Acessibilidade / Reportar erro

Preoperative prognostic factors in patients with ductal adenocarcinoma of the head of the pancreas

ABSTRACT

Objective:

To identify the preoperative prognostic factors influencing pancreatic cancer survival following curative resection in a single Brazilian institution.

Methods:

From 2005 to 2018, preoperative clinic, demographic and laboratory data were prospectively collected. Survival analysis was performed by the Kaplan-Meier method and the comparison between curves by the log-rank test. For multivariate analysis, the Cox regression model was used.

Results:

advanced age (p = 0.012) and previous history of cancer (p = 0.026) were the preoperative factors, according to the univariate analysis, that significantly impacted survival. Patients with preoperative serum levels of CA 19.9 from 38 to 554 U/mL had a 3.15 times higher chances of death (HR 3.15; 95% CI 1.01-9.82; p = 0.047), whereas patients with the marker above 554 U/mL were 3.96 times more likely than those with the normal level (HR 3.96; 95% CI 1.19-13.10; p = 0.024), by using the multivariate analysis. Patients with previous comorbidities had a 2.90 times higher chance of death than those without associated conditions (HR 2.90; 95% CI 1.10-7.67; p = 0.032).

Conclusion:

Preoperative factors related to the worst prognosis after pancreatic ductal adenocarcinoma resection were advanced age, presence of comorbidities, previous history of cancer and elevated preoperative serum CA 19.9.

Headings:
Carcinoma, Pancreatic Ductal; Pancreaticoduodenectomy; Prognosis; Survival Analysis

RESUMO

Objetivos:

Identificar fatores prognósticos pré-operatórios relacionados à sobrevida de pacientes com adenocarcinoma ductal da cabeça de pâncreas (ADCP) submetidos a tratamento cirúrgico com intenção curativa em uma única instituição brasileira.

Método:

No período de 2005 a 2018, dados clinicodemográficos e laboratoriais pré-operatórios foram prospectivamente coletados. A análise de sobrevida foi feita pelo método de Kaplan-Meier e a comparação entre as curvas pelo teste de log-rank. Para a análise multivariada utilizou-se o modelo de regressão de Cox.

Resultados:

Os fatores pré-operatórios com impacto significativo na sobrevida à análise univariada foram a idade maior ou igual a 70 anos (p=0,012) e história pessoal prévia positiva para câncer (p=0,026). A análise multivariada, pacientes com níveis séricos pré-operatórios de CA 19.9 de 38 a 554 U/ml apresentaram 3,15 vezes maior chance de óbito (HR 3,15; IC 95% 1,01 - 9,82; p=0,047), enquanto que os pacientes com o marcador acima de 554 U/ml evoluíram com chance 3,96 vezes maior de óbito que aqueles com a dosagem normal (HR 3,96; IC 95% 1,19 - 13,10; p=0,024). Pacientes com comorbidades prévias evoluíram com chance 2,90 vezes superior de óbito que doentes sem condições associadas (HR 2,90; IC 95% 1,10 - 7,67; p=0,032).

Conclusão:

O ADCP mostrou ser doença agressiva para a qual os fatores pré-operatórios de pior prognóstico foram idade avançada, presença de comorbidades, história prévia de câncer e nível sérico de CA 19.9 elevado no pré-operatório.

Descritores:
Carcinoma Ductal Pancreático; Pancreaticoduodenectomia; Prognóstico; Análise de Sobrevida

INTRODUCTION

Pancreatic cancer is the fourth leading cause of cancer death in the United States, where the incidence matches mortality11 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.,22 Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73-85.. In Brazil, it represents 2% of overall cancers and 4% of cancer deaths33 Brasil. Ministério da Saúde. Instituto Nacional de Câncer José Alencar Gomes da Silva - INCA, Rio de Janeiro. Tipo de câncer: Pâncreas. [publicação online]; 2013 [acesso em 28 ago 2018]. Disponível em: http://www2.inca.gov.br/wps/wcm/connect/tiposdecancer/site/home/pancreas
http://www2.inca.gov.br/wps/wcm/connect/...
. The long asymptomatic period contributes to the poor prognosis, which means that the diagnosis is made, in most cases, at an advanced stage. Two-thirds of patients have cancer located in the head of the pancreas, while one third in the body or tail22 Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73-85..

