Clinical necessity of the immunohistochemical reassessment of para‑aortic lymph nodes in resected pancreatic ductal adenocarcinoma

  • Authors:
    • Sung Hoon Choi
    • Se Hoon Kim
    • Jun Jeong Choi
    • Chang Moo Kang
    • Ho Kyoung Hwang
    • Woo Jung Lee
  • View Affiliations

  • Published online on: August 21, 2013     https://doi.org/10.3892/ol.2013.1539
  • Pages: 1189-1194
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Para‑aortic lymph node (PALN) metastasis is widely regarded as a systemic disease in cancer. Undetected PALN micrometastases during routine hematoxylin and eosin (HE) staining may be a cause of poor prognosis following a potentially curative pancreatectomy for pancreatic cancer. In the present study, paraffin‑embedded PALN tissue blocks from 99 patients who underwent a pancreatectomy were re‑evaluated by immunohistochemical staining using cytokeratin (CK)‑19. Patients with PALN metastasis were summarized according to the clinicopathological data. A total of 484 PALNs (median, 4.9 nodes per patient; range, 1‑19) were evaluated. PALN metastases were revealed in eight patients (8.1%) by routine HE staining of frozen section biopsies and in one patient (1.0%) by HE staining of a permanent section. Only one patient (1.0%) demonstrated micrometastasis by IHC; this patient did not display any adverse pathological characteristics and had a relatively favorable survival period of 41 months. The present study concluded that an additional reassessment for micrometastasis in PALNs using CK‑19 immunohistochemistry (IHC) is not a viable method for determining the survival outcome. A careful examination of a frozen section biopsy is sufficient for attempting curative surgery.

Introduction

Surgical resection is the only known curative option for pancreatic cancer. However, the majority of pancreatic cancers are usually diagnosed at advanced stages and only 15–20% of patients are candidates for a gross margin-negative pancreatectomy (R0) (1). Following a curative resection, distant metastases, particularly in the liver, local recurrence and peritoneal dissemination frequently occur and these patients succumb to their diseases (2,3) The reported five-year survival rate following surgical resection is 12.1–25.0% (15) and the overall survival rate is considered to be <5%, suggesting that pancreatic cancer is one of the most lethal gastrointestinal malignancies.

According to a previous survival analysis of resected pancreatic cancers, an R0 was shown to be an independent favorable prognostic factor (68). Over the past few decades, surgeons have attempted to achieve ideal R0 resections by extending the surgical margins in the hopes that clearing the surrounding soft tissue that contains malignant cells may improve the survival outcome. However, several significant prospective randomized control studies revealed that extending the margins did not result in an additional survival benefit over the standard surgery in resectable pancreatic cancer (912). It has been proposed that a possible underestimation of microscopic cancer spreading beyond the surgical field may contribute to a poor prognosis following a potentially curative surgical treatment (13).

Lymph node metastasis is common in pancreatic cancer (1416) and para-aortic lymph nodes (PALNs) are considered to be the final nodes in the systemic lymphatic circulation in periampullary cancer (17,18). Although there have only been a few studies on the oncological outcome according to PALN staging (1921), PALN involvement is known to be a poor prognostic factor in periampullary tumors (18). However, pancreatic surgeons may encounter clinical cases of potentially resectable pancreatic tumors with unexpected PALN metastasis that are only identified on intraoperative frozen section biopsies. Considering the expected poor prognosis in patients with unexpected PALN metastasis and the potential curative role of an R0 in pancreatic cancer, the decision to resect must be promptly determined in the operating theatre.

The present study aimed to develop an approach to this clinical dilemma. The oncological outcomes in patients with PALN metastasis that were detected by hematoxylin and eosin (HE) staining were analyzed in resected pancreatic tumors. Immunohistochemistry (IHC) with antibodies against cytokeratin (CK)-19 was used to detect the presence of PALN micrometastasis in resected pancreatic ductal adenocarcinoma. The role of curative surgery in resectable pancreatic cancer with incidentally identified PALN metastasis was further investigated using intraoperative frozen section biopsies.

