101
|
Radiotherapy for Resectable and Borderline Resectable Pancreas Cancer: When and Why? J Gastrointest Surg 2021; 25:843-848. [PMID: 33205307 DOI: 10.1007/s11605-020-04838-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 01/31/2023]
Abstract
The role of (chemo) radiation in the perioperative management of patients with resectable and borderline resectable pancreatic ductal adenocarcinoma is controversial. Herein, we review and interpret existing data relating to the ability of (chemo) radiation to "downstage" pancreatic tumors, delay recurrence, and prolong patients' survival. In sum, the evidence suggests that while neoadjuvant (chemo) radiation may impact pathologic metrics favorably, it rarely converts anatomically unresectable tumors to resectable ones. And while data do support the ability of (chemo)radiation to delay cancer progression, its ability to prolong longevity has not been confirmed. It is possible that (chemo)radiation is effective in prolonging the survival of select patients, but to date, this cohort remains undefined due to heterogeneity in both the populations studied and the regimens used to treat them. Based on our interpretation of existing data, we currently administer neoadjuvant and adjuvant (chemo)radiation selectively to patients with localized pancreatic cancer who we consider at highest risk for local progression. We may also use it as an alternative to pancreatectomy in patients who are poor candidates for surgery. Ultimately, the role of (chemo)radiation in these settings is evolving. Better studies of patients most likely to benefit from its local effects are necessary to clearly define its place within the perioperative treatment algorithms used for patients with localized pancreatic cancer.
Collapse
|
102
|
Hwang SH, Park MS. [Radiologic Evaluation for Resectability of Pancreatic Adenocarcinoma]. TAEHAN YONGSANG UIHAKHOE CHI 2021; 82:315-334. [PMID: 36238739 PMCID: PMC9431945 DOI: 10.3348/jksr.2021.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 11/25/2022]
Abstract
Imaging studies play an important role in the detection, diagnosis, assessment of resectability, staging, and determination of patient-tailored treatment options for pancreatic adenocarcinoma. Recently, for patients diagnosed with borderline resectable or locally advanced pancreatic cancers, it is recommended to consider curative-intent surgery following neoadjuvant or palliative therapy, if possible. This review covers how to interpret imaging tests and what to consider when assessing resectability, diagnosing distant metastasis, and re-assessing the resectability of pancreatic cancer after neoadjuvant or palliative therapy.
Collapse
|
103
|
Factors Predicting Response, Perioperative Outcomes, and Survival Following Total Neoadjuvant Therapy for Borderline/Locally Advanced Pancreatic Cancer. Ann Surg 2021; 273:341-349. [PMID: 30946090 DOI: 10.1097/sla.0000000000003284] [Citation(s) in RCA: 250] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To identify predictive factors associated with operative morbidity, mortality, and survival outcomes in patients with borderline resectable (BR) or locally advanced (LA) pancreatic ductal adenocarcinoma (PDAC) undergoing total neoadjuvant therapy (TNT). BACKGROUND The optimal preoperative treatment sequencing for BR/LA PDA is unknown. TNT, or systemic chemotherapy followed by chemoradiation (CRT), addresses both occult metastases and positive margin risks and thus is a potentially optimal strategy; however, factors predictive of perioperative and survival outcomes are currently undefined. METHODS We reviewed our experience in BR/LA patients undergoing resection from 2010 to 2017 following TNT assessing operative morbidity, mortality, and survival in order to define outcome predictors and response endpoints. RESULTS One hundred ninety-four patients underwent resection after TNT, including 123 (63%) BR and 71 (37%) LA PDAC. FOLFIRINOX or gemcitabine along with nab-paclitaxel were used in 165 (85%) and 65 (34%) patients, with 36 (19%) requiring chemotherapeutic switch before long-course CRT and subsequent resection. Radiologic anatomical downstaging was uncommon (28%). En bloc venous and/or arterial resection was required in 125 (65%) patients with 94% of patients achieving R0 margins. The 90-day major morbidity and mortality was 36% and 6.7%, respectively. Excluding operative mortalities, the median, 1-year, 2-year, and 3-year recurrence-free survival (RFS) [overall survival (OS)] rates were 23.5 (58.8) months, 65 (96)%, 48 (78)%, and 32 (62)%, respectively. Radiologic downstaging, vascular resection, and chemotherapy regimen/switch were not associated with survival. Only 3 factors independently associated with prolonged survival, including extended duration (≥6 cycles) chemotherapy, optimal post-chemotherapy CA19-9 response, and major pathologic response. Patients achieving all 3 factors had superior survival outcomes with a survival detriment for each failing factor. In a subset of patients with interval metabolic (PET) imaging after initial chemotherapy, complete metabolic response highly correlated with major pathologic response. CONCLUSION Our TNT experience in resected BR/LA PDAC revealed high negative margin rates despite low radiologic downstaging. Extended duration chemotherapy with associated biochemical and pathologic responses highly predicted postoperative survival. Potential modifications of initial chemotherapy treatment include extending cycle duration to normalize CA19-9 or achieve complete metabolic response, or consideration of chemotherapeutic switch in order to achieve these factors may improve survival before moving forward with CRT and subsequent resection.
Collapse
|
104
|
Holyoake DLP, Robinson M, Silva M, Grose D, McIntosh D, Sebag-Montefiore D, Radhakrishna G, Mukherjee S, Hawkins MA. SPARC, a phase-I trial of pre-operative, margin intensified, stereotactic body radiation therapy for pancreatic cancer. Radiother Oncol 2021; 155:278-284. [PMID: 33217498 DOI: 10.1016/j.radonc.2020.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/19/2020] [Accepted: 11/08/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Following resection of pancreatic cancer, risk of positive margins and local recurrence remain high, especially for borderline-resectable pancreatic cancer (BRPC). We aimed to establish the maximum tolerated dose of a margin-intensified five-fraction stereotactic body radiotherapy (SBRT) regimen designed to treat the region at risk. MATERIALS AND METHODS We conducted a prospective multicentre phase-1 rolling-six dose-escalation study. BRPC patients received pre-operative SBRT, with one dose to the primary tumour and an integrated boost to the region where tumour was in contact with vasculature. Four dose-levels were proposed, with starting dose 30 Gy to primary PTV and 45 Gy to boost volume (PTV_R), in five daily fractions. Primary endpoint was maximum tolerated dose (MTD), defined as highest dose where zero of three or one of six patients experienced dose-limiting toxicity (DLT). RESULTS Twelve patients were registered, eleven received SBRT. Radiotherapy was well tolerated with all treatment completed as scheduled. Dose was escalated one level up from starting dose without encountering any DLT (prescribed 32.5 Gy PTV, 47.5 Gy PTV_R). Nine serious adverse reactions or events occurred (seven CTCAE Grade 3, two Grade 4). Two patients went on to have surgical resection. Median overall survival for SBRT patients was 8.1 months. The study closed early when it was unable to recruit to schedule. CONCLUSION Toxicity of SBRT was low for the two dose-levels that were tested, but MTD was not established. Few patients subsequently underwent resection of pancreatic tumour after SBRT, and it is difficult to draw conclusions regarding the safety or toxicity of these therapies in combination.
Collapse
Affiliation(s)
- Daniel L P Holyoake
- Norfolk and Norwich University Hospitals NHS Foundation Trust, United Kingdom
| | - Maxwell Robinson
- Oxford University Hospitals NHS Foundation Trust, United Kingdom; CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, United Kingdom
| | - Michael Silva
- Oxford University Hospitals NHS Foundation Trust, United Kingdom
| | - Derek Grose
- The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - David McIntosh
- The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - David Sebag-Montefiore
- University of Leeds, United Kingdom; Leeds Cancer Centre, St James's University Hospital, United Kingdom
| | | | - Somnath Mukherjee
- Oxford University Hospitals NHS Foundation Trust, United Kingdom; CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, United Kingdom
| | - Maria A Hawkins
- Dept of Medical Physics & Biomedical Engineering, University College London, United Kingdom.
| |
Collapse
|
105
|
Induction Chemotherapy for Primarily Unresectable Locally Advanced Pancreatic Adenocarcinoma-Who Will Benefit from a Secondary Resection? ACTA ACUST UNITED AC 2021; 57:medicina57010077. [PMID: 33477505 PMCID: PMC7831047 DOI: 10.3390/medicina57010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/31/2020] [Accepted: 01/15/2021] [Indexed: 11/18/2022]
Abstract
Background and Objectives: An increasing number of patients (pts) with locally advanced pancreatic cancer (LAPC) are treated with an intensive neoadjuvant therapy to obtain a secondary curative resection. Only a certain number of patients benefit from this intention. The aim of this investigation was to identify prognostic factors which may predict a benefit for secondary resection. Materials and Methods: Survival time and clinicopathological data of pts with pancreatic cancer were prospective and consecutively collected in our Comprehensive Cancer Center Database. For this investigation, we screened for pts with primarily unresectable pancreatic cancer who underwent a secondary resection after receiving induction therapy in the time between March 2017 and May 2019. Results: 40 pts had a sufficient database to carry out a reliable analysis. The carbohydrate-antigen 19-9 (CA 19-9) level of the pts treated with induction therapy decreased by 44.7% from 4358.3 U/mL to 138.5 U/mL (p = 0.001). The local cancer extension was significantly reduced (p < 0.001), and the Eastern Cooperative Oncology Group (ECOG) performance status was lowered (p = 0.03). The median overall survival (mOS) was 20 months (95% CI: 17.2–22.9). Pts who showed a normal CA 19-9 level (<37 U/mL) at diagnosis and after neoadjuvant therapy or had a Body Mass Index (BMI) below 25 kg/m2 after chemotherapy had a significant prolonged overall survival (29 vs. 19 months, p = 0.02; 26 vs. 18 months, p = 0.04; 15 vs. 24 months, p = 0.01). Pts who still presented elevated CA 19-9 levels >400 U/mL after induction therapy did not profit from a secondary resection (24 vs. 7 months, p < 0.001). Nodal negativity as well as the performance of an adjuvant therapy lead to better mOS (25 vs. 15 months, p = 0.003; 10 vs. 25 months, p < 0.001). Conclusion: The pts in our investigation had different benefits from the multimodal treatment. We identified the CA 19-9 level at time of diagnosis and after neoadjuvant therapy as well as the preoperative BMI as predictive factors for overall survival. Furthermore, diagnostics of presurgical nodal status should gain more importance as nodal negativity is associated with better outcome.
Collapse
|
106
|
Radiographic Response of Vessel Involvement and Resectability After Neoadjuvant Chemoradiation in Patients With Locally Advanced Pancreatic Cancer. Am J Clin Oncol 2021; 43:776-783. [PMID: 32815856 DOI: 10.1097/coc.0000000000000746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVES Survival of patients with locally advanced pancreatic cancer (LAPC) is improved when neoadjuvant chemoradiation enables subsequent surgical resection. Here, the authors assess changes in vessel involvement as a possible indicator of resectability. METHODS Pancreatic gross tumor and all major abdominal vessels were contoured for 49 patients with unresectable LAPC before and after neoadjuvant chemoradiation. Changes were compared by paired t tests. Tumor-vessel relationships were automatically quantified using Medical Imaging Interaction Toolkit and examined for correlation with resectability and outcome. RESULTS Tumor volumes were significantly reduced by chemoradiation (41 to 33 mL, P<0.0001). Maximum circumferential vessel involvement decreased for most patients and was statistically significant for the superior mesenteric (P<0.003) and splenic veins (P<0.038). Resection was possible in some patients and correlated positively with survival (28 vs. 15 mo, r=0.40), a decrease in CA 19.9 levels (r=0.48), and reduced involvement of most vessels. Nevertheless, surgical resection with a successful detachment of tumor tissue from major vessels was also achieved in some patients who did not show improvement in radiographic vessel involvement, but rather a reduction in tumor volume and CA 19.9 levels. CONCLUSIONS The present analysis demonstrates that neoadjuvant chemoradiation can enable subsequent surgical resection in patients with LAPC. Complete resection substantially prolongs survival. Therefore, surgical exploration should be offered if vessel involvement is improved by chemoradiation and considered in radiographic unchanged vessel involvement if size and CA 19.9 levels decrease, as these factors may indicate resectable disease, too.
Collapse
|
107
|
Zhou IY, Montesi SB, Akam EA, Caravan P. Molecular Imaging of Fibrosis. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00077-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
108
|
Ahmed SA, Mourad AF, Hassan RA, Ibrahim MAE, Soliman A, Aboeleuon E, Elbadee OMA, Hetta HF, Jabir MA. Preoperative CT staging of borderline pancreatic cancer patients after neoadjuvant treatment: accuracy in the prediction of vascular invasion and resectability. Abdom Radiol (NY) 2021; 46:280-289. [PMID: 32488556 DOI: 10.1007/s00261-020-02605-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To assess the utility of MDCT tumor-vascular interface criteria for predicting vascular invasion and resectability in borderline pancreatic cancer (BRPC) patients after neoadjuvant therapy (NAT). METHODS This prospective study included 90 patients with BRPC who finished NAT, showed no progression in preoperative CTs and underwent surgery. Two radiologists independently assessed preoperative vessel-tumor interface criteria. The area under the ROC curve (AUC) was used to evaluate the diagnostic performance for predicting vascular invasions and resectability using surgical and pathological results as the gold standard. Inter-reader agreement was assessed using the κ coefficient. RESULTS Pathologic vascular invasion was confirmed in 47 (54.7%) veins and 14 (16.3%) arteries. R0 resection was achieved in (82.6%71/86) pancreatic resection. Using criteria of circumferential interface ≥ 180 degrees with contour deformity ≥ grade 3 and/or length of tumor contact > 2 cm to predict vascular invasion, the AUCs for the two readers were 0.85-0.88 for arterial invasion and 0.92-0.87 for venous invasion. Using criteria of circumferential interface ≤ 180° with contour deformity ≤ grade 2 and/or length of tumor contact < 2 cm to predict R0 resection, the AUCs was 0.85-0.86 for the two readers. The overall inter-reader agreement was good (κ = 0.75-0.80). The κ values for venous invasion, arterial invasion and R0 resection were 0.76, 0.78, and 0.80. CONCLUSION Tumor-vessel criteria demonstrated good diagnostic performance and reproducibility in the prediction of vascular invasion after NAT in BRPC. These criteria could be helpful in the prediction of R0 resection in cases with only venous involvement.