It predominantly affects individuals between 60 and 80 years of age, it is more frequent in men, and it is related to a component of family origin in about 10% of cases22 Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73-85.,44 Canto MI, Harinck F, Hruban RH, Offerhaus GJ, Poley JW, Kamel I, Nio Y, Schulick RS, Bassi C, Kluijt I, Levy MJ, Chak A, Fockens P, Goggins M, Bruno M; International Cancer of Pancreas Screening (CAPS) Consortium. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut. 2013;62(3):339-47. Erratum in: Gut. 2014;63(12):1978.,55 Klein AP, Brune KA, Petersen GM, Goggins M, Tersmette AC, Offerhaus GJ, et al. Prospective risk of pancreatic cancer in familial pancreatic cancer kindreds. Cancer Res. 2004;64(7):2634-8.. In addition to genetic factors, the increased risk among smokers, patients with chronic pancreatitis and type 2 diabetes is well established66 Iodice S, Gandini S, Maisonneuve P, Lowenfels AB. Tobacco and the risk of pancreatic cancer: a review and meta-analysis. Langenbecks Arch Surg. 2008;393(4):535-45.

7 Raimondi S, Lowenfels AB, Morselli-Labate AM, Maisonneuve P, Pezzilli R. Pancreatic cancer in chronic pancreatitis; aetiology, incidence, and early detection. Best Pract Res Clin Gastroenterol. 2010;24(3):349-58.

8 Bosetti C, Rosato V, Li D, Silverman D, Petersen GM, Bracci PM, et al. Diabetes, antidiabetic medications, and pancreatic cancer risk: an analysis from the International Pancreatic Cancer Case-Control Consortium. Ann Oncol. 2014;25(10):2065-72.
-99 Biadgo B, Abebe M. Type 2 diabetes mellitus and its association with the risk of pancreatic carcinogenesis: a review. Korean J Gastroenterol. 2016;67(4):168-77..

Only 10% to 20% of patients diagnosed with pancreatic cancer have a locoregional disease, which evolves with a global survival of about 30% in five years11 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.,22 Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73-85.. 52% of pancreatic neoplasms are diagnosed at an advanced stage and with metastasis and present with a 3% overall five-year survival11 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.. The overall survival for all stages of pancreatic cancer is about 8% in five years11 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.. Surgical resection is the only therapeutic option with a chance of cure22 Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73-85..

Among the periampular carcinomas, the ductal adenocarcinoma of the head of the pancreas (ADCP) has the worst prognosis. The survival of patients undergoing surgical interventions approximates that observed in patients with neoplasia of the papilla or distal choledochus, and belong to the pancreatobiliary subtype1010 Williams JL, Chan CK, Toste PA, Elliott IA, Vasquez CR, Sunjaya DB, et al. Association of histopathologic phenotype of periampullary adenocarcinomas with survival. JAMA Surg. 2017;152(1):82-8.,1111 da Silveira Santos JPL, Machado CJ, Junior EP, Rodrigues JBSR, Vidigal PT, Resende V. Immunohistochemical predictors for intestinal and pancreatobiliary types of adenocarcinoma of the ampulla of Vater. J Gastrointest Surg. 2018;22(7):1171-8..

When assessing surgical and postoperative data, lymph node involvement, larger lesions, undifferentiated tumors, angiolymphatic and perineural invasion as well as the non-association with adjuvant therapy are well-established predictors of a worse prognosis1212 Geer RJ, Brennan MF. Prognostic indicators for survival after resection of pancreatic adenocarcinoma. Am J Surg. 1993;165(1):68-72; discussion 72-3.

13 John BJ, Naik P, Ironside A, Davidson BR, Fusai G, Gillmore R, et al. Redefining the R1 resection for pancreatic ductal adenocarcinoma: tumour lymph nodal burden and lymph node ratio are the only prognostic factors associated with survival. HPB (Oxford). 2013;15(9):674-80.
-1414 Lim JE, Chien MW, Earle CC. Prognostic factors following curative resection for pancreatic adenocarcinoma: a population-based, linked database analysis of 396 patients. Ann Surg. 2003;237(1):74-85.. In order to allow early diagnosis, improve survival and prognosis of patients with pancreatic adenocarcinoma, screening methods are necessary. The identification of specific preoperative prognostic factors in different populations can also lead to improved survival in these patients.

