Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleClinical Studies

Preoperative Scoring System to Predict Prognosis in Patients Who Undergo Neoadjuvant Therapy for Pancreatic Cancer

KEN-ICHI OKADA, MANABU KAWAI, SEIKO HIRONO, KENSUKE TANIOKA, MOTOKI MIYAZAWA, YUJI KITAHATA, RYOHEI KOBAYASI, MASAKI UENO, SHINYA HAYAMI and HIROKI YAMAUE
Anticancer Research July 2020, 40 (7) 4033-4040; DOI: https://doi.org/10.21873/anticanres.14399
KEN-ICHI OKADA
1Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: okada@wakayama-med.ac.jp
MANABU KAWAI
1Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SEIKO HIRONO
1Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KENSUKE TANIOKA
2Clinical Study Support Center, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MOTOKI MIYAZAWA
1Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
YUJI KITAHATA
1Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
RYOHEI KOBAYASI
1Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MASAKI UENO
1Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SHINYA HAYAMI
1Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HIROKI YAMAUE
1Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: This study investigated the feasibility of an integrated scoring system of preoperative prognostic parameters using data from before/after neoadjuvant therapy in patients with borderline resectable pancreatic cancer (BRPC). Patients and Methods: We constructed and analyzed a prognostic scoring system using factors that were previously reported to be significant prognostic indicators or predictors of histological response. Results: We analyzed 28 consecutive patients with BRPC who underwent neoadjuvant therapy and subsequent surgical resection. Overall survival (OS) and recurrence free survival (RFS) were greater in patients with high scores (n=11) than in patients with low scores (n=17; log-rank test p=0.03/0.028). Pathological N0 status (p<0.05) and tumor cell destruction rate >50% (p<0.05) were found at a higher incidence among patients with high scores. Conclusion: OS and RFS can be predicted with an integrated scoring system that uses prognostic indices before/after neoadjuvant therapy for BRPC.

  • Apparent diffusion coefficient
  • neoadjuvant therapy
  • score
  • multimodality

Neoadjuvant therapy is an essential established strategy against various cancers (1-3). Efficacy of neoadjuvant therapy, meanwhile, can differ depending on the type of cancer and region. Such differences may be attributed to the lack of universal indicators for the assessment of the significance of neoadjuvant therapy. Pancreatic ductal adenocarcinoma (PDAC) ranks as the third leading cause of cancer-related deaths and is estimated to become the second leading cause in United States by 2030 (4). In PDAC, neoadjuvant therapy has been considered important for surgical strategy (5). Until now, the most easily implemented response determination method has been the evaluation of tumor diameter by contrast-enhanced CT imagery. However, the response of borderline resectable pancreatic cancer (BRPC) to neoadjuvant therapy is not always reflected by radiographic indicators, according to Katz et al. (6). Pancreatic cancer is typically a tumor accompanied by desmoplastic change, so it is thought that the tumor diameter does not change even if the cancer largely disappears.

Recently, various investigators have reported on the potential of tumor markers in laboratory tests or as parameters in imaging studies before and after neoadjuvant therapy to predict survival time or early recurrence. Sugiura et al. have reported a preoperative CA19-9 value ≥100 U/ml as a significant predictor of early recurrence and a poor prognosis after resection for PDAC (7). Meanwhile, Tsai et al. have reported that normalization of CA19-9 following neoadjuvant therapy, rather than the magnitude of change, was the strongest prognostic marker for long-term survival (8, 9). Yamamoto et al. introduced the preoperative pretreatment value of maximum standardized uptake value (SUV-max) ≥6.0 of 2-deoxy-2-[F-18] fluoro-D-glucose (FDG) positron emission tomography (FDG PET/CT) examination as systemic prognostic factor (10), and so did Akita et al. about a regression index >50% in SUV-max as local prognostic factor (11).

We have previously reported evaluation of apparent diffusion coefficient (ADC) value of whole tumor by diffusion-weighted magnetic resonance imaging (DW-MRI) before and after neoadjuvant therapy as an effective predictor of histological treatment effect (12, 13). When the residual cancer cells are histologically minimal, the prognosis of cancer is favorable (14). The weakness of the prediction of prognosis by a single parameter, however, is that it is affected by population bias. The current study, therefore, investigated the feasibility of an integrated scoring system of preoperative prognostic parameters using data from before and after neoadjuvant therapy. This prospective study uses patients with PDAC. We selected prognostic parameters that were previously reported as significant prognostic indicators or predictors of histological response (13). We constructed and analyzed the prognostic scoring system using the pre- and post-treatment values of CA19-9, SUV-max of FDG PET/CT examination, and whole tumor ADC values on DW-MRI. We compared overall survival (OS) and recurrence free survival (RFS) between patients with high scores and those with low scores. This trial is registered at the UMIN Clinical Trials Registry (UMIN000035672, 000028030) and at ClinicalTrials.gov (NCT02777463).

