Prediction of pathologic complete response prediction in patients with locally advanced esophageal squamous cell carcinoma treated with neoadjuvant immunochemotherapy: A real-world study

Authors

  • Jifeng Feng The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang province, China; Department of Thoracic Oncological Surgery, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China https://orcid.org/0000-0002-6983-1318
  • Liang Wang Department of Thoracic Oncological Surgery, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
  • Xun Yang Department of Thoracic Oncological Surgery, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China https://orcid.org/0000-0002-1056-4147
  • Qixun Chen Department of Thoracic Oncological Surgery, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
  • Xiangdong Cheng Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China

DOI:

https://doi.org/10.17305/bjbms.2022.7696

Keywords:

Esophageal squamous cell carcinoma, systemic immune-inflammation index, neutrophil lymphocyte ratio, prognostic nutritional index, lymphocyte monocyte ratio, platelet lymphocyte ratio, pathologic complete response

Abstract

As an emerging hotspot for patients with locally advanced esophageal squamous cell carcinoma (LA-ESCC), neoadjuvant immunochemotherapy (nICT) is safe and feasible. Pathological complete response (pCR) is considered to be an important therapeutic effect of neoadjuvant therapy. However, few studies have explored pCR predictors for nICT in LA-ESCC. The purpose of this study was to predict pCR after nICT in LA-ESCC by pretreatment clinical characteristics and hematological indexes. The primary endpoint was to explore the impacts on the predictors for pCR prediction. Clinical characteristics and hematological indexes including systemic immune-inflammation index (SII), neutrophil lymphocyte ratio (NLR), lymphocyte monocyte ratio (LMR), prognostic nutritional index (PNI) and platelet lymphocyte ratio (PLR) were conducted. A total of 150 LA-ESCC patients were enrolled in the current study. There were 14 (9.3%) female and 136 (90.7%) male patients. Fifty-two patients achieved pCR (34.7%). A higher pCR rate was found in low-NLR group (43.7% vs. 26.6%, P=0.028) and high-LMR group (43.8% vs. 21.3%, P=0.004), respectively. Differentiation [odds ratio (OR) =0.464, 95% confidence interval (CI)=0.259-0.830, P=0.010], LMR (OR=0.309, 95% CI=0.132-0.707, P=0.007) and cTNM (OR=0.225, 95% CI=0.115-0.441, P<0.001) were independent predictors for pCR. The nomogram for pCR prediction based on LMR, differentiation and cTNM stage had good discrimination performance and calibration coordination (C-index=0.779). The results of our study are of great significance for designing therapeutic strategies. Nomogram based on LMR, differentiation and cTNM may accurately and effectively predict pCR.

Downloads

Download data is not yet available.
Prediction of pathologic complete response prediction in patients with locally advanced esophageal squamous

Downloads

Additional Files

Published

2023-01-06

How to Cite

1.
Feng J, Wang L, Yang X, Chen Q, Cheng X. Prediction of pathologic complete response prediction in patients with locally advanced esophageal squamous cell carcinoma treated with neoadjuvant immunochemotherapy: A real-world study. Biomol Biomed [Internet]. 2023Jan.6 [cited 2023Mar.28];23(1):153-60. Available from: https://www.bjbms.org/ojs/index.php/bjbms/article/view/7696

Issue

Section

Translational and Clinical Research

Introduction

There were about 0.60 million new cases and 0.54 million deaths of esophageal cancer (EC) worldwide in 2020 [1]. EC was the sixth (320,000 new cases) leading cancer type and the fourth (300,000 deaths) most common cause of death in China in 2020 [2]. Therefore, more than half of the new cases and deaths of EC [mainly esophageal squamous cell carcinoma (ESCC), which accounts for more than 90%] occurred in China. Neoadjuvant chemotherapy (nCT) or chemoradiotherapy (nCRT) followed by surgery is the preferred therapy for locally advanced ESCC (LA-ESCC) [34]. However, the long-time survival for nCT or nCRT plus surgery in LA-ESCC is still unsatisfactory [5]. Recently, immunochemotherapy (ICT) has become one of the important regimens and has achieved remarkable results in advanced ESCC [67]. Following encouraging results in advanced ESCC, neoadjuvant ICT (nICT) has gained much attention. Clinical evidence reveal that nICT in LA-ESCC is safe and feasible [8–12].