In this regard, this study aims to identify preoperative prognostic factors related to the survival of patients with ADCP undergoing surgical treatment with curative intent, in a single Brazilian institution.

METHODS

This is a retrospective cohort study with data collected prospectively. The Research Ethics Committee of UFMG approved it under the number 23377113100005149. Initially, 116 patients who underwent pancreatectoduodenectomy or cephalic pancreatectomy were selected by the Liver, Bile Ducts, Pancreas and Spleen surgical team, at the Alfa Gastroenterology Institute, Hospital das Clínicas of UFMG, from 2005 to 2018. After fulfilling the inclusion and exclusion criteria, the final sample was 40 patients (Figure 1).

Figure 1
Patients’ distribution, according to the inclusion and exclusion criteria.

Data were collected by filling in specific protocols, analyzing medical records and clinical follow-up of the patients. Patients with ductal adenocarcinoma of pancreatic origin and cephalic location, who underwent laparotomy via cephalic pancreatectoduodenectomy or cephalic pancreatectomy with pyloric preservation, with curative intent, with or without resection of the superior mesenteric vein, associated with locoregional lymphadenectomy were included. Reconstruction of the transit was done with a single loop, and, in most cases, duct-to-mucosa end-to-side pancreaticojejunostomy, with or without a stent, was done. Adjuvant chemotherapy with gemcitabine was prescribed for all patients that underwent surgical resection.

The preoperative variables were: sex, age, smoking, alcoholism, previous history of cancer, family history of neoplasms, associated comorbidities and type 2 diabetes. Smokers were patients who reported smoking for ten years or more, and alcoholism was considered when the consumption of more than 30 grams of alcohol per day was informed. Preoperative serum levels of CA 19.9 (U/mL), CEA (ng/mL), total and direct bilirubin (mg/dL), albumin (g/dL), hemoglobin (g/dL), platelets (x103/µL) and leukocytes (x103/µL). Preoperative biliary drainage was done either by endoscopic biliary prosthesis or percutaneous biliary drainage. Data on survival were obtained either by outpatient follow-up, medical record checking or telephone contact with family members when patients did not attend follow up visits, and there was no notification of death.

Statistical analysis was performed using the software Stata® version 12 and IBM SPSS® (Chicago, IL, USA) version 23 for MAC (Macintosh®). Descriptive analysis of the data using measures of central tendency, dispersion and amplitude for continuous variables - mean and standard deviation (SD), median (50th percentile) and interquartile range (25th and 75th percentiles); minimum and maximum values were carried out. The descriptive analysis of categorical variables was performed by distributing the absolute and relative frequencies in each category. In order to test homogeneity, a Chi-square test (n> 4) or Fisher’s exact test (n <5) was performed, considering the existence of equality in the categories of variables, as a null hypothesis (H0). Thus, for variables with two categories, the null hypothesis contemplated a proportion (pr) equal to 50% in each category (H0: pr = 0.5); for three categories, H0: pr = 0.33; for four categories, H0: pr = 0.25.

The median survival time and the respective interquartile range (IIQ) were considered for patients in general and according to categories. Survival analysis was done by the Kaplan-Meier method, for which the dependent variable was death, concerning time, in months. In order to assess whether there was a difference between the survival curves, the log-rank test was used. Overall survival was assessed at 12, 18, 24 and 36 months. Deaths that occurred within 30 days of the postoperative period were excluded.

A multivariate regression model was done to assess the relationship between one or more factors of interest and patients’ survival. Based on the results of the univariate analysis, variables whose p values were equal to or less than 0.20 (p = 0.20) were selected as candidates for the final multivariate analysis. Cox regression was used to obtain the multivariate model. Survival analysis took into account the risk ratio or hazard ratio (HR), which is the chance of death divided by the chance of surviving along the entire spectrum of the survival curve. The level of significance was 5% (p <0.05).

RESULT (S)

Table 1 depicts the clinical and demographic characteristics and the median survival for the variables of interest. There was a predominance of females, with 25 women (62.5%). The patients’ age ranged from 34 to 89 years, with a mean of 60.1 years and a median of 59.5 years. Survival was significantly lower in patients aged 70 years or older, with a median of 12 months, while for younger patients, it was 27 months (p = 0.012). Figure 2 depicts the survival curve using the Kaplan-Meier method according to age.