Collapse
|
109
|
Castillo Tuñón JM, Valle Rodas ME, Botello Martínez F, Rojas Holguín A, López Guerra D, Santos Naharro J, Jaén Torrejímeno I, Blanco Fernández G. Implementation of a regional reference center in pancreatic surgery. Experience after 631 procedures. Cir Esp 2020; 99:S0009-739X(20)30313-4. [PMID: 33342520 DOI: 10.1016/j.ciresp.2020.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/01/2022]
Abstract
INTRODUCTION The main objective of this study is to determine if our unit meets the quality standards required by the scientific community from the reference centers for pancreatic surgery in terms of peri-operative results. The secondary objectives are to compare the different pancreatic surgery techniques performed in terms of early post-operative morbidity and mortality and to analyze the impact of the resections added in these terms. METHOD Descriptive, retrospective and single-center study, corresponding to the period 2006-2019. The results obtained were compared with the proposed quality standards, by Bassi et al. and Sabater et al., required from the reference centers in pancreatic surgery. The sample was divided according to surgical technique and compared in terms of early post-operative morbidity and mortality, studying the impact of extended vascular and visceral resections. All patients undergoing pancreatic surgery in our unit due to pancreatic, malignant and benign pathology were included, since it was implemented as a reference center. Emergency procedures were excluded. RESULTS 631 patients were analyzed. The values ??obtained in the quality standards are in range. The most frequent surgery was cephalic duodenopancreatectomy, which associated higher peri-operative morbidity and mortality rates (p ≤ 0.05). The extended vascular resections impacted the cephalic duodenopancreatectomy group, associating a longer mean stay (p = 0.01) and a higher rate of re-interventions (p = 0.02). CONCLUSIONS The experience accumulated allows to meet the required quality standards, as well as perform extended resections to pancreatectomy with good results in terms of post-operative morbi-mortality.
Collapse
|
110
|
Kunzmann V, Siveke JT, Algül H, Goekkurt E, Siegler G, Martens U, Waldschmidt D, Pelzer U, Fuchs M, Kullmann F, Boeck S, Ettrich TJ, Held S, Keller R, Klein I, Germer CT, Stein H, Friess H, Bahra M, Jakobs R, Hartlapp I, Heinemann V. Nab-paclitaxel plus gemcitabine versus nab-paclitaxel plus gemcitabine followed by FOLFIRINOX induction chemotherapy in locally advanced pancreatic cancer (NEOLAP-AIO-PAK-0113): a multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol 2020; 6:128-138. [PMID: 33338442 DOI: 10.1016/s2468-1253(20)30330-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND The optimal preoperative treatment for locally advanced pancreatic cancer is unknown. We aimed to compare the efficacy and safety of nab-paclitaxel plus gemcitabine with nab-paclitaxel plus gemcitabine followed by fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) as multidrug induction chemotherapy regimens in locally advanced pancreatic cancer. METHODS In this open-label, multicentre, randomised phase 2 study, done at 28 centres in Germany, eligible patients were adults (aged 18-75 years) with an Eastern Cooperative Oncology Group performance status of 0 or 1 and histologically or cytologically confirmed, treatment-naive locally advanced pancreatic adenocarcinoma, as determined by local multidisciplinary team review. After two cycles of nab-paclitaxel 125 mg/m2 plus gemcitabine 1000 mg/m2 (administered intravenously on days 1, 8, and 15 of each 28-day cycle), patients without progressive disease or unacceptable adverse events were randomly assigned (1:1) to receive either two additional cycles of nab-paclitaxel plus gemcitabine (nab-paclitaxel plus gemcitabine group) or four cycles of sequential FOLFIRINOX (oxaliplatin 85 mg/m2, leucovorin 400 mg/m2, irinotecan 180 mg/m2, fluorouracil 400 mg/m2 by intravenous bolus followed by a continuous intravenous infusion of 2400 mg/m2 for 46 h on day 1 of each 14-day cycle; sequential FOLFIRINOX group). Randomisation was done by the clinical research organisation on request of the trial centre using a permuted block design (block size 2 and 4). Patients, investigators, and study team members were not masked to treatment allocation. The primary endpoint was surgical conversion rate (complete macroscopic tumour resection) in the randomised population by intention-to-treat analysis, which was assessed by surgical exploration in all patients with at least stable disease after completion of induction chemotherapy. This trial is registered with ClinicalTrials.gov, NCT02125136. FINDINGS Between Nov 18, 2014, and April 27, 2018, 168 patients were registered and 130 were randomly assigned to either the nab-paclitaxel plus gemcitabine group (64 patients) or the sequential FOLFIRINOX group (66 patients). Surgical exploration after completed induction chemotherapy was done in 40 (63%) of 64 patients in the nab-paclitaxel plus gemcitabine group and 42 (64%) of 66 patients in the sequential FOLFIRINOX group. 23 patients in the nab-paclitaxel plus gemcitabine group and 29 in the sequential FOLFIRINOX group had complete macroscopic tumour resection, yielding a surgical conversion rate of 35·9% (95% CI 24·3-48·9) in the nab-paclitaxel plus gemcitabine group and 43·9% (31·7-56·7) in the sequential FOLFIRINOX group (odds ratio 0·72 [95% CI 0·35-1·45]; p=0·38). At a median follow-up of 24·9 months (95% CI 21·8-27·6), median overall survival was 18·5 months (95% CI 14·4-21·5) in the nab-paclitaxel plus gemcitabine group and 20·7 months (13·9-28·7) in the sequential FOLFIRINOX group (hazard ratio 0·86 [95% CI 0·55-1·36]; p=0·53). All other secondary efficacy endpoints, such as investigator-assessed progression-free survival, radiographic response rate, CA 19-9 response rate, and R0 resection rate, were not significantly different between the two treatment groups except for improved histopathological downstaging in evaluable resection specimens from the sequential FOLFIRINOX group (ypT1/2 stage: 20 [69%] of 29 patients in the sequential FOLFIRINOX group vs four [17%] of 23 patients in the nab-paclitaxel plus gemcitabine group, p=0·0003; ypN0 stage: 15 [52%] of 29 patients in the sequential FOLFIRINOX group vs four [17%] of 23 patients in the nab-paclitaxel plus gemcitabine group, p=0·02). Grade 3 or higher treatment-emergent adverse events during induction chemotherapy occurred in 35 (55%) of 64 patients in nab-paclitaxel plus gemcitabine group and in 35 (53%) of 66 patients in the sequential FOLFIRINOX group. The most common of which were neutropenia (18 [28%] in nab-paclitaxel plus gemcitabine group, 16 [24%] in the sequential FOLFIRINOX group), nausea and vomiting (two [3%] in nab-paclitaxel plus gemcitabine group, eight [12%] in the sequential FOLFIRINOX group), and bile duct obstruction with cholangitis (six [9%] in nab-paclitaxel plus gemcitabine group, seven [11%] in the sequential FOLFIRINOX group). No deaths were caused by treatment-related adverse events during the induction chemotherapy phase. INTERPRETATION Our findings suggest that nab-paclitaxel plus gemcitabine is similarly active and safe as nab-paclitaxel plus gemcitabine followed by FOLFIRINOX as multidrug induction chemotherapy regimens for locally advanced pancreatic cancer. Although conversion to resectability was achieved in about a third of patients, additional evidence is required to determine whether this translates into improved overall survival. FUNDING Celgene.
Collapse
Affiliation(s)
- Volker Kunzmann
- Department of Internal Medicine II, Medical Oncology, Comprehensive Cancer Center Mainfranken Würzburg, University Hospital Würzburg, Würzburg, Germany.
| | - Jens T Siveke
- Department of Medical Oncology and Division of Solid Tumour Translational Oncology, West German Cancer Center, University Medicine Essen, Essen, Germany
| | - Hana Algül
- Comprehensive Cancer Center Munich, Klinikum rechts der Isar, Department of Internal Medicine II, Technical University Munich, Munich, Germany
| | - Eray Goekkurt
- North-German Trial Center for Innovative Oncology, Hematology-Oncology Practice Eppendorf, Hamburg, Germany
| | - Gabriele Siegler
- Department of Internal Medicine 5, Hematology and Medical Oncology, Paracelsus Medical University, Nürnberg, Germany
| | - Uwe Martens
- Department of Internal Medicine III, SLK-Clinics Heilbronn, Heilbronn, Germany
| | - Dirk Waldschmidt
- Department of Gastroenterology and Hepatology, University Hospital Cologne, Cologne, Germany
| | - Uwe Pelzer
- Division of Oncology and Hematology, Charite Campus Mitte and Charite Campus Virchow Klinikum, Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, Berlin Germany
| | - Martin Fuchs
- Clinic for Gastroenterology, Hepatology and Gastrointestinal-Oncology, München Klinik Bogenhausen, Munich, Germany
| | - Frank Kullmann
- Department of Internal Medicine I, Kliniken Nordoberpfalz, Klinikum Weiden, Weiden, Germany
| | - Stefan Boeck
- Department of Medical Oncology and Comprehensive Cancer Center, Ludwig Maximilians University - Grosshadern, Munich, Germany
| | - Thomas J Ettrich
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Swantje Held
- Department of Biometrics, ClinAssess, Leverkusen, Germany
| | - Ralph Keller
- Clinical Research, Arbeitsgemeinschaft für Internistische Onkologie Studien, Berlin, Germany
| | - Ingo Klein
- Department of Surgery I, Comprehensive Cancer Center Mainfranken Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Christoph-Thomas Germer
- Department of Surgery I, Comprehensive Cancer Center Mainfranken Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Hubert Stein
- Department of Surgery, Paracelsus Medical University, Nürnberg, Germany
| | - Helmut Friess
- Department of Surgery, School of Medicine, Technical University Munich, Munich, Germany
| | - Marcus Bahra
- Department of Surgery, Charite Campus Mitte and Charite Campus Virchow Klinikum, Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, Berlin Germany
| | - Ralf Jakobs
- Department of Internal Medicine C, Gastroenterology and Gastrointestinal Oncology, Klinikum Ludwigshafen, Ludwigshafen, Germany
| | - Ingo Hartlapp
- Department of Internal Medicine II, Medical Oncology, Comprehensive Cancer Center Mainfranken Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Volker Heinemann
- Department of Medical Oncology and Comprehensive Cancer Center, Ludwig Maximilians University - Grosshadern, Munich, Germany
| | | |
Collapse
|
111
|
Yasuta S, Kobayashi T, Aizawa H, Takahashi S, Ikeda M, Konishi M, Kojima M, Kuno H, Uesaka K, Morinaga S, Miyamoto A, Toyama H, Takakura N, Sugimachi K, Takayama W. Relationship between surgical R0 resectability and findings of peripancreatic vascular invasion on CT imaging after neoadjuvant S-1 and concurrent radiotherapy in patients with borderline resectable pancreatic cancer. BMC Cancer 2020; 20:1184. [PMID: 33267820 PMCID: PMC7709301 DOI: 10.1186/s12885-020-07698-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/26/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Borderline resectable pancreatic cancer (BRPC) is frequently associated with positive surgical margins and a poor prognosis because the tumor is in contact with major vessels. This study evaluated the relationship between the margin-negative (R0) resection rate and findings indicating peripancreatic vascular invasion on multidetector computed tomography (MDCT) imaging after neoadjuvant chemoradiotherapy (NACRT) in patients with BRPC. METHODS Twenty-nine BRPC patients who underwent laparotomy after neoadjuvant S-1 with concurrent radiotherapy were studied retrospectively. Peripancreatic major vessel invasion was evaluated based on the length of tumor-vessel contact on MDCT. The R0 resection rates were compared between the progression of vascular invasion (PVI) group and the non-progression of vascular invasion (NVI) group. RESULTS There were 3 patients with partial responses (10%), 25 with stable disease (86%), and 1 with progressive disease (3%) according to the RECISTv1.1 criteria. Regarding vascular invasion, 9 patients (31%) were classified as having PVI, and 20 patients (69%) were classified as having NVI. Of the 29 patients, 27 (93%) received an R0 resection, and all the PVI patients received an R0 resection (9/9; R0 resection rate = 100%) while 90% (18/20) of the NVI patients underwent an R0 resection. The exact 95% confidence interval of risk difference between those R0 resection rates was - 10.0% [- 31.7-20.4%]. CONCLUSIONS Patients with BRPC after NACRT achieved high R0 resection rates regardless of the vascular invasion status. BRPC patients can undergo R0 resections unless progressive disease is observed after NACRT. TRIAL REGISTRATION UMIN-CTR, UMIN000009172 . Registered 23 October 2012.