Materials and methods

Study design

The present study retrospectively investigated patients who underwent a surgical resection for pancreatic ductal adenocarcinoma between January 1999 and December 2009 at Yonsei University Health System (Seoul, South Korea). During the study period, a total of 1,119 patients were diagnosed with pancreatic ductal adenocarcinoma and 171 patients (15.3%) underwent grossly curative pancreatectomies. Of these patients, 99 with available healthy paraffin-embedded tissue blocks of PALN were re-evaluated using IHC. This study was approved by the ethics committee of Yonsei University Health System (Seoul, Korea).

Surgery and staging

A pancreaticoduodenectomy or distal pancreatectomy with splenectomy was performed, which included a resection of the main pancreatic tumor, the associated regional lymph nodes, the retroperitoneal soft tissue and the PALNs, which allowed for pathological staging. The surgical margins, including the bile duct, pancreatic duct, peripancreatic soft tissue adjacent to the superior mesenteric artery (retroperitoneal margin), duodenum or stomach, were evaluated grossly and microscopically in order to determine their status. These surgical margins, with the exception of the retroperitoneal margin, were analyzed using frozen-sections. If the margin was positive for invasive carcinoma, an additional resection was performed. A pancreatic resection margin without evidence of invasive carcinoma was considered an R0. The final margin status was noted in the permanent pathology report. The TMN stage was evaluated based on the American Joint Committee on Cancer (AJCC) Cancer Staging Manual, 7th edition (23).

IHC

Three serial sectional cuts from formalin-fixed and paraffin-embedded blocks were studied for IHC. Each of the 4-μm sections with 6-μm intervals was prepared for IHC staining with CK-19 (M0888 mouse, monoclonal, 1:100; Dako, Copenhagen, Denmark). The IHC was detected using a dextran polymer-based, biotin-free visualization system (Envision kit; Dako). Previous HE-stained sections were re-evaluated for metastasis and the results were compared with the IHC-stained sections by an experienced pathologist. Lymph node micrometastasis was defined as metastatic tumor cells that were detected by IHC evaluation using antibodies against CK, but were missed by routine histological examination using HE staining.

Statistical analysis

The primary goal of the study was to investigate the clinical significance of PALN micrometastasis in resected pancreatic ductal adenocarcinoma. The cumulative survival rates according to overall lymph node metastasis (pN stage) and PALN metastasis (LN16) were analyzed. The clinicopathological characteristics of the patients with PALN metastasis were summarized. By reviewing the medical records, recurrence and survival data were obtained. Overall survival was defined as the interval between surgery and mortality or the final follow-up visit. The cumulative survival rate was calculated using the Kaplan-Meier method. A log-rank test was used to ascertain the statistically significant differences. P<0.05 was considered to indicate a statistically significant difference.

Results

Characteristics of patients and resected ductal adenocarcinoma

Among the 99 patients (39 females and 60 males), the mean age was 61.3 years (range, 39–78). Ductal adenocarcinoma was confirmed by microscopic examination in all patients. A total of 21 (21.2%) patients underwent a conventional pancreaticoduodenectomy, 64 (64.6%) underwent a pylorus-preserving pancreaticoduodenectomy, 13 (13.1%) underwent a distal pancreatectomy with splenectomy and one (1%) was treated with a total pancreatectomy. Three patients were diagnosed with pT1 pancreatic cancer, six with pT2, 85 with pT3 and five with pT4. The median tumor size was 2.6 cm (range, 0.5–8.5). There were no R2 resections recorded in the medical records. An R1 resection was performed on 18 patients (18.2%) and an R0 was performed on 81 patients (81.8%).