Patients and Methods

Patients. This study was approved by the Wakayama Medical University Hospital (WMUH) Institutional Review Board (No. 2484, 2092). It is a prospective study defining its protocol treatment as completion of neoadjuvant chemotherapy following pancreatectomy. Patients and methods have been described previously, including a CONSORT diagram (13). During the study period, 31 patients were enrolled. Of them, two did not undergo surgery for disease progression according to restaging imaging studies and one did not complete neoadjuvant therapy because of repeated cholecystitis. Finally, 28 consecutive patients were included in the study, who underwent surgery 3-8 weeks after the final administration of chemotherapy and were all recommended to receive postoperative adjuvant chemotherapy by systemic administration of oral S-1. Resectability status was defined according to National Comprehensive Cancer Network (NCCN) criteria, version 2.2015 (15). During the study period, patients with BRPC received neoadjuvant nab-paclitaxel plus gemcitabine therapy (13). As we have previously reported (12), routine imaging studies included staging and restaging of CT scans, enhanced magnetic resonance imaging (MRI), DW-MRI, and 2-deoxy-2-[F-18] fluoro-D-glucose (FDG) positron emission tomography (FDG PET/CT) prior to and after neoadjuvant therapy.

Neoadjuvant therapy. During the study period all enrolled patients were scheduled for 2 cycles of neoadjuvant nab-paclitaxel plus gemcitabine therapy. One cycle of regimen for BRPC comprised a 30 min intravenous infusion of nab-paclitaxel at a dose of 125 mg/m2, followed by a 30 min intravenous infusion of gemcitabine at a dose of 1000 mg/m2, on days 1, 8 and 15. All patients were then given a week of rest before a second identical cycle (16, 17).

Follow-up. After the registered patients discharged, they were observed as outpatients once per month for six months and then every three months thereafter. Laboratory study including serum CA19-9 levels and other general blood tests were performed at every visit. Dynamic computed tomography was performed every 1-3 months. Patients with elevated tumor markers or lesions suspicious for malignancy underwent FDG PET/CT examination or enhanced magnetic resonance imaging.

Endpoints. The primary endpoint of this prospective study was to confirm the feasibility of a prognostic scoring system with prognostic indices of pancreatic carcinoma. Secondary endpoints included analysis of overall survival and recurrence free survival between patients with high scores and those with low scores.

Definition of prognostic indices and scoring points. We determined six favorable parameters. Three systemically-weighted factors were determined as: value of CA19-9 <100 U/ml prior to neoadjuvant therapy (7), normal post-treatment value of CA 19-9 (8, 9), SUV-max value of FDG PET/CT <6.0 prior to neoadjuvant therapy (10). Three locally-weighted factors were determined as: reduction index of post/pre-treatment SUV-max value >50.0% (11), pre-treatment whole tumor ADC value >1.20×10−3 mm2/s (12), and post-treatment whole tumor ADC value >1.40×10−3 mm2/s (12, 13). Regarding the value of CA 19-9, a patient with Lewisa-b- type was included in this study. The collected data was assessed as favorable or non-favorable; favorable factors were counted as one point each and finally were evaluated as an integrated total point value between zero and six. An integrated point score ≥4 was categorized as a high score, and <4 as a low score (Table I) (7-13).

Diffusion MRI. Diffusion MRI procedures were the same as we have been previously reported (12, 13). Patients underwent MRI within three weeks before the start of neoadjuvant therapy and three weeks after the last administration. The ADC map was constructed using diffusion weighted images at b=0, 50, and 1000 s/mm2. Mean ADC values for each tumor were automatically calculated on the image of ADC map. The same MRI system/scanner, (Intera Achieva 3.0T, Philips Medical Systems, Andover, MA, USA) was used for all patients analyzed in this study. Regions of interest (ROI) were determined by a single experienced MR radiologist as 3-4 of the largest cross-sectional areas representing the whole tumor, as previously reported (12, 13).

18F-fluorodeoxyglucose (FDG) positron emission tomography (PET)/CT. Patients underwent PET/CT and MRI on the same day. PET studies and calculation of tumor SUVmax were performed as previously reported (13). The same PET/CT scanner (SET-3000BCT/L; Shimadzu, Kyoto, Japan) was used for all patients.

Ethical approval. The protocol for this research project has been approved by the Ethics Committee of the institution and it conforms to the provisions of the Declaration of Helsinki. Informed consent was obtained from all individual participants included in the study.

