Open Access

Pretherapeutic factors predicting conversion surgery in unresectable pancreatic ductal adenocarcinoma: A retrospective study

  • Authors:
    • Toshiya Higashi
    • Katsutoshi Murase
    • Daichi Watanabe
    • Takuma Ishihara
    • Ryoma Yokoi
    • Masashi Kuno
    • Masahiro Fukada
    • Takuji Iwashita
    • Jesse Yu Tajima
    • Shigeru Kiyama
    • Yoshihiro Tanaka
    • Naoki Okumura
    • Masahito Shimizu
    • Nobuhisa Matsuhashi
  • View Affiliations

  • Published online on: February 22, 2024     https://doi.org/10.3892/ol.2024.14304
  • Article Number: 171
  • Copyright: © Higashi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Recently, conversion surgery (CS) has been reported to improve the prognosis in patients with unresectable pancreatic ductal adenocarcinoma (UR‑PDAC) with a favorable response to intense chemotherapy or chemoradiotherapy. However, few pretherapeutic parameters predict the attainability of CS in patients with UR‑PDAC. The present study aimed to explore the pretherapeutic predictors for the attainability of CS in patients with UR‑PDAC. The present study retrospectively evaluated 130 patients with UR‑PDAC treated at Gifu University Hospital (Gifu, Japan) from January 2015 to December 2021. Survival analysis was performed using the Simon and Makuch‑modified Kaplan‑Meier method. The hazard ratio (HR) was estimated using a time‑varying Cox regression model. The association between each predictor and CS was evaluated using the univariate analysis and age‑adjusted Fine‑Gray sub‑distribution hazard model. The bootstrap bias‑corrected area under the receiver operating characteristic curve analysis for predicting CS was used to assess the cut‑off values for each predictor. The cumulative incidence rate was calculated with CS as the outcome when divided into two groups based on the cut‑off value of each pretherapeutic predictor. Among the 130 patients included in the analysis, only 14 (11%) underwent CS. The median survival time was significantly longer in patients who underwent CS compared with patients without CS (56.3 vs. 14.1 months; P<0.001). The age‑adjusted Fine‑Gray sub‑distribution hazard regression showed that the total protein (TP) [HR 2.81, 95% confidence interval (CI) 1.19‑6.65; P=0.018], neutrophil‑to‑lymphocyte ratio (NLR) (HR 0.53, 95% CI 0.31‑0.90; P=0.020), and lymphocyte‑to‑monocyte ratio (LMR) (HR 1.28, 95% CI 1.07‑1.53; P=0.006) were significantly associated with CS. Moreover, TP ≥6.8, NLR <2.84 and LMR ≥3.87 were associated with a higher cumulative incidence of CS. In conclusion, pretherapeutic TP, NLR and LMR are clinically feasible biomarkers for predicting the attainability of CS in patients with UR‑PDAC.

Introduction

Globally, pancreatic ductal adenocarcinoma (PDAC) is the seventh leading cause of cancer-related deaths among men and women (1). Unfortunately, 80–85% of patients present with unresectable PDAC (UR-PDAC) [unresectable locally advanced cancer (UR-LA) or unresectable cancer with distant metastasis (UR-M)] and have a poor prognosis (2). However, the recent implementation of intense regimens, such as FOLFIRINOX (3) and gemcitabine + nab-paclitaxel (GnP) (4), has provided better clinical response rates, and the number of patients who can expect long-term survival is increasing. Moreover, the number of long-term survival cases after such intense chemotherapy or chemoradiotherapy followed by conversion surgery (CS) has increased in recent years (5). Currently, the National Comprehensive Cancer Network Guidelines (6) suggest that CS is an option for patients with UR-LA if resectable after a successful multidisciplinary treatment. Conversely, some reports (7,8) have shown that CS may prolong the prognosis of UR-M patients, but the actual benefit of CS in patients with UR-M remains controversial.

Previous studies (916) have reported several prognostic factors associated with the survival time of CS or the recurrence after CS in patients with UR-PDAC. However, few pretherapeutic parameters have been reported to predict the attainability of CS in patients with UR-PDAC. Therefore, this study aimed to explore the pretherapeutic factors predicting the attainability of CS in patients with UR-PDAC. In this study, we focused on examining predictors for CS rather than the overall survival (OS). The reason was that the association of intermediate treatment, including CS, from the start of treatment to death is too strong to ensure that baseline factors are accurate in their prediction for OS. Previous reports have shown that CS is beneficial for survival in patients with UR-PDAC. Therefore, by examining factors that predict the attainability of CS, we considered that these factors could influence the prognosis of patients with UR-PDAC.

Materials and methods

Study design

This retrospective study included patients treated at a single tertiary care center (Gifu University Hospital, Gifu, Japan) from January 2015 to December 2021, who were radiologically diagnosed with UR-PDAC according to the Classification of Pancreatic Cancer by the Japan Pancreas Society (4th English Edition) (17). We excluded patients who were lost to follow-up, underwent only the best supportive care, were not diagnosed with PDAC, and patients with UR-PDAC and other malignant tumors (Fig. 1). The participants provided informed consent by an opt-out option before enrollment in the study. The study was conducted following the human and ethical principles of the Declaration of Helsinki, and the study protocol was approved by the Institutional Review Board of the Gifu University Hospital (approval number: 2022-285).