Retrieved PALN assessment in resected pancreatic cancer

A total of 484 PALNs (mean, 4.9 nodes per patient; range, 1–19) were evaluated from the available PALN blocks in 99 patients (Fig. 1). A total of 13 PALNs (2.7%) demonstrated metastasis with HE staining in nine patients (eight patients in frozen sections and one patient in a permanent section). All the eight patients who demonstrated PALN metastasis on the HE staining of frozen sections showed a pattern of clustered gland formation and desmoplasia, which occupied the entire involved node (Fig. 2A). The histology of the patient who had PALN metastasis confirmed by HE staining of the permanent section, which was not detected in the frozen section, revealed a pattern of scattered small gland formation without desmoplasia (Fig. 2B). IHC reassessment for all 484 PALNs revealed that only one additional patient immunohistochemically demonstrated micrometastasis in a PALN that was not detected otherwise. The CK-19 staining revealed micrometastases in an ~46-μm isolated pattern (Fig. 2C).

Oncological outcomes of pancreatic cancer with PALN metastasis

The survival rate did not significantly differ based on the pN staging in the present study. The median survival time was 34 months (95% CI, 24.3–43.7) for the pN0 tumors and 30 months (95% CI,: 20.3–39.7) for the pN1 tumors (P=0.223; Fig. 3A). However, a statistically significant difference was observed in the survival time based on PALN involvement, confirmed by routine HE evaluation. The median survival time was 31 months (95% CI, 23.9–38.1) in patients without PALN metastases versus 17 months (95% CI, 12.8–21.6) for patients with PALN metastases (P=0.008; Fig. 3B).

Table I shows the characteristics of the patients with PALN metastasis. Cases 1–8 were the patients who showed PALN metastasis on HE staining of the frozen sections. These patients exhibited large metastatic tumors in the PALNs (median size, 2 mm; range, 0.8–12 mm) with clustered gland patterns and extensive desmoplasia. Case 9, who had PALN metastasis confirmed by HE staining of the permanent section, had relatively small metastatic tumors in the PALNs with a pattern of scattered small gland formation and no desmoplasia. Lastly, case 10, who exhibited PALN detected only by IHC staining, possessed extremely small isolated metastatic tumors in the PALNs. All patients who exhibited PALN metastasis confirmed by HE staining (cases 1–9) had other aggressive tumor characteristics, including other regional lymph node metastasis (N1) and lymphovascular and/or perineural invasions, and one patient demonstrated positive resection margins (R1). Case 10 exhibited less aggressive adverse pathological features. The median survival time of cases 1–8 was 17.5 months (range, 10–39). The survival times of cases 9 and 10 were 34 and 41 months, respectively.

Table I

Summary of patients with PALN metastasis.

Table I

Summary of patients with PALN metastasis.

CaseAge, years /genderSurgeryR statusInitial stageTumor size, cmPALN, positive/totalPALN size and character, mmLVI/PNIRecurrence time, monthsRecurrence patternSurvival, months
160/MPDR1IIB (T3N1M0)2.01/5 (F)2 GL+D+/+4Liver, Rp, ureter18
253/MPPPDR0IIB (T3N1M0)4.02/4 (F)1.3, 1.2 GL+D−/+11Bone, lung mesentery19
366/MPPPDR0IIB (T3N1M0)2.24/5 (F)10, 2, 1, 0.9 GL+D+/−12Rp, lung17
463/FPDR0IIB (T3N1M0)2.51/6 (F)2 GL+D−/+9Rp20
578/FDPR0IIB (T3N1M0)7.02/3 (F)2.3, 1.2 GL+D+/−8Para-aortic area39
673/FPPPDR0IIB (T3N1M0)2.01/3 (F)8 GL+D−/+8Liver, para-aortic area10
762/MPPPDR0IIB (T3N1M0)3.21/3 (F)12 GL+D+/+4Liver, para-aortic area15
875/FPPPDR0IIB(T3N1M0)2.51/5 (F)0.8 GL+D+/+6Peritoneal seeding, Rp11
955/FPPPDR0IIA (T3N1M0)1.31/11 (P)0.3 scattered GL+/−18 Para-aorticarea34
1051/FPPPDR0IIA (T3N0M0)1.81/4 (I)0.046 isolated24Lung, spine, Rp41

[i] PALN, para-aortic lymph node; PD, pancreatoduodenectomy; PPPD, pylorus-preserving pancreatoduodenectomy; DP, distal pancreatosplenectomy; (F), frozen biopsy with routine HE staining; (P), permanent pathological report with HE staining; (I), immunohistochemically demonstrated; GL, gland formation; +D, with desmoplasia; LVI, lymphovascular invasion; PNI, perineural invasion; Rp, retroperitoneum; HE. hematoxylin and eosin; R, residual tumor; R0, no residual tumor; R1, microscopic residual tumor; LN, lymph node.

