducting a prospective multicenter study would enhance the robustness and applicability of the findings. Moreover, the study lacked pathological data and molecular characteristics of included patients, including major mismatch repair status, human epidermal growth factor receptor 2 status, and PD‐L1 expression, were insufficient. Despite the above mentioned limitations, this scoring system has huge prospects for clinical application but warrants further validation in larger populations and extension to other tumor types.
5. CONCLUSION
In the rapidly evolving landscape of tumor therapy, the need for a concise prognostic marker is paramount. Our findings indicate that the MPS represents a cost‐effective and straightforward clinical prognostic model for advanced GC. This prognostic scoring system can serve as a valuable reference for clinical decision‐making and holds promising prospects for application in medical practice. Nonetheless, it is crucial to emphasize that further studies with larger sample sizes are indispensable to validate the reliability and robustness of our findings.
AUTHOR CONTRIBUTIONS
Mingyu Wan: Data curation (equal); formal analysis (equal); writing – original draft (equal). Yongfeng Ding: Data curation (equal); formal analysis (equal); writing – original draft (equal). Xiaolu Ma: Formal analysis (equal). Xiaoyu Chen: Investigation (equal). Xin Xu: Project administration (equal). Chenyu Mao: Project administration (equal). Jiong Qian: Supervision (equal). Cheng Xiao: Investigation (equal). Haiping Jiang: Validation (equal). Yulong Zheng: Validation (equal). Lisong Teng: Conceptualization (equal). Nong Xu: Conceptualization (equal); funding acquisition (equal); writing – review and editing (equal).
FUNDING INFORMATION
This work was supported by grants from Natural Science Foundation of Zhejiang Province (LQ19H160031).
CONFLICT OF INTEREST STATEMENT
The authors report no conflicts of interest in this work.
ETHICS STATEMENT
The study was conducted in accordance with the Declaration of Helsinki, the protocol was approved by Ethics Committee of Zhejiang University (reference number: 2022–871). Individual consent was needless for this retrospective analysis.
Supporting information
Figure S1.
Figure S2.
Figure S3.
Figure S4.
ACKNOWLEDGMENTS
All the authors thank the support and assistance provided by the First Affiliated Hospital of Zhejiang University School of Medicine.
Wan M, Ding Y, Ma X, et al. The Memorial Sloan Kettering Prognostic Score: Correlation with survival in patients with advanced gastric cancer. Cancer Med. 2023;12:19656‐19666. doi: 10.1002/cam4.6608
Mingyu Wan and Yongfeng Ding have contributed equally to this work.
DATA AVAILABILITY STATEMENT
Data supported the findings of the study could be acquired from the corresponding author by reasonable request.
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(3):209‐249. [DOI] [PubMed] [Google Scholar]
- 2. Kono K, Nakajima S, Mimura K. Current status of immune checkpoint inhibitors for gastric cancer. Gastric Cancer. 2020;23(4):565‐578. [DOI] [PubMed] [Google Scholar]
- 3. Deng K, Yang L, Hu B, Wu H, Zhu H, Tang C. The prognostic significance of pretreatment serum CEA levels in gastric cancer: a meta‐analysis including 14651 patients. PloS One. 2015;10(4):e0124151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Inoue M, Sano T, Kuchiba A, Taniguchi H, Fukagawa T, Katai H. Long‐term results of gastrectomy for alpha‐fetoprotein‐producing gastric cancer. Br J Surg. 2010;97(7):1056‐1061. [DOI] [PubMed] [Google Scholar]
