1851
|
Petrillo A, Pappalardo A, Pompella L, Tirino G, Calabrese F, Laterza MM, Caterino M, Ventriglia A, Orditura M, Conzo G, Molino C, Ciardiello F, Biglietto M, De Vita F. Nab-paclitaxel plus gemcitabine as first line therapy in metastatic pancreatic cancer patients relapsed after gemcitabine adjuvant treatment. Med Oncol 2019; 36:83. [PMID: 31444639 DOI: 10.1007/s12032-019-1306-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 08/15/2019] [Indexed: 12/27/2022]
Abstract
Nab-paclitaxel plus gemcitabine (Nab-Gem) represents one of the standard regimen for first-line treatment of metastatic pancreatic adenocarcinoma (mPDAC). However, few data are available in mPDAC relapsed after gemcitabine as adjuvant treatment. Our study aims to evaluate the efficacy and feasibility of Nab-Gem as first-line treatment for mPDAC patients previously treated with adjuvant treatment. We retrospectively analyzed the safety and efficacy data of 36 patients, who received first-line Nab-Gem after gemcitabine as adjuvant treatment. All patients received gemcitabine after radical surgery. Median disease-free survival was 12 months (95% CI 9.7-14.3); at relapse, all patients received Nab-Gem. We observed an objective response rate and disease control rate of 11.1% and 63.9%, respectively. With a median follow-up of 47 months, median progression-free survival was 5 months (95% CI 1.0-9.0), whereas median overall survival (OS) was 13 months (95% CI 5.5-20.5). Median OS was higher in patients with a relapse ≥ 7 months after the end of adjuvant treatment than in patients relapsed < 7 months (14 vs. 8 months, respectively, p: 0.52). Our results show that first-line Nab-Gem is feasible and effective in patients previously treated with gemcitabine as adjuvant treatment.
Collapse
Affiliation(s)
- Angelica Petrillo
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy.
| | - Annalisa Pappalardo
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Luca Pompella
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Giuseppe Tirino
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | | | - Maria Maddalena Laterza
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Marianna Caterino
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Anna Ventriglia
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Michele Orditura
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Giovanni Conzo
- Department of Cardiothoracic Sciences, University of Study of Campania "L. Vanvitelli", Via Sergio Pansini 5, 80131, Naples, Italy
| | - Carlo Molino
- Department of Oncological Surgery, A.O.R.N. "A. Cardarelli", Naples, Italy
| | - Fortunato Ciardiello
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Maria Biglietto
- Division of Medical Oncology, AORN, "A Cardarelli", Naples, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy.
| |
Collapse
|
1852
|
Wei AC, Ou FS, Shi Q, Carrero X, O'Reilly EM, Meyerhardt J, Wolff RA, Kindler HL, Evans DB, Deshpande V, Misdraji J, Tamm E, Sahani D, Moore M, Newman E, Merchant N, Berlin J, Goff LW, Pisters P, Posner MC. Perioperative Gemcitabine + Erlotinib Plus Pancreaticoduodenectomy for Resectable Pancreatic Adenocarcinoma: ACOSOG Z5041 (Alliance) Phase II Trial. Ann Surg Oncol 2019; 26:4489-4497. [PMID: 31418130 DOI: 10.1245/s10434-019-07685-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND There is considerable interest in a neoadjuvant approach for resectable pancreatic ductal adenocarcinoma (PDAC). This study evaluated perioperative gemcitabine + erlotinib (G+E) for resectable PDAC. METHODS A multicenter, cooperative group, single-arm, phase II trial was conducted between April 2009 and November 2013 (ACOSOG Z5041). Patients with biopsy-confirmed PDAC in the pancreatic head without evidence of involvement of major mesenteric vessels (resectable) were eligible. Patients (n = 123) received an 8-week cycle of G+E before and after surgery. The primary endpoint was 2-year overall survival (OS), and secondary endpoints included toxicity, response, resection rate, and time to progression. Resectability was assessed retrospectively by central review. The study closed early due to slow accrual, and no formal hypothesis testing was performed. RESULTS Overall, 114 patients were eligible, consented, and initiated protocol treatment. By central radiologic review, 97 (85%) of the 114 patients met the protocol-defined resectability criteria. Grade 3+ toxicity was reported in 60% and 79% of patients during the neoadjuvant phase and overall, respectively. Twenty-two of 114 (19%) patients did not proceed to surgery; 83 patients (73%) were successfully resected. R0 and R1 margins were obtained in 67 (81%) and 16 (19%) resected patients, respectively, and 54 patients completed postoperative G+E (65%). The 2-year OS rate for the entire cohort (n = 114) was 40% (95% confidence interval [CI] 31-50), with a median OS of 21.3 months (95% CI 17.2-25.9). The 2-year OS rate for resected patients (n = 83) was 52% (95% CI 41-63), with a median OS of 25.4 months (95% CI 21.8-29.6). CONCLUSIONS For resectable PDAC, perioperative G+E is feasible. Further evaluation of neoadjuvant strategies in resectable PDAC is warranted with more active systemic regimens.
Collapse
Affiliation(s)
- Alice C Wei
- University Health Network-Princess Margaret Hospital, Toronto, ON, Canada. .,Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Fang-Shu Ou
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Qian Shi
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Xiomara Carrero
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Hedy L Kindler
- University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | | | | | - Joseph Misdraji
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Eric Tamm
- MD Anderson Cancer Center, Houston, TX, USA
| | - Dushyant Sahani
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Malcolm Moore
- University Health Network-Princess Margaret Hospital, Toronto, ON, Canada
| | - Elliot Newman
- New York University Langone Medical Center, New York, NY, USA
| | - Nipun Merchant
- University of Miami Miller School of Medicine-Sylvester Cancer Center, Miami, FL, USA
| | - Jordan Berlin
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laura W Goff
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | |
Collapse
|
1853
|
Khorana AA, McKernin SE, Berlin J, Hong TS, Maitra A, Moravek C, Mumber M, Schulick R, Zeh HJ, Katz MHG. Potentially Curable Pancreatic Adenocarcinoma: ASCO Clinical Practice Guideline Update. J Clin Oncol 2019; 37:2082-2088. [PMID: 31180816 DOI: 10.1200/jco.19.00946] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2019] [Indexed: 12/17/2023] Open
Abstract
PURPOSE The purpose of this guideline update is to incorporate recently reported practice-changing evidence into ASCO's recommendations on potentially curable pancreatic adenocarcinoma. METHODS ASCO convened an Expert Panel to evaluate data from PRODIGE 24/CCTG PA.6, a phase III, multicenter, randomized clinical trial of postoperative leucovorin calcium, fluorouracil, irinotecan hydrochloride, and oxaliplatin (FOLFIRINOX) versus gemcitabine alone, presented at the 2018 ASCO Annual Meeting. In addition, PubMed was searched for additional papers that may influence the existing recommendations. RECOMMENDATIONS The Expert Panel only updated Recommendation 4.1 as a result of the practice-changing data. Recommendation 4.1 states that all patients with resected pancreatic adenocarcinoma who did not receive preoperative therapy should be offered 6 months of adjuvant chemotherapy in the absence of medical or surgical contraindications. The modified combination regimen of 5-fluorouracil, oxaliplatin, and irinotecan (mFOLFIRINOX; oxaliplatin 85 mg/m2, leucovorin 400 mg/m2, irinotecan 150 mg/m2 D1, and 5-fluorouracil 2.4 g/m2 over 46 hours every 14 days for 12 cycles) is now preferred in the absence of concerns for toxicity or tolerance; alternatively, doublet therapy with gemcitabine and capecitabine or monotherapy with gemcitabine alone or fluorouracil plus folinic acid alone can be offered.Additional information can be found at www.asco.org/gastrointestinal-cancer-guidelines.