Discussion

Based on the results of the present study, additional IHC using CK-19 antibodies to detect PALN micrometastases in resected pancreatic ductal adenocarcinoma is not an appropriate method to predict prognosis. While the rate of PALN metastasis has been reported to be 6–26% (2427), there may be a surgical selection bias that is affecting these numbers. For example, pancreatectomies are generally not performed when PALN metastasis is strongly suggested in pre-operative imaging studies, with observations that include large and conglomerated lymph nodes in the retroperitoneal para-aortic area. As a result, only nine patients (9.1%) exhibited PALN metastases in routine intraoperative frozen section biopsies and permanent pathological reports, and only one additional patient (1.0%) was shown to have PALN micrometastasis upon CK-19 staining. Therefore, the incidence of PALN metastasis was observed to be 10.1%. An accurate staging of PALN metastasis conducted by a careful pathological examination with routine HE staining is considered to be sufficient for tumor staging and is useful in predicting prognosis.

Due to the infrequency of resectable pancreatic ductal adenocarcinoma, only a few studies have demonstrated the clinical significance of PALN metastasis in resected pancreatic cancer. Doi et al(20) analyzed the clinicopathological factors in patients with short-term survival who underwent margin-negative radical extended pancreaticoduodenectomy. PALN metastasis was concluded to be the only independent factor for poor prognosis and ~85% of patients with PALN metastasis succumbed within one year. Shimada et al(28) reported that PALN metastasis was the definitive predictor of recurrence and a shorter survival outcome (<12 months). Yoshida et al(27) also identified the clinicopathological features and surgical outcomes of PALN-positive periampullary adenocarcinoma, and recommended performing intraoperative PALN sampling for frozen section biopsies. The study concluded that radical pancreatectomy with extended soft tissue clearance should not be performed in PALN-positive patients due to a poor oncological outcome. According to the data set of the present study, all eight patients whose frozen section biopsies confirmed PALN metastasis eventually developed tumor recurrences within 12 months of surgery and seven succumbed to the disease within two years (median survival, 17.5 months; Table I). These eight patients had significantly shorter survival periods compared with those patients with regional lymph node metastasis without PALN involvement (P<0.008; Fig. 2B).

Notably, a few cases of long-term survival in patients with PALN metastasis in resected pancreatic cancer have been reported (21,29). Although, as noted previously, studies suggest a poor outcome in PALN-positive resected pancreatic cancer, the majority do not consider the associated characteristics of PALN metastasis. Shimada et al(28), however, observed that PALN metastasis was notably associated with elevated CA 19–9 levels, a larger tumor size and positive surgical margins. Peritoneal cytology was correlated with PALN metastasis in the study (P=0.09). Furthermore, Yamada, et al(30) concluded that radical surgery may still have value for certain populations of patients with PALN metastasis, such as those aged 60 years or older, patients with tumors of <4 cm and those without portal vein involvement. The study also suggested that patients with one lymph node positive for PALN metastasis tended to have an improved prognosis compared with those with two or more positive PALN metastases (P=0.14).

Although the present data set was rather small and selection bias may have been a factor, it suggests a potential role for curative surgery in certain patient groups with resectable pancreatic cancer and PALN involvement. In spite of the poor prognosis in patients who demonstrated PALN metastases observed in routine HE staining, relatively longer survivals were noted in several patients (Table I). Considering a poor median survival time of 5–11 months for unresectable pancreatic tumors without distant metastases (31), the oncological outcomes of the patients included in the present study are thought to be significant, given that curative surgery is the only intervention that may lead to long-term survival in pancreatic cancer. Given the current state of continually improving surgical techniques, perioperative management and adjuvant therapies for pancreatic cancer, the findings of the present study may expand the role of surgery in managing pancreatic cancer, even when PALN metastasis is unexpectedly identified intraoperatively.