Collapse
Affiliation(s)
- Sho Yasuta
- Department of Hepato-biliary Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tatsushi Kobayashi
- Department of Diagnostic Radiology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| | - Hidetoshi Aizawa
- Department of Surgery, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Shinichiro Takahashi
- Department of Hepato-biliary Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masafumi Ikeda
- Department of Hepato-biliary Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masaru Konishi
- Department of Hepato-biliary Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Motohiro Kojima
- Division of Pathology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hirofumi Kuno
- Department of Diagnostic Radiology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Katsuhiko Uesaka
- Department of Hepato-biliary Pancreatic Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Soichiro Morinaga
- Department of Hepato-Biliary and Pancreatic Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Atsushi Miyamoto
- Department of Hepato-Biliary-Pancreatic Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Hirochika Toyama
- Department of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | - Keishi Sugimachi
- Department of Hepato-Biliary-Pancreatic Surgery, National Hospital Organization Kyusyu Cancer Center, Fukuoka, Japan
| | - Wataru Takayama
- Department of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, Chiba, Japan
| |
Collapse
|
112
|
The Utility of EUS-FNA to Determine Surgical Candidacy in Patients with Pancreatic Cancer after Neoadjuvant Therapy. J Gastrointest Surg 2020; 24:2807-2813. [PMID: 31823316 DOI: 10.1007/s11605-019-04415-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/16/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND In patients with borderline resectable pancreatic cancer (BRPC) or locally advanced pancreatic cancer (LAPC) who undergo neoadjuvant therapy, CT imaging is the standard of care for restaging. However, differentiating residual tumor from post-treatment inflammation with CT is unreliable. The diagnosis of periarterial soft tissue cuffing (PSTC) near major vessels is key to guiding resectability. The goal of this study was to assess the utility of EUS-FNA in determining the etiology of PSTC in BRPC or LAPC after neoadjuvant treatment. METHODS We performed a retrospective analysis of patients referred for EUS-FNA of PSTC following downstaging therapy for LAPC or BRPC at our tertiary medical center. Negative EUS-FNA cytology results were compared with surgical pathology after resection. Patients with positive EUS-FNA cytology results were either followed clinically or results were compared to surgical pathology if surgery was attempted despite the positive cytology. RESULTS Fourteen patients were included in the study of whom four had positive cytology. Two of these patients had progression of disease, and two had attempted resection with positive surgical pathology (100% true positives). All ten patients with negative cytology underwent attempted surgical resection. Nine patients (90%) achieved negative margins, and one patient (10%) had a positive surgical margin. The sensitivity, specificity, and accuracy of EUS-FNA for determining resectability were 80%, 100%, and 92.9%, respectively. CONCLUSIONS In this series of patients with BRPC or LAPC and persistent PSTC after downstaging neoadjuvant treatment, EUS-FNA accurately determined surgical resectability and should be considered as part of the evaluation of such patients.
Collapse
|
113
|
Therapeutic response assessment in pancreatic ductal adenocarcinoma: society of abdominal radiology review paper on the role of morphological and functional imaging techniques. Abdom Radiol (NY) 2020; 45:4273-4289. [PMID: 32936417 DOI: 10.1007/s00261-020-02723-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/17/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is the third leading cause of cancer-related death in the United States and is projected to be the second by 2030. Systemic combination chemotherapy is considered an essential first-line treatment for the majority of patients with PDA, in both the neoadjuvant and palliative settings. In addition, a number of novel therapies are being tested in clinical trials for patients with advanced PDA. In all cases, accurate and timely assessment of treatment response is critical to guide therapy, reduce drug toxicities and cost from a failing therapy, and aid adaptive clinical trials. Conventional morphological imaging has significant limitations, especially in the context of determining primary tumor response and resectability following neoadjuvant therapies. In this article, we provide an overview of current therapy options for PDA, highlight several morphological imaging findings that may be helpful to reduce over-staging following neoadjuvant therapy, and discuss a number of emerging imaging, and non-imaging, tools that have shown promise in providing a more precise quantification of disease burden and treatment response in PDA.
Collapse
|
114
|
Yu Y, Zheng P, Chen Y, Wang B, Paul ME, Tao P, Wang D, Li H, Gu B, Gao L, Wang D, Chen H. Advances and challenges of neoadjuvant therapy in pancreatic cancer. Asia Pac J Clin Oncol 2020; 17:425-434. [PMID: 33164329 DOI: 10.1111/ajco.13504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/10/2020] [Indexed: 01/06/2023]
Abstract
Pancreatic cancer has been becoming the second cause of cancer death in the western world, and its disease burden has increased. Neoadjuvant therapy is one of the current research hotspots in the field of pancreatic cancer, aiming to improve the surgical rate and prognosis of pancreatic cancer. Based on the latest evidence, this review discussed neoadjuvant therapy in pancreatic cancer from the following three aspects: patient selection, protocols selection of neoadjuvant therapy, and treatment response evaluation and resectability prediction. A big controversy existed on the indications of neoadjuvant treatment, but it was agreed that any patient who is likely to achieve R0 resection due to neoadjuvant therapy should be the targeted population. A variety of chemotherapy regimens were tried for neoadjuvant therapy in pancreatic cancer, and FOLFIRINOX and Nab-Paclitaxel plus Gemcitabine are two preferred regimens at present. It was challenging to evaluate treatment response and predict resectability after neoadjuvant therapy, although imaging by CT is widely used. Based on new findings of the remarkable performance of several chemotherapy regimens with or without radiotherapy, the neoadjuvant indications of pancreatic cancer have extended in recent years. However, it is still a challenge to assess the neoadjuvant treatment response and determine the timing of surgery.
Collapse
Affiliation(s)
- Yang Yu
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Peng Zheng
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Yajing Chen
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Bofang Wang
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Maswikiti Ewetse Paul
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Pengxian Tao
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Dengfeng Wang
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Haiyuan Li
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Baohong Gu
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Lei Gao
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Dan Wang
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Second Clinical Medical College of Lanzhou University, Lanzhou, P. R. China
| | - Hao Chen
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, P. R. China.,The Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, P. R. China
| |
Collapse
|
115
|
Rykina-Tameeva N, Nahm CB, Mehta S, Gill AJ, Samra JS, Mittal A. Neoadjuvant therapy for pancreatic cancer changes the composition of the pancreatic parenchyma. HPB (Oxford) 2020; 22:1631-1636. [PMID: 32247587 DOI: 10.1016/j.hpb.2020.03.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 01/16/2020] [Accepted: 03/08/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Postoperative pancreatic fistula (POPF) remains a prominent complication following pancreatic cancer resections. The primary aim of this study was to evaluate the histological changes that occur in the pancreas due to neoadjuvant therapy (NAT) by comparing the acinar, collagen and fat scores in resected PDAC specimens of patients who did and did not receive NAT. Secondary aims included (1) the difference in rates of POPF in PDAC patients who received NAT versus upfront resection; and (2) the association between acinar/collagen/fat scores and the development of POPF. METHODS Consecutive patients who underwent pancreaticoduodenectomy for PDAC, with and without NAT were included for analysis. Acinar, collagen and fat scores were determined from histology slides of the pancreatic resection margin. RESULTS One hundred and thirty-four patients were included. There was a significant decrease in the median acinar score (48 vs 23, p = 0.003) and increase in the collagen score (28 vs 50, p = 0.011) for patients who received NAT and a significant correlation with the number of cycles of NAT. This study found no statistical difference between NAT and the development of POPF. CONCLUSION The use of NAT in the treatment of PDAC changes the composition of the pancreas.
Collapse
Affiliation(s)
- Nadya Rykina-Tameeva
- Upper Gastrointestinal Surgical Unit, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Christopher B Nahm
- Upper Gastrointestinal Surgical Unit, Royal North Shore Hospital, St. Leonards, NSW, Australia; Northern Clinical School, Faculty of Medical and Health Sciences, The University of Sydney, NSW, Australia
| | - Shreya Mehta
- Upper Gastrointestinal Surgical Unit, Royal North Shore Hospital, St. Leonards, NSW, Australia; Northern Clinical School, Faculty of Medical and Health Sciences, The University of Sydney, NSW, Australia.
| | - Anthony J Gill
- Northern Clinical School, Faculty of Medical and Health Sciences, The University of Sydney, NSW, Australia; Cancer Diagnosis and Pathology Group, Kolling Institute, The University of Sydney, NSW, Australia; Australian Pancreatic Centre, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Jaswinder S Samra
- Upper Gastrointestinal Surgical Unit, Royal North Shore Hospital, St. Leonards, NSW, Australia; Northern Clinical School, Faculty of Medical and Health Sciences, The University of Sydney, NSW, Australia; Australian Pancreatic Centre, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Anubhav Mittal
- Upper Gastrointestinal Surgical Unit, Royal North Shore Hospital, St. Leonards, NSW, Australia; Northern Clinical School, Faculty of Medical and Health Sciences, The University of Sydney, NSW, Australia; Australian Pancreatic Centre, Royal North Shore Hospital, St. Leonards, NSW, Australia
| |
Collapse
|
116
|
Ye C, Sadula A, Ren S, Guo X, Yuan M, Yuan C, Xiu D. The prognostic value of CA19-9 response after neoadjuvant therapy in patients with pancreatic cancer: a systematic review and pooled analysis. Cancer Chemother Pharmacol 2020; 86:731-740. [PMID: 33047181 DOI: 10.1007/s00280-020-04165-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 10/03/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly aggressive and refractory disease, with disappointing 5-year survival rates. Regarding the wide application of neoadjuvant treatment in patients with PC, how the post-neoadjuvant Carbohydrate antigen 19-9 (CA19-9) response could translate into a survival benefit is not clearly understood. We aimed to evaluate the correlation of the CA19-9 response with overall survival (OS) in patients with PC receiving neoadjuvant therapy. METHODS An extensive electronic search in PubMed, Embase, and the Cochrane Library was performed to identify relevant articles, from which data relevant to independent correlations of the CA19-9 response with overall survival (OS) were extracted for analysis. A random-effects model was used to calculate the pooled hazard ratios (HRs) with their corresponding 95% confidence intervals (CIs). RESULTS Altogether, 17 eligible studies were identified in the systematic review. Pooled analysis showed that CA19-9 response > 50% (HR, 0.43; 95% CI 0.29-0.56; P < 0.001) and normalization of CA19-9 (HR, 0.52; 95% CI 0.42-0.63; P < 0.001) after neoadjuvant treatment are significantly associated with promising overall survival. The results also showed that optimal CA19-9 response after neoadjuvant treatment was significantly related to a favorable prognosis (HR = 0.49, 95% CI 0.42-0.55, P < 0.001; I2 = 45.1%, P = 0.04). Subgroup analysis revealed there were no prognostic difference between CA19-9 > 50% and normalization of CA19-9 after neoadjuvant treatment (P = 0.338), but the duration of neoadjuvant chemotherapy over 4 months was significantly associated with expanded postoperative survival (P = 0.013). CONCLUSIONS Serum CA19‑9 is valuable in determining the effect of neoadjuvant treatment in patients with PC. Post-neoadjuvant CA19-9 response > 50% or CA19-9 normalization was related to a more promising overall survival, suggesting that optimal CA19-9 response may be a suitable prognostic index to guide treatment decisions.
Collapse
Affiliation(s)
- Chen Ye
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan Bei Lu, Haidian District, Beijing, People's Republic of China
| | - Abuduhaibaier Sadula
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan Bei Lu, Haidian District, Beijing, People's Republic of China
| | - Siqian Ren
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan Bei Lu, Haidian District, Beijing, People's Republic of China
| | - Xin Guo
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan Bei Lu, Haidian District, Beijing, People's Republic of China
| | - Meng Yuan
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan Bei Lu, Haidian District, Beijing, People's Republic of China
| | - Chunhui Yuan
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan Bei Lu, Haidian District, Beijing, People's Republic of China.
| | - Dianrong Xiu
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan Bei Lu, Haidian District, Beijing, People's Republic of China.
| |
Collapse
|
117
|
Vega EA, Kutlu OC, Salehi O, James D, Alarcon SV, Herrick B, Krishnan S, Kozyreva O, Conrad C. Preoperative Chemotherapy for Pancreatic Cancer Improves Survival and R0 Rate Even in Early Stage I. J Gastrointest Surg 2020; 24:2409-2415. [PMID: 32394126 DOI: 10.1007/s11605-020-04601-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 04/06/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND While preoperative chemotherapy for patients with stage II-III pancreatic adenocarcinoma (PDAC) is frequently practiced, its impact on very early PDAC (stage I) remains unclear today. MATERIAL AND METHODS Patients undergoing pancreatectomy for PDAC between 2010 and 2016 were identified in the National Cancer Database. Early-stage patients (IA-IB) with complete oncologic and clinical information and more than 30-day survival were included. The effect of preoperative chemotherapy on margin status was assessed with binary logistic regression. Following correction for confounders, the effect of therapy sequencing was assessed via comparison of preoperative, postoperative, perioperative (pre- and post-operative) chemotherapy, and surgery only using Cox regression. RESULTS Of 4785 patients, 688 (14.4%) were stage IA, and 4197 (87.7%) IB. The rate of preoperative chemotherapy was only 8.8%. Rate of margin positivity was lower for preoperative chemotherapy (12.3% vs 19.7%). After correcting for confounders, the risk of a positive margin was lower in preoperative chemotherapy (odd ratio [OR] 0.703, p = 0.042). Cox regression showed a significant overall survival advantage for preoperative (hazard ratio [HR] 0.784, p = 0.002), postoperative (HR 0.618, p < 0.001), and perioperative (HR 0.601, p < 0.001) chemotherapy compared with surgery alone. There was no significant difference in survival between chemotherapy groups but a trend towards optimal survival for preoperative chemotherapy. CONCLUSION Despite preoperative chemotherapy vs surgery alone resulting in improved R0 rates and overall survival even in stage I PDAC, it is rarely practiced. The results presented here suggest that preoperative chemotherapy should be strongly considered in all patients with resectable PDAC, including very early PDAC.