Collapse
Affiliation(s)
| | | | | | | | - Anirban Maitra
- 5The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | |
Collapse
|
1854
|
|
1855
|
Ma SJ, Oladeru OT, Miccio JA, Iovoli AJ, Hermann GM, Singh AK. Association of Timing of Adjuvant Therapy With Survival in Patients With Resected Stage I to II Pancreatic Cancer. JAMA Netw Open 2019; 2:e199126. [PMID: 31411712 PMCID: PMC6694394 DOI: 10.1001/jamanetworkopen.2019.9126] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/24/2019] [Indexed: 12/14/2022] Open
Abstract
IMPORTANCE Surgery followed by adjuvant chemotherapy or chemoradiation is widely used to treat resectable pancreatic cancer. Although studies suggest initiation of adjuvant therapy within 12 weeks of surgery, there is no clear time interval associated with better survival. OBJECTIVE To evaluate the ideal timing of adjuvant therapy for patients with stage I to II resected pancreatic cancer. DESIGN, SETTING, AND PARTICIPANTS This cohort study included 7548 patients with stage I to II resected pancreatic cancer (5453 with adjuvant therapy; 2095 without adjuvant therapy) from the National Cancer Database from 2004 to 2015. Data were collected from January 2014 to December 2015 and analyzed from December 2018 to May 2019. EXPOSURES Adjuvant chemotherapy or chemoradiation at various time intervals. MAIN OUTCOMES AND MEASURES Overall survival (OS). RESULTS A total of 7548 patients (3770 male [49.9%]; median [interquartile range] age, 67 [59-74] years) were identified from the National Cancer Database. Among 5453 patients with adjuvant therapy, a Cox model with restricted cubic splines identified the lowest mortality risk when adjuvant therapy was started 28 to 59 days after surgery. Patients were divided into early (n = 269, adjuvant therapy initiated within <28 days), reference (n = 3048, adjuvant therapy initiated within 28-59 days), and late (n = 2136, adjuvant therapy initiated after >59 days) interval cohorts. Median (interquartile range) overall follow-up was 38.6 (24.6-62.0) months. Compared with the reference interval cohort on multivariable analysis, both the early cohort (hazard ratio, 1.17; 95% CI, 1.02-1.35; P = .03) and the late cohort (hazard ratio, 1.09; 95% CI, 1.02-1.17; P = .008) were associated with worse mortality. Similarly, the reference interval cohort had improved OS compared with the early cohort in 268 propensity-matched pairs (2-year OS, 52.5% [95% CI, 46.7%-59.0%] vs 45.1% [95% CI, 39.5%-51.6%]; P = .02) and compared with the late cohort in 2042 propensity-matched pairs (2-year OS, 51.3% [95% CI, 49.1%-53.6%] vs 45.4% [95% CI, 43.3%-47.7%]; P = .01). Patients who received adjuvant therapy more than 12 weeks after surgery (n = 683) had improved OS compared with surgery alone in both multivariable analysis (hazard ratio, 0.75; 95% CI, 0.66-0.85; P < .001) and 655 propensity-matched pairs (2-year OS, 47.2% [95% CI, 43.5%-51.3%] vs 38.0% [95% CI, 34.4%-42.0%]; P < .001). CONCLUSIONS AND RELEVANCE Patients with stage I to II pancreatic cancer who commenced adjuvant therapy within 28 to 59 days after primary surgical resection had improved survival outcomes compared with those with adjuvant therapy before 28 days or after 59 days. Patients who recovered slowly from surgery still benefited from delayed adjuvant therapy initiated more than 12 weeks after surgery compared with patients who underwent surgery only.
Collapse
Affiliation(s)
- Sung Jun Ma
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | - Joseph A. Miccio
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Austin J. Iovoli
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo-State University of New York (SUNY), Buffalo
| | - Gregory M. Hermann
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Anurag K. Singh
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
1856
|
Ma SJ, Iovoli AJ, Hermann GM, Prezzano KM, Singh AK. Duration of chemotherapy prior to chemoradiation affects survival outcomes for resected stage I-II or unresected stage III pancreatic cancer. Cancer Med 2019; 8:4110-4123. [PMID: 31183965 PMCID: PMC6675727 DOI: 10.1002/cam4.2326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND For resected early stage pancreatic cancer, RTOG 9704 evaluated the outcome of 3 weeks of postoperative chemotherapy (C) followed by chemoradiation (CRT) and further C. For unresectable locally advanced pancreatic cancer, a recent literature review of prospective studies showed that the duration of induction C prior to CRT can impact survival. However, the ideal duration of C prior to CRT remains unclear for these patient cohorts. This National Cancer Database (NCDB) study was performed to compare the outcome of various durations of C prior to CRT. METHODS The NCDB was queried for resected primary stage I-II, cT1-3N0-1M0, and unresected stage III, cT4N0-1M0 pancreatic adenocarcinoma treated with C + CRT (2004-2015). Cohorts I-II and III included stage I-II and stage III cases, respectively. Patients were stratified by short (short C) and long duration (long C) of chemotherapy based on their median durations. Baseline patient, tumor, and treatment characteristics were examined. The primary endpoint was overall survival (OS). Kaplan-Meier analysis, multivariable Cox proportional hazards method, and propensity score matching were used. RESULTS Among 1577 patients, cohort I-II had 839 patients and cohort III had 738 patients. The longer duration of chemotherapy prior to CRT showed improved OS in the multivariate analysis in both cohort I-II (hazards ratio [HR] 0.72, P < 0.001) and cohort III (HR 0.83, P = 0.03). Using 1:1 propensity score matching, 610 patients for cohort I-II and 542 patients for cohort III were matched. After matching, long C remained statistically significant for improved OS compared with short C in both cohort I-II (median OS 26.1 vs 21.9 months; P = 0.003) and cohort III (median OS 16.7 vs 14.2; P = 0.02). CONCLUSION Our NCDB study using propensity score-matched analysis showed a survival benefit for using the longer duration of chemotherapy compared to the shorter duration for both resected stage I-II and unresected stage III pancreatic cancer.