According to the present results, all eight patients with PALN metastasis confirmed in frozen section biopsies showed large metastatic tumor sizes and clustered gland patterns with extensive desmoplasia. However, one patient exhibited PALN metastasis that was only identified in routine HE staining of the permanent section, but was missed in the frozen section. This section demonstrated a relatively small size gland pattern without desmoplasia or extensive involvement of the node (Fig. 2). In spite of these histological differences, all of the patients possessed other adverse pathological factors including regional lymph node metastasis, perineural invasion and lymphovascular invasion (Table I). The patient whose PALN metastasis was only detected with IHC (case 10) demonstrated a small, isolated metastasis with less aggressive adverse pathological features. This patient had a relatively long-term survival of 41 months. Notably, despite a small sample size, a correlation was observed between the tumor burden of PALN metastasis and the survival outcome. A small PALN metastasis, which is either undetectable in frozen biopsy or only noticeable in immunostaining, may lead to a longer survival time.

The present study defined lymph node micrometastasis as metastatic tumor cells that are only detectable by IHC staining. However, a universal definition and clinical significance of micrometastasis for pancreatic cancer is still lacking. There are more established characterizations of micrometastasis in breast (32), esophageal (33,34), stomach (35) and colon cancers (36,37) Therefore, the micrometastasis of pancreatic cancer requires further study.

In summary, PALN metastasis that is undetected by routine HE staining in frozen section biopsy, but identified using CK-19 immunostaining, may indicate a relatively lower tumor burden. This may be associated with less aggressive behavior and a favorable prognosis in pancreatic cancer. Therefore, routine HE staining is thought to be sufficient for predicting prognosis. Additionally, in cases where PALN metastasis is unexpectedly identified in intraoperative frozen section biopsies, patients may benefit from curative radical surgery with aggressive adjuvant chemotherapy. However, further large volume investigations are warranted to validate this issue.

Acknowledgements

This study was supported by the Institute of Gastroenterology, Yonsei University Health System, Seoul, South Korea.

References

1 

Kayahara M, Nagakawa T, Ueno K, Ohta T, Takeda T and Miyazaki I: An evaluation of radical resection for pancreatic cancer based on the mode of recurrence as determined by autopsy and diagnostic imaging. Cancer. 72:2118–2123. 1993. View Article : Google Scholar : PubMed/NCBI

2 

Sener SF, Fremgen A, Menck HR and Winchester DP: Pancreatic cancer: a report of treatment and survival trends for 100,313 patients diagnosed from 1985–1995, using the National Cancer Database. J Am Coll Surg. 189:1–7. 1999.PubMed/NCBI

3 

Sperti C, Pasquali C, Piccoli A and Pedrazzoli S: Recurrence after resection for ductal adenocarcinoma of the pancreas. World J Surg. 21:195–200. 1997. View Article : Google Scholar : PubMed/NCBI

4 

Han SS, Jang JY, Kim SW, Kim WH, Lee KU and Park YH: Analysis of long-term survivors after surgical resection for pancreatic cancer. Pancreas. 32:271–275. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Sohn TA, Yeo CJ, Cameron JL, et al: Resected adenocarcinoma of the pancreas-616 patients: results, outcomes, and prognostic indicators. J Gastrointest Surg. 4:567–579. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Howard TJ, Krug JE, Yu J, et al: A margin-negative R0 resection accomplished with minimal postoperative complications is the surgeon’s contribution to long-term survival in pancreatic cancer. J Gastrointest Surg. 10:1338–1346. 2006.PubMed/NCBI

7 

Raut CP, Tseng JF, Sun CC, et al: Impact of resection status on pattern of failure and survival after pancreaticoduodenectomy for pancreatic adenocarcinoma. Ann Surg. 246:52–60. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Winter JM, Cameron JL, Campbell KA, et al: 1423 pancreaticoduodenectomies for pancreatic cancer: A single-institution experience. J Gastrointest Surg. 10:1199–1211. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Riall TS, Cameron JL, Lillemoe KD, et al: Pancreaticoduodenectomy with or without distal gastrectomy and extended retroperitoneal lymphadenectomy for periampullary adenocarcinoma - part 3: update on 5-year survival. J Gastrointest Surg. 9:1191–1206. 2005.