Collapse
Affiliation(s)
- Eduardo A Vega
- Department of Surgery, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Onur C Kutlu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Omid Salehi
- Department of Surgery, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Daria James
- Department of Surgery, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Sylvia V Alarcon
- Department of Medical Oncology & Hematology, St. Elizabeth's Medical Center and Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard School of Medicine, Boston, MA, USA
| | - Beth Herrick
- Department of Radiation Oncology, St. Elizabeth's Medical Center, University of Massachusetts Medical School, Boston, MA, USA
| | - Sandeep Krishnan
- Department of Gastroenterology, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Olga Kozyreva
- Department of Medical Oncology & Hematology, St. Elizabeth's Medical Center and Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard School of Medicine, Boston, MA, USA
| | - Claudius Conrad
- Department of Surgery, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA. .,General Surgery and Surgical Oncology, Hepato-Pancreato-Biliary Surgery, St. Elizabeth's Medical Center, Tufts University School of Medicine, 11 Nevins St., Suite 201, Brighton, MA, 02135, USA.
| |
Collapse
|
118
|
Abstract
Importance In the past few decades, there has been rapid advancements in imaging technologies that have become irreplaceable in the pre-operative assessment of patients with pancreatic tumors. Modern imaging modalities, including computed tomography (CT) and endoscopic ultrasound (EUS), can provide critical information of the absence or presence of metastatic disease in pancreatic cancer, as well as details on the local extent and resectability, allowing for the selection of stage appropriate treatments and pre-operatively determined surgical approach. Objective The aim of this review is to discuss staging, resectability, and imaging for patients with pancreatic tumors. Evidence Review A literature review was performed of articles relevant to the topics of staging, resectability, and imaging of pancreatic tumors. Imaging modalities included CT, EUS, magnetic resonance imaging (MRI), positron emission tomography (PET), antibody-based and narrow band imaging. Findings CT pancreas protocol combined with EUS serve as the primary modalities in diagnosis, staging, and surgical planning in patients with pancreatic tumors. MRI is an alternative to CT with near equivalent utility in the pre-operative setting. In some circumstances, PET-CT may be a cost-effective initial study to detect distant disease. Conclusions and Relevance Current imaging technologies play a critical role in the evaluation of patients with pancreatic tumors. Advances in the past 3 decades in imaging technologies have revolutionized the process of assessment of stage and resectability in patients with pancreatic tumors. Future imaging technologies will address current limitation in the evaluation of occult metastatic disease.
Collapse
|
119
|
First report on the feasibility of a permanently implantable uni-directional planar low dose rate brachytherapy sheet for patients with resectable or borderline resectable pancreatic cancer. Brachytherapy 2020; 20:207-217. [PMID: 32978081 DOI: 10.1016/j.brachy.2020.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Margin negative resection in pancreatic cancer remains the only curative option but is challenging, especially with the retroperitoneal margin. Intraoperative radiation therapy (IORT) can improve rates of local control but requires specially designed facilities and equipment. This retrospective review describes initial results of a novel implantable mesh of uni-directional low dose rate (LDR) Pd-103 sources (sheet) used to deliver a focal margin-directed high-dose boost in patients with concern for close or positive margins. METHODS Eleven consecutive patients from a single institution with resectable or borderline resectable pancreatic cancer with concern for positive margins were selected for sheet placement and retrospectively reviewed. Procedural outcomes, including the time to implant the device and complications, and clinical outcomes, including survival and patterns of failure, are reported. A dosimetric comparison of the LDR sheet with hypothetical stereotactic body radiotherapy (SBRT) boost is reported. RESULTS One patient had a resectable disease, and 10 patients had a borderline resectable disease and underwent neoadjuvant treatment. Sheet placement added 15 min to procedural time with no procedural or sheet-related complications. At a median follow up of 13 months, 64% (n = 7) of patients are alive and 55% (n = 6) are disease-free. Compared to a hypothetical SBRT boost, the LDR sheet delivered a negligible dose to kidneys, liver, and spinal cord with a 50% reduction in max dose to the small bowel. CONCLUSION This is the first report of the use of an implantable uni-directional LDR brachytherapy sheet in patients with resected pancreatic cancer with concern for margin clearance, with no associated toxicity and favorable clinical outcomes.
Collapse
|
120
|
Implementation of a standardized approach to borderline resectable pancreatic cancer in a multisite community oncology program. Surg Open Sci 2020; 2:25-31. [PMID: 32954245 PMCID: PMC7482012 DOI: 10.1016/j.sopen.2020.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 06/21/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Background Treatment paradigms for borderline resectable pancreatic cancer are evolving with increasing use of neoadjuvant chemotherapy and neoadjuvant chemoradiation. Variations in the definition of borderline resectable pancreatic cancer and neoadjuvant approaches have made standardizing care for borderline resectable pancreatic cancer difficult. We report an effort to standardize management of borderline resectable pancreatic cancer throughout Sanford Health, a large community oncology network. Methods Starting in October 2013, cases of pancreatic adenocarcinoma without known metastatic disease were categorized as borderline resectable pancreatic cancer if they met ≥ 1 of the following criteria: (1) abutment of superior mesenteric, common hepatic, or celiac arteries with < 180° involvement, (2) venous involvement deemed potentially suitable for reconstruction, and/or (3) biopsy-proven lymph node involvement. Patients with borderline resectable pancreatic cancer were treated with neoadjuvant chemotherapy followed by reimaging and surgery if venous involvement had improved; if disease remained borderline resectable, patients underwent neoadjuvant chemoradiation and surgical exploration as long as reimaging did not reveal evidence of progressive disease. Results Forty-three patients from October 2013 to April 2017 were diagnosed with borderline resectable pancreatic cancer. Twelve of 42 (29%) patients proceeded to surgical exploration directly after neoadjuvant chemotherapy; 23 (55%) received neoadjuvant chemoradiation. Overall, 28/43 (65%) underwent exploration with 19 (44%) able to undergo resection. Of those, 14/19 (74%) attained R0 resection and 11/19 (58%) were pathologic N0. No pretreatment or treatment variables were associated with resection rates; resection was the only variable associated with survival. Conclusion This report demonstrates the feasibility of implementing a standardized approach to borderline resectable pancreatic cancer across multiple sites over a wide geographic area. Adherence to protocol therapies was good and surgical outcomes are similar to many reported series.
Collapse
|
121
|
Colloca GA, Venturino A, Guarneri D. Tumor growth kinetics by CA 19-9 in patients with unresectable pancreatic cancer receiving chemotherapy: A retrospective analysis. Pancreatology 2020; 20:1189-1194. [PMID: 32747196 DOI: 10.1016/j.pan.2020.07.397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Recently, measures of tumor growth kinetics calculated by carbohydrate antigen 19-9 (CA 19-9) determinations after cytotoxic chemotherapy (CHT) have been reported as effective prognostic indicators in locally-advanced unresectable and metastatic pancreatic adenocarcinoma (mPDAC). The study aims to evaluate the prognostic role of tumor kinetics measured by CA 19-9 in patients with mPDAC, measuring it by three different ways. METHODS Patients with mPDAC receiving a first-line CHT between 2009 and 2017 were identified, and those for whom CA 19-9 data were available were enrolled. Three CA 19-9-related variables were calculated: CA 19-9 related reduction rate (RR) and tumor growth rate (G), after 8 weeks of CHT, tumor growth and inflammation index (TGII), after 90 days of CHT. The relationships with the outcome were analysed, and a Cox model has been build with each of the three variables. RESULTS Of 118 patients only 48 were eligible for the analysis. RR, G, or TGII appear as significant prognostic factors, and, after multivariate analysis, a reduction rate of 20% the baseline or more was associated with good survival (HR 0.321; CIs 0.156-0.661) as well as a G > -0.4%/day (HR 2.114; CIs 1.034-4.321), whereas TGII >190 was not correlated with the outcome (HR 1.788; CIs 0.789-4.055). CONCLUSIONS In patients with mPDAC, after 8 weeks of first-line CHT, CA 19-9-related tumor reduction or growth rate appear as valuable prognostic factors.
Collapse
|
122
|
Rhee H, Park MS. The Role of Imaging in Current Treatment Strategies for Pancreatic Adenocarcinoma. Korean J Radiol 2020; 22:23-40. [PMID: 32901458 PMCID: PMC7772381 DOI: 10.3348/kjr.2019.0862] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/30/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
In pancreatic cancer, imaging plays an essential role in surveillance, diagnosis, resectability evaluation, and treatment response evaluation. Pancreatic cancer surveillance in high-risk individuals has been attempted using endoscopic ultrasound (EUS) or magnetic resonance imaging (MRI). Imaging diagnosis and resectability evaluation are the most important factors influencing treatment decisions, where computed tomography (CT) is the preferred modality. EUS, MRI, and positron emission tomography play a complementary role to CT. Treatment response evaluation is of increasing clinical importance, especially in patients undergoing neoadjuvant therapy. This review aimed to comprehensively review the role of imaging in relation to the current treatment strategy for pancreatic cancer, including surveillance, diagnosis, evaluation of resectability and treatment response, and prediction of prognosis.
Collapse
Affiliation(s)
- Hyungjin Rhee
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Suk Park
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
123
|
Multiparametric MRI for prediction of treatment response to neoadjuvant FOLFIRINOX therapy in borderline resectable or locally advanced pancreatic cancer. Eur Radiol 2020; 31:864-874. [PMID: 32813104 DOI: 10.1007/s00330-020-07134-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/10/2020] [Accepted: 07/31/2020] [Indexed: 01/02/2023]
Abstract
OBJECTIVES To identify multiparametric MRI biomarkers to predict the tumor response to neoadjuvant FOLFIRINOX therapy in patients with borderline resectable (BR) or locally advanced (LA) pancreatic ductal adenocarcinoma (PDAC). METHODS From May 2016 to March 2018, adult patients with BR or LA PDAC were prospectively enrolled in this study. They received eight cycles of FOLFIRINOX therapy and underwent multiparametric MRI twice (at baseline and after the second cycle). MRI evaluations included dynamic contrast-enhanced MRI, intravoxel incoherent motion diffusion-weighted imaging, and assessment of T2* relaxivity (R2*) and the change in T1 relaxivity (ΔR1, equilibrium phase R1 minus non-enhanced R1) of the tumors. Factors to predict the responders determined by the best overall response during FOLFIRINOX therapy and those to predict progression-free survival (PFS) and overall survival (OS) were evaluated using multivariable logistic regression and the Cox proportional hazard model. RESULTS Forty-one patients (mean age, 60.3 years ± 9.3; 24 men) were included. Among the clinical and MRI factors, the baseline ΔR1 (adjusted odds ratio, 31.07; p = 0.008) was the only independent predictor for tumor response. The baseline ΔR1 was also an independent predictor for PFS (adjusted hazard ratio, 0.40; p = 0.033) along with R0 resection. The use of a cutoff ΔR1 value of ≥ 1.31 s-1 enabled prognostic stratification (median PFS, 16.0 months vs.10.0 months; p = 0.029; median OS, 34.9 months vs. 16.6 months; p = 0 .023, respectively). CONCLUSIONS The baseline tumor ΔR1 value may be useful to predict tumor response and survival in patients with BR or LA PDAC receiving FOLFIRINOX neoadjuvant therapy. KEY POINTS • Baseline ΔR1 was an independent predictor for tumor response (adjusted odds ratio, 31.07; p = 0.008) and progression-free survival (adjusted hazard ratio, 0.40; p = 0.033) in patients with borderline resectable or locally advanced pancreatic ductal adenocarcinoma receiving neoadjuvant FOLFIRINOX therapy. • The criterion of baseline ΔR1 value ≥ 1.31 s-1 allowed for the prediction of favorable tumor response and survival outcome after neoadjuvant FOLFIRINOX therapy.
Collapse
|
124
|
A Single-Institution Experience of Induction 5-Fluorouracil, Leucovorin, Irinotecan, and Oxaliplatin Followed by Surgery Versus Consolidative Radiation for Borderline and Locally Advanced Unresectable Pancreatic Cancer. Pancreas 2020; 49:904-911. [PMID: 32658074 DOI: 10.1097/mpa.0000000000001592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES In the 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) era, the benefit of surgery versus definitive radiation for borderline resectable (BR) and locally advanced (LA) unresectable pancreatic ductal adenocarcinoma (PDAC) is not well defined. Our primary objective was to identify the survival impact of surgery for BR and LA unresectable PDAC treated with induction FOLFIRINOX. METHODS We performed a single-center retrospective review of BR and LA PDAC treated with FOLFIRINOX from 2010 to 2018. The overall survival of surgery and consolidative radiotherapy was estimated in the Kaplan-Meier method and compared via the log-rank test. Subgroup analyses were conducted for BR and LA patients. RESULTS We identified 101 BR and LA PDAC patients treated with induction FOLFIRINOX (41 surgeries and 60 consolidative radiotherapies). Surgery patients were 68.3% (28/41) BR and 31.7% (13/41) LA, whereas consolidative radiotherapy patients were 30% (18/60) BR and 70% (42/60) LA. The R0 resection rate was 100%, and 46.3% (19/41) received preoperative radiation. Median overall survival of surgery versus consolidative radiotherapy was 42.3 versus 19.6 months, respectively (P < 0.001). On multivariate analysis, surgery associated with improved survival. CONCLUSIONS Surgery after induction FOLFIRINOX is feasible and has a clinically meaningful survival benefit in BR and LA PDAC.
Collapse
|
125
|
Abstract
OPINION STATEMENT Patients with borderline resectable pancreatic ductal adenocarcinoma (BR PDAC) should receive preoperative chemotherapy with or without radiation therapy, with the intent to eradicate occult metastatic cancer cells, to select patients with a "locally dominant cancer phenotype" for whom local therapies might be most effective, and to reduce the anatomic extent of tumors to facilitate surgical resection. The administration of preoperative therapy may also be a useful strategy to deliver the maximum load of chemotherapy to patients with BR PDAC, since as many as half of patients will never qualify for adjuvant treatments following pancreatectomy due to postoperative morbidity or disease progression. Patients with BR PDAC should be categorized at diagnosis on the basis of anatomical, biological, and conditional criteria and should be offered induction systemic chemotherapy with close monitoring for toxicity, followed by administration of (chemo)radiation in selected cases. Patients should be restaged after systemic therapy and, if used, (chemo)radiation. Patients who continue to show disease response or disease stability without signs of progression should be considered for pancreatectomy; better measures of response to therapy are needed.