Statistical analysis. Descriptive statistics were calculated to examine the demographic characteristics of the study population. Survival times between the date of registration and death from any cause (without discrimination between deaths resulting from PDAC and from other causes) was taken as the outcome. Survival curves were calculated by the Kaplan-Meier method and comparison of survival curves was based on the log-rank test. Univariate analyses (Chi-square test) were primarily used for selecting variables based on a p-value <0.05. The significant variables and clinically effective factors were subjected to forward logistic regression analysis to determine the net effect for each predictor while controlling the effects of the other factors. The defined prognostic indices were compared between surviving and non-surviving patients, and the presence/absence of metastasis or recurrence between patients with high and low scores by Chi-squared test for categorical variables. All analyses were performed using the statistical software package SPSS II (version 18.0; SPSS, Tokyo, Japan) and JMP ver. 13.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Prognostic scoring system with prognostic indexes of pancreatic carcinoma.

Results

Patient characteristics. Between June 2016 and November 2018, 28 consecutive patients with borderline resectable pancreatic cancer (BRPC) underwent neoadjuvant therapy and subsequent surgical resection. Table II shows the characteristics of the 28 analyzed patients with BRPC. There was one patient with Lewisa-b- type. No patients required metal biliary stent insertion during the period of neoadjuvant chemotherapy. There was no mortality of the registered patients.

Prognostic indices. Figure 1 shows the actual integration score of each prognostic index by individual registered patients. The mean of the actual integration score was 2.9±1.5 (0-5), and the median score was 3 (Figure 2).

Validity of cutoff value for high score. ROC curve was calculated to determine the cutoff value of the actual integration score so that the two groups could be discriminated. The area under the curve for high scores discriminating the two groups was 0.718 (95% confidential interval, 0.538-0.898). Among candidate cutoff values, the final cutoff value was selected using Youden's index. Finally, we adopted 4 as the cutoff value in this study, and total score ≥4 was considered as high score (sensitivity 52.4%, specificity 100%).

Survival. Median follow-up time was 16 months (range=7-36 months). The estimated 1-year, 2-year, and 3-year survival rates were 89%, 72%, and 58% respectively, and the estimated median survival time (MST) was 16 months in all patients, including four cases of mortality. Estimated median recurrence-free survival time (RFS) was 16 months (range=5-36 months). Patients presented recurrence in local sites (n=6), in the liver (n=10), in the lungs (n=1), in the lymph node (n=1), and in the peritoneum (n=5) with some overlap. Table III shows univariate and multivariate analysis of prognostic indices for survival using the six prognostic indices and integrated total score. OS and RFS were greater in patients with high scores (n=11) than in patients with low scores (n=17; log-rank test p=0.03/0.028; Figures 3 and 4).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

Demographics and characteristics of patients with BRPC.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

The actual integration score of each prognostic index by individual registered patients.

Assessment for independence of prognostic factors. We performed multivariate analysis of prognostic indices and clinicopathological factors for overall survival to investigate the independence of clinically effective factors with the integrated total score. However, integrated total score was not an independent prognostic factor as other clinically effective factors (Table III).

Clinicopathological features of the high score group. The relationship between high/low scores and the significant clinicopathological features is shown in Table IV. Pathological N0 status (p=0.019) and histological response of more than Evans grade IIb (tumor cell destruction rate >50%) (p=0.019) were found in significantly higher incidence in patients with high scores.

Discussion

The present study was considered to be reliable prognostic analysis with a multimodality scoring system using the total and cutoff score of prognostic indices. OS and RFS of patients with high scores were significantly better than those in patients with low scores. To our knowledge, this is the only model that utilizes such preoperative predictors and the first to describe a multimodality scoring system for predicting prognosis in patients who undergo neoadjuvant therapy for PDAC. This concept may apply to various other kinds of cancer that require preoperative therapy.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

The mean of the actual integration score was 2.9±1.46 (0-5), and the median score was 3. Y-axis represents the integrated total score, and x-axis represents number of cases.

Originally, it is difficult to predict the overall survival rate by assessing preoperative treatment alone, because the overall survival rate depends on many other variables, such as race, surgical techniques, and regimen of chemotherapy (18). In this context, it is possible to preoperatively predict the long-term results by treatment effect if the method of evaluation of treatment response is appropriate. A universal treatment response assessment method with good accuracy is therefore required.