Table 1
Clinical and demographic variables and median survival of patients with ductal adenocarcinoma of the head of the pancreas undergoing surgical treatment with curative intent.

Figure 2
Kaplan-Meier survival curve in patients that underwent curative surgical treatment for ductal adenocarcinoma of the head of the pancreas, according to age. Curves equality test: log-rank ( p = 0.012 ).

Smokers predominated (60.0%), while 45.0% of patients reported a history of alcoholism. Of the total number of patients, 27 had associated comorbidities (67.5%) and, despite the discrepancy between median survival (30 months versus 13 months for patients with and without associated conditions, respectively), this was not a statistic prognostic factor (p = 0.084). The most prevalent comorbidity was systemic arterial hypertension (16), followed by type 2 diabetes (15), and seven patients had both comorbidities. Three patients had chronic pancreatitis. All elderly patients had at least one associated comorbidity. Six patients were classified as ASA I (15.0%), the majority as ASA II (31 patients/77.5%) and three ASA III (7.5 %).

Seventy-five percent of patients were able to reply the question regarding a family history of cancer, of which half was positive, two of them with pancreatic cancer. Seven patients (three men and four women) had previously had other types of cancer, and these patients had a median survival of 10 months, while the others, which represented 82.5% of the sample, had significantly higher survival rates, with a median of 27 months (p = 0.026). Figure 3 depicts y the Kaplan-Meier survival curve according to this variable. Among men, prostate cancer predominated (three patients). Among women, previous neoplasms were colorectal cancer, meningioma, gallbladder and breast cancer. Nonetheless, the patient with the longest survival (27 months) was the one with previous breast cancer.

Figure 3
Kaplan-Meier survival cure in patients who underwent curative surgical treatment for ductal adenocarcinoma of the head of the pancreas, according to the previous history of cancer. Curves equality test: log-rank ( p = 0.026 ).

The preoperative laboratory tests and their relationship to median survival are shown in Table 2.

Table 2
Preoperative laboratory tests and median survival in patients who underwent curative surgical treatment for ductal adenocarcinoma of the head of the pancreas.

The mean CA 19.9 was 1,186.0 U/mL, with a standard deviation of 2,080.8 U/mL, ranging from 2.0 to 9,760.0 U/mL. The median serum CA 19.9 was 281.0 U/mL, and 25% of the patients were in 25th percentile) with levels below 40.8 U/mL, while 25% (percentile 75) were above 1,532.0 U/mL. 27.2% of the patients had the marker within normal limits.

The mean preoperative CEA was 5.19 ng/mL (SD 4.8 ng/mL), with a median of 3.05 ng/mL. Before the surgical procedure, 80.0% of patients presented a CEA within the reference values. The mean serum albumin was 3.29 g/dL, the median was 3.40 g/dL and just over half of the patients were within the reference values.

Nine patients (23.7%) underwent preoperative biliary decompression, which did not affect survival (p = 0.398). The mean total preoperative serum bilirubin was 11.92 mg/dL (SD 9.23 mg/dL) and the median was 11.9 mg/dL. 25% of patients in the first quartile had a CEA less than 2.3 mg/dL, while the other 25.0%, in the last quartile, had values higher than 20.6 mg/dL. There was no statistical difference between the groups with and without values above 20.0 mg/dL (p = 0.651). Preoperative hematimetric parameters were not a significant prognostic factor.

According to the multivariate analysis, comorbidities and high preoperative CA 19.9 were the variables that proved to be independent prognostic factors, with statistical significance (Table 3).

Table 3
Multivariate and Hazard Ratio (HR) analysis of patients with ductal adenocarcinoma of the head of the pancreas who underwent curative surgical treatment.

Patients with preoperative serum CA 19.9 ranging from 38 to 554 U/mL had 3.15 times greater chance of death than those within the normal range (HR 3.15; 95% CI 1.01-9.82; p = 0.047), while patients with levels over 554 U/mL presented with four times higher risk of death (HR 3.96; 95% CI 1.19-13.10; p = 0.024).

Patients with previous comorbidities had a death risk almost three times higher than patients without other associated conditions (HR 2.90; 95% CI 1.10-7.67; p = 0.032).