Collapse
|
126
|
Distal pancreas-coeliac axis resection with preoperative selective embolization of the coeliac axis: a single high-volume centre experience. Langenbecks Arch Surg 2020; 405:635-645. [PMID: 32683485 DOI: 10.1007/s00423-020-01919-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/24/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Patients with ductal adenocarcinoma in the body and/or tail of the pancreas with involvement of the common hepatic artery and/or celiac axis have until recently been considered unresectable. In selected cases, distal pancreatectomy (DP) with en bloc celiac axis resection (DP-CAR) may be an option to achieve R0 resection. METHODS Patients with tumours in the body and/or tail of the pancreas locally advanced with involvement of the common hepatic artery and/or celiac axis, with no distant metastases, were evaluated for DP-CAR procedures. Preoperative embolization was performed 10-14 days prior to surgery to enhance collateral arterial supply for the liver and stomach. RESULTS A total of 21 patients went through DP-CAR of whom 15 were preoperatively embolized. R0 resection vas achieved in 76% of the patients comparable to our standard distal pancreatectomies. The DP-CAR patients had a significant longer postoperative hospital stay, but no difference in major complications, including pancreatic fistulas compared with our standard distal pancreatectomies. No 30 nor 90 days postoperative mortality were recorded. Median survival in patients who underwent DP and DP-CAR procedures was 24.0 and 23.5 months, respectively (P > 0.5). CONCLUSION Outcomes after DP-CAR are comparable to standard distal pancreatectomies. DP-CAR after preoperative embolization is feasible and may in selected patients be a good option for treating patients with tumours in the body and/or tail of the pancreas with central arterial involvement.
Collapse
|
127
|
Azab B, Macedo FI, Chang D, Ripat C, Franceschi D, Livingstone AS, Yakoub D. The Impact of Prolonged Chemotherapy to Surgery Interval and Neoadjuvant Radiotherapy on Pathological Complete Response and Overall Survival in Pancreatic Cancer Patients. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2020; 14:1179554920919402. [PMID: 32669884 PMCID: PMC7336830 DOI: 10.1177/1179554920919402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 03/04/2020] [Indexed: 01/15/2023]
Abstract
Background: We aimed to study the impact of neoadjuvant chemotherapy to surgery (NCT-S)
interval and neoadjuvant radiotherapy (NRT) on pathological complete
response (pCR) and overall survival (OS) in pancreatic cancer (pancreatic
ductal adenocarcinoma [PDAC]). Methods: National Cancer Data Base (NCDB)–pancreatectomy patients who underwent
NCT/NRT were included. The NCT-S interval was divided into time quintiles in
weeks: 8 to 11, 12 to 14, 15 to 19, 20 to 29, and >29 weeks. Results: A total of 2093 patients with NCT were included with median follow-up of
74 months and 71% NRT. The pCR rate was 2.1% with higher median OS compared
with non-pCR (41 vs 19 months, P = .03). The pCR rate
increased with longer NCT-S interval (quintiles: 1%, 1.6%, 1.7%, 3%, and 6%,
P < .001, respectively). In logistic regression, NRT
(odds ratio [OR] = 2.5, 95% confidence interval [CI]: 1.1-6.1,
P = .03) and NCT-S >29 weeks (OR = 6.1, 95%
CI = 2.02-18.50, P < .001) were predictive of increased
pCR. The prolonged NCT-S interval and pCR were independent predictors of OS,
whereas NRT was not. Conclusions: Longer NCT-S interval and pCR were independent predictors of improved OS in
patients with PDAC. The NRT predicted increased pCR but not OS.
Collapse
Affiliation(s)
- Basem Azab
- Surgical Oncology, Sentara Healthcare, Sentara CarePlex Hospital, Hampton, VA, USA
| | - Francisco Igor Macedo
- Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David Chang
- Virginia Oncology Associate, Hampton, VA, USA
| | - Caroline Ripat
- Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dido Franceschi
- Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alan S Livingstone
- Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Danny Yakoub
- Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
128
|
Schawkat K, Manning MA, Glickman JN, Mortele KJ. Pancreatic Ductal Adenocarcinoma and Its Variants: Pearls and Perils. Radiographics 2020; 40:1219-1239. [PMID: 32678699 DOI: 10.1148/rg.2020190184] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), an epithelial neoplasm derived from the pancreatic ductal tree, is the most common histologic type of pancreatic cancer and accounts for 85%-95% of all solid pancreatic tumors. As a highly lethal malignancy, it is the seventh leading cause of cancer death worldwide and is responsible for more than 300 000 deaths per year. PDAC is highly resistant to current therapies, affording patients a 5-year overall survival rate of only 7.2%. It is characterized histologically by its highly desmoplastic stroma embedding tubular and ductlike structures. On images, it typically manifests as a poorly defined hypoenhancing mass, causing ductal obstruction and vascular involvement. Little is known about the other histologic subtypes of PDAC, mainly because of their rarity and lack of specific patterns of disease manifestation. According to the World Health Organization, these variants include adenosquamous carcinoma, colloid carcinoma, hepatoid carcinoma, medullary carcinoma, signet ring cell carcinoma, undifferentiated carcinoma with osteoclast-like giant cells, and undifferentiated carcinoma. Depending on the subtype, they can confer a better or even worse prognosis than that of conventional PDAC. Thus, awareness of the existence and differentiation of these variants on the basis of imaging and histopathologic characteristics is crucial to guide clinical decision making for optimal treatment and patient management.
Collapse
Affiliation(s)
- Khoschy Schawkat
- From the Division of Abdominal Imaging, Department of Radiology (K.S., K.J.M.), and Department of Pathology (J.N.G.), Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115; Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (K.S.); and American Institute for Radiologic Pathology, Silver Spring, Md, and MedStar Georgetown University Hospital, Washington, DC (M.A.M.)
| | - Maria A Manning
- From the Division of Abdominal Imaging, Department of Radiology (K.S., K.J.M.), and Department of Pathology (J.N.G.), Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115; Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (K.S.); and American Institute for Radiologic Pathology, Silver Spring, Md, and MedStar Georgetown University Hospital, Washington, DC (M.A.M.)
| | - Jonathan N Glickman
- From the Division of Abdominal Imaging, Department of Radiology (K.S., K.J.M.), and Department of Pathology (J.N.G.), Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115; Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (K.S.); and American Institute for Radiologic Pathology, Silver Spring, Md, and MedStar Georgetown University Hospital, Washington, DC (M.A.M.)
| | - Koenraad J Mortele
- From the Division of Abdominal Imaging, Department of Radiology (K.S., K.J.M.), and Department of Pathology (J.N.G.), Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115; Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland (K.S.); and American Institute for Radiologic Pathology, Silver Spring, Md, and MedStar Georgetown University Hospital, Washington, DC (M.A.M.)
| |
Collapse
|
129
|
Prognostic Impact of the Neutrophil-to-Lymphocyte Ratio in Borderline Resectable Pancreatic Ductal Adenocarcinoma Treated with Neoadjuvant Chemoradiotherapy Followed by Surgical Resection. World J Surg 2020; 43:3153-3160. [PMID: 31549202 DOI: 10.1007/s00268-019-05159-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Increasing evidence suggests that cancer-associated inflammation, as indicated by markers such as the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and modified Glasgow Prognostic Score (mGPS), predicts poor outcomes in pancreatic cancer. In this study, the associations between systemic inflammation markers and survival were examined in borderline resectable pancreatic ductal adenocarcinoma (BR-PDAC) patients who underwent neoadjuvant chemoradiotherapy (NACRT) followed by surgical resection. METHODS From April 2009 to December 2017, 119 patients diagnosed with BR-PDAC and receiving NACRT followed by radical surgery were included in this retrospective study. The associations between the pre- and post-NACRT NLR, PLR, mGPS, and clinicopathological characteristics, as well as their predictive values for survival outcomes, were analyzed. This study was approved by an institutional review board at Yokohama City University (B180600049). RESULTS On multivariate analysis with a Cox's proportional hazards regression model, post-NACRT NLR ≥3 (p = 0.040; hazard ratio, 2.24; 95% CI 1.28-3.91) and lymph node metastasis (p = 0.002; hazard ratio, 2.33; 95% CI 1.36-3.99) were significantly associated with shorter overall survival. The median survival time was 22.0 months for patients with post-NACRT NLR ≥3 and 45.0 months for patients with post-NACRT NLR <3 (p = 0.028). CONCLUSIONS The NLR following NACRT might predict survival in BR-PDAC patients. Patients with an elevated post-NACRT NLR or positive lymph node metastasis may be candidates for stronger adjuvant therapies.
Collapse
|
130
|
Molina G, Clancy TE, Tsai TC, Lam M, Wang J. Racial Disparity in Pancreatoduodenectomy for Borderline Resectable Pancreatic Adenocarcinoma. Ann Surg Oncol 2020; 28:1088-1096. [PMID: 32651695 DOI: 10.1245/s10434-020-08717-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previous studies have found racial disparity in pancreatectomies for resectable pancreatic adenocarcinoma. The aim of this study was to investigate if racial disparities were worse in the performance of pancreaticoduodenectomy for borderline resectable pancreatic adenocarcinoma. METHODS This study used the National Cancer Database (2004-2016) and included patients with non-metastatic and head of the pancreas borderline resectable pancreatic adenocarcinoma. Multivariable, Poisson regression models with robust standard errors evaluated the relative risk (RR) of undergoing a pancreaticoduodenectomy among non-White patients (Black, Asian, and non-White Hispanic) compared with White patients. A Poisson regression model with hospital fixed effects was performed to evaluate if findings were due to within-hospital or between-hospital variation. Interaction between race and neoadjuvant therapy was also evaluated. RESULTS There were 15,482 patients (median age 68 years, interquartile range 60-76 years; 48.6% male) with borderline resectable pancreatic adenocarcinoma who were predominantly White (84.3%, n = 13,058; non-White, 15.7%, n = 2424). Overall, 18.4% (n = 2853) had a pancreatic resection. Non-White patients had a significantly lower likelihood of undergoing a pancreatic resection for borderline resectable pancreatic adenocarcinoma when compared with White patients (RR 0.75, 95% confidence interval 0.68-0.83; p < 0.001). These findings persisted in the hospital fixed-effects model. In the interaction analysis, there were no significant differences in the likelihood of pancreatic resection if patients received neoadjuvant therapy. CONCLUSIONS Non-White patients were 25% less likely to undergo a pancreatic resection for borderline resectable pancreatic adenocarcinoma compared with White patients. This racial disparity was due to variation in care within-hospitals and disappeared if non-White patients were treated with neoadjuvant therapy.
Collapse
Affiliation(s)
- George Molina
- Division of Surgical Oncology, Department of Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.,Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
| | - Thomas E Clancy
- Division of Surgical Oncology, Department of Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.,Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
| | - Thomas C Tsai
- Division of General and Gastrointestinal Surgery, Department of Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Miranda Lam
- Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA.,Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jiping Wang
- Division of Surgical Oncology, Department of Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA. .,Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA.
| |
Collapse
|
131
|
Yamada S, Fujii T, Yamamoto T, Takami H, Yoshioka I, Yamaki S, Sonohara F, Shibuya K, Motoi F, Hirano S, Murakami Y, Inoue H, Hayashi M, Murotani K, Kitayama J, Ishikawa H, Kodera Y, Sekimoto M, Satoi S. Phase I/II study of adding intraperitoneal paclitaxel in patients with pancreatic cancer and peritoneal metastasis. Br J Surg 2020; 107:1811-1817. [PMID: 32638367 PMCID: PMC7689756 DOI: 10.1002/bjs.11792] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/27/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Intraperitoneal chemotherapy using paclitaxel is considered an experimental approach for treating peritoneal carcinomatosis. This study aimed to determine the recommended dose, and to evaluate the clinical efficacy and safety, of the combination of intravenous gemcitabine, intravenous nab-paclitaxel and intraperitoneal paclitaxel in patients with pancreatic cancer and peritoneal metastasis. METHODS The frequencies of dose-limiting toxicities were evaluated, and the recommended dose was determined in phase I. The primary endpoint of the phase II analysis was overall survival rate at 1 year. Secondary endpoints were antitumour effects, symptom-relieving effects, safety and overall survival. RESULTS The recommended doses of intravenous gemcitabine, intravenous nab-paclitaxel and intraperitoneal paclitaxel were 800, 75 and 20 mg/m2 respectively. Among 46 patients enrolled in phase II, the median time to treatment failure was 6·0 (range 0-22·6) months. The response and disease control rates were 21 of 43 and 41 of 43 respectively. Ascites disappeared in 12 of 30 patients, and cytology became negative in 18 of 46. The median survival time was 14·5 months, and the 1-year overall survival rate was 61 per cent. Conversion surgery was performed in eight of 46 patients, and those who underwent resection survived significantly longer than those who were not treated surgically (median survival not reached versus 12·4 months). Grade 3-4 haematological toxicities developed in 35 of 46 patients, whereas non-haematological adverse events occurred in seven patients. CONCLUSION Adding intraperitoneal paclitaxel had clinical efficacy with acceptable tolerability.
Collapse
Affiliation(s)
- S Yamada
- Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - T Fujii
- Department of Surgery and Science Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - T Yamamoto
- Department of Surgery, Kansai Medical University, Hirakata, Japan
| | - H Takami
- Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - I Yoshioka
- Department of Surgery and Science Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - S Yamaki
- Department of Surgery, Kansai Medical University, Hirakata, Japan
| | - F Sonohara
- Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - K Shibuya
- Department of Surgery and Science Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - F Motoi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - S Hirano
- Department of Surgery, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Y Murakami
- Department of Gastroenterological Surgery II, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - H Inoue
- Department of Hepatobiliary-pancreatic and Breast Surgery, Ehime University Graduate School of Medicine, Ehime, Fukuoka, Japan
| | - M Hayashi
- Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - K Murotani
- Biostatistics Centre, Graduate School of Medicine, Kurume University, Fukuoka, Japan
| | - J Kitayama
- Department of Gastrointestinal Surgery, Jichi Medical University, Tochigi, Japan
| | - H Ishikawa
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Y Kodera
- Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - M Sekimoto
- Department of Surgery, Kansai Medical University, Hirakata, Japan
| | - S Satoi
- Department of Surgery, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
132
|
Bouchart C, Navez J, Closset J, Hendlisz A, Van Gestel D, Moretti L, Van Laethem JL. Novel strategies using modern radiotherapy to improve pancreatic cancer outcomes: toward a new standard? Ther Adv Med Oncol 2020; 12:1758835920936093. [PMID: 32684987 PMCID: PMC7343368 DOI: 10.1177/1758835920936093] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive solid tumours with an estimated 5-year overall survival rate of 7% for all stages combined. In this highly resistant disease that is located in the vicinity of many radiosensitive organs, the role of radiotherapy (RT) and indications for its use in this setting have been debated for a long time and are still under investigation. Although a survival benefit has yet to be clearly demonstrated for RT, it is the only technique, other than surgery, that has been demonstrated to lead to local control improvement. The adjuvant approach is now strongly challenged by neoadjuvant treatments that could spare patients with rapidly progressive systemic disease from unnecessary surgery and may increase free margin (R0) resection rates for those eligible for surgery. Recently developed dose-escalated RT treatments, designed either to maintain full-dose chemotherapy or to deliver a high biologically effective dose, particularly to areas of contact between the tumour and blood vessels, such as hypofractionated ablative RT (HFA-RT) or stereotactic body RT (SBRT), are progressively changing the treatment landscape. These modern strategies are currently being tested in prospective clinical trials with encouraging preliminary results, paving the way for more effective treatment combinations using novel targeted therapies. This review summarizes the current literature regarding the use of RT for the treatment of primary PDAC, describes the limitations of conventional RT, and discusses the emerging role of dose-escalated RT and heavy-particle RT.