It is difficult to determine the response to treatment before excision. Currently, the golden standard for determining the response to preoperative treatment is considered to be the histologic death of the tumor cells in the resected specimen. Chua et al. have conducted a systematic review of 17 clinical studies (977 cases) of preoperative chemoradiotherapy for resectable pancreatic cancer between 2000-2010 (19, 20). Pathological complete response was, therefore, obtained at a relatively low rate of 5-15%, partial response was reported as 33-60%, and the minimal response as 38-42%. Since, both, Shimosato and Evans classifications require evaluation by an experienced pathologist, there is a possibility that the institutions are limited, and there is a tendency toward subjective evaluation because it cannot be quantified (21). The most easily implemented response assessment method is the evaluation of tumor diameter by imaging in contrast-enhanced CT. However, according to Evans et al., no cases have shown more than 50% tumor reduction by NACRT (21). Pancreatic cancer is originally a tumor accompanied by desmoplastic change, so it is thought that the cancer diameter does not significantly change even if, to a large extent, the cancer disappears. FDG PET/CT shows uptake of FDG into cells, so it is expected that reduction of tumor cells will reduce FDG accumulation. In esophageal cancer and malignant lymphoma, there are many reports that SUV in PET examinations are useful for response evaluation. Akita et al. have reported the usefulness of PET in pancreatic cancer, and stated that groups with a ≥50% or more decrease in SUV-max before and after NACRT were significantly related to Evans grade III and IV, and the long-term results were also significantly better. The reliability may be lowered, however, if the control of diabetes mellitus is complicated as a peculiarity of pancreatic cancer. If pancreatitis is complicated, FDG may be accumulated in the whole pancreas and it may not be suitable for evaluation. Detection of trace markers by blood tests may compensate for the shortcomings of diagnostic imaging. Regarding tumor markers, CA19-9 has been used, but there may be unreliability owing to biliary obstruction and the possibility that it will not be elevated depending on the race of patients. Each of the above prognostic indices have individual weaknesses in independently predicting prognosis.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Comparison of overall survival curves according to the integrated scoring index. Overall survival rate was significantly higher in patients with high index ≥4 (solid line, n=11) than those with low index <4 (dotted line, n=17) (log-rank test).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Comparison of recurrence-free survival curves according to the integrated scoring index. Recurrence free survival rate was significantly higher in patients with high index ≥4 (solid line, n=11) compared to those with low index <4 (dotted line, n=17) (log-rank test).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table III.

Univariate and multivariate analysis of prognostic indexes for overall survival.

In a recent study, Dreyer et al. aimed to define preoperative clinical and molecular characteristics that would allow better patient selection for operative resection for PDAC without neoadjuvant chemotherapy or radiotherapy (22). Among their patients with poor preoperative nomogram scores using existing risk factors, approximately 50% died within a year of resection. In the present study, we used predictors prior to and after neoadjuvant therapy and demonstrated that high scores were negatively associated with lymph node metastasis and the patients with higher than grade IIb cancer had better histological response to neoadjuvant therapy. Similarly, Perri et al. have also revealed radiographic and serologic change in two predictors of pathologic major response to preoperative therapy for pancreatic cancer (23).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table IV.

High score group and clinicopathological features in pancreatic cancer.

Our results may be limited by selection bias of prognostic indices, the small number of patients, short follow-up period, and the total score ≥4 as high score, which revealed low sensitivity. Differences between populations in resectability categories that produced each prognostic index may be also considered to be a bias in the present study. The real question in clinical situations is whether this is just a prognostic indicator, or if there will be a definitive role of this value in changing therapy. How this analysis will be used to improve therapy or to change the planned approach requires validation.

However, the strength of the multimodality scoring system is that, when applied to different populations, the weakness of each parameter will be compensated by its combination with other parameters and differences in the predictive ability due to population bias for each population will be corrected. The predictive role of the scoring system may be useful in algorithmic treatment approach to neoadjuvant therapy for BRPC as well as resectable pancreatic cancer (24-26). Recently, new surrogate markers obtained by liquid biopsy have attracted attention (27, 28), and it is expected that such novel markers will be gradually developed and used for decision-making regarding preoperative treatment within an integrated scoring system and treatment response assessment on clinical setting during the neoadjuvant therapy. These stronger markers are expected to dramatically improve sensitivity, specificity, and accuracy of the scoring system. This may more precisely predict site/time of recurrence and prognosis after neoadjuvant therapy. A future collaborative study with a larger patient population is therefore needed to confirm the feasibility of this integrated scoring system in predicting prognosis in patients with PDAC.

In conclusion, prediction of OS and RFS with the integrated scoring system using preoperative prognostic indices before/after neoadjuvant therapy for PDAC in this study was feasible. Patients with high scores had favorable survival compared with those with low scores. The poorer prognosis in patients with low scores may be related to more advanced tumors.

Acknowledgements

The Authors would like to thank Benjamin Phillis at the Clinical Study Support Center, Wakayama Medical University Hospital, for proofreading and editing the manuscript.

Footnotes

  • Authors' Contributions

    Study concept and design: KO; Acquisition of data: MM, YK, RK, MU, SH; Analysis and interpretation of data: KT (Statistical analysis); Drafting of manuscript: KO, MK, SH; Critical revision of manuscript: HY.