During the follow-up period, which ranged from three to 43 months, 76.3% of the patients died. The median survival was 18.4 months (SD 12.7 months) and the median survival was 14.5 months (IIQ 7; 28). The overall survival rate at 12, 18, 24 and 36 months was 66.0%, 52.8%, 45.8% and 19.1%, respectively. Figure 4 indicates the overall Kaplan-Meier survival curve.

Figure 4
Global Kaplan-Meier survival curve for patients with ductal adenocarcinoma of the head of the pancreas who underwent curative surgical treatment.

DISCUSSION

Pancreatic cancer is a tumor with an unfavorable prognosis, being the fourth leading cause of cancer death in the United States11 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.,22 Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73-85.. In the 70s of the last century, curative intent operations in patients with tumors of the head of the pancreas did not represent a gain in survival compared to the palliative surgical approach, adding, however, postoperative morbidity1515 Shapiro TM. Adenocarcinoma of the pancreas: a statistical analysis of biliary bypass vs Whipple resection in good risk patients. Ann Surg. 1975;182(6):715-21.. Currently, surgical resection represents the only therapeutic option with a chance of cure. However, early diagnosis of a localized and resectable disease occurs in only 10% to 20% of patients11 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.,22 Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73-85.. The overall five-year survival reaches 32% when patients with pancreatic head cancer are diagnosed and treated at an early stage. When there is regional dissemination, this rate drops to 12%, reaching 3% in five years in patients with distant metastasis at diagnosis11 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30..

In this study, advanced age was identified as a prognostic factor with a lower survival rate in patients aged 70 years or older. Petrou et al.1616 Petrou A, Soonawalla Z, Silva MA, Manzelli A, Moris D, Tabet PP, et al. Prognostic indicators following curative pancreatoduodenectomy for pancreatic carcinoma: A retrospective multivariate analysis of a single centre experience. J BUON. 2016;21(4):874-82. evaluated a total of 101 patients who underwent duodenopancreatectomy due to periampular tumors and indicated that patients older than 65 years were twice as likely to die during follow-up (p = 0.037). However, some authors have not identified age as a prognostic factor1212 Geer RJ, Brennan MF. Prognostic indicators for survival after resection of pancreatic adenocarcinoma. Am J Surg. 1993;165(1):68-72; discussion 72-3.,1717 Salmiheimo A, Mustonen H, Stenman UH, Puolakkainen P, Kemppainen E, Seppänen H, et al. Systemic inflammatory response and elevated tumour markers predict worse survival in resectable pancreatic ductal adenocarcinoma. PLoS One. 2016;11(9):e0163064..

The coexistence of comorbidities, found in 67.5% of our sample, was an important prognostic factor, with a significant impact on survival. Patients with associated diseases had an almost three times higher chance of early death than the others. Our sample had a prevalence of comorbidities two to three times higher than that found in the literature, which ranges from 20.0% to 30.0% of patients undergoing pancreatic resection1414 Lim JE, Chien MW, Earle CC. Prognostic factors following curative resection for pancreatic adenocarcinoma: a population-based, linked database analysis of 396 patients. Ann Surg. 2003;237(1):74-85.,1818 Nathan H, Cameron JL, Choti MA, Schulick RD, Pawlik TM. The volume-outcomes effect in hepato-pancreato-biliary surgery: hospital versus surgeon contributions and specificity of the relationship. J Am Coll Surg. 2009;208(4):528-38.. Lim et al.1414 Lim JE, Chien MW, Earle CC. Prognostic factors following curative resection for pancreatic adenocarcinoma: a population-based, linked database analysis of 396 patients. Ann Surg. 2003;237(1):74-85. allocated operated pancreatic cancer patients into three groups: without comorbidities, one comorbidity and two or more comorbidities. Most patients had no comorbidities (77.0%) and, when comparing the groups, this factor did not significantly affect survival.