Measures

The demographic and clinical variables included age, sex, body mass index (BMI), tumor location, tumor size, carbohydrate antigen 19-9 (CA19-9), total protein (TP), albumin, C-reactive protein, hemoglobin, neutrophil, lymphocyte, monocyte, prognostic nutritional index (18), modified Glasgow prognostic score (19), neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), and platelet-to-lymphocyte ratio. These variables were measured before the initial treatment. The tumor size was measured using pretherapeutic multidetector CT. Response Evaluation Criteria in Solid Tumors version 1.1 (20) was used for radiologic tumor response evaluation.

Outcome

In this study, the primary outcome was CS, defined as surgical resection following chemotherapy or chemoradiotherapy for patients initially diagnosed with UR-PDAC. At our institute, tumor resectability is determined case-by-case through discussions among surgeons, physicians, and radiologists. We considered surgical exploration if the eligibility criteria met after chemotherapy or chemoradiotherapy were as follows: i) Decrease or normalization of tumor markers; ii) Clinical response (stable disease/partial response/complete response) on multidetector CT; iii) Technically resectable on imaging; iv) Decreased or absent accumulation of the primary tumor on fluorine-18-fluorodeoxyglucose positron emission tomography (FGD-PET); v) In cases of UR-M, metastases shrank or disappeared on MDCT with no accumulation on FGD-PET; vi) No appearance of new metastatic sites; and vii) Fine performance status (0–1).

Statistical analysis

The patient characteristics are presented as frequencies and percentages for categorical variables, and as medians with interquartile ranges or means with standard deviations for continuous variables. When comparing the OS between patients with CS and non-CS, we treated CS as a time-varying covariate to avoid immortal time bias. The survival rate was estimated using the Simon and Makuch modified Kaplan-Meier method (21,22). The hazard ratio (HR) was estimated using a time-varying Cox regression model. The association between each pretherapeutic predictor and CS was evaluated using the univariable and age-adjusted Fine-Gray sub-distribution hazard model, considering death and disease progression as competing risks. No correction for the multiplicity of statistically significant tests was performed as the analysis was exploratory. The bootstrap bias-corrected area under the receiver operating characteristic curve (AUC-ROC) was reported as a measure of the predictive performance of the pretherapeutic predictors. In total, 10,000 bootstrap samples were generated, and the AUCs obtained from each ROC were averaged to calculate the bootstrap AUC-ROC. The cut-off values for predicting the CS obtained from ROC were determined based on the Youden Index. An appropriate cut-off value for predicting CS was calculated by averaging the thresholds obtained from each ROC from the bootstrap sample. The cumulative incidence rate was calculated with CS as the outcome when divided into two groups based on the cut-off value of each pretherapeutic predictor. Fisher's exact test was performed for categorical variables. The Mann-Whitney U test was applied for the comparison of continuous variables. All P-values are two-sided, with the significance level set at P<0.05. All analyses, including only data for patients with assessed pretherapeutic predictor variables, were performed using R 4.2.2 software (The R Project for Statistical Computing).

Results

Patient characteristics

This study retrospectively analyzed a total of 130 consecutive patients with UR-PDAC who were treated at our institute. Table I summarizes the patients' characteristics; 66 patients (51%) were men, with a median age of 69 years. We used multidetector CT or laparotomy to diagnose 36 patients (28%) with UR-PDAC and 94 patients (72%) with UR-M. In patients with UR-LA, the median size of the main lesion was 3.7 cm at the initial diagnosis, and the median CA19-9 level before the initial treatment was 746 U/ml. The most frequently used first-line treatment regimen was modified FOLFIRINOX (50%), followed by GnP (31%). In patients with UR-M, the median size of the main lesion was 3.4 cm at the initial diagnosis, and the median CA19-9 level before the initial treatment was 888 U/ml. The most frequently used first-line treatment regimen was modified FOLFIRINOX (62%), followed by GnP (30%).

Table I.

Patients' characteristics.

Table I.

Patients' characteristics.