  • Conflicts of Interest

    All Authors declare no conflicts of interest in relation to this study.

  • Received May 10, 2020.
  • Revision received June 2, 2020.
  • Accepted June 3, 2020.
  • Copyright© 2020, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved

References

  1. ↵
    1. Ando N,
    2. Kato H,
    3. Igaki H,
    4. Shinoda M,
    5. Ozawa S,
    6. Shimizu H,
    7. Nakamura T,
    8. Yabusaki H,
    9. Aoyama N,
    10. Kurita A,
    11. Ikeda K,
    12. Kanda T,
    13. Tsujinaka T,
    14. Nakamura K,
    15. Fukuda H
    : A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol 19: 68-74, 2012. PMID: 21879261. DOI: 10.1245/s10434-011-2049-9
    OpenUrlCrossRefPubMed
    1. Swedish Rectal Cancer Trial,
    2. Cedermark B,
    3. Dahlberg M,
    4. Glimelius B,
    5. Påhlman L,
    6. Rutqvist LE,
    7. Wilking N
    : Improved survival with preoperative radiotherapy in resectable rectal cancer. N Engl J Med 336: 980-987, 1997. PMID: 9091798. DOI: 10.1056/NEJM199704033361402
    OpenUrlCrossRefPubMed
  2. ↵
    1. Abraha I,
    2. Aristei C,
    3. Palumbo I,
    4. Lupattelli M,
    5. Trastulli S,
    6. Cirocchi R,
    7. De Florio R,
    8. Valentini V
    : Preoperative radiotherapy and curative surgery for the management of localised rectal carcinoma. Cochrane Database Syst Rev 10: CD002102, 2018. DOI: 10.1002/14651858.CD002102.pub3
  3. ↵
    1. Siegel RL,
    2. Miller KD,
    3. Jemal A
    : Cancer statistics, 2020. CA Cancer J Clin 70: 7-30, 2020. PMID: 31912902. DOI: 10.3322/caac.21590
    OpenUrlCrossRefPubMed
  4. ↵
    1. Motoi F,
    2. Kosuge T,
    3. Ueno H,
    4. Yamaue H,
    5. Satoi S,
    6. Sho M,
    7. Honda G,
    8. Matsumoto I,
    9. Wada K,
    10. Furuse J,
    11. Matsuyama Y,
    12. Unno M,
    13. Study Group of Preoperative Therapy for Pancreatic Cancer (Prep),
    14. Japanese Study Group of Adjuvant Therapy for Pancreatic cancer (JSAP)
    : Randomized phase II/III trial of neoadjuvant chemotherapy with gemcitabine and S-1 versus upfront surgery for resectable pancreatic cancer (Prep-02/JSAP05). Jpn J Clin Oncol 49: 190-194, 2019. PMID: 30608598. DOI: 10.1093/jjco/hyy190
    OpenUrl
  5. ↵
    1. Katz MH,
    2. Fleming JB,
    3. Bhosale P,
    4. Varadhachary G,
    5. Lee JE,
    6. Wolff R,
    7. Wang H,
    8. Abbruzzese J,
    9. Pisters PW,
    10. Vauthey JN,
    11. Charnsangavej C,
    12. Tamm E,
    13. Crane CH,
    14. Balachandran A
    : Response of borderline resectable pancreatic cancer to neoadjuvant therapy is not reflected by radiographic indicators. Cancer 118: 5749-5756, 2012. PMID: 22605518. DOI: 10.1002/cncr.27636
    OpenUrlCrossRefPubMed
  6. ↵
    1. Sugiura T,
    2. Uesaka K,
    3. Kanemoto H,
    4. Mizuno T,
    5. Sasaki K,
    6. Furukawa H,
    7. Matsunaga K,
    8. Maeda A
    : Serum CA19-9 is a significant predictor among preoperative parameters for early recurrence after resection of pancreatic adenocarcinoma. J Gastrointest Surg 16: 977-985, 2012. PMID: 22411488. DOI: 10.1007/s11605-012-1859-9
    OpenUrlCrossRefPubMed
  7. ↵
    1. Tsai S,
    2. George B,
    3. Wittmann D,
    4. Ritch PS,
    5. Krepline AN,
    6. Aldakkak M,
    7. Barnes CA,
    8. Christians KK,
    9. Dua K,
    10. Griffin M,
    11. Hagen C,
    12. Hall WA,
    13. Erickson BA,
    14. Evans DB
    : Importance of normalization of CA19-9 levels following neoadjuvant therapy in patients with localized pancreatic cancer. Ann Surg 271: 740-747, 2020. PMID: 30312198. DOI: 10.1097/SLA.0000000000003049
    OpenUrl
  8. ↵
    1. Tzeng CW,
    2. Balachandran A,
    3. Ahmad M,
    4. Lee JE,
    5. Krishnan S,
    6. Wang H,
    7. Crane CH,
    8. Wolff RA,
    9. Varadhachary GR,
    10. Pisters PW,
    11. Aloia TA,
    12. Vauthey JN,
    13. Fleming JB,
    14. Katz MH
    : Serum carbohydrate antigen 19-9 represents a marker of response to neoadjuvant therapy in patients with borderline resectable pancreatic cancer. HPB (Oxford) 16: 430-438, 2014. PMID: 23991810. DOI: 10.1111/hpb.12154
    OpenUrlCrossRefPubMed
  9. ↵
    1. Yamamoto T,
    2. Sugiura T,
    3. Mizuno T,
    4. Okamura Y,
    5. Aramaki T,
    6. Endo M,
    7. Uesaka K