Collapse
Affiliation(s)
- Sung J. Ma
- Department of Radiation MedicineRoswell Park Comprehensive Cancer CenterBuffaloNY
| | - Austin J. Iovoli
- Jacobs School of Medicine and Biomedical SciencesUniversity at Buffalo, The State University of New YorkBuffaloNY
| | - Gregory M. Hermann
- Department of Radiation MedicineRoswell Park Comprehensive Cancer CenterBuffaloNY
| | - Kavitha M. Prezzano
- Department of Radiation MedicineRoswell Park Comprehensive Cancer CenterBuffaloNY
| | - Anurag K. Singh
- Department of Radiation MedicineRoswell Park Comprehensive Cancer CenterBuffaloNY
| |
Collapse
|
1857
|
Janssen QP, Buettner S, Suker M, Beumer BR, Addeo P, Bachellier P, Bahary N, Bekaii-Saab T, Bali MA, Besselink MG, Boone BA, Chau I, Clarke S, Dillhoff M, El-Rayes BF, Frakes JM, Grose D, Hosein PJ, Jamieson NB, Javed AA, Khan K, Kim KP, Kim SC, Kim SS, Ko AH, Lacy J, Margonis GA, McCarter MD, McKay CJ, Mellon EA, Moorcraft SY, Okada KI, Paniccia A, Parikh PJ, Peters NA, Rabl H, Samra J, Tinchon C, van Tienhoven G, van Veldhuisen E, Wang-Gillam A, Weiss MJ, Wilmink JW, Yamaue H, Homs MYV, van Eijck CHJ, Katz MHG, Groot Koerkamp B. Neoadjuvant FOLFIRINOX in Patients With Borderline Resectable Pancreatic Cancer: A Systematic Review and Patient-Level Meta-Analysis. J Natl Cancer Inst 2019; 111:782-794. [PMID: 31086963 PMCID: PMC6695305 DOI: 10.1093/jnci/djz073] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 12/19/2018] [Accepted: 04/22/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND FOLFIRINOX is a standard treatment for metastatic pancreatic cancer patients. The effectiveness of neoadjuvant FOLFIRINOX in patients with borderline resectable pancreatic cancer (BRPC) remains debated. METHODS We performed a systematic review and patient-level meta-analysis on neoadjuvant FOLFIRINOX in patients with BRPC. Studies with BRPC patients who received FOLFIRINOX as first-line neoadjuvant treatment were included. The primary endpoint was overall survival (OS), and secondary endpoints were progression-free survival, resection rate, R0 resection rate, and grade III-IV adverse events. Patient-level survival outcomes were obtained from authors of the included studies and analyzed using the Kaplan-Meier method. RESULTS We included 24 studies (8 prospective, 16 retrospective), comprising 313 (38.1%) BRPC patients treated with FOLFIRINOX. Most studies (n = 20) presented intention-to-treat results. The median number of administered neoadjuvant FOLFIRINOX cycles ranged from 4 to 9. The resection rate was 67.8% (95% confidence interval [CI] = 60.1% to 74.6%), and the R0-resection rate was 83.9% (95% CI = 76.8% to 89.1%). The median OS varied from 11.0 to 34.2 months across studies. Patient-level survival data were obtained for 20 studies representing 283 BRPC patients. The patient-level median OS was 22.2 months (95% CI = 18.8 to 25.6 months), and patient-level median progression-free survival was 18.0 months (95% CI = 14.5 to 21.5 months). Pooled event rates for grade III-IV adverse events were highest for neutropenia (17.5 per 100 patients, 95% CI = 10.3% to 28.3%), diarrhea (11.1 per 100 patients, 95% CI = 8.6 to 14.3), and fatigue (10.8 per 100 patients, 95% CI = 8.1 to 14.2). No deaths were attributed to FOLFIRINOX. CONCLUSIONS This patient-level meta-analysis of BRPC patients treated with neoadjuvant FOLFIRINOX showed a favorable median OS, resection rate, and R0-resection rate. These results need to be assessed in a randomized trial.