VariableTotal (n=130)UR-LA (n=36)UR-M (n=94)
Male sex, n (%)66 (51)20 (56)46 (49)
Median age, years (IQR)69 (62–74)71 (68–76)68 (62–73)
Tumor location Ph, n (%)56 (43)23 (64)33 (35)
Median tumor size at diagnosis, cm (IQR)3.5 (2.8–4.4)3.7 (2.9–4.1)3.4 (2.7–4.4)
Median CA19-9, U/ml (IQR)877 (140–5,375)746 (169–2,426)888 (124–7,793)
Mean TP, g/dl (SD)6.5 (0.6)6.6 (0.5)6.5 (0.6)
Median albumin, g/dl (IQR)3.8 (3.5–4.1)4.0 (3.5–4.3)3.8 (3.5–4.0)
Median total cholesterol, mg/dl (IQR)177 (154–208)172 (139–204)178 (158–211)
Median LDH, IU/l (IQR)186 (159–230)179 (159–220)189 (160–240)
Median CRP, mg/dl (IQR)0.45 (0.14–1.56)0.38 (0.10–1.12)0.47 (0.15–2.31)
Mean hemoglobin, g/dl (SD)12.1 (1.3)12.0 (1.4)12.1 (1.3)
Median neutrophils, cells/µl (IQR)4,200 (2,961–5,333)3,415 (2,773–4,975)4,280 (3,268–5,445)
Median lymphocytes, cells/µl (IQR)1,266 (1,000–1,528)1,369 (1,151–1,570)1,210 (972–1,508)
Median monocytes, cells/µl (IQR)401 (308–513)411 (318–532)399 (298–512)
Median platelets, ×104/µl (IQR)23.1 (18.0–29.2)20.0 (17.3–27.8)23.4 (18.5–29.2)
Median PNI IQR38.9 (35.7–41.8)40.2 (35.7–43.8)38.9 (35.7–41.3)
Modified GPS, 0/1/260/52/1817/16/343/36/15
Median NLR (IQR)3.10 (2.25–4.38)2.65 (1.95–3.66)3.42 (2.55–4.70)
Median LMR (IQR)3.16 (2.33–4.17)3.25 (2.37–4.08)3.15 (2.26–4.23)
Median PLR (IQR)178 (133–263)156 (123–223)189 (144–274)
First-line treatment
  mFFX/GnP/GEM/S-1/Other76/39/11/2/218/11/4/1/258/28/7/1/0
Best response based on imaging studies
  CR/PR/SD/PD/unknown1/45/53/29/20/14/14/7/11/31/39/22/1

[i] UR-LA, unresectable locally advanced; UR-M, unresectable metastatic; IQR, interquartile range; Ph, pancreatic head; CA19-9, carbohydrate antigen 19-9; TP, total protein; SD, standard deviation; LDH, lactate dehydrogenase; CRP, C-reactive protein; PNI, prognostic nutritional index; GPS, Glasgow prognostic score; NLR, neutrophil-to-lymphocyte ratio; LMR, lymphocyte-to-monocyte ratio; PLR, platelet-to-lymphocyte ratio; mFFX, modified FOLFIRINOX; GnP, gemcitabine + nab-paclitaxel; GEM, gemcitabine; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease.

Clinical characteristics and outcome of conversion surgery

During this study, CS was performed for six patients with UR-LA and eight patients with UR-M (Table II). The first-line treatment regimens were modified FOLFIRINOX for 12 cases and GnP for two cases. The median duration of first-line treatment was 7.4 months. Treatment responses included stable disease in four cases, partial response in nine, and complete response in one. In all cases, the preoperative CA19-9 was lower than before first-line treatment, with unresectable factors disappearing or shrinking afterwards. We performed pancreaticoduodenectomy for six patients, distal pancreatectomy (DP) for six, DP with en-bloc celiac axis resection for one, and total pancreatectomy for one patient. One patient initially presenting with liver metastases underwent partial hepatectomy. There were no residual cancer cells in the resected liver. R0 (no residual tumor) resection was achieved in 12 patients (86%), and 11 received postoperative chemotherapy. Recurrence was confirmed in eight patients (57%). The median OS in patients who underwent CS was estimated as 56.3 months and was significantly longer than that in all patients without CS (median OS of 14.1 months, HR 0.08, 95% CI 0.03–0.21; P<0.001) (Fig. 2A). In patients with UR-LA and UR-M, the CS group had significantly longer OS than the non-CS group (UR-LA: 64.9 vs. 12.3 months; HR 0.05, 95% CI 0.01–0.28; P=0.001 and UR-M: 56.3 vs. 14.2 months; HR 0.12, 95% CI 0.03–0.40; P=0.001, respectively) (Fig. 2B). The median OS from CS was estimated as 44.4 months in patients with UR-LA and 62.8 months in patients with UR-M.

Table II.

Clinical characteristics and outcome of 14 patients who underwent conversion surgery.

Table II.

Clinical characteristics and outcome of 14 patients who underwent conversion surgery.