Collapse
Affiliation(s)
- Christelle Bouchart
- Department of Radiation-Oncology, Institut Jules Bordet, Boulevard de Waterloo, 121, Brussels, 1000, Belgium
| | - Julie Navez
- Department of Hepato-Biliary-Pancreatic Surgery, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean Closset
- Department of Hepato-Biliary-Pancreatic Surgery, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Alain Hendlisz
- Department of Gastroenterology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Dirk Van Gestel
- Department of Radiation-Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Luigi Moretti
- Department of Radiation-Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Luc Van Laethem
- Department of Gastroenterology, Hepatology and Digestive Oncology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
133
|
Truty MJ, Colglazier JJ, Mendes BC, Nagorney DM, Bower TC, Smoot RL, DeMartino RR, Cleary SP, Oderich GS, Kendrick ML. En Bloc Celiac Axis Resection for Pancreatic Cancer: Classification of Anatomical Variants Based on Tumor Extent. J Am Coll Surg 2020; 231:8-29. [DOI: 10.1016/j.jamcollsurg.2020.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/04/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
|
134
|
Tuli R, David J, Lobaugh S, Zhang Z, O'Reilly EM. Duration of therapy for locally advanced pancreatic cancer: Does it matter? Cancer Med 2020; 9:4572-4580. [PMID: 32368871 PMCID: PMC7333837 DOI: 10.1002/cam4.3081] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Evidence-based recommendations on duration of multiagent systemic therapy for LAPC are lacking. Herein, we assess the impact of duration of combination systemic therapy on survival of patients with LAPC. METHODS The National Cancer Database was interrogated to identify patients with untreated LAPC diagnosed from 2004 to 2014. Patients treated with ≥ 1 month of multiagent chemotherapy (MAC) and ≥ 6 months of follow-up were included. Kaplan-Meier survival curves were generated to examine OS of each MAC duration group. Univariable and multivariable Cox proportional hazards regression was used to examine the association between OS with demographic and clinical variables. Statistical computations were performed using SAS Software Version 9.4. RESULTS Of the 3410 patients, 1114 met inclusion criteria. Median age was 64 years. Median treatment duration was 3.2 months (range 1-19.8). Median follow-up was 23.5 months (range 3-120). Median OS of all patients was 9.4 months (95% CI: 8.7-10.1). Median OS of patients receiving ≥ 1-4 months, >4-6 months and > 6 months of MAC was 8.4 months (95% CI: 7.7-9), 10.2 months (95% CI: 9-11.8), and 12.8 months (95% CI 11.6-16). Twelve-month survival was 37% for patients receiving ≥ 1-4 months, 43% for > 4-6 months, and 56% for > 6 months. Female sex (P = .02), higher median household income (P = .03), and longer duration of MAC (P < .001) were independently associated with improved OS following multivariable analysis. CONCLUSION This analysis in LAPC patients suggests that combination systemic therapy regimens of 6 months or more may optimize survival outcomes. Further investigation on the duration of systemic therapy question in LAPC is needed.
Collapse
Affiliation(s)
- Richard Tuli
- Department of Radiation Oncology, David M. Rubenstein Center for Pancreatic Cancer ResearchMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - John David
- Department of Radiation Oncology, David M. Rubenstein Center for Pancreatic Cancer ResearchMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Stephanie Lobaugh
- Department of Epidemiology‐BiostatisticsMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Zhigang Zhang
- Department of Epidemiology‐BiostatisticsMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Eileen M. O'Reilly
- Gastrointestinal Oncology ServiceDepartment of Medicine, David M. Rubenstein Center for Pancreatic Cancer ResearchMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| |
Collapse
|
135
|
Erstad DJ, Sojoodi M, Taylor MS, Jordan VC, Farrar CT, Axtell AL, Rotile NJ, Jones C, Graham-O'Regan KA, Ferreira DS, Michelakos T, Kontos F, Chawla A, Li S, Ghoshal S, Chen YCI, Arora G, Humblet V, Deshpande V, Qadan M, Bardeesy N, Ferrone CR, Lanuti M, Tanabe KK, Caravan P, Fuchs BC. Fibrotic Response to Neoadjuvant Therapy Predicts Survival in Pancreatic Cancer and Is Measurable with Collagen-Targeted Molecular MRI. Clin Cancer Res 2020; 26:5007-5018. [PMID: 32611647 DOI: 10.1158/1078-0432.ccr-18-1359] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 04/05/2019] [Accepted: 06/26/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE To evaluate the prognostic value of posttreatment fibrosis in human PDAC patients, and to compare a type I collagen targeted MRI probe, CM-101, to the standard contrast agent, Gd-DOTA, for their abilities to identify FOLFIRINOX-induced fibrosis in a murine model of PDAC. EXPERIMENTAL DESIGN Ninety-three chemoradiation-treated human PDAC samples were stained for fibrosis and outcomes evaluated. For imaging, C57BL/6 and FVB mice were orthotopically implanted with PDAC cells and FOLFIRINOX was administered. Mice were imaged with Gd-DOTA and CM-101. RESULTS In humans, post-chemoradiation PDAC tumor fibrosis was associated with longer overall survival (OS) and disease-free survival (DFS) on multivariable analysis (OS P = 0.028, DFS P = 0.047). CPA increased the prognostic accuracy of a multivariable logistic regression model comprised of previously established PDAC risk factors [AUC CPA (-) = 0.76, AUC CPA (+) = 0.82]. In multiple murine orthotopic PDAC models, FOLFIRINOX therapy reduced tumor weight (P < 0.05) and increased tumor fibrosis by collagen staining (P < 0.05). CM-101 MR signal was significantly increased in fibrotic tumor regions. CM-101 signal retention was also increased in the more fibrotic FOLFIRINOX-treated tumors compared with untreated controls (P = 0.027), consistent with selective probe binding to collagen. No treatment-related differences were observed with Gd-DOTA imaging. CONCLUSIONS In humans, post-chemoradiation tumor fibrosis is associated with OS and DFS. In mice, our MR findings indicate that translation of collagen molecular MRI with CM-101 to humans might provide a novel imaging technique to monitor fibrotic response to therapy to assist with prognostication and disease management.
Collapse
Affiliation(s)
- Derek J Erstad
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Mozhdeh Sojoodi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Martin S Taylor
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Veronica Clavijo Jordan
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Christian T Farrar
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Andrea L Axtell
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nicholas J Rotile
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Chloe Jones
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Katherine A Graham-O'Regan
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Diego S Ferreira
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Theodoros Michelakos
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Filippos Kontos
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Akhil Chawla
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shen Li
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sarani Ghoshal
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yin-Ching Iris Chen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Gunisha Arora
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nabeel Bardeesy
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael Lanuti
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kenneth K Tanabe
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Peter Caravan
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Bryan C Fuchs
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
136
|
Okada K, Murakami Y, Uemura K, Kondo N, Nakagawa N, Seo S, Otsuka H, Takahashi S. Neoadjuvant therapy for pancreatic cancer: an intention-to-treat analysis. Langenbecks Arch Surg 2020; 405:623-633. [PMID: 32592044 DOI: 10.1007/s00423-020-01914-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/22/2020] [Indexed: 01/13/2023]
Abstract
PURPOSE This study aimed to reassess the duration of neoadjuvant therapy (NAT) for patients with borderline resectable pancreatic cancer (BRPC). METHODS The medical records of patients with BRPC who received NAT before intended curative resection were retrospectively reviewed. Patient demographics, clinicopathological factors, and prognostic factors for overall survival were analyzed. The serum carbohydrate antigen (CA) 19-9 level was examined monthly during NAT. RESULTS A total of 118 patients with BRPC were enrolled. The median survival time and 5-year overall survival were 28.0 months and 31%, respectively. Three months after NAT, the CA19-9 levels were normal in 57% of the patients, and 92% underwent resection. Multivariate analysis showed that radiological partial response (hazard ratio (HR), 0.53; 95% confidence interval (CI), 0.26-0.99; p = 0.047); a normal CA19-9 level after NAT (HR, 0.30; 95% CI, 0.22-0.66; p = 0.006); and tumor resection (HR, 0.29; 95% CI, 0.13-0.67; p = 0.005) were independent predictors of better survival. The median CA19-9 level and the rate of normal CA19-9 levels before and after NAT were 256 (interquartile range (IQR), 23-1197) U/mL and 33%, and 27 (IQR, 7-176) U/mL and 57%, respectively. CONCLUSION A normal CA19-9 level after NAT was an independent predictor of better survival in patients with BRPC. A longer NAT duration might contribute to improved prognosis of patients with elevated CA19-9 levels.
Collapse
Affiliation(s)
- Kenjiro Okada
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Yoshiaki Murakami
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kenichiro Uemura
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8551, Japan
| | - Naru Kondo
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8551, Japan
| | - Naoya Nakagawa
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shingo Seo
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hiroyuki Otsuka
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinya Takahashi
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
137
|
Al Abbas AI, Zenati M, Reiser CJ, Hamad A, Jung JP, Zureikat AH, Zeh HJ, Hogg ME. Serum CA19-9 Response to Neoadjuvant Therapy Predicts Tumor Size Reduction and Survival in Pancreatic Adenocarcinoma. Ann Surg Oncol 2020; 27:2007-2014. [PMID: 31898105 PMCID: PMC7996002 DOI: 10.1245/s10434-019-08156-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND The optimal cutoffs for carbohydrate antigen 19-9 (CA19-9) response after neoadjuvant therapy (NT) for pancreatic adenocarcinoma (PDAC) are not well characterized. This study aimed to analyze the relationship of serum CA19-9 to other markers of response and to identify thresholds correlating to outcomes. METHODS A retrospective review of resected PDAC patients from 2010 to 2017 at an academic tertiary referral center was conducted. RESULTS The analysis enrolled 250 subjects. Normalization and multiple cutoff points for CA19-9 response were assessed. Normalization was not associated with improved survival (35.17 vs. 29.43 months; p = 0.173). Although a response 45% or higher was associated with longer survival (35 vs. 20 months; p = 0.018), a response of 85% or higher was optimal (55.7 vs. 25.97 months; p < 0.0001). A response of 85% or higher remained a strong independent predictor of survival [hazard ratio (HR), 0.47; p = 0.007]. Subjects with a response of 85% or higher had received more NT cycles [3 (range 2-6) vs. 3 (range 2-4) cycles; p = 0.006] and fewer adjuvant cycles [4 (range 3-6) vs. 5 (range 3-6) cycles; p = 0.027]. Reduction in T-size correlated with a drop in CA19-9 and a size reduction of 25% or higher (56.97 vs. 28.17 months; p = 0.016) improved survival. A serum CA19-9 response of 85% or higher was a strong independent predictor of a reduction in T-size of 25% or higher (HR 2.40; p = 0.007). CONCLUSION A CA19-9 response of 85% or higher is the optimal threshold for predicting survival. It is predictive of T-size reduction. Future NT trials should incorporate CA19-9 response as an end point.
Collapse
Affiliation(s)
- Amr I Al Abbas
- University of Pittsburgh, Pittsburgh, PA, USA
- University of Texas Southwestern, Dallas, TX, USA
| | | | | | - Ahmad Hamad
- University of Pittsburgh, Pittsburgh, PA, USA
- Ohio State University, Columbus, OH, USA
| | - Jae Pil Jung
- University of Pittsburgh, Pittsburgh, PA, USA
- Andong Medical Group Hospital, Andong-si, Gyeongbuk, Korea
| | | | - Herbert J Zeh
- University of Pittsburgh, Pittsburgh, PA, USA
- University of Texas Southwestern, Dallas, TX, USA
| | - Melissa E Hogg
- University of Pittsburgh, Pittsburgh, PA, USA.