Collapse
Affiliation(s)
| | - Stefan Buettner
- See the Notes section for the full list of authors’ affiliations
| | - Mustafa Suker
- See the Notes section for the full list of authors’ affiliations
| | - Berend R Beumer
- See the Notes section for the full list of authors’ affiliations
| | - Pietro Addeo
- See the Notes section for the full list of authors’ affiliations
| | | | - Nathan Bahary
- See the Notes section for the full list of authors’ affiliations
| | | | - Maria A Bali
- See the Notes section for the full list of authors’ affiliations
| | - Marc G Besselink
- See the Notes section for the full list of authors’ affiliations
| | - Brian A Boone
- See the Notes section for the full list of authors’ affiliations
| | - Ian Chau
- See the Notes section for the full list of authors’ affiliations
| | - Stephen Clarke
- See the Notes section for the full list of authors’ affiliations
| | - Mary Dillhoff
- See the Notes section for the full list of authors’ affiliations
| | | | - Jessica M Frakes
- See the Notes section for the full list of authors’ affiliations
| | - Derek Grose
- See the Notes section for the full list of authors’ affiliations
| | - Peter J Hosein
- See the Notes section for the full list of authors’ affiliations
| | - Nigel B Jamieson
- See the Notes section for the full list of authors’ affiliations
| | - Ammar A Javed
- See the Notes section for the full list of authors’ affiliations
| | - Khurum Khan
- See the Notes section for the full list of authors’ affiliations
| | - Kyu-Pyo Kim
- See the Notes section for the full list of authors’ affiliations
| | - Song Cheol Kim
- See the Notes section for the full list of authors’ affiliations
| | - Sunhee S Kim
- See the Notes section for the full list of authors’ affiliations
| | - Andrew H Ko
- See the Notes section for the full list of authors’ affiliations
| | - Jill Lacy
- See the Notes section for the full list of authors’ affiliations
| | | | | | - Colin J McKay
- See the Notes section for the full list of authors’ affiliations
| | - Eric A Mellon
- See the Notes section for the full list of authors’ affiliations
| | | | - Ken-Ichi Okada
- See the Notes section for the full list of authors’ affiliations
| | | | - Parag J Parikh
- See the Notes section for the full list of authors’ affiliations
| | - Niek A Peters
- See the Notes section for the full list of authors’ affiliations
| | - Hans Rabl
- See the Notes section for the full list of authors’ affiliations
| | - Jaswinder Samra
- See the Notes section for the full list of authors’ affiliations
| | | | | | | | | | - Matthew J Weiss
- See the Notes section for the full list of authors’ affiliations
| | | | - Hiroki Yamaue
- See the Notes section for the full list of authors’ affiliations
| | | | | | - Matthew H G Katz
- See the Notes section for the full list of authors’ affiliations
| | | |
Collapse
|
1858
|
Khorana AA, McKernin SE, Katz MH. Potentially Curable Pancreatic Adenocarcinoma: ASCO Clinical Practice Guideline Update Summary. J Oncol Pract 2019. [DOI: 10.1200/jop.19.00253] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
1859
|
König A, Ellenrieder V, Nitschmann S. Neuer Meilenstein in der adjuvanten Therapie des Pankreaskarzinoms. Internist (Berl) 2019; 60:881-884. [DOI: 10.1007/s00108-019-0628-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
1860
|
Affiliation(s)
- Andrew H Ko
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco.,UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco
| | - Eric K Nakakura
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco.,Division of General Surgery, Section of Surgical Oncology and Hepatopancreaticobiliary Surgery, Department of Surgery, University of California, San Francisco
| |
Collapse
|
1861
|
Stødkilde‐Jørgensen H, Laustsen C, Hansen ESS, Schulte R, Ardenkjaer‐Larsen JH, Comment A, Frøkiær J, Ringgaard S, Bertelsen LB, Ladekarl M, Weber B. Pilot Study Experiences With Hyperpolarized [1‐
13
C]pyruvate MRI in Pancreatic Cancer Patients. J Magn Reson Imaging 2019; 51:961-963. [DOI: 10.1002/jmri.26888] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 12/16/2022] Open
Affiliation(s)
| | - Christoffer Laustsen
- MR Research Centre, Department of Clinical MedicineAarhus University Aarhus Denmark
| | | | | | - Jan Henrik Ardenkjaer‐Larsen
- Department of Health TechnologyTechnical University of Denmark Kgs Lyngby Denmark
- General Electric Healthcare Brøndby Denmark
| | | | - Jørgen Frøkiær
- Department of Clinical MedicineAarhus University Aarhus Denmark
| | - Steffen Ringgaard
- MR Research Centre, Department of Clinical MedicineAarhus University Aarhus Denmark
| | | | - Morten Ladekarl
- Department of OncologyClinical Cancer Research Center, Aalborg University Hospital Denmark
| | - Britta Weber
- Danish Centre for Particle TherapyAarhus University Hospital Denmark
- Department of OncologyAarhus University Hospital Denmark
| |
Collapse
|
1862
|
Del Chiaro M, Schulick RD. It is time for a biological definition of locally advanced pancreatic cancer. Br J Surg 2019; 106:1257. [DOI: 10.1002/bjs.11249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 05/03/2019] [Indexed: 11/08/2022]
Affiliation(s)
- M Del Chiaro
- Department of Surgery, Division of Surgical Oncology, University of Colorado, Anschutz Medical Campus, Denver, Colorado, USA
| | - R D Schulick
- Department of Surgery, Division of Surgical Oncology, University of Colorado, Anschutz Medical Campus, Denver, Colorado, USA
| |
Collapse
|
1863
|
Karamitopoulou E, Gloor B. Clinical Scenarios Emerging from Combined Immunophenotypic, Molecular and Morphologic Analysis of Pancreatic Cancer: The Good, the Bad and the Ugly Scenario. Cancers (Basel) 2019; 11:E968. [PMID: 31295960 PMCID: PMC6678850 DOI: 10.3390/cancers11070968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with increasing incidence and dismal prognosis. The composition of the immune cell infiltrates in the tumor microenvironment (TME) and the dynamic interplay between cancer- and immune cells can influence and/or be influenced by tumor-intrinsic characteristics like molecular profiles and tumor cell morphology. The combined analyses of pancreatic cancer by using morphologic, genetic, and immunologic features help us understand the significant heterogeneity of the TME and recognize the different mechanisms of immune evasion. Moreover, this information may lead to the identification of novel biomarkers for more precise patient stratification and therapy guidance.
Collapse
Affiliation(s)
- Eva Karamitopoulou
- Pancreatic Cancer Research Group, Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland.
| | - Beat Gloor
- Department of Visceral Surgery, Insel University Hospital, University of Bern, Freiburgstrasse 18, CH-3010 Bern, Switzerland.
| |
Collapse
|
1864
|
Paredes AZ, Hyer JM, Tsilimigras DI, Mehta R, Sahara K, White S, Dillhoff ME, Ejaz A, Cloyd JM, Pawlik TM. Hospice utilization among Medicare beneficiaries dying from pancreatic cancer. J Surg Oncol 2019; 120:624-631. [PMID: 31290170 DOI: 10.1002/jso.25623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 06/21/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Use of hospice services among patients with pancreatic cancer following pancreatic resection remains unknown. METHODS Patients with pancreatic cancer who underwent resection were identified in the Medicare Standard Analytic Files. Outcomes included overall hospice use, early hospice enrollment (≥4 weeks before death), late hospice enrollment (initiation within 3 days of death), and Medicare expenditures. RESULTS Among the 4369 deceased individuals, three-fourths of patients (n = 3252, 74.4%) used hospice at the time of death. Patients who did not use hospice were more likely to be male, have a complication on index admission and receive life sustaining treatments on subsequent admissions (P < .05). Only one-third (32.2%) of patients initiated hospice services early. Medicare expenditure during the last month of life was $10 000 lower among patients who initialized hospice services at least 1 month before death versus within 3 days of death (late: $10 581 [$5454-$17 200], early: $221 [$46-$733]; P < .001) CONCLUSION: While three-fourths of patients utilized hospice services after pancreatic resection, only one-third of patients initiated hospice services at least one-month before death. Late hospice use was associated with higher Medicare expenditures during the last month of life. Further research is needed to understand barriers to early hospice utilization.