Figure 1.: The flow diagram of selection of eligible LA-ESCC patients who received nICT followed by radical resection. Based on the inclusion and exclusion criteria, a total of 150 patients were enrolled. LA-ESCC: locally advanced esophageal squamous cell carcinoma; nICT: neoadjuvant immunochemotherapy; pCR: Pathological complete response.

Pathological complete response (pCR) is considered to be an important therapeutic effect of neoadjuvant therapy [13]. However, no reliable indicators can be used to predict pCR for neoadjuvant therapy in ESCC. Hematological indexes, such as neutrophil to lymphocyte ratio (NLR), systemic immune-inflammation index (SII), platelet to lymphocyte ratio (PLR), prognostic nutritional index (PNI), and lymphocyte to monocyte ratio (LMR), were related to pCR and prognosis in several cancers after neoadjuvant treatment [14–17]. As an emerging treatment pattern in LA-ESCC with nICT, however, there are few studies regarding predictors for pCR prediction. Recently, a study including 64 cases of ESCC explored the associations between hematological indexes and pCR in nICT [18]. Another study including 79 cases of ESCC established a nomogram to predict tumor regression grade (TRG), but not for pCR, in nICT [19]. However, the sample sizes of these two studies were small. Moreover, the predictors for pCR in nICT are still unclear in LA-ESCC patients.

The knowledge of predictors for pCR will be of great value in designing treatment strategies. Therefore, we aimed to predict pCR in nICT with pretreatment clinical characteristics and hematological indexes in LA-ESCC. Moreover, a nomogram model in LA-ESCC was also developed to predict pCR in nICT.

Materials and methods

Study design and patient selection

The study enrolled LA-ESCC patients receiving nICT (camrelizumab plus chemotherapy) followed by surgery in the period between June 2019 and December 2021. The inclusion criteria for the retrospective study involved the following: (1) ESCC confirmed by histopathology; (2) LA-ESCC with clinical TNM (cTNM) stage II-IVA; (3) neoadjuvant camrelizumab combined with chemotherapy; (4) radical resection (R0) after nICT; and (5) complete medical records. Patients with any infectious, autoimmune, or hematologic disease were excluded. Patients with other previous or synchronous cancers were also excluded. The detailed criteria are shown in Figure 1. Finally, 150 LA-ESCC patients were enrolled in the current study.

Treatment strategies

Prior to surgery, all patients in the current study received two cycles of nICT. Carboplatin (5 mg/ml per minute for the area under the curve) on day 1, albumin-bound paclitaxel (100 mg/m2) on days 1 and 8, and camrelizumab (200 mg) on day 1 were administered intravenously every three weeks. Clinical effect evaluation was performed after two therapy cycles. The open or laparo-thoracoscopic McKeown or Ivor Lewis procedures with twofield lymphadenectomy were performed within 4–6 weeks after the completion of nICT [2021]. The clinical TNM stage was determined according to the eighth AJCC/UICC TNM staging system [22]. The pCR was defined as no evidence of residual tumor cells (ypT0N0M0) [23].

Hematological indexes definition

The medical data were retrospectively collected, such as pretreatment clinical characteristics and hematological indexes from medical records. The blood indexes were obtained within one week before nICT, such as neutrophils, lymphocytes, platelets, monocytes, albumin, and so on. The pretreatment blood indexes were defined as neutrophils divided by lymphocytes (NLR), platelets divided by lymphocytes (PLR), and lymphocytes divided by monocytes (LMR), respectively. The PNI and SII were defined as follows: PNI is measured by preoperative [10×albumin (g/dl)] + [0.005 × lymphocytes (/mm3)] [16]. The SII was defined as platelets × neutrophil/lymphocytes [17]. The reportings of clinicopathological variables and hematological indicators were conducted in accordance with the REMARK guidelines [24].

Ethical statement

This study was conducted in accordance with the Declaration of Helsinki. Informed consent was signed by each patient in the current study. The ethics committee of Zhejiang Cancer Hospital approved the study (IRB-2020-183).