Due to the advancement of cancer treatments and improved survival of these patients, the risk of developing second primary neoplasias is increasing1919 Wood ME, Vogel V, Ng A, Foxhall L, Goodwin P, Travis LB. Second malignant neoplasms: assessment and strategies for risk reduction. J Clin Oncol. 2012;30(30):3734-45.,2020 Donin NM, Kwan L, Lenis AT, Drakaki A, Chamie K. Second primary lung cancer in United States Cancer Survivors, 1992-2008. Cancer Causes Control. 2019;30(5):465-75.. These patients are especially susceptible to it due to genetic issues, exposure to common risk factors, as well as the late effect of chemotherapy and radiotherapy treatments2020 Donin NM, Kwan L, Lenis AT, Drakaki A, Chamie K. Second primary lung cancer in United States Cancer Survivors, 1992-2008. Cancer Causes Control. 2019;30(5):465-75.. Pancreatic cancer, as the second primary neoplasm, had a more aggressive outcome in our series. The 33 patients who did not have a history of other previous neoplasia had a median survival significantly higher than the others (27 months versus 10 months, respectively, p = 0.026). In a recent Asian study, He et al.2121 He C, Zhang Y, Cai Z, Lin X. Effect of prior cancer on survival outcomes for patients with pancreatic adenocarcinoma: a propensity score analysis. BMC Cancer. 2019;19(1):509. indicated no significant difference in survival or mortality when comparing patients with pancreatic cancer with and without a previous history of neoplasia.

CA 19.9 is considered the best tumor marker for the evaluation and monitoring of patients with pancreatic ductal adenocarcinoma, with a sensitivity of 70.0% to 90.0% and specificity around 90.0%, being useful as a prognostic factor and tumor recurrence marker2222 Duffy MJ, Sturgeon C, Lamerz R, Haglund C, Holubec VL, Klapdor R, et al. Tumor markers in pancreatic cancer: a European Group on Tumor Markers (EGTM) status report. Ann Oncol. 2010;21(3):441-7.,2323 Fong ZV, Winter JM. Biomarkers in pancreatic cancer: diagnostic, prognostic, and predictive. Cancer J. 2012;18(6):530-8.. Dong et al.2424 Dong Q, Yang XH, Zhang Y, Jing W, Zheng LQ, Liu YP, et al. Elevated serum CA19-9 level is a promising predictor for poor prognosis in patients with resectable pancreatic ductal adenocarcinoma: a pilot study. World J Surg Oncol. 2014;12:171. indicated that patients with serum CA 19.9 equal to or higher than 338.45 U/mL presented with shorter survival compared to the group with lower values ​​(HR 1.96; 95% CI 1.24-3.09; p = 0.004). Another study, with 147 patients, found a preoperative median of 122 U/mL and indicated that patients with serum levels equal to or less than 40 U/mL had a five-time higher chance of survival in five years (OR 5.02; 95% CI 1.68-16.48; p = 0.0036)2525 Kimura K, Amano R, Nakata B, Yamazoe S, Hirata K, Murata A, et al. Clinical and pathological features of five-year survivors after pancreatectomy for pancreatic adenocarcinoma. World J Surg Oncol. 2014;12:360.. In the study by Salmiheimo et al.1717 Salmiheimo A, Mustonen H, Stenman UH, Puolakkainen P, Kemppainen E, Seppänen H, et al. Systemic inflammatory response and elevated tumour markers predict worse survival in resectable pancreatic ductal adenocarcinoma. PLoS One. 2016;11(9):e0163064., patients with preoperative values of CA 19.9 equal to or higher than 555 U/mL had almost twice the chance of earlier death during follow-up (HR 1.91; 95% CI 1.18-3.08 ; p = 0.008). In our analysis, we used similar cut-off levels, and the chance of death during follow-up was 3.96 times greater than for the patients within the normal range (HR 3, 96; 95% CI 1.19-13.10; p = 0.024).

The type of chemotherapy our patients underwent was similar to what has been recommended: gemcitabine as the first line of treatment. In 2007, after the publication of a relevant randomized clinical trial, it was proven that adjuvant chemotherapy with gemcitabine delays disease recurrence and increases survival when compared to the operation2626 Oettle H, Post S, Neuhaus P, Gellert K, Langrehr J, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA. 2007;297(3):267-77.. Ten years later, a new study published by the European pancreatic cancer study group indicated that the combination of gemcitabine and capecitabine, after pancreatic cancer surgical treatment, increased the median survival by 2.5 months compared to gemcitabine alone, suggesting that this should be the ideal adjuvant chemotherapy regimen2727 Neoptolemos JP, Palmer DH, Ghaneh P, Psarelli EE, Valle JW, Halloran CM, Faluyi O, O’Reilly DA, Cunningham D, Wadsley J, Darby S, Meyer T, Gillmore R, Anthoney A, Lind P, Glimelius B, Falk S, Izbicki JR, Middleton GW, Cummins S, Ross PJ, Wasan H, McDonald A, Crosby T, Ma YT, Patel K, Sherriff D, Soomal R, Borg D, Sothi S, Hammel P, Hackert T, Jackson R, Büchler MW; European Study Group for Pancreatic Cancer. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, phase 3 trial. Lancet. 2017;389(10073):1011-24..