Collapse
Affiliation(s)
- Anghela Z Paredes
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - J Madison Hyer
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Diamantis I Tsilimigras
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Rittal Mehta
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Kota Sahara
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Susan White
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Mary E Dillhoff
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Aslam Ejaz
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Jordan M Cloyd
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Timothy M Pawlik
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio.,Division of Health Information Management and Systems, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
1865
|
|
1866
|
Fernandes GDS, Pereira A. Development of new therapies for metastatic pancreatic cancer: are they better than FOLFIRINOX? ESMO Open 2019; 4:e000537. [PMID: 31354965 PMCID: PMC6615872 DOI: 10.1136/esmoopen-2019-000537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 01/05/2023] Open
Affiliation(s)
| | - Allan Pereira
- Division of Medical Oncology, Hospital Sirio-Libanes, Brasília, Brazil
| |
Collapse
|
1867
|
Survival of patients with borderline resectable pancreatic cancer who received neoadjuvant therapy and surgery. Surgery 2019; 166:277-285. [PMID: 31272811 DOI: 10.1016/j.surg.2019.05.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/30/2019] [Accepted: 05/13/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND It is difficult to successfully deliver multimodality therapy to patients with operable pancreatic cancer. Data on the natural history of such efforts are necessary for physicians to guide shared decision-making with patients and families. We report the survival of consecutive patients with borderline resectable pancreatic cancer who received neoadjuvant therapy before surgery. METHODS Data regarding demographics, neoadjuvant therapy, surgery, pathology, and survival duration were abstracted on consecutive patients with borderline resectable pancreatic cancer diagnosed between 2009 and 2017 and not treated on available clinical trials. Borderline resectable pancreatic cancer was defined based on ≥1 of the following: local tumor anatomy, pretreatment serum carbohydrate antigen 19-9 >2,000 U/mL, and the presence of radiographic lesions indeterminate for metastases. RESULTS Neoadjuvant therapy was delivered to 185 patients with borderline resectable pancreatic cancer who were not enrolled in competing clinical trials; 13 (7%) patients received chemoradiation, 12 (7%) received chemotherapy, and 160 (86%) received both. Of the 185 patients, 115 (62%) completed all intended neoadjuvant therapy and surgery; 81 (70%) of 115 underwent pancreaticoduodenectomy; and vascular reconstruction was performed in 51 (44%). A margin negative resection was achieved in 111 (97%) of 115 patients, and 83 (72%) were node negative. Median overall survival for all 185 patients was 20 months; 31 months for the 115 patients who completed all neoadjuvant therapy and surgery as compared to 13 months for the 70 patients who were not resected (P < .0001). CONCLUSION After neoadjuvant therapy, surgical resection was performed in 62% of patients with borderline resectable pancreatic cancer. Those who normalized preoperative serum carbohydrate antigen 19-9 and had node negative pathology achieved the longest survival. To further improve median survival for all patients, we are incorporating adaptive approaches to neoadjuvant therapy sequencing based on objective assessments of response.
Collapse
|
1868
|
Asano D, Nara S, Kishi Y, Esaki M, Hiraoka N, Tanabe M, Shimada K. A Single-Institution Validation Study of Lymph Node Staging By the AJCC 8th Edition for Patients with Pancreatic Head Cancer: A Proposal to Subdivide the N2 Category. Ann Surg Oncol 2019; 26:2112-2120. [PMID: 31037440 DOI: 10.1245/s10434-019-07390-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND According to the revised staging of the American Joint Committee on Cancer, 8th edition (AJCC8), the N category in pancreatic ductal adenocarcinoma is classified as N0 (0), N1 (1-3), and N2 (≥ 4) based on the number of metastatic lymph nodes (LNs). This study aimed to validate this classification and analyze cutoff values of metastatic LN numbers. METHODS Patients with pancreatic head ductal adenocarcinoma who underwent pancreaticoduodenectomy at our institution between 2005 and 2016 without preoperative therapy were retrospectively analyzed. The patients were staged by AJCC8, and prognostic analyses were performed. The best cutoff value for the metastatic LN number was determined by the minimum P value approach. RESULTS In 228 of 309 patients, LN metastases were found (median number of examined LNs, 41). The median survival time (MST) was 56 months in the N0 group, 34 months in the N1 group, and 20 months in the N2 group (N0 vs N1: P = 0.023; N1 vs N2: P < 0.001). The best cutoff number of metastatic LNs was 4 for patients with LN metastases and 7 for patients with N2 disease. The MST for patients with four to six positive nodes (N2a) was significantly longer than for those with seven or more positive nodes (N2b) (24.0 vs 19.1 months: P = 0.012). For N2b patients, conventional adjuvant chemotherapy did not show survival benefits (P = 0.133), and overall survival did not differ significantly from that for patients with para-aortic LN metastasis (P = 0.562). CONCLUSION The N staging of AJCC8 was valid. Clinicians should regard N2b as similar to distant LN metastasis, and more intensive adjuvant therapy may be indicated for this group.
Collapse
Affiliation(s)
- Daisuke Asano
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Nara
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan.