    : Preoperative FDG-PET predicts early recurrence and a poor prognosis after resection of pancreatic adenocarcinoma. Ann Surg Oncol 22: 677-684, 2015. PMID: 25190125. DOI: 10.1245/s10434-014-4046-2
    OpenUrl
  10. ↵
    1. Akita H,
    2. Takahashi H,
    3. Ohigashi H,
    4. Tomokuni A,
    5. Kobayashi S,
    6. Sugimura K,
    7. Miyoshi N,
    8. Moon JH,
    9. Yasui M,
    10. Omori T,
    11. Miyata H,
    12. Ohue M,
    13. Fujiwara Y,
    14. Yano M,
    15. Ishikawa O,
    16. Sakon M
    : FDG-PET predicts treatment efficacy and surgical outcome of pre-operative chemoradiation therapy for resectable and borderline resectable pancreatic cancer. Eur J Surg Oncol 43: 1061-1067, 2017. PMID: 28389044. DOI: 10.1016/j.ejso.2017.03.015
    OpenUrl
  11. ↵
    1. Okada KI,
    2. Hirono S,
    3. Kawai M,
    4. Miyazawa M,
    5. Shimizu A,
    6. Kitahata Y,
    7. Ueno M,
    8. Hayami S,
    9. Kojima F,
    10. Yamaue H
    : Value of apparent diffusion coefficient prior to neoadjuvant therapy is a predictor of histologic response in patients with borderline resectable pancreatic carcinoma. J Hepatobiliary Pancreat Sci 24: 161-168, 2017. PMID: 28064462. DOI: 10.1002/jhbp.430
    OpenUrl
  12. ↵
    1. Okada KI,
    2. Kawai M,
    3. Hirono S,
    4. Kojima F,
    5. Tanioka K,
    6. Terada M,
    7. Miyazawa M,
    8. Kitahata Y,
    9. Iwahashi Y,
    10. Ueno M,
    11. Hayami S,
    12. Murata SI,
    13. Shimokawa T,
    14. Yamaue H
    : Diffusion-weighted MRI predicts the histologic response for neoadjuvant therapy in patients with pancreatic cancer: a prospective study (DIFFERENT trial). Langenbecks Arch Surg 405: 23-33, 2020. PMID: 31993737. DOI: 10.1007/s00423-020-01857-4
    OpenUrl
  13. ↵
    1. Chatterjee D,
    2. Katz MH,
    3. Rashid A,
    4. Varadhachary GR,
    5. Wolff RA,
    6. Wang H,
    7. Lee JE,
    8. Pisters PW,
    9. Vauthey JN,
    10. Crane C,
    11. Gomez HF,
    12. Abbruzzese JL,
    13. Fleming JB,
    14. Wang H
    : Histologic grading of the extent of residual carcinoma following neoadjuvant chemoradiation in pancreatic ductal adenocarcinoma: a predictor for patient outcome. Cancer 118: 3182-3190, 2012. PMID: 22028089. DOI: 10.1002/cncr.26651
    OpenUrlCrossRefPubMed
  14. ↵
    1. National Comprehensive Cancer Network
    . NCCN practice guidelines for pancreatic cancer, version 2. 2015 Available at: http://www.nccn.org/professionals/physician_gls/PDF/pancreatic.pdf [Last Accessed January 15, 2016]
  15. ↵
    1. Von Hoff DD,
    2. Ramanathan RK,
    3. Borad MJ,
    4. Laheru DA,
    5. Smith LS,
    6. Wood TE,
    7. Korn RL,
    8. Desai N,
    9. Trieu V,
    10. Iglesias JL,
    11. Zhang H,
    12. Soon-Shiong P,
    13. Shi T,
    14. Rajeshkumar NV,
    15. Maitra A,
    16. Hidalgo M
    : Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol 29: 4548-4554, 2011. PMID: 21969517. DOI: 10.1200/JCO.2011.36.5742
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Okada KI,
    2. Hirono S,
    3. Kawai M,
    4. Miyazawa M,
    5. Shimizu A,
    6. Kitahata Y,
    7. Ueno M,
    8. Hayami S,
    9. Yamaue H
    : Phase I study of nab-paclitaxel plus gemcitabine as neoadjuvant therapy for borderline resectable pancreatic cancer. Anticancer Res 37: 853-858, 2017. PMID: 28179342. DOI: 10.21873/anticanres.11389
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Truty MJ,
    2. Kendrick ML,
    3. Nagorney DM,
    4. Smoot RL,
    5. Cleary SP,
    6. Graham RP,
    7. Goenka AH,
    8. Hallemeier CL,
    9. Haddock MG,
    10. Harmsen WS,
    11. Mahipal A,
    12. McWilliams RR,
    13. Halfdanarson TR,
    14. Grothey AF
    : Factors predicting response, perioperative outcomes, and survival following total neoadjuvant therapy for borderline/locally advanced pancreatic cancer. Ann Surg, 2019. PMID: 30946090. DOI: 10.1097/SLA.0000000000003284
  18. ↵
    1. Chua TC,
    2. Saxena A
    : Preoperative chemoradiation followed by surgical resection for resectable pancreatic cancer: a review of current results. Surg Oncol 20: e161-168, 2011. PMID: 21704510. DOI: 10.1016/j.suronc.2011.05.003
    OpenUrlPubMed
  19. ↵
    1. Tang K,
    2. Lu W,
    3. Qin W,
    4. Wu Y