Statistical analysis

Statistical analyses were performed by R software (version 4.1.2), SPSS 20.0, and Medcalc 17.6. Student’s t-tests (normal distribution) or Mann–Whitney U-tests (non-normal distribution) were used for continuous variables. Chi-square or Fisher’s exact tests were carried out to analyze categorical variables. The optimum cut-off values for PLR, NLR, LMR, SII, and PNI were performed with the cutoff finder [25]. To better understand the predictive ability for pCR, the AUCs were compared by ROC curves. Odds ratios (ORs) and 95% confidence intervals (CIs) in logistic regression analyses were carried out to identify the predictors for pCR. A nomogram was built for pCR prediction and assessed the discrimination performance and calibration coordination. The internal validation was performed by a calibration curve. The decision curve analysis (DCA) and AUC were calculated to quantify the ability of pCR prediction. All tests were two-sided, and P-value <0.05 indicated statistical significance.

Data availability

The datasets used in the study are available from the corresponding author on reasonable request.

Results

Patient characteristics

A total of 150 LA-ESCC patients were recruited in this study. There were 14 (9.3%) female and 136 (90.7%) male patients. The mean age was 62.7 ± 6.7 years. Most patients were diagnosed at the stage of cT3 (65.4%), cN1 (59.3%), and cTNM III (54.7%), respectively. Fifty-two patients achieved pCR (34.7%). The detailed baseline characteristics, as well as hematological indicators are shown in Table 1.

Table 1: Baseline characteristics for 150 LA-ESCC patients
Characteristics Value
Age (mean ± SD, years) 62.7 ± 6.7
Sex (male/female, %) 136 (90.7)/14 (9.3)
ECOG-PS (0/1, %) 115 (76.7)/35 (23.3)
BMI (mean ± SD, Kg/m2) 20.16 ± 2.60
Tumor location (upper/middle/lower, %) 13 (8.7)/84 (56.0)/53 (35.3)
Differentiation (well/moderate/poor, %) 23 (15.3)/70 (46.7)/57 (38.0)
Hypertension history (yes/no, %) 41 (27.3)/109 (72.7)
Diabetes history (yes/no, %) 16 (10.7)/134 (89.3)
Smoking history (yes/no, %) 100 (66.7)/50 (33.3)
Drinking history (yes/no, %) 106 (70.7)/44 (29.6)
cT stage (T2/T3/T4a, %) 26 (17.3)/98 (65.4)/26 (17.3)
cN stage (N0/N1/N2/N3, %) 26 (17.3)/89 (59.3)/31 (20.7)/4 (2.7)
cTNM stage (II/III/Iva, %) 42 (28.0)/82 (54.7)/26 (17.3)
pCR (yes/no, %) 52 (34.7)/98 (65.3)
Inflammatory and nutritional indexes
Neutrophils (mean ± SD, 10ˆ9/L) 4.93 ± 1.71
Lymphocytes (mean ± SD, 10ˆ9/L) 1.60 ± 0.52
Monocytes (mean ± SD, 10ˆ9/L) 0.46 ± 0.16
Platelets (mean ± SD, 10ˆ9/L) 236.9 ± 73.1
Albumin (mean ± SD, g/dL) 4.11 ± 3.43
NLR (mean ± SD) 3.36 ± 1.63
PLR (mean ± SD) 162.9 ± 72.9
LMR (mean ± SD) 3.80 ± 1.55
PNI (mean ± SD) 49.05 ± 4.42
SII (mean ± SD) 828.7 ± 580.1

LA-ESCC: locally advanced esophageal squamous cell carcinoma; SD: standard deviation; ECOG-PS: eastern cooperative oncology group performance status; BMI: body mass index; TNM: tumor node metastasis; pCR: pathological complete response; NLR: neutrophil to lymphocyte ratio; PLR: platelet to lymphocyte ratio; LMR: lymphocyte to monocyte ratio; SII: systemic immune-inflammation index; PNI: prognostic nutritional index.