The number of patients is a limitation of our study. The sample, which represents the experience of a single institution, may have been responsible for the lack of statistical significance regarding some variables.

CONCLUSION

The prognostic factors related to ductal adenocarcinoma of the head of the pancreas after pancreatectoduodenectomy were older age, previous history of cancer, associated comorbidities and elevated CA 19.9 levels. These factors were associated with decreased overall survival, which was, for the entire series, 66.0%, 52.8%, 45.8% and 19.1% at 12, 18, 24 and 36 months, respectively. Despite the current advances, the treatment of pancreatic cancer is still a challenge.

  • Financing source: None

REFERÊNCIAS

  • 1
    Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30.
  • 2
    Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73-85.
  • 3
    Brasil. Ministério da Saúde. Instituto Nacional de Câncer José Alencar Gomes da Silva - INCA, Rio de Janeiro. Tipo de câncer: Pâncreas. [publicação online]; 2013 [acesso em 28 ago 2018]. Disponível em: http://www2.inca.gov.br/wps/wcm/connect/tiposdecancer/site/home/pancreas
    » http://www2.inca.gov.br/wps/wcm/connect/tiposdecancer/site/home/pancreas
  • 4
    Canto MI, Harinck F, Hruban RH, Offerhaus GJ, Poley JW, Kamel I, Nio Y, Schulick RS, Bassi C, Kluijt I, Levy MJ, Chak A, Fockens P, Goggins M, Bruno M; International Cancer of Pancreas Screening (CAPS) Consortium. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut. 2013;62(3):339-47. Erratum in: Gut. 2014;63(12):1978.
  • 5
    Klein AP, Brune KA, Petersen GM, Goggins M, Tersmette AC, Offerhaus GJ, et al. Prospective risk of pancreatic cancer in familial pancreatic cancer kindreds. Cancer Res. 2004;64(7):2634-8.
  • 6
    Iodice S, Gandini S, Maisonneuve P, Lowenfels AB. Tobacco and the risk of pancreatic cancer: a review and meta-analysis. Langenbecks Arch Surg. 2008;393(4):535-45.
  • 7
    Raimondi S, Lowenfels AB, Morselli-Labate AM, Maisonneuve P, Pezzilli R. Pancreatic cancer in chronic pancreatitis; aetiology, incidence, and early detection. Best Pract Res Clin Gastroenterol. 2010;24(3):349-58.
  • 8
    Bosetti C, Rosato V, Li D, Silverman D, Petersen GM, Bracci PM, et al. Diabetes, antidiabetic medications, and pancreatic cancer risk: an analysis from the International Pancreatic Cancer Case-Control Consortium. Ann Oncol. 2014;25(10):2065-72.
  • 9
    Biadgo B, Abebe M. Type 2 diabetes mellitus and its association with the risk of pancreatic carcinogenesis: a review. Korean J Gastroenterol. 2016;67(4):168-77.
  • 10
    Williams JL, Chan CK, Toste PA, Elliott IA, Vasquez CR, Sunjaya DB, et al. Association of histopathologic phenotype of periampullary adenocarcinomas with survival. JAMA Surg. 2017;152(1):82-8.
  • 11
    da Silveira Santos JPL, Machado CJ, Junior EP, Rodrigues JBSR, Vidigal PT, Resende V. Immunohistochemical predictors for intestinal and pancreatobiliary types of adenocarcinoma of the ampulla of Vater. J Gastrointest Surg. 2018;22(7):1171-8.
  • 12
    Geer RJ, Brennan MF. Prognostic indicators for survival after resection of pancreatic adenocarcinoma. Am J Surg. 1993;165(1):68-72; discussion 72-3.
  • 13
    John BJ, Naik P, Ironside A, Davidson BR, Fusai G, Gillmore R, et al. Redefining the R1 resection for pancreatic ductal adenocarcinoma: tumour lymph nodal burden and lymph node ratio are the only prognostic factors associated with survival. HPB (Oxford). 2013;15(9):674-80.
  • 14