    : Neoadjuvant therapy for patients with borderline resectable pancreatic cancer: A systematic review and meta-analysis of response and resection percentages. Pancreatology 16: 28-37, 2016. PMID: 26687001. DOI: 10.1016/j.pan.2015.11.007
    OpenUrl
  20. ↵
    1. Evans DB,
    2. Rich TA,
    3. Byrd DR,
    4. Cleary KR,
    5. Connelly JH,
    6. Levin B,
    7. Charnsangavej C,
    8. Fenoglio CJ,
    9. Ames FC
    : Preoperative chemoradiation and pancreaticoduodenectomy for adenocarcinoma of the pancreas. Arch Surg 127: 1335-1339, 1992. PMID: 1359851. DOI: 10.1001/archsurg.1992.01420110083017
    OpenUrlCrossRefPubMed
  21. ↵
    1. Dreyer SB,
    2. Pinese M,
    3. Jamieson NB,
    4. Scarlett CJ,
    5. Colvin EK,
    6. Pajic M,
    7. Johns AL,
    8. Humphris JL,
    9. Wu J,
    10. Cowley MJ,
    11. Chou A,
    12. Nagrial AM,
    13. Chantrill L,
    14. Chin VT,
    15. Jones MD,
    16. Moran-Jones K,
    17. Carter CR,
    18. Dickson EJ,
    19. Samra JS,
    20. Merrett ND,
    21. Gill AJ,
    22. Kench JG,
    23. Duthie F,
    24. Miller DK,
    25. Cooke S,
    26. Aust D,
    27. Knösel T,
    28. Rümmele P,
    29. Grützmann R,
    30. Pilarsky C,
    31. Nguyen NQ,
    32. Musgrove EA,
    33. Bailey PJ,
    34. McKay CJ,
    35. Biankin AV,
    36. Chang DK,
    37. Australian Pancreatic Cancer Genome Initiative,
    38. Glasgow Precision Oncology Laboratory
    : Precision oncology in surgery: Patient selection for operable pancreatic cancer. Ann Surg, 2018. PMID: 30570546. DOI: 10.1097/SLA.0000000000003143
  22. ↵
    1. Perri G,
    2. Prakash L,
    3. Wang H,
    4. Bhosale P,
    5. Varadhachary GR,
    6. Wolff R,
    7. Fogelman D,
    8. Overman M,
    9. Pant S,
    10. Javle M,
    11. Koay E,
    12. Herman J,
    13. Kim M,
    14. Ikoma N,
    15. Tzeng CW,
    16. Lee JE,
    17. Katz MHG
    : Radiographic and serologic predictors of pathologic major response to preoperative therapy for pancreatic cancer. Ann Surg, 2019. PMID: 31274655. DOI: 10.1097/SLA.0000000000003442
  23. ↵
    1. Cassinotto C,
    2. Mouries A,
    3. Lafourcade JP,
    4. Terrebonne E,
    5. Belleannée G,
    6. Blanc JF,
    7. Lapuyade B,
    8. Vendrely V,
    9. Laurent C,
    10. Chiche L,
    11. Wagner T,
    12. Sa-Cunha A,
    13. Gaye D,
    14. Trillaud H,
    15. Laurent F,
    16. Montaudon M
    : Locally advanced pancreatic adenocarcinoma: reassessment of response with CT after neoadjuvant chemotherapy and radiation therapy. Radiology 273: 108-116, 2016. PMID: 24960211. DOI: 10.1148/radiol.14132914
    OpenUrl
    1. Mellon EA,
    2. Jin WH,
    3. Frakes JM,
    4. Centeno BA,
    5. Strom TJ,
    6. Springett GM,
    7. Malafa MP,
    8. Shridhar R,
    9. Hodul PJ,
    10. Hoffe SE
    : Predictors and survival for pathologic tumor response grade in borderline resectable and locally advanced pancreatic cancer treated with induction chemotherapy and neoadjuvant stereotactic body radiotherapy. Acta Oncol 56: 391-397, 2017. PMID: 27885876. DOI: 10.1080/0284186X.2016.1256497
    OpenUrl
  24. ↵
    1. Gemenetzis G,
    2. Groot VP,
    3. Blair AB,
    4. Laheru DA,
    5. Zheng L,
    6. Narang AK,
    7. Fishman EK,
    8. Hruban RH,
    9. Yu J,
    10. Burkhart RA,
    11. Cameron JL,
    12. Weiss MJ,
    13. Wolfgang CL,
    14. He J
    : Survival in locally advanced pancreatic cancer after neoadjuvant therapy and surgical resection. Ann Surg 270: 340-347, 2019. PMID: 29596120. DOI: 10.1097/SLA.0000000000002753
    OpenUrl
  25. ↵
    1. Xie ZB,
    2. Yao L,
    3. Jin C,
    4. Fu DL
    . Circulating tumor cells in pancreatic cancer patients: efficacy in diagnosis and value in prognosis. Discov Med 22:121-128, 2016. PMID: 27755967.
    OpenUrl
  26. ↵
    1. Gemenetzis G,
    2. Groot VP,
    3. Yu J,
    4. Ding D,
    5. Teinor JA,
    6. Javed AA,
    7. Wood LD,
    8. Burkhart RA,
    9. Cameron JL,
    10. Makary MA,
    11. Weiss MJ,
    12. He J,
    13. Wolfgang CL
    : Circulating tumor cells dynamics in pancreatic adenocarcinoma correlate with disease status: results of the prospective CLUSTER study. Ann Surg 268: 408-420, 2018. PMID: 30080739. DOI: 10.1097/SLA.0000000000002925
    OpenUrl
PreviousNext
Back to top