No.Age, yearsSexTumor locationUR status (UR factor)First-line treatmentDuration, monthsRECISTCA19-9 before/after treatment, U/mlUR factor after treatmentSurgeryREvans gradePost-operative therapyRecurrence siteTime to recurrence from CS, monthsPrognosis, monthsa
172MPhLA (SMV)mFFX8.0PR1,462/43DisappearedPDR0IIbS-1Remnant pancreas20Dead (66/58)
273MPhLA (CHA, PHA)mFFX3.7SD354/89DisappearedDP-CARR0IIaNoneNone Dead (45/41)
358MPhLA (Ao)mFFX6.8PR171/12ShrankPDR0IIbS-1Local29Alive (52/45)
456FPhLA (SMA, SMV)mFFX13.4PR5,451/116ShrankPD+PVRR0IVNoneLung14Alive (56/43)
570FPhLA (CHA, PHA)GnP9.8PR228/20ShrankPD+PVR+HARR0IIbS-1Remnant pancreas18Alive (50/40)
657MPhLA (Ao)mFFX6.8PR10,890/26DisappearedPDR0IIbS-1None Alive (22/15)
772MPtM (eLN)mFFX2.8SD24,469/3,333ShrankDPR1IIamFFXLN50Dead (67/64)
863MPbM (Per)mFFX4.5SD481/370DisappearedDPR0IIaS-1Lung, LN24Dead (35/31)
959MPbM (Per)mFFX24.0PR250/17DisappearedDPR0IIamFFXPER19Dead (57/33)
1063FPbM (Liver-Multi, Bone-Multi)mFFX23.3PR4,379/81DisappearedDPR0IIbS-1None Alive (69/46)
1179MPhtM (Per)GnP4.6SD159/52DisappearedTPR1IIaS-1Liver5Dead (20/16)
1261FPhM (Liver-Multi)mFFX26.0CR10,424/36DisappearedPD+HepR0IVNoneNone Alive (63/37)
1362MPbM (eLN)mFFX9.1PR96/48ShrankDPR0IIbmFFXNone Alive (24/15)
1459MPtM (eLN)mFFX5.5PR13/12ShrankDPR0IVmFFXNone Alive (24/18)

a Months after initial treatment/after conversion surgery. Ph, pancreatic head; Pt, pancreatic tail; Pb, pancreatic body; Pht, pancreatic head and tail; UR, unresectable; LA, locally advanced lesion; SMV, superior mesenteric vein; CHA, common hepatic artery; PHA, proper hepatic artery; Ao, aorta; SMA, superior mesenteric artery; M, metastatic lesion; eLN, extraregional lymph node; Per, peritoneum; Multi, multiple metastatic sites; mFFX, modified FOLFIRINOX; GnP, gemcitabine + nab-paclitaxel; RECIST, response evaluation criteria in solid tumors; PR, partial response; SD, stable disease; CR, complete response; PD, pancreaticoduodenectomy; DP-CAR, distal pancreatectomy with en bloc celiac axis resection; PVR, portal vein resection; HAR, hepatic artery resection; DP, distal pancreatectomy; TP, total pancreatectomy; Hep, hepatectomy; R, residual tumor; R0, no microscopic residual on resected margin; R1, microscopic residual on resected margin; LN, lymph node; CS, conversion surgery; OS, overall survival.

Association between pretherapeutic parameters and the attainability of CS

Age-adjusted Fine-Gray regression showed that TP (HR 2.81; P=0.018), NLR (HR 0.53; P=0.020), and LMR (HR 1.28; P=0.006) were significant pretherapeutic markers predicting the attainability of CS (Table III). The cut-off value of each marker for CS based on the ROC curve is presented in Table IV. The optimal cut-off values for useful parameters using the bootstrap ROC curve were 6.8 (AUC=0.70), 2.84 (AUC=0.74), and 3.87 (AUC=0.72) for TP, NLR, and LMR, respectively. Fig. 3 shows the cumulative incidence of these parameters with each cut-off value using the bootstrap ROC curve. The percentage of patients who underwent CS was higher in the TP ≥6.8 (20%, 9/45), NLR <2.84 (20%, 10/51), and LMR ≥3.87 (22%, 9/41) groups compared with the TP <6.8 (6%, 5/85), NLR ≥2.84 (5%, 4/79), and LMR <3.87 (6%, 5/89) groups, respectively. In patients who underwent CS, the changes in TP, NLR, and LMR before and after first-line treatment were 0.6 (95% CI 0.28–0.86), 0.27 (95% CI-0.55–1.08), and 1.27 (95% CI 0.28–2.25), respectively. In patients who did not undergo CS, the changes in TP, NLR, and LMR between and after first-line treatment were 0.3 (95% CI 0.20–0.40), 0.31 (95% CI-0.54–1.16), and 0.25 (95% CI-0.34–0.83), respectively.

Table III.

Pretherapeutic predictors for the attainability of conversion surgery (univariate analysis and age-adjusted Fine-Gray sub-distribution hazard regression).

Table III.

Pretherapeutic predictors for the attainability of conversion surgery (univariate analysis and age-adjusted Fine-Gray sub-distribution hazard regression).