    Lim JE, Chien MW, Earle CC. Prognostic factors following curative resection for pancreatic adenocarcinoma: a population-based, linked database analysis of 396 patients. Ann Surg. 2003;237(1):74-85.
  • 15
    Shapiro TM. Adenocarcinoma of the pancreas: a statistical analysis of biliary bypass vs Whipple resection in good risk patients. Ann Surg. 1975;182(6):715-21.
  • 16
    Petrou A, Soonawalla Z, Silva MA, Manzelli A, Moris D, Tabet PP, et al. Prognostic indicators following curative pancreatoduodenectomy for pancreatic carcinoma: A retrospective multivariate analysis of a single centre experience. J BUON. 2016;21(4):874-82.
  • 17
    Salmiheimo A, Mustonen H, Stenman UH, Puolakkainen P, Kemppainen E, Seppänen H, et al. Systemic inflammatory response and elevated tumour markers predict worse survival in resectable pancreatic ductal adenocarcinoma. PLoS One. 2016;11(9):e0163064.
  • 18
    Nathan H, Cameron JL, Choti MA, Schulick RD, Pawlik TM. The volume-outcomes effect in hepato-pancreato-biliary surgery: hospital versus surgeon contributions and specificity of the relationship. J Am Coll Surg. 2009;208(4):528-38.
  • 19
    Wood ME, Vogel V, Ng A, Foxhall L, Goodwin P, Travis LB. Second malignant neoplasms: assessment and strategies for risk reduction. J Clin Oncol. 2012;30(30):3734-45.
  • 20
    Donin NM, Kwan L, Lenis AT, Drakaki A, Chamie K. Second primary lung cancer in United States Cancer Survivors, 1992-2008. Cancer Causes Control. 2019;30(5):465-75.
  • 21
    He C, Zhang Y, Cai Z, Lin X. Effect of prior cancer on survival outcomes for patients with pancreatic adenocarcinoma: a propensity score analysis. BMC Cancer. 2019;19(1):509.
  • 22
    Duffy MJ, Sturgeon C, Lamerz R, Haglund C, Holubec VL, Klapdor R, et al. Tumor markers in pancreatic cancer: a European Group on Tumor Markers (EGTM) status report. Ann Oncol. 2010;21(3):441-7.
  • 23
    Fong ZV, Winter JM. Biomarkers in pancreatic cancer: diagnostic, prognostic, and predictive. Cancer J. 2012;18(6):530-8.
  • 24
    Dong Q, Yang XH, Zhang Y, Jing W, Zheng LQ, Liu YP, et al. Elevated serum CA19-9 level is a promising predictor for poor prognosis in patients with resectable pancreatic ductal adenocarcinoma: a pilot study. World J Surg Oncol. 2014;12:171.
  • 25
    Kimura K, Amano R, Nakata B, Yamazoe S, Hirata K, Murata A, et al. Clinical and pathological features of five-year survivors after pancreatectomy for pancreatic adenocarcinoma. World J Surg Oncol. 2014;12:360.
  • 26
    Oettle H, Post S, Neuhaus P, Gellert K, Langrehr J, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA. 2007;297(3):267-77.
  • 27
    Neoptolemos JP, Palmer DH, Ghaneh P, Psarelli EE, Valle JW, Halloran CM, Faluyi O, O’Reilly DA, Cunningham D, Wadsley J, Darby S, Meyer T, Gillmore R, Anthoney A, Lind P, Glimelius B, Falk S, Izbicki JR, Middleton GW, Cummins S, Ross PJ, Wasan H, McDonald A, Crosby T, Ma YT, Patel K, Sherriff D, Soomal R, Borg D, Sothi S, Hammel P, Hackert T, Jackson R, Büchler MW; European Study Group for Pancreatic Cancer. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, phase 3 trial. Lancet. 2017;389(10073):1011-24.

Publication Dates

  • Publication in this collection
    03 June 2020
  • Date of issue
    2020

History

  • Received
    23 Sept 2019
  • Accepted
    11 Dec 2019
Colégio Brasileiro de Cirurgiões Rua Visconde de Silva, 52 - 3º andar, 22271- 090 Rio de Janeiro - RJ, Tel.: +55 21 2138-0659, Fax: (55 21) 2286-2595 - Rio de Janeiro - RJ - Brazil
E-mail: revista@cbc.org.br