In this issue

Anticancer Research: 40 (7)
Anticancer Research
Vol. 40, Issue 7
July 2020
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Preoperative Scoring System to Predict Prognosis in Patients Who Undergo Neoadjuvant Therapy for Pancreatic Cancer
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
1 + 0 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Preoperative Scoring System to Predict Prognosis in Patients Who Undergo Neoadjuvant Therapy for Pancreatic Cancer
KEN-ICHI OKADA, MANABU KAWAI, SEIKO HIRONO, KENSUKE TANIOKA, MOTOKI MIYAZAWA, YUJI KITAHATA, RYOHEI KOBAYASI, MASAKI UENO, SHINYA HAYAMI, HIROKI YAMAUE
Anticancer Research Jul 2020, 40 (7) 4033-4040; DOI: 10.21873/anticanres.14399

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Preoperative Scoring System to Predict Prognosis in Patients Who Undergo Neoadjuvant Therapy for Pancreatic Cancer
KEN-ICHI OKADA, MANABU KAWAI, SEIKO HIRONO, KENSUKE TANIOKA, MOTOKI MIYAZAWA, YUJI KITAHATA, RYOHEI KOBAYASI, MASAKI UENO, SHINYA HAYAMI, HIROKI YAMAUE
Anticancer Research Jul 2020, 40 (7) 4033-4040; DOI: 10.21873/anticanres.14399
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Patients and Methods
    • Results
    • Discussion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Efficacy and Safety of Lenvatinib After Progression on First-line Atezolizumab Plus Bevacizumab Treatment in Advanced Hepatocellular Carcinoma Patients
  • Efficacy and Safety of Platinum-based Chemotherapy With Bevacizumab Followed by Bevacizumab Maintenance for Recurrent Ovarian, Fallopian Tube, and Primary Peritoneal Cancer During PARP Inhibitor Therapy: A Multicenter Retrospective Study
  • Real-world Data of Palliative First-line Checkpoint Inhibitor Therapy for Head and Neck Cancer
Show more Clinical Studies

Similar Articles

Keywords

  • apparent diffusion coefficient
  • neoadjuvant therapy
  • score
  • multimodality
Anticancer Research

© 2023 Anticancer Research

Powered by HighWire