Baseline characteristics grouped by pCR

According to the cut-off finder, the optimal cut-off levels of PLR, NLR, LMR, PNI, and SII were 150.3, 2.935, 3.27, 50.15, and 672, respectively (Figure 2). The levels of hematological indexes grouped by pCR are shown in Figure 3A. The values of NLR (2.98 ± 1.06 vs. 3.56 ± 1.83, P = 0.015) and LMR (4.18 ± 1.60 vs. 3.60 ± 1.49, P = 0.027) in patients who achieved pCR were significantly lower and higher than those with non-pCR, respectively. Significantly higher pCR rates were found in low-NLR group (43.7% vs. 26.6%, P = 0.028) and high-LMR group (43.8% vs. 21.3%, P = 0.004) (Figure 3B). ROC curves for pCR prediction based on hematological indexes are shown in Figure 3C-D. Regarding clinical characteristics, a significantly higher pCR rate was found in well differentiation, early cT stage, early cN stage, and early cTNM stage (Table 2).

Figure 2.: The optimal cutoff value achieved for hematological indicators. (A) Distribution for NLR-, PLR-, LMR-, SII-, and PNI-based cutoff optimization. (B) Cutoff optimization for NLR, PLR, LMR, SII, and PNI by correlation with pCR prediction. (C) Waterfall plot regarding cutoff value for NLR, PLR, LMR, SII, and PNI. NLR: neutrophil lymphocyte ratio; PLR: platelet to lymphocyte ratio; LMR: lymphocyte to monocyte ratio; SII: systemic immune-inflammation; PNI: prognostic nutritional index.
Figure 3.: The violin plots, histograms, and ROC curves. (A) The violin plots about the values of hematological indicators grouped by pCR. (B) The pCR rates grouped by hematological indexes. (C) ROC curve for pCR prediction based on continuous hematological indicators. (D) ROC curve for pCR prediction based on categorical hematological indicators. pCR: Pathological complete response; NLR: Neutrophil lymphocyte ratio; PLR: Platelet to lymphocyte ratio; LMR: Lymphocyte to monocyte ratio; SII: Systemic immune-inflammation; PNI: Prognostic nutritional index.

Predictors of pCR with logistic analyses

The results of univariate and multivariate logistic regression analyses are shown in Table 3. Univariate analyses revealed that NLR, LMR, cTNM stage, cT stage, cN stage, and differentiation were the predictors of pCR. Multivariate analyses then reported that patients in high-LMR group (OR = 0.309, 95% CI = 0.132–0.707, P = 0.007), well differentiation (OR = 0.464, 95% CI = 0.259–0.830, P = 0.010) and early cTNM stage (OR = 0.225, 95% CI = 0.115–0.441, P < 0.001) were more inclined to achieve pCR after nICT.

Table 2: Comparison of the baseline characteristics in LA-ESCC grouped by pCR
pCR (n = 52) Non-pCR (n = 98) P-value
Age (≤60/>60, years) 16 (30.8)/36 (69.2) 38 (38.8)/60 (61.2) 0.331
Sex (female/male) 7 (13.5)/45 (86.5) 7 (7.1)/91 (92.9) 0.205
ECOG-PS (0/1) 40 (76.9)/12 (23.1) 75 (76.5)/23 (23.5) 0.957
BMI (≤20/>20, kg/m2) 28 (53.8)/24 (46.2) 47 (48.0)/51 (52.0) 0.493
Tumor location (U/M/L) 4 (7.7)/32(61.5)/16(30.8) 9 (9.2)/52(53.1)/37(37.7) 0.226
Differentiation (W/M/P) 15(28.9)/23(44.2)/14(26.9) 8 (8.2)/47(48.0)/43(43.8) 0.002
Hypertension history (N/Y) 42(80.8)/10 (19.2) 67(68.4)/31 (31.6) 0.105
Diabetes history (N/Y) 48 (92.3)/4 (7.7) 86 (87.8)/12 (12.2) 0.390
Smoking history (N/Y) 21 (40.4)/31 (59.6) 29 (29.6)/69 (70.4) 0.182
Drinking history (N/Y) 18 (34.6)/34 (65.4) 26 (26.5)/72 (73.5) 0.301
cT stage (T2/T3/T4a) 18 (34.6)/32 (61.5)/2 (3.9) 8 (8.2)/66 (67.3)/24 (24.5) <0.001
cN stage (N0/N1/N2/N3) 12(23.1)/35(67.3)/5(9.6)/0(0) 14(14.3)/54(55.1)/26(26.5)/4(4.1) 0.027
cTNM stage (II/III/IVa) 28 (53.8)/22 (42.3)/2 (3.9) 14 (14.3)/60 (61.2)/24 (24.5) <0.001
NLR (≤2.935/>2.935) 31 (59.6)/21 (40.4) 40 (40.8)/58 (59.2) 0.028
PLR (≤150.3/>150.3) 33 (63.5)/19 (36.5) 51 (52.0)/47 (48.0) 0.180
LMR (>3.27/≤3.27) 39 (75.0)/13 (25.0) 50 (51.0)/48 (49.0) 0.004
PNI (>50.15/≤50.15) 29 (55.8)/23 (44.2) 41 (41.8)/57 (58.2) 0.104
SII (≤672/>672) 29 (55.8)/23 (44.2) 48 (49.0)/50 (51.0) 0.428