| | - Yoji Kishi
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Minoru Esaki
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Nobuyoshi Hiraoka
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuaki Shimada
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| |
Collapse
|
1869
|
Doussot A, Bouvier A, Santucci N, Lequeu JB, Cheynel N, Ortega-Deballon P, Rat P, Facy O. Pancreatic ductal adenocarcinoma and paraaortic lymph nodes metastases: The accuracy of intraoperative frozen section. Pancreatology 2019; 19:710-715. [PMID: 31174978 DOI: 10.1016/j.pan.2019.05.465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/11/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatoduodenectomy for pancreatic ductal adenocarcinoma (PDAC) with paraaortic lymph nodes metastases (PALN +) is associated with poor survival. Still, there are no current guidelines advocating systematic detection of PALN+. METHODS All consecutive patients who underwent surgical exploration/resection with concurrent paraaortic (group 16) lymphadenectomy for PDAC between 2009 and 2016 were considered for inclusion. Resection was systematically aborted in case of intraoperative PALN + detection. Diagnostic performance of preoperative imaging upon blind review and intraoperative PALN dissection with frozen section (FS) for PALN detection were evaluated. Additionally, the prognostic significance of PALN + on overall survival (OS) was analyzed. RESULTS Over the study period, among 129 patients undergoing surgery for PDAC, 113 had intraoperative PALN dissection with FS analysis. Median number of resected PALN was 3 (range, 1-15). Overall, PALN+ was found in 19 patients (16.8%). Upon blind review, preoperative imaging performed poorly for PALN + detection with a low agreement between imaging and final pathology (Kappa-Cohen index<0.2). In contrast, PALN FS showed high detection performances and strong agreement with final pathology (Kappa-Cohen index = 0.783, 95%CI 0.779-0.867, p < 0.001). Regarding survival outcomes, there was no difference between patients with PALN+ and patients not resected in the setting of liver metastases or locally unresectable disease found at exploration (p = 0.708). CONCLUSIONS Before PD for PDAC, intraoperative PALN dissection and FS analysis yields accurate PALN assessment and allows appropriate patient selection. This should be routinely performed and aborting resection should be strongly considered in case of PALN+.
Collapse
Affiliation(s)
- Alexandre Doussot
- Department of Digestive Surgical Oncology, University Hospital of Dijon, France; Department of Digestive Surgical Oncology - Liver Transplantation Unit, University Hospital of Besançon, France.
| | - Aurélie Bouvier
- Department of Digestive Surgical Oncology, University Hospital of Dijon, France
| | - Nicolas Santucci
- Department of Digestive Surgical Oncology, University Hospital of Dijon, France
| | | | - Nicolas Cheynel
- Department of Digestive Surgical Oncology, University Hospital of Dijon, France
| | | | - Patrick Rat
- Department of Digestive Surgical Oncology, University Hospital of Dijon, France
| | - Olivier Facy
- Department of Digestive Surgical Oncology, University Hospital of Dijon, France
| |
Collapse
|
1870
|
Voss N, Izbicki JR, Nentwich MF. Oligometastases in pancreatic cancer (Synchronous resections of hepatic oligometastatic pancreatic cancer: Disputing a principle in a time of safe pancreatic operations in a retrospective multicenter analysis). Ann Gastroenterol Surg 2019; 3:373-377. [PMID: 31346576 PMCID: PMC6635688 DOI: 10.1002/ags3.12255] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 12/25/2022] Open
Abstract
The aim of the present review was to analyze the current data on surgery of synchronous liver metastases in pancreatic ductal adenocarcinoma (PDAC) in curative intent. A review of the literature was carried out to identify the current international concepts regarding surgery of liver metastases of PDAC and, furthermore, we addressed the current challenges of resection of liver metastases of PDAC. Resection of liver metastases in PDAC may provide survival benefit without compromising safety and quality of life in a highly selected group of patients.
Collapse
Affiliation(s)
- Nina Voss
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Michael F. Nentwich
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
1871
|
Panda A, Garg I, Johnson GB, Truty MJ, Halfdanarson TR, Goenka AH. Molecular radionuclide imaging of pancreatic neoplasms. Lancet Gastroenterol Hepatol 2019; 4:559-570. [DOI: 10.1016/s2468-1253(19)30081-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/26/2019] [Accepted: 03/02/2019] [Indexed: 02/07/2023]
|
1872
|
Sakaguchi T, Valente R, Tanaka K, Satoi S, Del Chiaro M. Surgical treatment of metastatic pancreatic ductal adenocarcinoma: A review of current literature. Pancreatology 2019; 19:672-680. [PMID: 31285145 DOI: 10.1016/j.pan.2019.05.466] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/05/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND There is no international consensus concerning the role of surgical treatment of metastatic pancreatic ductal adenocarcinoma (mPDAC), but favorable prognoses can be expected for highly selected patients. METHODS A comprehensive literature search of the PubMed and Cochrane databases was conducted using combinations of keywords to 4 July 2018. Eligible studies were those reporting on patients with histologically confirmed mPDAC undergoing surgery with curative intent. We excluded case reports with fewer than five patients, insufficient descriptions of survival data, and palliative or cytoreductive surgery as well as studies that assessed para-aortic lymph node metastasis or peritoneal washing cytology. RESULTS Thirteen studies were deemed eligible, and six studies were identified from their references. The studies involved 428 patients who underwent surgical resection for liver metastases (n = 343), lung metastases (n = 57), and peritoneal dissemination (n = 28). Median overall survival (OS) in patients with synchronous liver metastases who underwent conversion surgery following favorable response to initial chemotherapy was 27 or 34 months, and peritoneum metastases was 28 months. Median OS after the initial treatment was varied from 51 to 121 months in metachronous lung metastasis and from 24 to 40 months in metachronous liver metastasis, respectively. CONCLUSION Encouraging OS was indicated in patients with synchronous mPDAC of liver and peritoneum who underwent conversion surgery. Metastasectomy for metachronous lung and liver oligometastases could be considered a practical treatment option.
Collapse
Affiliation(s)
| | - Roberto Valente
- Division of Surgery, HPB Disease Unit, Karolinska University Hospital, Stockholm, Sweden.
| | - Kimitaka Tanaka
- Department of Gastroenterological Surgery II, Hokkaido University, Faculty of Medicine, Sapporo, Japan
| | - Sohei Satoi
- Department of Surgery, Kansai Medical University, Osaka, Japan
| | - Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Denver, USA
| |
Collapse
|
1873
|
Cherkassky L, Jarnagin W. Selecting treatment sequence for patients with incidental gallbladder cancer: a neoadjuvant approach versus upfront surgery. Updates Surg 2019; 71:217-225. [PMID: 31254234 DOI: 10.1007/s13304-019-00670-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 06/22/2019] [Indexed: 02/07/2023]
Abstract
At MSKCC, over 50% of the patients presenting with gallbladder cancer have been diagnosed incidentally following elective cholecystectomy for presumed benign disease. While traditional management of incidental gallbladder cancer (IGBC) dictates re-resection with the ultimate goal of achieving cure, surgical decision-making must take into account that this malignancy is characterized by poor tumor biology with frequent distant recurrence. Since early and frequent distant recurrence is the most common cause of surgical failure, the surgical oncologist's goal should be to selectively re-resect only those patients most likely to benefit from an operation. The astute surgeon recognizes the high-risk patients who likely have micrometastatic disease at the time of diagnosis and alters the treatment sequence, delivering neoadjuvant chemotherapy. This strategy acts as a selection tool, as those progressing at distant sites during therapy are spared the morbidity and mortality of surgery and furthermore has the potential to treat micrometastatic disease. However, a chemotherapy first approach must be applied selectively since a poor response risks local progression to unresectability and a decrease in functional status that comes from the toxicities of dual agent chemotherapy that can impair surgical candidacy. To balance these risks and benefits, two other criteria for a neoadjuvant approach must be met: i) reliable identification of those patients who are at high risk of distant recurrence and who are, therefore, most likely to benefit from a systemic therapy first approach and ii) availability of effective chemotherapy options. In this review, we will outline the data and judgement we use to select a treatment sequence at our institution.