10 

Farnell MB, Pearson RK, Sarr MG, et al; Pancreas Cancer Working Group. A prospective randomized trial comparing standard pancreatoduodenectomy with pancreatoduodenectomy with extended lymphadenectomy in resectable pancreatic head adenocarcinoma. Surgery. 138:618–630. 2005. View Article : Google Scholar

11 

Pedrazzoli S, DiCarlo V, Dionigi R, et al: Standard versus extended lymphadenectomy associated with pancreatoduodenectomy in the surgical treatment of adenocarcinoma of the head of the pancreas: a multicenter, prospective, randomized study. Lymphadenectomy Study Group. Ann Surg. 228:508–517. 1998. View Article : Google Scholar

12 

Iqbal N, Lovegrove RE, Tilney HS, et al: A comparison of pancreaticoduodenectomy with extended pancreaticoduodenectomy: a meta-analysis of 1909 patients. Eur J Surg Oncol. 35:79–86. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Kurahara H, Takao S, Maemura K, et al: Impact of lymph node micrometastasis in patients with pancreatic head cancer. World J Surg. 31:483–492. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Geer RJ and Brennan MF: Prognostic indicators for survival after resection of pancreatic adenocarcinoma. Am J Surg. 165:68–73. 1993. View Article : Google Scholar : PubMed/NCBI

15 

Trede M, Schwall G and Saeger HD: Survival after pancreatoduodenectomy. 118 consecutive resections without an operative mortality. Ann Surg. 211:447–458. 1990. View Article : Google Scholar : PubMed/NCBI

16 

Yeo CJ, Cameron JL, Lillemoe KD, et al: Pancreaticoduodenectomy for cancer of the head of the pancreas. 201 patients. Ann Surg. 221:721–733. 1995. View Article : Google Scholar : PubMed/NCBI

17 

Sobin LH and Wittekind C: TNM Classification of Malignant Tumours (UICC). 5th edition. John Wiley & Sons; New York, NY: 1997

18 

Japanese Pancreas Society. Classification of Pancreatic Carcinoma. 1st English edition. Kanehara huppan; Tokyo, Japan: 1996

19 

Yoshiaki M, Kenichiro U, Takeshi S, et al: Prognostic impact of para-aortic lymph node metastasis in pancreatic ductal adenocarcinoma. World J Surg. 34:1900–1907. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Doi R, Kami K, Ito D, et al: Prognostic implication of para-aortic lymph node metastasis in resectable pancreatic cancer. World J Surg. 31:147–154. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Sakai M, Nakao A, Kaneko T, et al: Para-aortic lymph node metastasis in carcinoma of the head of the pancreas. Surgery. 137:606–611. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Andersen HB, Baden H, Brahe NE and Burcharth F: Pancreaticoduodenectomy for periampullary adenocarcinoma. J Am Coll Surg. 179:545–552. 1994.PubMed/NCBI

23 

Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL and Trotti A: AJCC cancer Staging Manual. 7th edition. Springer; New York, NY: 2010

24 

Ishikawa O, Ohigashi H, Sasaki Y, et al: Practical grouping of positive lymph nodes in pancreatic head cancer treated by an extended pancreatectomy. Surgery. 121:244–249. 1997. View Article : Google Scholar : PubMed/NCBI

25 

Nakao A, Harada A, Nonami T, et al: Lymph node metastases in carcinoma of the head of the pancreas region. Br J Surg. 82:399–402. 1995. View Article : Google Scholar : PubMed/NCBI

26 

Yoshida T, Aramaki M, Matsumoto T, Morii Y, Sasaki A and Kitano S: The pattern of lymphatic spread in carcinoma of the distal bile duct. Int Surg. 83:124–127. 1998.PubMed/NCBI