LA-ESCC: locally advanced esophageal squamous cell carcinoma; pCR: pathological complete response; ECOG-PS: eastern cooperative oncology group performance status; BMI: body mass index; Y/N: yes/no; NLR: neutrophil to lymphocyte ratio; PLR: platelet to lymphocyte ratio; LMR: lymphocyte to monocyte ratio; PNI: prognostic nutritional index; SII: systemic immune-inflammation index; TNM: tumor node metastasis; U/M/L: upper/middle/lower; W/M/P: well/moderate/poor.

Table 3: Logistic univariate and multivariate analyses of predictors for pCR in LA-ESCC
Univariate analyses Multivariate analyses
OR (95% CI) P-value OR (95% CI) P-value
Age (years, >60/≤60) 1.425 (0.679–2.914) 0.332
Sex (male/female) 0.495 (0.163–1.494) 0.212
ECOG-PS (1/0) 0.978 (0.441–2.170) 0.957
BMI (kg/m2, >20/≤20) 0.853 (0.435–1.673) 0.644
Tumor location (L/M/U) 0.862 (0.496–1.499) 0.589
Differentiation (P/M/W) 0.449 (0.269–0.748) 0.002 0.464 (0.259–0.830) 0.010
Hypertension history (Y/N) 0.515 (0.229–1.157) 0.108
Diabetes history (Y/N) 0.597 (0.183–1.954) 0.394
Smoking history (Y/N) 0.620 (0.307–1.254) 0.184
Drinking history (Y/N) 0.682 (0.330–1.410) 0.302
cT stage (T4a/T3/T2) 0.201 (0.096–0.419) <0.001
cN stage (N+/N0) 0.462 (0.268–0.797) 0.006
cTNM stage (IVa/III/II) 0.193 (0.100–0.374) <0.001 0.225 (0.115–0.441) <0.001
NLR (>2.935/≤2.935) 0.467 (0.236–0.927) 0.029
PLR (>150.3/≤150.3) 0.625 (0.313–1.245) 0.181
LMR (≤3.27/>3.27) 0.347 (0.165–0.729) 0.005 0.309 (0.132–0.707) 0.007
PNI (≤50.15/>50.15) 0.570 (0.289–1.125) 0.105
SII (>672/≤672) 0.761 (0.387–1.496) 0.429

LA-ESCC: locally advanced esophageal squamous cell carcinoma; pCR: pathological complete response; ECOG-PS: eastern cooperative oncology group performance status; U/M/L: upper/middle/lower; W/M/P: well/moderate/poor; Y/N: yes/no; BMI: body mass index; TNM: tumor node metastasis; NLR: neutrophil lymphocyte ratio; PLR: platelet to lymphocyte ratio; LMR: lymphocyte to monocyte ratio; PNI: prognostic nutritional index; SII: systemic immune-inflammation index; OR: odds ratio; CI: confidence interval.

Establishment of a nomogram to predict pCR

A predictive nomogram in LA-ESCC with the C-index of 0.779 including differentiation, cTNM, and LMR was established to predict pCR in nICT (Figure 4A). The calibration of the nomogram was carried out internally by bootstrap sampling (n = 1000), indicating that the model was well calibrated (Figure 4B). Good predictive ability and clinical applicability of the nomogram model for pCR prediction were found in ROC curve (AUC = 0.803) and DCA curve (Figure 4C-D).