Univariate analysisAge-adjusted


VariableHR (95% CI)P-valueHR (95% CI)P-value
CA19-91.00 (1.00–1.00)0.2601.00 (1.00–1.00)0.270
TP2.59 (1.09–6.15)0.0312.81 (1.19–6.65)0.018
Albumin5.22 (0.90–30.3)0.0664.65 (0.75–28.8)0.099
Total cholesterol1.00 (0.99–1.01)0.9101.00 (0.99–1.01)0.820
LDH0.99 (0.99–1.00)0.1300.99 (0.99–1.00)0.079
CRP0.72 (0.49–1.06)0.0970.75 (0.54–1.04)0.085
Hemoglobin1.09 (0.81–1.46)0.5801.09 (0.80–1.47)0.600
Neutrophil1.00 (1.00–1.00)0.0271.00 (1.00–1.00)0.029
Lymphocyte1.00 (1.00–1.00)0.0301.00 (1.00–1.00)0.017
Monocyte1.00 (0.99–1.00)0.3101.00 (1.00–1.00)0.320
Platelet1.01 (1.00–1.01)<0.0011.01 (1.00–1.01)<0.001
PNI1.18 (1.00–1.40)0.0551.17 (0.98–1.39)0.081
Modified GPS0.68 (0.31–1.49)0.3400.75 (0.35–1.59)0.450
NLR0.53 (0.31–0.91)0.0200.53 (0.31–0.90)0.020
LMR1.27 (1.07–1.51)0.0071.28 (1.07–1.53)0.006
PLR1.00 (1.00–1.00)0.0241.00 (1.00–1.00)0.022

[i] CA19-9, carbohydrate antigen 19-9; TP, total protein; LDH, lactate dehydrogenase; CRP, C-reactive protein; PNI, prognostic nutritional index; GPS, Glasgow prognostic score; NLR, neutrophil-to-lymphocyte ratio; LMR, lymphocyte-to-monocyte ratio; PLR, platelet-to-lymphocyte ratio.

Table IV.

Cutoff value and predictive performance for each marker based on ROC curves.

Table IV.

Cutoff value and predictive performance for each marker based on ROC curves.

Usual methodBootstrap method


VariableAUC95% CISensitivitySpecificityCutoff valueAUCSensitivitySpecificityCutoff value
TP0.700.54–0.860.860.506.60.700.760.666.8
NLR0.740.61–0.870.860.553.080.740.830.652.84
LMR0.720.59–0.860.640.774.120.720.770.703.87

[i] AUC, area under the ROC curve; HR, hazard ratio; CI, confidence interval; TP, total protein; NLR, neutrophil-to-lymphocyte ratio; LMR, lymphocyte-to-monocyte ratio.

Differences in the course of treatment for each parameter

There was no difference in the first-line treatment regimens or treatment-related adverse events for each parameter (Table V). However, patients with NLR <2.84 and LMR ≥3.87 had significantly longer overall treatment time than those with NLR ≥2.84 (P=0.006) and LMR <3.87 (P<0.001), respectively.

Table V.

Summary of the course of treatment for each parameter.

Table V.

Summary of the course of treatment for each parameter.

VariableTP <6.8 (n=85)TP ≥6.8 (n=45)P-valueNLR <2.84 (n=51)NLR ≥2.84 (n=79)P-valueLMR <3.87 (n=89)LMR ≥3.87 (n=41)P-value
First-line treatment 0.335 0.359 0.286
  mFFX/GnP/51/24/0/8/225/15/2/3/0 30/16/2/3/046/23/0/8/2 53/24/1/10/123/15/1/1/1
  S-1/GEM/Other
≥Grade 3 toxicity75 (88.2)37 (82.2)0.49844 (86.3)68 (86.1)0.99977 (86.5)35 (85.4)0.999
adverse event,
n (%)
Median overall11.210.80.85413.38.50.0068.515.8<0.001
treatment time,(4.8–18.3)(3.7–20.0) (7.8–23.0)(2.8–17.5) (3.2–16.8)(8.7–28.5)
months (IQR)

[i] TP, total protein; NLR, neutrophil-to-lymphocyte ratio; LMR, lymphocyte-to-monocyte ratio; mFFX, modified FOLFIRINOX; GnP, gemcitabine + nab-paclitaxel; GEM, gemcitabine; IQR, interquartile range.

Discussion

In this study, the median OS was significantly longer in patients who underwent CS than in all patients without CS. The study also indicated that patients who underwent CS with UR-LA and UR-M had a significant difference in the median OS compared to patients who did not undergo CS. Therefore, this study demonstrated that CS significantly impacted the prognosis of patients with UR-PDAC. We examined the factors that predict the attainability of CS based on the pretherapeutic parameters in patients with UR-PDAC. As a result, TP ≥6.8, NLR <2.84, and LMR ≥3.87 were associated with a higher cumulative incidence of CS.

The three parameters evaluated in this study were associated with nutritional status and cancer-related inflammation. TP is a predictor of postoperative prognosis in lung cancer (23) and retroperitoneal sarcoma (24). TP decreases with the progression of the disease because of malnutritional and inflammatory status, cachexia, and increased intracellular catabolism by cancer cells. Furthermore, hypoproteinemia is associated with decreased tolerance to chemotherapy (25).

NLR is a leading inflammation-related marker in various cancers and is valuable for predicting the prognosis of UR-PDAC (2628). LMR also predicts poor outcomes in patients with UR-PDAC undergoing chemotherapy (29). Moreover, both NLR and LMR are associated with nutritional status (30,31). The features of cancer-related inflammation include the following: malignant cell proliferation and survival, inflammatory cell infiltration and production of inflammatory mediators in tumor tissues, tissue remodeling, promotion of tissue repair and angiogenesis, disruption of adaptive immune responses, and altered responses to chemotherapeutic agents (32). Therefore, TP, NLR, and LMR are important indicators of nutritional status, cancer progression, and tolerability of chemotherapy.