- Walgreens Building, Department of Surgery 2539, Northsore University HealthSystem, 2650 Ridge Road, Evanston, IL, 60201, USA.
| |
Collapse
|
138
|
Yokose T, Kitago M, Matsusaka Y, Masugi Y, Shinoda M, Yagi H, Abe Y, Oshima G, Hori S, Endo Y, Toyama K, Iwabuchi Y, Takemura R, Ishii R, Nakahara T, Okuda S, Jinzaki M, Kitagawa Y. Usefulness of 18 F-fluorodeoxyglucose positron emission tomography/computed tomography for predicting the prognosis and treatment response of neoadjuvant therapy for pancreatic ductal adenocarcinoma. Cancer Med 2020; 9:4059-4068. [PMID: 32281301 PMCID: PMC7300404 DOI: 10.1002/cam4.3044] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/20/2020] [Accepted: 03/21/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The Response Evaluation Criteria in Solid Tumors (RECIST) for computed tomography (CT) is preoperatively used to evaluate therapeutic effects. However, it does not reflect the pathological treatment response (PTR) of pancreatic ductal adenocarcinoma (PDAC). The Positron Emission Tomography Response Criteria in Solid Tumors (PERCIST) for positron emission tomography (PET)/CT is effective in other cancers. This study aimed to confirm the usefulness of PERCIST and the prognostic utility of PET/CT for PDAC. METHODS Forty-two consecutive patients with PDAC who underwent neoadjuvant therapy (NAT) and pancreatectomy at our institution between 2014 and 2018 were retrospectively analyzed. We evaluated the treatment response and prognostic significance of PET/CT parameters and other clinicopathological factors. RESULTS Twenty-two patients who underwent PET/CT both before and after NAT with the same protocol were included. RECIST revealed stable disease and partial response in 20 and 2 cases, respectively. PERCIST revealed stable metabolic disease, partial metabolic response, and complete metabolic response in 8, 9, and 5 cases, respectively. The PTR was G3, G2, and G1 in 8, 12, and 2 cases, respectively. For comparing the concordance rates between PTR and each parameter, PERCIST (72.7% [16/22]) was significantly superior to RECIST (36.4% [8/22]) (P = .017). The area under the curve survival values of PET/CT parameters were 0.777 for metabolic tumor volume (MTV), 0.500 for maximum standardized uptake value, 0.554 for peak standardized uptake value corrected for lean body mass, and 0.634 for total lesion glycolysis. A 50% cut-off value for the MTV reduction rate yielded the largest difference in survival between responders and nonresponders. On multivariate analysis, MTV reduction rates < 50% were independent predictors for relapse-free survival (hazard ratio [HR], 3.92; P = .044) and overall survival (HR, 14.08; P = .023). CONCLUSIONS PERCIST was more accurate in determining NAT's therapeutic effects for PDAC than RECIST. MTV reduction rates were independent prognostic factors for PDAC.
Collapse
Affiliation(s)
- Takahiro Yokose
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Minoru Kitago
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yohji Matsusaka
- Department of RadiologyKeio University School of MedicineTokyoJapan
| | - Yohei Masugi
- Department of PathologyKeio University School of MedicineTokyoJapan
| | - Masahiro Shinoda
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Hiroshi Yagi
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yuta Abe
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Go Oshima
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Shutaro Hori
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yutaka Endo
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Kenji Toyama
- Department of RadiologyKeio University School of MedicineTokyoJapan
| | - Yu Iwabuchi
- Department of RadiologyKeio University School of MedicineTokyoJapan
| | - Ryo Takemura
- Biostatistics Unit, Clinical and Translational Research CenterKeio University HospitalTokyoJapan
| | - Ryota Ishii
- Biostatistics Unit, Clinical and Translational Research CenterKeio University HospitalTokyoJapan
| | - Tadaki Nakahara
- Department of RadiologyKeio University School of MedicineTokyoJapan
| | - Shigeo Okuda
- Department of RadiologyKeio University School of MedicineTokyoJapan
| | - Masahiro Jinzaki
- Department of RadiologyKeio University School of MedicineTokyoJapan
| | - Yuko Kitagawa
- Department of SurgeryKeio University School of MedicineTokyoJapan
| |
Collapse
|
139
|
Wolfe AR, Prabhakar D, Yildiz VO, Cloyd JM, Dillhoff M, Abushahin L, Alexandra Diaz D, Miller ED, Chen W, Frankel WL, Noonan A, Williams TM. Neoadjuvant-modified FOLFIRINOX vs nab-paclitaxel plus gemcitabine for borderline resectable or locally advanced pancreatic cancer patients who achieved surgical resection. Cancer Med 2020; 9:4711-4723. [PMID: 32415696 PMCID: PMC7333854 DOI: 10.1002/cam4.3075] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/02/2020] [Accepted: 04/05/2020] [Indexed: 12/11/2022] Open
Abstract
We conducted an institutional study to compare the clinical and pathological efficacy between the neoadjuvant therapy (NAT)‐modified FOLFIRINOX (mFOLF) vs nanoparticle albumin–bound paclitaxel plus gemcitabine (nab‐P/G) for borderline resectable pancreatic cancer (BRPC) and locally advanced pancreatic cancer (LAPC) patients who completed resection. The study retrospectively enrolled patients with pathologically confirmed BRPC or LAPC from 2010 to 2018 at our institution. The survival rates were determined by the Kaplan‐Meier method and log‐rank test was used to test differences. Cox's proportional hazard model was used to assess survival with respect to covariates. Seventy‐two patients who completed at least two cycles of neoadjuvant chemotherapy and surgical resection were included, with 52 (72.2%) patients receiving mFOLF and 20 (27.8%) receiving nab‐P/G. Patients treated with mFOLF had statistically higher rates of RECIST 1.1 partial or complete response (16/52 vs 1/20, P = .028). Additionally, mFOLF patients had greater pathological tumor size reduction, fewer positive lymph nodes, and higher treatment response grade compared to the nab‐P/G patients (all P < .05). The median overall survival was 33.3 months vs 27.1 months (P = .105), and distant metastasis‒free survival (DMFS) was 21.3 months vs 14.6 months (P = .042) in the mFOLF vs nab‐P/G groups, respectively. On multivariate analysis, mFOLF (hazard ratio, 0.428; 95% confidence interval [CI], 0.186‐0.987) and abnormal postoperative CA 19‐9 (hazard ratio, 2.47; 95% CI, 1.06‐5.76) were associated with DMFS. Among patients with BRPC and LAPC who complete surgical resection, neoadjuvant mFOLF was associated with improved pathological and clinical outcomes compared with nab‐P/G.
Collapse
Affiliation(s)
- Adam R Wolfe
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Dhivya Prabhakar
- Department of Medical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Vedat O Yildiz
- Department of Biomedical Informatics, Ohio State College of Medicine, Columbus, OH, USA
| | - Jordan M Cloyd
- Department of Surgical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Mary Dillhoff
- Department of Surgical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Laith Abushahin
- Department of Medical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Dayssy Alexandra Diaz
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Eric D Miller
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Wei Chen
- Department of Pathology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Wendy L Frankel
- Department of Pathology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Anne Noonan
- Department of Medical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Terence M Williams
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
140
|
Simpson G, Spieler B, Dogan N, Portelance L, Mellon EA, Kwon D, Ford JC, Yang F. Predictive value of 0.35 T magnetic resonance imaging radiomic features in stereotactic ablative body radiotherapy of pancreatic cancer: A pilot study. Med Phys 2020; 47:3682-3690. [DOI: 10.1002/mp.14200] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/31/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Garrett Simpson
- Department of Radiation Oncology University of Miami Miami FL 33136 USA
| | - Benjamin Spieler
- Department of Radiation Oncology University of Miami Miami FL 33136 USA
| | - Nesrin Dogan
- Department of Radiation Oncology University of Miami Miami FL 33136 USA
| | | | - Eric A. Mellon
- Department of Radiation Oncology University of Miami Miami FL 33136 USA
| | - Deukwoo Kwon
- Department of Radiation Oncology University of Miami Miami FL 33136 USA
| | - John C. Ford
- Department of Radiation Oncology University of Miami Miami FL 33136 USA
| | - Fei Yang
- Department of Radiation Oncology University of Miami Miami FL 33136 USA
| |
Collapse
|
141
|
Perri G, Katz MHG. ASO Author Reflections: The Sequential Radiographic Effects of Preoperative Chemotherapy and (Chemo)Radiation on Tumor Anatomy in Patients with Localized Pancreatic Cancer. Ann Surg Oncol 2020; 27:3948-3949. [PMID: 32358700 DOI: 10.1245/s10434-020-08531-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Indexed: 11/18/2022]
Affiliation(s)
- Giampaolo Perri
- Department of General and Pancreatic Surgery, Verona University Hospital, Verona, Italy
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
142
|
Perri G, Prakash LR, Katz MHG. Response to Preoperative Therapy in Localized Pancreatic Cancer. Front Oncol 2020; 10:516. [PMID: 32351893 PMCID: PMC7174698 DOI: 10.3389/fonc.2020.00516] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/23/2020] [Indexed: 12/22/2022] Open
Abstract
Evaluation of response to preoperative therapy for patients with pancreatic adenocarcinoma has been historically difficult. Therefore, preoperative regimens have generally been selected on the basis of baseline data such as radiographic stage and serum CA 19-9 level and then typically administered for a pre-specified duration as long as 6 months or more. The decision to proceed with resection following preoperative therapy likewise has rested upon the absence of disease progression rather than evidence for tumor response. This article reviews the basis for the evaluation of therapeutic response after preoperative therapy for pancreatic cancer in the existing scientific literature, and providing updates and new perspectives.
Collapse
Affiliation(s)
- Giampaolo Perri
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Laura R Prakash
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
143
|
Perri G, Prakash L, Malleo G, Caravati A, Varadhachary GR, Fogelman D, Pant S, Koay EJ, Herman J, Maggino L, Milella M, Kim M, Ikoma N, Tzeng CW, Salvia R, Lee JE, Bassi C, Katz MHG. The Sequential Radiographic Effects of Preoperative Chemotherapy and (Chemo)Radiation on Tumor Anatomy in Patients with Localized Pancreatic Cancer. Ann Surg Oncol 2020; 27:3939-3947. [PMID: 32266574 PMCID: PMC7471157 DOI: 10.1245/s10434-020-08427-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND The incidence and magnitude of indicators of radiographic response of pancreatic cancer to systemic chemotherapy and (chemo)radiation administered prior to anticipated pancreatectomy are unclear. METHODS Sequential computed tomography scans of 226 patients with localized pancreatic cancer who received chemotherapy consisting of 5-fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFIRINOX) or gemcitabine and nanoparticle albumin-bound paclitaxel (GA) with or without (chemo)radiation and who subsequently underwent surgery with curative intent from January 2010 to December 2018 at The University of Texas MD Anderson Cancer Center and Verona University Hospital were re-reviewed and compared. RESULTS Overall, 141 patients (62%) received FOLFIRINOX, 70 (31%) received GA, and 15 (7%) received both; 164 patients (73%) received preoperative (chemo)radiation following chemotherapy and prior to surgery; and 151 (67%), 70 (31%), and 5 (2%) patients had Response Evaluation Criteria in Solid Tumors (RECIST) stable disease, partial response, and progressive disease, respectively. The tumors of 29% of patients with borderline resectable or locally advanced cancer were downstaged after preoperative therapy. Radiographic downstaging was more common with chemotherapy than with (chemo)radiation (24% vs. 6%; p = 0.04), and the median tumor volume loss after chemotherapy was significantly greater than that after (chemo)radiation (28% vs. 17%; p < 0.01). CONCLUSIONS Less than one-third of patients treated with FOLFIRINOX or GA with or without (chemo)radiation experienced either RECIST partial response or radiographic downstaging prior to surgery. The incidence of tumor downstaging was higher and the magnitude of tumor volume loss was greater following chemotherapy than after (chemo)radiation.
Collapse
Affiliation(s)
- Giampaolo Perri
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laura Prakash
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giuseppe Malleo
- Department of General and Pancreatic Surgery, Verona University Hospital, Verona, Italy
| | - Andrea Caravati
- Department of General and Pancreatic Surgery, Verona University Hospital, Verona, Italy
| | - Gauri R Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Fogelman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Herman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laura Maggino
- Department of General and Pancreatic Surgery, Verona University Hospital, Verona, Italy
| | - Michele Milella
- Department of Gastrointestinal Medical Oncology, Verona University Hospital, Verona, Italy
| | - Michael Kim
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naruhiko Ikoma
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ching-Wei Tzeng
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roberto Salvia
- Department of General and Pancreatic Surgery, Verona University Hospital, Verona, Italy
| | - Jeffrey E Lee
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Claudio Bassi
- Department of General and Pancreatic Surgery, Verona University Hospital, Verona, Italy
| | - Matthew H G Katz
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
144
|
Systematic Analysis of Accuracy in Predicting Complete Oncological Resection in Pancreatic Cancer Patients-Proposal of a New Simplified Borderline Resectability Definition. Cancers (Basel) 2020; 12:cancers12040882. [PMID: 32260453 PMCID: PMC7226508 DOI: 10.3390/cancers12040882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/20/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Borderline resectability in pancreatic cancer (PDAC) is currently debated. Methods: Patients undergoing pancreatic resections for PDAC were identified from a prospectively maintained database. As new borderline criteria, the presence of any superior mesenterico-portal vein alteration (SMPV) and perivascular stranding of the superior mesenteric artery (SMA) was evaluated in preoperative imaging. The accuracy of established radiological borderline criteria as compared to the new borderline criteria in predicting R status (sensitivity/negative predictive value) and overall survival was assessed. (3) Results: 118 patients undergoing pancreatic resections for PDAC from 2013 to 2018 were identified. Forty-three (36.4%) had radiological perivascular SMA stranding and 55 (46.6%) had SMPV alterations. Interrater reliability was 90% for SMA stranding and 87% for SMPV alterations. The new borderline definition including SMPV alterations and perivascular SMA stranding was the best predictor of conventional R status (p = 0.040, sensitivity 53%, negative predictive value 81%) and Leeds/Wittekind circumferential margin status (p = 0.050, sensitivity 73%, negative predictive value 79%) as compared to established borderline resectability definition criteria. Perivascular SMA stranding qualified as an independent negative prognostic parameter (HR 3.066, 95% CI 1.078-5.716, p = 0.036). Conclusion: The radiological evaluation of any SMPV alteration and perivascular SMA stranding predicts R status and overall survival in PDAC patients, and may serve to identify potential candidates for neoadjuvant therapy.