Figure 4.: Nomogram for pCR prediction. (A) A predictive nomogram with the C-index of 0.779 including differentiation, cTNM, and LMR was established. Using a calibration plot with bootstrap sampling (n = 1000), the calibration revealed an acceptable agreement regarding pCR prediction internally (B); ROC (C), and DCA (D) indicated a good clinical applicability of the model in predicting pCR. cTNM; DCA: decision curve analysis; LMR: lymphocyte monocyte ratio; pCR: pathological complete response.

Discussion

We used several pretreatment clinical characteristics and hematological indexes to predict pCR. The study initially revealed that differentiation, cTNM, and LMR were independent predictors for pCR. Moreover, a nomogram model in LA-ESCC receiving nICT may accurately and effectively predict pCR. Our study focuses on the predictors from hematological indicators in predicting pCR before nICT. The results of this study will bring an important assessment of pCR using various pretreatment indexes before nICT in LA-ESCC patients.

Neoadjuvant treatment (nCT or nCRT) was recommended by the CSCO and NCCN guidelines [34]. Immunotherapy has been a relatively modern innovation in the treatment of cancer in recent years. Based on the KEYNOTE and ATTRACTION studies, immunotherapy significantly improved the outcomes in patients with advanced ESCC. Therefore, immunotherapy was approved for first-line treatment for advanced ESCC [67]. Following encouraging results in the advanced ESCC, nICT has gained much attention. Recently, nICT has had various promising outcomes in patients with LA-ESCC, such as high R0 resection rate, low adverse effects, high pCR rate, and limited postoperative complications [8–12]. However, most published studies focused on the efficiency and safety as well as pCR of nICT. Moreover, few studies have been conducted regarding predictors of pCR in LA-ESCC receiving nICT.

It is well known that patients with pCR after neoadjuvant therapy have a significantly long time of survival. Therefore, the prediction of pCR after neoagjuvant treatment has been a research hotspot in recent years. A variety of studies on ESCC reported that NLR, LMR, and PLR were correlated with pCR and prognosis [14–17]. A study including 87 cases of LA-ESCC reported that LMR was significantly higher in pCR patients compared to non-pCR patients and confirmed as an independent predictor in nCRT [14]. Another study demonstrated that pre-nCRT NLR and post-nCRT PLR were associated with pCR in 306 ESCC patients with nCRT [15]. Moreover, similar results were also found in PLR and SII in 311 ESCC patients who received nCRT [16]. These evidence indicate that peripheral blood parameters may have a certain significance for pCR prediction in ESCC with nCRT.

Recently, a study including 64 cases of LA-ESCC explored the relations between several hematological indicators and pCR in nICT [18]. However, the study was in a small sample, and the authors only focused on the pCR prediction between baseline and post-treatment indexes. Another study established a nomogram to predict TRG, but not for pCR, in 79 cases of LA-ESCC [19]. The authors developed a TRG prediction model and revealed that the changes in albumin and pretreatment white blood cells were significantly related to TRG. However, these two studies have some limitations. Firstly, the sample sizes of these two studies were small. Secondly, the predictors for pCR in nICT are still unclear. Thirdly, these two studies analyzed the blood indexes at baseline and after nICT treatment, but ignored the possibility of nICT itself might have some influence on these indicators after nICT. Fourthly, the above two studies included different immune checkpoint inhibitors that could influence the results. Our study included only one immune checkpoint inhibitor (camrelizumab) and initially revealed that LMR was an independent predictor of pCR. Results from our study provided new insights into nICT for LA-ESCC.

The exact mechanism between LMR and pCR remains unknown. There are some hypotheses on this issue. Firstly, lymphocyte can inhibit tumor cell proliferation and migration by inducing tumor cell apoptosis, which plays an important role in tumor immune surveillance and defense [2627]. Secondly, monocytes, especially tumor-associated macrophages, can reshape extracellular matrix, inhibit specific antitumor immunity and promote tumor proliferation, angiogenesis, progression, and metastasis by generating a series of cytokines [2829]. These reasons may explain why patients with higher LMR have higher pCR rates.