Collapse
Affiliation(s)
- Leonid Cherkassky
- Clinical Fellow in Complex General Surgical Oncology, Department of Surgical Oncology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - William Jarnagin
- Leslie Blumgart Chair in Surgery, Department of Surgical Oncology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
1874
|
Hall WA, Goodman KA. Radiation therapy for pancreatic adenocarcinoma, a treatment option that must be considered in the management of a devastating malignancy. Radiat Oncol 2019; 14:114. [PMID: 31242912 PMCID: PMC6595558 DOI: 10.1186/s13014-019-1277-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/14/2019] [Indexed: 01/05/2023] Open
Abstract
Clinical outcomes for patients with pancreatic adenocarcinoma (PAC) remain dismal. Local recurrences, proportions of margin positive surgical resections, and overall survival outcomes remain inferior in PAC than any other solid tumor. This stems from a current standard of care management approach that needs to be inspired and transformed with modern treatment techniques and novel therapeutic options. Radiation therapy has historically been a central component in the treatment of pancreatic adenocarcinoma; however, the role of radiation therapy has been called into question based on the publication of clinical trials with conflicting results. We present an overview of the rationale for radiation therapy in resectable, borderline resectable, and unresectable pancreatic adenocarcinoma. We further present a summary of emerging clinical data and future directions to improve outcomes in this devastating malignancy.
Collapse
Affiliation(s)
- William A Hall
- Department of Radiation Oncology, Medical College of Wisconsin, Wauwatosa, USA.
| | - Karyn A Goodman
- Department of Radiation Oncology, University of Colorado, Aurora, CO, USA
| |
Collapse
|
1875
|
Yang F, Jin C, Fu DL, Warshaw AL. Modified FOLFIRINOX for resected pancreatic cancer: Opportunities and challenges. World J Gastroenterol 2019; 25:2839-2845. [PMID: 31249443 PMCID: PMC6589737 DOI: 10.3748/wjg.v25.i23.2839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer death worldwide. Adjuvant chemotherapy has been developed based on the experiences made with palliative chemotherapy, and advocated to improve long-term survival of patients with this disease. However, the optimal chemotherapeutic regimen remains controversial. Recently, Conroy et al demonstrated the impressive benefits of modified FOLFIRINOX over gemcitabine alone in the multicenter Partenariat de Recherche en Oncologie Digestive 24 (PRODIGE-24) trial. The remarkable results mark a new milestone in treating resectable pancreatic cancer and have now changed the standard of care for this patient population. In this commentary, we discuss an issue of difference of tumor grade between the PRODIGE-24 trial and previous phase III trials. We also discuss potential biomarkers predicting therapeutic response to modified FOLFIRINOX. Finally, we summarize several ongoing clinical trials of replacing part of the FOLFIRINOX regimen with Xeloda/S-1/nanoliposomal irinotecan for pancreatic cancer.
Collapse
Affiliation(s)
- Feng Yang
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chen Jin
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - De-Liang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Andrew L Warshaw
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| |
Collapse
|
1876
|
Nakano Y, Kitago M, Shinoda M, Yagi H, Abe Y, Takano K, Oshima G, Takeuch A, Endo Y, Kitagawa Y. Prognostic significance of the postoperative level and recovery rate of serum albumin in patients with curatively resected pancreatic ductal adenocarcinoma. Mol Clin Oncol 2019; 11:270-278. [PMID: 31384459 DOI: 10.3892/mco.2019.1883] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 05/30/2019] [Indexed: 12/17/2022] Open
Abstract
The aim of the present study was to investigate the clinicopathological features and prognostic factors associated with pre- and postoperative serum albumin levels in patients with curatively resected pancreatic ductal adenocarcinoma (PDAC). To achieve this, the data of patients who underwent pancreatectomy for PDAC between January 1995 and March 2016 were retrospectively reviewed, and the pre- and postoperative serum albumin levels at postoperative months (POMs) 3, 6, and 12 were evaluated. The serum albumin recovery rate was also investigated. A total of 196 patients were enrolled in the present study. In the multivariate Cox regression analysis, lymph node metastasis [hazard ratio (HR): 1.65; P=0.022], serum albumin level at POM 12 (≥3.9 g/dl; HR: 0.60; P=0.017), and serum albumin recovery rate at POM 12 (≥1.00; HR: 0.60; P=0.017) were independent prognostic factors for disease-free survival. Lymph node metastasis (HR: 1.79; P=0.013) and serum albumin level at POM 12 (≥3.9 g/dl) (HR: 0.60; P=0.033) were independent prognostic factors for overall survival. These results indicated that the postoperative level and recovery rate of serum albumin are potential biomarkers for predicting the prognosis of patients with curatively resected PDAC. However, further studies are required in order to investigate the survival benefit of increasing postoperative serum albumin levels in these patients.
Collapse
Affiliation(s)
- Yutaka Nakano
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masahiro Shinoda
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yuta Abe
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kiminori Takano
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Go Oshima
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ayano Takeuch
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yutaka Endo
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
1877
|
Buscail E, Maulat C, Muscari F, Chiche L, Cordelier P, Dabernat S, Alix-Panabières C, Buscail L. Liquid Biopsy Approach for Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2019; 11:cancers11060852. [PMID: 31248203 PMCID: PMC6627808 DOI: 10.3390/cancers11060852] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/01/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Pancreatic cancer is a public health problem because of its increasing incidence, the absence of early diagnostic tools, and its aggressiveness. Despite recent progress in chemotherapy, the 5-year survival rate remains below 5%. Liquid biopsies are of particular interest from a clinical point of view because they are non-invasive biomarkers released by primary tumours and metastases, remotely reflecting disease burden. Pilot studies have been conducted in pancreatic cancer patients evaluating the detection of circulating tumour cells, cell-free circulating tumour DNA, exosomes, and tumour-educated platelets. There is heterogeneity between the methods used to isolate circulating tumour elements as well as the targets used for their identification. Performances for the diagnosis of pancreatic cancer vary depending of the technique but also the stage of the disease: 30–50% of resectable tumours are positive and 50–100% are positive in locally advanced and/or metastatic cases. A significant prognostic value is demonstrated in 50–70% of clinical studies, irrespective of the type of liquid biopsy. Large prospective studies of homogeneous cohorts of patients are lacking. One way to improve diagnostic and prognostic performances would be to use a combined technological approach for the detection of circulating tumour cells, exosomes, and DNA.