Collapse
|
145
|
Reni M, Peretti U, Zanon S, Macchini M, Balzano G, Mazza E, Tamburrino D, Orsi G, Arcidiacono PG, Falconi M, Gianni L. Time to CA19-9 nadir: a clue for defining optimal treatment duration in patients with resectable pancreatic ductal adenocarcinoma. Cancer Chemother Pharmacol 2020; 85:641-650. [PMID: 32157412 DOI: 10.1007/s00280-020-04047-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/19/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Defining optimal treatment duration in patients with resectable pancreatic ductal adenocarcinoma (PDAC) receiving primary chemotherapy is an unmet need. The role of time to CA19-9 nadir and of nadir magnitude was explored in this study. PATIENTS AND METHODS The databases of our institution's prospective trials were queried to speculate on the time to maximum chemotherapy response. Patients with pathologically proven, metastatic (N = 356) or non-metastatic non-resected (N = 163) PDAC and elevated baseline (> 34 UI/mL) CA19-9 were analyzed. Survival curves were estimated using the Kaplan-Meier method and compared by means of the log-rank test for analyses including at least 45 patients. Multivariable Cox proportional hazards model was used to estimate clinical features for their association with OS. All probability values were from two-sided tests. RESULTS Time to CA19-9 nadir was ≥ 4 months in 184 of 346 (53%) metastatic and 121 of 163 (74%) non-metastatic patients (p = 0.002). The likelihood of a later nadir was higher with taxane-based chemotherapy as compared to taxane-free combinations (73% versus 56%; p = 0.02). Both metastatic and non-metastatic patients had significantly longer survival when nadir occurred later. Patients with a larger CA19-9 nadir magnitude had significantly longer survival. Metastatic patients with CA19-9 reduced by < 50%, 50-89%, or > 89% and had a median survival of 7.4, 9.8, and 14.7 months, respectively (p ≤ 0.001 for all comparisons). The corresponding figures for non-metastatic patients were 10.6; 17.0; and 18.7 months, respectively (p ≤ 0.02 for < 50% versus 50-89% or > 89%; p = 0.14 for 50-89% versus > 89%). Multivariable analyses showed that time to CA19-9 nadir but not CA19-9 nadir magnitude was independently predictive of survival. CONCLUSION The present study suggests that a 4-6 months program might be a more suitable candidate for prospective assessment in comparison to shorter pre-defined period in patients who are candidates to surgery after primary chemotherapy.
Collapse
Affiliation(s)
- Michele Reni
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| | - Umberto Peretti
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Silvia Zanon
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Gianpaolo Balzano
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Elena Mazza
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Domenico Tamburrino
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Giulia Orsi
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
- Università "Vita E Salute", Via Olgettina 58, 20132, Milan, Italy
| | - Luca Gianni
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
146
|
Al Abbas AI, Hogg ME. ASO Author Reflections: Serum CA19-9 Utility in Pancreatic Adenocarcinoma in Patients Undergoing Neoadjuvant Therapy. Ann Surg Oncol 2020; 27:2015-2016. [PMID: 32172332 DOI: 10.1245/s10434-020-08342-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Indexed: 11/18/2022]
Affiliation(s)
- Amr I Al Abbas
- University of Pittsburgh, Pittsburgh, PA, USA.,University of Texas Southwestern, Dallas, TX, USA
| | - Melissa E Hogg
- University of Pittsburgh, Pittsburgh, PA, USA. .,Walgreens Building, Department of Surgery 2539, Northshore University Health System, Evanston, IL, USA.
| |
Collapse
|
147
|
Tanaka M, Inoue Y, Matsueda K, Hiratsuka M, Muto M, Kawakatsu S, Ono Y, Mise Y, Ishizawa T, Ito H, Takahashi Y, Takazawa Y, Saiura A. Three-Dimensional Fixation: Pathological Protocol Following Pancreaticoduodenectomy with Portal Vein Resection for Pancreatic Cancer. J Gastrointest Surg 2020; 24:619-626. [PMID: 30937709 DOI: 10.1007/s11605-019-04203-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/08/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Although existing histopathologic protocols for pancreatic cancer have been standardized, the relevance between prognosis and resection margin clearance is still controversial. Reconstruction of specimens as in situ to appropriately assess the margin is desirable in these protocols. METHODS The three-dimensional fixation protocol defined specimen handling of pancreaticoduodenectomy (PD) with portal vein (PV) resection. The superior mesenteric artery (SMA) margin of the specimen was tidily fixed around an artificial SMA as if in an in situ setting. In this prospective study, patients undergoing PD with PV resection for pancreatic cancer in 2016 were enrolled. To evaluate the feasibility of the three-dimensional fixation protocol, the SMA margin distance and PV involvement of tumor assessed by computed tomography (CT) were compared with those assessed by pathology. RESULTS Thirty-three patients with/without preoperative chemotherapy were enrolled. The entire cohort did not present with high-quality diagnostic assessment of the medial margins around SMA and PV (correct estimation, 58% and 73%, respectively). In contrast, in 16 patients undergoing upfront surgery, the concordance value of the SMA margin, which assesses the agreement between CT and pathology measures, was 0.48 (moderate agreement). The PV involvement examined by imaging was significantly associated with that by pathology (P = 0.013). CONCLUSIONS The three-dimensional fixation protocol was applicable to all cases undergoing PD with PV resection. Focusing on the patients with upfront surgery demonstrated the feasibility of accurate pathological assessment of medial margins. We propose this protocol as a promising standard for the assessment of true surgical margin status.
Collapse
Affiliation(s)
- Masayuki Tanaka
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yosuke Inoue
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Kiyoshi Matsueda
- Department of Diagnostic Imaging, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makiko Hiratsuka
- Department of Diagnostic Imaging, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mariko Muto
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shoji Kawakatsu
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yoshihiro Ono
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yoshihiro Mise
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takeaki Ishizawa
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Hiromichi Ito
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yu Takahashi
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yutaka Takazawa
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akio Saiura
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| |
Collapse
|
148
|
Murthy P, Zenati MS, Al Abbas AI, Rieser CJ, Bahary N, Lotze MT, Zeh HJ, Zureikat AH, Boone BA. Prognostic Value of the Systemic Immune-Inflammation Index (SII) After Neoadjuvant Therapy for Patients with Resected Pancreatic Cancer. Ann Surg Oncol 2020; 27:898-906. [PMID: 31792715 PMCID: PMC7879583 DOI: 10.1245/s10434-019-08094-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The systemic immune-inflammation index (SII), calculated using absolute platelet, neutrophil, and lymphocyte counts, has recently emerged as a predictor of survival for patients with pancreatic ductal adenocarcinoma (PDAC) when assessed at diagnosis. Neoadjuvant therapy (NAT) is increasingly used in the treatment of PDAC. However, biomarkers of response are lacking. This study aimed to determine the prognostic significance of SII before and after NAT and its association with the pancreatic tumor biomarker carbohydrate-antigen 19-9 (CA 19-9). METHODS This study retrospectively analyzed all PDAC patients treated with NAT before pancreatic resection at a single institution between 2007 and 2017. Pre- and post-NAT lab values were collected to calculate SII. Absolute pre-NAT, post-NAT, and change in SII after NAT were evaluated for their association with clinical outcomes. RESULTS The study analyzed 419 patients and found no significant correlation between pre-NAT SII and clinical outcomes. Elevated post-NAT SII was an independent, negative predictor of overall survival (OS) when assessed as a continuous variable (hazard ratio [HR], 1.0001; 95% confidence interval [CI] 1.00003-1.00014; p = 0.006). Patients with a post-NAT SII greater than 900 had a shorter median OS (31.9 vs 26.1 months; p = 0.050), and a post-NAT SII greater than 900 also was an independent negative predictor of OS (HR, 1.369; 95% CI 1.019-1.838; p = 0.037). An 80% reduction in SII independently predicted a CA 19-9 response after NAT (HR, 4.22; 95% CI 1.209-14.750; p = 0.024). CONCLUSION Post-treatment SII may be a useful prognostic marker in PDAC patients receiving NAT.
Collapse
Affiliation(s)
- Pranav Murthy
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mazen S Zenati
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amr I Al Abbas
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Caroline J Rieser
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nathan Bahary
- Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Herbert J Zeh
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Amer H Zureikat
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian A Boone
- Department of Surgery, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
149
|
Kulkarni NM, Soloff EV, Tolat PP, Sangster GP, Fleming JB, Brook OR, Wang ZJ, Hecht EM, Zins M, Bhosale PR, Arif-Tiwari H, Mannelli L, Kambadakone AR, Tamm EP. White paper on pancreatic ductal adenocarcinoma from society of abdominal radiology's disease-focused panel for pancreatic ductal adenocarcinoma: Part I, AJCC staging system, NCCN guidelines, and borderline resectable disease. Abdom Radiol (NY) 2020; 45:716-728. [PMID: 31748823 DOI: 10.1007/s00261-019-02289-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive gastrointestinal malignancy with a poor 5-year survival rate. Accurate staging of PDAC is an important initial step in the development of a stage-specific treatment plan. Different staging systems/consensus statements convened by different societies and academic practices are currently used. The most recent version of the American Joint Committee on Cancer (AJCC) tumor/node/metastases (TNM) staging system for PDAC has shifted its focus from guiding management to assessing prognosis. In order to preoperatively define the resectability of PDAC and to guide management, additional classification systems have been developed. The National Comprehensive Cancer Network (NCCN) guidelines, one of the most commonly used systems, provide recommendations on the management and the determination of resectability for PDAC. The NCCN divides PDAC into three categories of resectability based on tumor-vessel relationship: 'resectable,' 'borderline resectable,' and 'unresectable'. Among these, the borderline disease category is of special interest given its evolution over time and the resulting variations in the definition and the associated recommendations for management between different societies. It is important to be familiar with the evolving criteria, and treatment and follow-up recommendations for PDAC. In this article, the most current AJCC staging (8th edition), NCCN guidelines (version 2.2019-April 9, 2019), and challenges and controversies in borderline resectable PDAC are reviewed.
Collapse
Affiliation(s)
- Naveen M Kulkarni
- Department of Radiology, Medical College of Wisconsin, 9200 W Wisconsin Ave, Milwaukee, WI, 53226, USA.
| | - Erik V Soloff
- Department of Radiology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Parag P Tolat
- Department of Radiology, Medical College of Wisconsin, 9200 W Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Guillermo P Sangster
- Department of Radiology, LSU Health - Shreveport Ochsner-LSU Health - Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Jason B Fleming
- Gastrointestinal Oncology, Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Olga R Brook
- Department of Radiology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Shapiro 4, Boston, MA, 02215-5400, USA
| | - Zhen Jane Wang
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 505 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Elizabeth M Hecht
- Department of Radiology, Columbia University Medical Center, 622 W 168th St, PH1-317, New York, NY, 10032, USA
| | - Marc Zins
- Department of Radiology, Groupe Hospitalier Paris Saint-Joseph, 185 Rue Raymond Losserand, 75014, Paris, France
| | - Priya R Bhosale
- Abdominal Imaging Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1473, Houston, TX, 77030-400, USA
| | - Hina Arif-Tiwari
- Department of Radiology, University of Arizona College of Medicine, 1501 N. Campbell Ave., P.O. Box 245067, Tucson, AZ, 85724, USA
| | | | - Avinash R Kambadakone
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, White 270, Boston, MA, 02114, USA
| | - Eric P Tamm
- Abdominal Imaging Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1473, Houston, TX, 77030-400, USA
| |
Collapse
|
150
|
Kulkarni NM, Mannelli L, Zins M, Bhosale PR, Arif-Tiwari H, Brook OR, Hecht EM, Kastrinos F, Wang ZJ, Soloff EV, Tolat PP, Sangster G, Fleming J, Tamm EP, Kambadakone AR. White paper on pancreatic ductal adenocarcinoma from society of abdominal radiology's disease-focused panel for pancreatic ductal adenocarcinoma: Part II, update on imaging techniques and screening of pancreatic cancer in high-risk individuals. Abdom Radiol (NY) 2020; 45:729-742. [PMID: 31768594 DOI: 10.1007/s00261-019-02290-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive gastrointestinal malignancy with a poor 5-year survival rate. Its high mortality rate is attributed to its aggressive biology and frequently late presentation. While surgical resection remains the only potentially curative treatment, only 10-20% of patients will present with surgically resectable disease. Over the past several years, development of vascular bypass graft techniques and introduction of neoadjuvant treatment regimens have increased the number of patients who can undergo resection with a curative intent. While the role of conventional imaging in the detection, characterization, and staging of patients with PDAC is well established, its role in monitoring treatment response, particularly following neoadjuvant therapy remains challenging because of the complex anatomic and histological nature of PDAC. Novel morphologic and functional imaging techniques (such as DECT, DW-MRI, and PET/MRI) are being investigated to improve the diagnostic accuracy and the ability to measure response to therapy. There is also a growing interest to detect PDAC and its precursor lesions at an early stage in asymptomatic patients to increase the likelihood of achieving cure. This has led to the development of pancreatic cancer screening programs. This article will review recent updates in imaging techniques and the current status of screening and surveillance of individuals at a high risk of developing PDAC.
Collapse
Affiliation(s)
- Naveen M Kulkarni
- Department of Radiology, Medical College of Wisconsin, 9200 W Wisconsin Ave, Milwaukee, WI, 53226, USA.
| | | | - Marc Zins
- Department of Radiology, Groupe Hospitalier Paris Saint-Joseph, 185 rue Raymond Losserand, 75014, Paris, France
| | - Priya R Bhosale
- Abdominal Imaging Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1473, Houston, TX, 77030-400, USA
| | - Hina Arif-Tiwari
- Department of Medical Imaging, University of Arizona College of Medicine, 1501 N. Campbell Ave, P.O. Box 245067, Tucson, AZ, 85724, USA
| | - Olga R Brook
- Department of Radiology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Shapiro 4, Boston, MA, 02215-5400, USA
| | - Elizabeth M Hecht
- Department of Radiology, Columbia University Medical Center, 622 W 168th St, PH1-317, New York, NY, 10032, USA
| | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Cancer, 161 Fort Washington Avenue, Suite: 862, New York, NY, 10032, USA
| | - Zhen Jane Wang
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 505 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Erik V Soloff
- Department of Radiology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Parag P Tolat
- Department of Radiology, Medical College of Wisconsin, 9200 W Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Guillermo Sangster
- Department of Radiology, Ochsner LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Jason Fleming
- Gastrointestinal Oncology, Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Eric P Tamm
- Abdominal Imaging Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1473, Houston, TX, 77030-400, USA
| | - Avinash R Kambadakone
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, White 270, Boston, MA, 02114, USA
| |
Collapse
|