Our study developed an integrative nomogram model to predict pCR. Recent studies have reported that nomogram was a good method to predict factors in various cancers [1530–32]. Ajani et al. [30] and Toxopeus et al. [31] conducted two nomograms to predict pCR after nCRT in ESCC. They indicated that clinical data analyzed using a logistic regression model had a high probability of pCR prediction. The same results were also found in our study. The nomogram based on LMR, differentiation, and cTNM stage in LA-ESCC had good discrimination performance and calibration coordination for pCR prediction in nICT. Our study allows clinicians to use the model in their daily work to predict individual pCR in LA-ESCC before nICT.

Limitations should be acknowledged. Firstly, this was a retrospective study, which required attention to bias in data selection and collection. Secondly, the current study was a single-center study. Thirdly, hematological indexes may be influenced by various other factors. Fourthly, the nomogram model lacks external validation. Fifthly, the follow-up time for the current study was too short. Therefore, there was a lack of recurrence prediction. Finally, the basic biological and mechanisms regarding hematological indicators have not been thoroughly elucidated. Although limitations existed, our model may accurately and effectively predict pCR in LA-ESCC patients receiving nICT. Results from our study provided new insights for patients with LA-ESCC receiving nICT.

Conclusion

Pretreatment LMR, differentiation, and cTNM stage are the predictors of pCR. Our study on nICT in LA-ESCC is of great significance for the current treatment. Nomogram based on LMR, differentiation, and cTNM stage in LA-ESCC may accurately and effectively predict pCR.

Acknowledgments

The authors thank all the patients and their families who participated in this study.

Conflicts of interest: The authors declare no conflicts of interest.

Funding: The study was supported by grants from Zhejiang Medical and Health Science and Technology Project (2017KY237, 2018KY022). The study was also supported by Zhejiang TCM Science and Technology Project (2020ZB036, 2021ZB034 and 2022ZB051).