This study demonstrated that three pretherapeutic markers reflecting nutritional and inflammatory status were associated with the attainability of CS in patients with UR-PDAC during treatment. Moreover, patients with low NLR and high LMR had significantly longer overall treatment time than those with high NLR and low LMR, respectively. This may indicate that patients with better nutritional status and lower systemic inflammatory response were more likely to receive relatively long-term treatment because they tolerate chemotherapy better and their cancer progresses is slower. Consequently, these patients are often able to receive adequate doses of chemotherapy, potentially leading to CS. Nutrition and inflammation levels are known to affect the risk of severe toxicity during cancer chemotherapy and the overall progression of the disease. Malnutrition can impact the absorption, protein binding, hepatic metabolism, and renal elimination of drugs and their metabolites (33). In malnourished patients, a reduced concentration of plasma protein levels can heighten the toxicity risk from drugs that bind strongly to proteins, such as prednisolone, etoposide, cisplatinum, paclitaxel, and metabolites of irinotecan (34). Additionally, several studies have linked inflammation-based scores with the prognosis of several types of malignances (35). Inflammation is causally related to cancer development, through processes that involve genotoxicity, aberrant tissue repair, proliferative responses, invasion, and metastasis (36). Elevated systemic inflammation can also directly enhance cancer cell metastases in other parts of the body and accelerate the overall progression of the disease (15). While we found no significant link between these pre-treatment markers and adverse events during chemotherapy or chemoradiotherapy, assessing patients' nutritional and inflammatory status before surgery for UR-PDAC might be vital. This assessment aids in selecting appropriate treatments and identifying patients with advanced disease or those unsuitable for surgery, potentially improving patient outcomes.

Our study had some limitations. First, this was a retrospective study conducted at a single institution. Second, the number of enrolled patients, particularly those who underwent CS, was relatively small. As a result, the outcomes could not be separately evaluated between UR-LA and UR-M, although these two conditions might be different. Therefore, sufficiently large cohorts should be analyzed in future multicenter collaborative studies. Third, in this study, the criteria for surgical indications of CS were defined as described above. However, the surgical indications for CS have not been clearly defined currently. Furthermore, the efficacy of CS in patients with UR-M remains controversial. Further studies are needed to determine the efficacy and surgical indications for CS.

In conclusion, our findings suggest that pretherapeutic TP, NLR, and LMR in patients with UR-PDAC are predictive factors for the attainability of CS. These parameters may be useful to predict which patients with UR-PDAC will likely undergo CS during multidisciplinary therapy.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

TH, KM, DW, TIs, RY, MK, MF, TIw, JYT, SK, YT, NO, MS and NM conceived the study concept and study design. TH made the initial proposal for this study, collected and analyzed the data, and wrote and edited the manuscript. TH, KM, RY, MK, MF, TIw, JYT, SK, YT and NO treated and monitored the patients. TH, DW and TIs conducted the statistical analyses. KM, MS, and NM revised and supervised the study. TH, KM and NM confirm the authenticity of all the raw data. All authors read and approved the final manuscript, and agreed to be accountable for the content of this work.

Ethics approval and consent to participate

The study protocol was approved by the Institutional Review Board of the Gifu University Hospital (approval number: 2022-285; Gifu, Japan). Participants provided informed consent by an opt-out option before study enrollment.

Patient consent for publication

Not applicable.

Competing interests

NM received grants to his institute from Taiho Pharma and Yakult Honsha. All the other authors have no conflicts of interest.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Vincent A, Herman J, Schulick R, Hruban RH and Goggins M: Pancreatic cancer. Lancet. 378:607–620. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C, et al: FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 364:1817–1825. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, et al: Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 369:1691–1703. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Satoi S, Yamaue H, Kato K, Takahashi S, Hirono S, Takeda S, Eguchi H, Sho M, Wada K, Shinchi H, et al: Role of adjuvant surgery for patients with initially unresectable pancreatic cancer with a long-term favorable response to non-surgical anti-cancer treatments: Results of a project study for pancreatic surgery by the Japanese society of hepato-biliary-pancreatic surgery. J Hepatobiliary Pancreat Sci. 20:590–600. 2013. View Article : Google Scholar : PubMed/NCBI

6 

National Comprehensive Cancer Network, . NCCN Clinical practice guidelines in oncology: pancreatic adenocarcinoma. https://www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdfAugust 29–2023