- 5. Fuchs CS, Muro K, Tomasek J, et al. Prognostic factor analysis of overall survival in gastric cancer from two phase III studies of second‐line ramucirumab (REGARD and RAINBOW) using pooled patient data. J Gastric Cancer. 2017;17(2):132‐144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Boger C, Behrens HM, Mathiak M, Krüger S, Kalthoff H, Röcken C. PD‐L1 is an independent prognostic predictor in gastric cancer of Western patients. Oncotarget. 2016;7(17):24269‐24283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Wang F, Wei XL, Wang FH, et al. Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemo‐refractory gastric cancer treated with toripalimab, a PD‐1 antibody in phase Ib/II clinical trial NCT02915432. Ann Oncol. 2019;30(9):1479‐1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Pietrantonio F, Miceli R, Raimondi A, et al. Individual patient data meta‐analysis of the value of microsatellite instability as a biomarker in gastric cancer. J Clin Oncol. 2019;37(35):3392‐3400. [DOI] [PubMed] [Google Scholar]
- 9. Zhang XJ, Cui L, Ye G, et al. Gastric juice microRNA‐421 is a new biomarker for screening gastric cancer. Tumor Biol. 2012;33(6):2349‐2355. [DOI] [PubMed] [Google Scholar]
- 10. Park SJ, Jang HR, Kim M, et al. Epigenetic alteration of CCDC67 and its tumor suppressor function in gastric cancer. Carcinogenesis. 2012;33(8):1494‐1501. [DOI] [PubMed] [Google Scholar]
- 11. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420(6917):860‐867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646‐674. [DOI] [PubMed] [Google Scholar]
- 13. Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor‐promoting chronic inflammation: a magic bullet? Science. 2013;339(6117):286‐291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer‐related inflammation. Nature. 2008;454(7203):436‐444. [DOI] [PubMed] [Google Scholar]
- 15. Nakanishi Y, Masuda T, Yamaguchi K, et al. Albumin–globulin ratio is a predictive biomarker of antitumor effect of anti‐PD‐1 antibody in patients with non‐small cell lung cancer. Int J Clin Oncol. 2020;25(1):74‐81. [DOI] [PubMed] [Google Scholar]
- 16. Gupta D, Lis CG. Pretreatment serum albumin as a predictor of cancer survival: a systematic review of the epidemiological literature. Nutr J. 2010;9(1):69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Oñate‐Ocaña LF, Aiello‐Crocifoglio V, Gallardo‐Rincón D, et al. Serum albumin as a significant prognostic factor for patients with gastric carcinoma. Ann Surg Oncol. 2007;14(2):381‐389. [DOI] [PubMed] [Google Scholar]
- 18. Fujii T, Tokuda S, Nakazawa Y, et al. Implications of low serum albumin as a prognostic factor of long‐term outcomes in patients with breast cancer. In Vivo. 2020;34(4):2033‐2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Lebenthal JM, Zheng J, Glare PA, O'Reilly EM, Yang AC, Epstein AS. Prognostic value of the Memorial Sloan Kettering Prognostic Score in metastatic pancreatic adenocarcinoma. Cancer. 2021;127(10):1568‐1575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Lebenthal J, Zheng J, Glare PA, O'Reilly EM, Epstein AS. Prognostic value of the Memorial Sloan Kettering (MSK) prognostic score (MPS) in pancreatic ductal adenocarcinoma (PDAC). J Clin Oncol. 2019;37(31):133. [Google Scholar]
- 21. Wang YQ, Jin C, Zheng HM, et al. A novel prognostic inflammation score predicts outcomes in patients with ovarian cancer. Clin Chim Acta. 2016;456:163‐169. [DOI] [PubMed] [Google Scholar]
- 22. Costa T, Nogueiro J, Ribeiro D, Viegas P, Santos‐Sousa H. Impact of serum albumin concentration and neutrophil‐lymphocyte ratio score on gastric cancer prognosis. Langenbecks Arch Surg. 2023;408(1):57. [DOI] [PubMed] [Google Scholar]
- 23. Sun XW, Wang J, Liu J, Chen S, Liu X. Albumin concentrations plus neutrophil lymphocyte ratios for predicting overall survival after curative resection for gastric cancer. Onco Targets Ther. 2016;9:4661‐4669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Guner A, Cho M, Kim YM, Cheong JH, Hyung WJ, Kim HI. Prognostic value of postoperative neutrophil and albumin: reassessment one month after gastric cancer surgery. Front Oncol. 2021;11:633924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Huamán MO, Cerna‐Barco J, Correa‐López LE, Beltran‐Garate B, De La Cruz ‐Vargas JA. Albumina e índice neutrófilo‐linfocito como predictores de estadío tumoral en pacientes con cáncer gástrico. Revista de la Facultad de Medicina Humana. 2020;20(2):268‐275. [Google Scholar]