References

  1. , , , , , (). Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries.. CA Cancer J Clin. https://doi.org/10.3322/caac.21660
  2. , , , , (). Changing profiles of cancer burden worldwide and in China: a secondary analysis of the global cancer statistics 2020.. Chin Med J (Engl). https://doi.org/10.1097/CM9.0000000000001474
  3. , , , , , (). Preoperative chemoradiotherapy for esophageal or junctional cancer.. N Engl J Med. https://doi.org/10.1056/NEJMoa1112088
  4. , , , , , (). A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907).. Ann Surg Oncol. https://doi.org/10.1245/s10434-011-2049-9
  5. , , , , (). Recent progress in the neoadjuvant treatment strategy for locally advanced esophageal cancer.. Cancers (Basel). https://doi.org/10.3390/cancers13205162
  6. , , , , , (). Randomized phase III KEYNOTE-181 study of pembrolizumab versus chemotherapy in advanced esophageal cancer.. J Clin Oncol. https://doi.org/10.1200/JCO.20.01888
  7. , , , , , (). Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial.. Lancet Oncol. https://doi.org/10.1016/S1470-2045(19)30626-6
  8. , , , , , (). The safety and efficacy of neoadjuvant PD-1 inhibitor with chemotherapy for locally advanced esophageal squamous cell carcinoma.. J Gastrointest Oncol. https://doi.org/10.21037/jgo-20-599
  9. , , , , , (). Efficacy and safety of neoadjuvant chemotherapy and immunotherapy in locally resectable advanced esophageal squamous cell carcinoma.. J Thorac Dis.. https://doi.org/10.21037/jtd-21-340
  10. , , , , , (). Neoadjuvant immunotherapy combined chemotherapy followed by surgery versus surgery alone for locally advanced esophageal squamous cell carcinoma: a propensity score-matched study.. Front Oncol. https://doi.org/10.3389/fonc.2021.797426
  11. , , , , , (). Neoadjuvant programmed death-1 blockade plus chemotherapy in locally advanced esophageal squamous cell carcinoma.. Ann Transl Med. https://doi.org/10.21037/atm-21-3352
  12. , , , , , (). Neoadjuvant programmed cell death 1 blockade combined with chemotherapy for resectable esophageal squamous cell carcinoma.. J Immunother Cancer. https://doi.org/10.1136/jitc-2021-003497
  13. , , , , , (). (Neo)adjuvant chemoradiotherapy is beneficial to the long-term survival of locally advanced esophageal squamous cell carcinoma: a network meta-analysis.. World J Surg. https://doi.org/10.1007/s00268-021-06301-2
  14. , , , , (). Lymphocyte-monocyte ratio as a predictive marker for pathological complete response to neoadjuvant therapy in esophageal squamous cell carcinoma.. Transl Cancer Res. https://doi.org/10.21037/tcr-19-2849
  15. , , , , , (). Prediction of pathologic response to neoadjuvant chemoradiotherapy in patients with esophageal squamous cell carcinoma incorporating hematological biomarkers.. Cancer Res Treat. https://doi.org/10.4143/crt.2020.594
  16. , , (). Predicting prognosis and adverse events by hematologic markers in patients with locally advanced esophageal squamous cell carcinoma treated with neoadjuvant chemoradiotherapy.. Cancer Manag Res. https://doi.org/10.2147/CMAR.S257058
  17. , (). Change in maximal esophageal wall thickness provides prediction of survival and recurrence in patients with esophageal squamous cell carcinoma after neoadjuvant chemoradiotherapy and surgery.. Cancer Manag Res. https://doi.org/10.2147/CMAR.S295646
  18. , , , , , (). Combining serum inflammation indexes at baseline and post treatment could predict pathological efficacy to anti-PD-1 combined with neoadjuvant chemotherapy in esophageal squamous cell carcinoma.. J Transl Med. https://doi.org/10.1186/s12967-022-03252-7
  19. , , , , , (). A clinical nomogram for predicting tumor regression grade in esophageal squamous-cell carcinoma treated with immune neoadjuvant immunotherapy.. Ann Transl Med. https://doi.org/10.21037/atm-22-78
  20. , , , , , (). Ivor Lewis vs Mckeown esophagectomy: analysis of operative outcomes from the ACS NSQIP database.. Gen Thorac Cardiovasc Surg. https://doi.org/10.1007/s11748-020-01290-w
  21. , , , , , (). Effectiveness and safety of minimally invasive Ivor Lewis and McKeown oesophagectomy in Chinese patients with stage IA-IIIB oesophageal squamous cell cancer: a multicentre, non-interventional and observational study.. Interact Cardiovasc Thorac Surg. https://doi.org/10.1093/icvts/ivaa038
  22. , , , , , (). Recommendations for pathologic staging (pTNM) of cancer of the esophagus and esophagogastric junction for the 8th edition AJCC/UICC staging manuals.. Dis Esophagus. https://doi.org/10.1111/dote.12533
  23. , , , , , (). Posttherapy pathologic stage predicts survival in patients with esophageal carcinoma receiving preoperative chemoradiation.. Cancer. https://doi.org/10.1002/cncr.20916
  24. , , , , (). Reporting recommendations for tumor marker prognostic studies (REMARK): an abridged explanation and elaboration.. J Natl Cancer Inst. https://doi.org/10.1093/jnci/djy088
  25. , , , , , (). Cutoff finder: a comprehensive and straightforward Web application enabling rapid biomarker cutoff optimization.. PLoS One. https://doi.org/10.1371/journal.pone.0051862
  26. , , (). Inflammatory markers in cancer immunotherapy.. Biology (Basel). https://doi.org/10.3390/biology10040325
  27. , (). Lymphocyte surface molecules as immune activation biomarkers.. Clin Biochem. https://doi.org/10.1016/j.clinbiochem.2015.07.099
  28. , , (). Monocyte heterogeneity and functions in cancer.. J Leukoc Biol. https://doi.org/10.1002/JLB.4RI0818-311R
  29. , , , (). Monocytes in the tumor microenvironment.. Annu Rev Pathol. https://doi.org/10.1146/annurev-pathmechdis-012418-013058
  30. , , , , , (). Clinical parameters model for predicting pathologic complete response following preoperative chemoradiation in patients with esophageal cancer.. Ann Oncol. https://doi.org/10.1093/annonc/mds210
  31. , , , , , (). Nomogram for predicting pathologically complete response after neoadjuvant chemoradiotherapy for oesophageal cancer.. Radiother Oncol. https://doi.org/10.1016/j.radonc.2015.04.028
  32. , , , , , (). Novel prognostic nomograms based on inflammation-related markers for patients with hepatocellular carcinoma underwent hepatectomy.. Cancer Res Treat. https://doi.org/10.4143/crt.2018.657