7 

Zhou Y, Liao S, You J and Wu H: Conversion surgery for initially unresectable pancreatic ductal adenocarcinoma following induction therapy: A systematic review of the published literature. Updates Surg. 74:43–53. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Tanaka M, Heckler M, Mihaljevic AL, Sun H, Klaiber U, Heger U, Buchler MW and Hackert T: CT response of primary tumor and CA19-9 predict resectability of metastasized pancreatic cancer after FOLFIRINOX. Eur J Surg Oncol. 45:1453–1459. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Wang M, Zhu P, Chen Z and Yang L: Conversion therapy, palliative chemotherapy and surgery, which of these is the best treatment for locally advanced and advanced pancreatic cancer? Anticancer Drugs. 33:e686–e691. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Yanagimoto H, Satoi S, Yamamoto T, Yamaki S, Hirooka S, Kotsuka M, Ryota H, Ishida M, Matsui Y and Sekimoto M: Benefits of conversion surgery after multimodal treatment for unresectable pancreatic ductal adenocarcinoma. Cancers (Basel). 12:14282020. View Article : Google Scholar : PubMed/NCBI

11 

Ushida Y, Inoue Y, Oba A, Mie T, Ito H, Ono Y, Sato T, Ozak a, Sasaki T, Saiura A, et al: Optimizing indications for conversion surgery based on analysis of 454 consecutive Japanese cases with unresectable pancreatic cancer who received modified FOLFIRINOX or gemcitabine plus nab-paclitaxel: A single-center retrospective study. Ann Surg Oncol. 29:5038–5050. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Takano N, Yamada S, Sonohara F, Inokawa Y, Takami H, Hayashi M, Koike M, Fujii T and Kodera Y: The impact of early tumor shrinkage on conversion surgery and the survival in patients with unresectable locally advanced pancreatic cancer. Surg Today. 51:1099–1107. 2021. View Article : Google Scholar : PubMed/NCBI

13 

Lee M, Kang JS, Kim H, Kwon W, Lee SH, Ryu JK, Kim YT, Oh DY, Chie EK and Jang JY: Impact of conversion surgery on survival in locally advanced pancreatic cancer patients treated with FOLFIRINOX chemotherapy. J Hepatobiliary Pancreat Sci. 30:111–121. 2023. View Article : Google Scholar : PubMed/NCBI

14 

Mataki Y, Kurahara H, Idichi T, Tanoue K, Hozaka Y, Kawasaki Y, Iino S, Maemura K, Shinchi H and Ohtsuka T: Clinical benefits of conversion surgery for unresectable pancreatic ductal adenocarcinoma: A single-institution, retrospective analysis. Cancers (Basel). 13:10572021. View Article : Google Scholar : PubMed/NCBI

15 

Kokumai T, Aoki S, Mizuma M, Maeda S, Ohtsuka H, Nakagawa K, Morikawa T, Motoi F, Kamei T and Unno M: Prognostic value of an inflammation-based nutritional score for patients with initially unresectable pancreatic adenocarcinoma undergoing conversion surgery following chemo-/radiotherapy. Surg Today. 51:1682–1693. 2021. View Article : Google Scholar : PubMed/NCBI

16 

Tsuchiya N, Matsuyama R, Murakami T, Yabushita Y, Sawada Y, Kumamoto T and Endo I: Role of conversion surgery for unresectable pancreatic cancer after long-term chemotherapy. World J Surg. 44:2752–2760. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Japan Pancreas Society, . Classification of Pancreatic Carcinoma (4th English Edition). Kanehara & Co.; Tokyo, Japan: 2017, http://www.suizou.org/pdf/Classification_of_Pancreatic_Carcinoma_4th_Engl_ed.pdfAugust 30–2023

18 

Kanda M, Fujii T, Kodera Y, Nagai S, Takeda S and Nakao A: Nutritional predictors of postoperative outcome in pancreatic cancer. Br J Surg. 98:268–274. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Toiyama Y, Miki C, Inoue Y, Tanaka K, Mohri Y and Kusunoki M: Evaluation of an inflammation-based prognostic score for the identification of patients requiring postoperative adjuvant chemotherapy for stage II colorectal cancer. Exp Ther Med. 2:95–101. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, et al: New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur J Cancer. 45:228–247. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Simon R and Makuch RW: A non-parametric graphical representation of the relationship between survival and the occurrence of an event: Application to responder versus non-responder bias. Stat Med. 3:35–44. 1984. View Article : Google Scholar : PubMed/NCBI

22 

Schultz LR, Peterson EL and Breslau N: Graphing survival curve estimates for time-dependent covariates. Int J Methods Psychiatr Res. 11:68–74. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Cao J, Luo F, Zeng K, Ma W, Lu F, Huang Y, Zhang L and Zhao H: Predictive value of high preoperative serum total protein and elevated hematocrit in patients with non-small-cell lung cancer after radical resection. Nutr Cancer. 74:3533–3545. 2022. View Article : Google Scholar : PubMed/NCBI

24 

Yao Y, Wang Z, Yong L, Yao Q, Tian X, Wang T, Yang Q, Hao C and Zhou T: Longitudinal and time-to-event modeling for prognostic implications of radical surgery in retroperitoneal sarcoma. CPT Pharmacometrics Syst Pharmacol. 11:1170–1182. 2022. View Article : Google Scholar : PubMed/NCBI