- 26. Sun J, Chen X, Gao P, et al. Can the neutrophil to lymphocyte ratio be used to determine gastric cancer treatment outcomes? A systematic review and meta‐analysis. Dis Markers. 2016;2016:7862469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Shimada H, Takiguchi N, Kainuma O, et al. High preoperative neutrophil‐lymphocyte ratio predicts poor survival in patients with gastric cancer. Gastric Cancer. 2010;13(3):170‐176. [DOI] [PubMed] [Google Scholar]
- 28. Ajani JA, D'Amico TA, Bentrem DJ, et al. Gastric cancer, version 2.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2022;20(2):167‐192. [DOI] [PubMed] [Google Scholar]
- 29. Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357(9255):539‐545. [DOI] [PubMed] [Google Scholar]
- 30. Behrens C, Feng L, Kadara H, et al. Expression of interleukin‐1 receptor‐associated kinase‐1 in non‐small cell lung carcinoma and preneoplastic lesions. Clin Cancer Res. 2010;16(1):34‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Zhou Q, Bucher C, Munger ME, et al. Depletion of endogenous tumor‐associated regulatory T cells improves the efficacy of adoptive cytotoxic T‐cell immunotherapy in murine acute myeloid leukemia. Blood. 2009;114(18):3793‐3802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883‐899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16(7):431‐446. [DOI] [PubMed] [Google Scholar]
- 34. Fridlender ZG, Sun J, Kim S, et al. Polarization of tumor‐associated neutrophil phenotype by TGF‐β: “N1” versus “N2” TAN. Cancer Cell. 2009;16(3):183‐194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Piccard H, Muschel RJ, Opdenakker G. On the dual roles and polarized phenotypes of neutrophils in tumor development and progression. Crit Rev Oncol Hematol. 2012;82(3):296‐309. [DOI] [PubMed] [Google Scholar]
- 36. Ménétrier‐Caux C, Ray‐Coquard I, Blay JY, Caux C. Lymphopenia in cancer patients and its effects on response to immunotherapy: an opportunity for combination with cytokines? J Immunother Cancer. 2019;7(1):85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Rivadeneira DB, DePeaux K, Wang Y, et al. Oncolytic viruses engineered to enforce leptin expression reprogram tumor‐infiltrating T cell metabolism and promote tumor clearance. Immunity. 2019;51(3):548‐560.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Kim N, Kim HK, Lee K, et al. Single‐cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat Commun. 2020;11(1):2285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Cézé N, Thibault G, Goujon G, et al. Pre‐treatment lymphopenia as a prognostic biomarker in colorectal cancer patients receiving chemotherapy. Cancer Chemother Pharmacol. 2011;68(5):1305‐1313. [DOI] [PubMed] [Google Scholar]
- 40. Ray‐Coquard I, Cropet C, Van Glabbeke M, et al. Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res. 2009;69(13):5383‐5391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Tanriverdi O. A discussion of serum albumin level in advanced‐stage hepatocellular carcinoma: a medical oncologist's perspective. Med Oncol. 2014;31(11):282. [DOI] [PubMed] [Google Scholar]
- 42. Seve P, Ray‐Coquard I, Trillet‐Lenoir V, et al. Low serum albumin levels and liver metastasis are powerful prognostic markers for survival in patients with carcinomas of unknown primary site. Cancer. 2006;107(11):2698‐2705. [DOI] [PubMed] [Google Scholar]
- 43. Lis CG, Grutsch JF, Vashi PG, Lammersfeld CA. Is serum albumin an independent predictor of survival in patients with breast cancer? JPEN J Parenter Enteral Nutr. 2003;27(1):10‐15. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Figure S1.
Figure S2.
Figure S3.
Figure S4.
Data Availability Statement
Data supported the findings of the study could be acquired from the corresponding author by reasonable request.