27 

Yoshida T, Matsumoto T, Sasaki A, Shibata K, Aramaki M and Kitano S: Outcome of paraaortic node-positive pancreatic head and bile duct adenocarcinoma. Am J Surg. 187:736–740. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Shimada K, Sakamoto Y, Sano T and Kosuge T: The role of paraaortic lymph node involvement on early recurrence and survival after macroscopic curative resection with extended lymphadenectomy for pancreatic carcinoma. J Am Coll Surg. 203:345–352. 2006. View Article : Google Scholar

29 

Kayahara M, Nagakawa T, Ohta T, et al: Analysis of paraaortic lymph node involvement in pancreatic carcinoma: a significant indication for surgery? Cancer. 85:583–590. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Yamada S, Nakao A, Fujii T, et al: Pancreatic cancer with paraaortic lymph node metastasis: a contraindication for radical surgery? Pancreas. 38:e13–e17. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Moertel CG, Frytak S, Hahn RG, et al: Therapy of locally unresectable pancreatic carcinoma: a randomized comparison of high dose (6000 rads) radiation alone, moderate dose radiation (4000 rads + 5-fluorouracil), and high dose radiation + 5-fluorouracil: The Gastrointestinal Tumor Study Group. Cancer. 48:1705–1710. 1981.PubMed/NCBI

32 

McGuckin MA, Cummings MC, Walsh MD, Hohn BG, Bennett IC and Wright RG: Occult axillary node metastases in breast cancer: their detection and prognostic significance. Br J Cancer. 73:88–95. 1996. View Article : Google Scholar : PubMed/NCBI

33 

Jiao X, Eslami A, Ioffe O, et al: Immunohistochemistry analysis of micrometastasis in pretreatment lymph nodes from patients with esophageal cancer. Ann Thorac Surg. 76:996–1000. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Tanabe T, Nishimaki T, Watanabe H, et al: Immunohistochemically detected micrometastasis in lymph nodes from superficial esophageal squamous cell carcinoma. J Surg Oncol. 82:153–159. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Ishigami S, Natsugoe S, Tokuda K, et al: Clinical impact of micrometastasis of the lymph node in gastric cancer. Am Surg. 69:573–577. 2003.PubMed/NCBI

36 

Bilchik AJ, Hoon DS, Saha S, et al: Prognostic impact of micrometastases in colon cancer: interim results of a prospective multicenter trial. Ann Surg. 246:568–577. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Bosch RC: Interview with Dr. Ramón Colomer Bosch, President of the Spanish Society of Medical Oncology (SEOM) by Enrique Garcia Jorda. Clin Transl Oncol. 10:310–312. 2008.

Related Articles

Journal Cover

November 2013
Volume 6 Issue 5

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
Choi SH, Kim SH, Choi JJ, Kang CM, Hwang HK and Lee WJ: Clinical necessity of the immunohistochemical reassessment of para‑aortic lymph nodes in resected pancreatic ductal adenocarcinoma. Oncol Lett 6: 1189-1194, 2013
APA
Choi, S.H., Kim, S.H., Choi, J.J., Kang, C.M., Hwang, H.K., & Lee, W.J. (2013). Clinical necessity of the immunohistochemical reassessment of para‑aortic lymph nodes in resected pancreatic ductal adenocarcinoma. Oncology Letters, 6, 1189-1194. https://doi.org/10.3892/ol.2013.1539
MLA
Choi, S. H., Kim, S. H., Choi, J. J., Kang, C. M., Hwang, H. K., Lee, W. J."Clinical necessity of the immunohistochemical reassessment of para‑aortic lymph nodes in resected pancreatic ductal adenocarcinoma". Oncology Letters 6.5 (2013): 1189-1194.
Chicago
Choi, S. H., Kim, S. H., Choi, J. J., Kang, C. M., Hwang, H. K., Lee, W. J."Clinical necessity of the immunohistochemical reassessment of para‑aortic lymph nodes in resected pancreatic ductal adenocarcinoma". Oncology Letters 6, no. 5 (2013): 1189-1194. https://doi.org/10.3892/ol.2013.1539