25 

Abbasi B, Hayat A, Lyons M, Gupta A and Gupta S: Serum protein and electrolyte imbalances are associated with chemotherapy induced neutropenia. Heliyon. 8:e099492022. View Article : Google Scholar : PubMed/NCBI

26 

Iwai N, Okuda T, Sakagami J, Harada T, Ohara T, Taniguchi M, Sakai H, Oka K, Hara T, Tsuji T, et al: Neutrophil to lymphocyte ratio predicts prognosis in unresectable pancreatic cancer. Sci Rep. 10:187582020. View Article : Google Scholar : PubMed/NCBI

27 

Allen J, Cernik C, Bajwa S, Al-Rajabi R, Saeed A, Baranda J, Williamson S, Sun W and Kasi A: Association of neutrophil, platelet, and lymphocyte ratios with the prognosis in unresectable and metastatic pancreatic cancer. J Clin Med. 9:32832020. View Article : Google Scholar : PubMed/NCBI

28 

Terao T, Kumagi T, Hyodo I, Yokota T, Azemoto N, Miyata H, Kuroda T, Ohno Y, Tanaka Y, Shibata N, et al: Simple prognostic markers for optimal treatment of patients with unresectable pancreatic cancer. Medicine (Baltimore). 100:e275912021. View Article : Google Scholar : PubMed/NCBI

29 

Shimizu T, Taniguchi K, Asakuma M, Tomioka A, Inoue Y, Komeda K, Hirokawa F and Uchiyama K: Lymphocyte-to-monocyte ratio and prognostic nutritional index predict poor prognosis in patients on chemotherapy for unresectable pancreatic cancer. Anticancer Res. 39:2169–2176. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Kaya T, Acikgoz SB, Yildirim M, Nalbant A, Altas AE and Cinemre H: Association between neutrophil-to-lymphocyte ratio and nutritional status in geriatric patients. J Clin Lab Anal. 33:e226362019. View Article : Google Scholar : PubMed/NCBI

31 

Nakamura Y, Imada A, Fukugaki A, Kanto S, Yamaura T, Kinjo Y and Kuroda N: Association of nutritional risk and systemic inflammation with survival in patients with colorectal cancer who underwent curative surgery. Clin Nutr ESPEN. 49:417–424. 2022. View Article : Google Scholar : PubMed/NCBI

32 

Mantovani A, Allavena P, Sica A and Balkwill F: Cancer-related inflammation. Nature. 454:436–444. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Santarpia L, Contaldo F and Pasanisi F: Nutritional screening and early treatment of malnutrition in cancer patients. J Cachexia Sarcopenia Muscle. 2:27–35. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Murry DJ, Riva L and Poplack DG: Impact of nutrition on pharmacokinetics of anti-neoplastic agents. Int J Cancer. (Suppl 11):48–51. 1998. View Article : Google Scholar : PubMed/NCBI

35 

Ikuta S, Sonoda T, Aihara T, Nakajima T and Yamanaka N: The preoperative modified Glasgow prognostic score for the prediction of survival after pancreatic cancer resection following non-surgical treatment of an initially unresectable disease. Contemp Oncol (Pozn). 22:229–235. 2018.PubMed/NCBI

36 

Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C and Flavell RA: Inflammation-induced cancer: Crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 13:759–771. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2024
Volume 27 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Higashi T, Murase K, Watanabe D, Ishihara T, Yokoi R, Kuno M, Fukada M, Iwashita T, Tajima JY, Kiyama S, Kiyama S, et al: Pretherapeutic factors predicting conversion surgery in unresectable pancreatic ductal adenocarcinoma: A retrospective study. Oncol Lett 27: 171, 2024
APA
Higashi, T., Murase, K., Watanabe, D., Ishihara, T., Yokoi, R., Kuno, M. ... Matsuhashi, N. (2024). Pretherapeutic factors predicting conversion surgery in unresectable pancreatic ductal adenocarcinoma: A retrospective study. Oncology Letters, 27, 171. https://doi.org/10.3892/ol.2024.14304
MLA
Higashi, T., Murase, K., Watanabe, D., Ishihara, T., Yokoi, R., Kuno, M., Fukada, M., Iwashita, T., Tajima, J. Y., Kiyama, S., Tanaka, Y., Okumura, N., Shimizu, M., Matsuhashi, N."Pretherapeutic factors predicting conversion surgery in unresectable pancreatic ductal adenocarcinoma: A retrospective study". Oncology Letters 27.4 (2024): 171.
Chicago
Higashi, T., Murase, K., Watanabe, D., Ishihara, T., Yokoi, R., Kuno, M., Fukada, M., Iwashita, T., Tajima, J. Y., Kiyama, S., Tanaka, Y., Okumura, N., Shimizu, M., Matsuhashi, N."Pretherapeutic factors predicting conversion surgery in unresectable pancreatic ductal adenocarcinoma: A retrospective study". Oncology Letters 27, no. 4 (2024): 171. https://doi.org/10.3892/ol.2024.14304