Collapse
Affiliation(s)
- Etienne Buscail
- INSERM U1035, Bordeaux University, 33000 Bordeaux, France.
- Department of Digestive Surgery, Bordeaux University Hospital, 33600 Pessac, France.
| | - Charlotte Maulat
- Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France.
- Department of Digestive Surgery, Toulouse University Hospital, 31059 Toulouse, France.
| | - Fabrice Muscari
- Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France.
- Department of Digestive Surgery, Toulouse University Hospital, 31059 Toulouse, France.
| | - Laurence Chiche
- INSERM U1035, Bordeaux University, 33000 Bordeaux, France.
- Department of Digestive Surgery, Bordeaux University Hospital, 33600 Pessac, France.
| | - Pierre Cordelier
- Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France.
| | | | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), Montpellier Hospital and University of Montpellier, 34295 Montpellier, France.
| | - Louis Buscail
- Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France.
- Department of Gastroenterology and Pancreatology, Toulouse University Hospital, 31059 Toulouse, France.
| |
Collapse
|
1878
|
Goetze RG, Buchholz SM, Ou N, Zhang Q, Patil S, Schirmer M, Singh SK, Ellenrieder V, Hessmann E, Lu QB, Neesse A. Preclinical Evaluation of 1,2-Diamino-4,5-Dibromobenzene in Genetically Engineered Mouse Models of Pancreatic Cancer. Cells 2019; 8:cells8060563. [PMID: 31181844 PMCID: PMC6627568 DOI: 10.3390/cells8060563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/04/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is highly resistant to standard chemo- and radiotherapy. Recently, a new class of non-platinum-based halogenated molecules (called FMD compounds) was discovered that selectively kills cancer cells. Here, we investigate the potential of 1,2-Diamino-4,5-dibromobenzene (2Br-DAB) in combination with standard chemotherapy and radiotherapy in murine and human PDAC. Methods: Cell viability and colony formation was performed in human (Panc1, BxPC3, PaTu8988t, MiaPaCa) and three murine LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre (KPC) pancreatic cancer cell lines. In vivo, preclinical experiments were conducted in LSL-KrasG12D/+;p48-Cre (KC) and KPC mice using 2Br-DAB (7 mg/kg, i.p.), +/- radiation (10 × 1.8 Gy), gemcitabine (100 mg/kg, i.p.), or a combination. Tumor growth and therapeutic response were assessed by high-resolution ultrasound and immunohistochemistry. Results: 2Br-DAB significantly reduced cell viability in human and murine pancreatic cancer cell lines in a dose-dependent manner. In particular, colony formation in human Panc1 cells was significantly decreased upon 25 µM 2Br-DAB + radiation treatment compared with vehicle control (p = 0.03). In vivo, 2Br-DAB reduced tumor frequency in KC mice. In the KPC model, 2Br-DAB or gemcitabine monotherapy had comparable therapeutic effects. Furthermore, the combination of gemcitabine and 2Br-DAB or 2Br-DAB and 18 Gy irradiation showed additional antineoplastic effects. Conclusions: 2Br-DAB is effective in killing pancreatic cancer cells in vitro. 2Br-DAB was not toxic in vivo, and additional antineoplastic effects were observed in combination with irradiation.
Collapse
Affiliation(s)
- Robert G Goetze
- Department of Gastroenterology and Gastrointestinal Oncology, University Medicine Goettingen, 37075 Goettingen, Germany.
| | - Soeren M Buchholz
- Department of Gastroenterology and Gastrointestinal Oncology, University Medicine Goettingen, 37075 Goettingen, Germany.
| | - Ning Ou
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON N2L 3G1, Canada.
| | - Qinrong Zhang
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON N2L 3G1, Canada.
| | - Shilpa Patil
- Department of Gastroenterology and Gastrointestinal Oncology, University Medicine Goettingen, 37075 Goettingen, Germany.
| | - Markus Schirmer
- Department of Radiotherapy and Radiation Oncology, University Medicine Goettingen, 37075 Goettingen, Germany.
| | - Shiv K Singh
- Department of Gastroenterology and Gastrointestinal Oncology, University Medicine Goettingen, 37075 Goettingen, Germany.
| | - Volker Ellenrieder
- Department of Gastroenterology and Gastrointestinal Oncology, University Medicine Goettingen, 37075 Goettingen, Germany.
| | - Elisabeth Hessmann
- Department of Gastroenterology and Gastrointestinal Oncology, University Medicine Goettingen, 37075 Goettingen, Germany.
| | - Qing-Bin Lu
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON N2L 3G1, Canada.
| | - Albrecht Neesse
- Department of Gastroenterology and Gastrointestinal Oncology, University Medicine Goettingen, 37075 Goettingen, Germany.
| |
Collapse
|
1879
|
Preclinical Modelling of PDA: Is Organoid the New Black? Int J Mol Sci 2019; 20:ijms20112766. [PMID: 31195689 PMCID: PMC6600483 DOI: 10.3390/ijms20112766] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a malignancy of the exocrine pancreas with the worst prognosis among all solid tumours, and soon to become the second leading cause of cancer-related deaths. A more comprehensive understanding of the molecular mechanisms underlying this disease is crucial to the development of diagnostic tools as well as to the identification of more effective therapies. High-frequency mutations in PDA occur in “undruggable” genes, and molecular subtyping based on bulk transcriptome analysis does not yet nominate valid therapeutic intervention strategies. Genome-wide sequencing studies have also demonstrated a considerable intra- and inter-patient’s genetic heterogeneity, which further complicate this dire scenario. More than in other malignancies, functionalization of the PDA genome and preclinical modelling at the individual patient level appear necessary to substantially improve survival rates for pancreatic cancer patients. Traditional human PDA models, including monolayer cell cultures and patient-derived xenografts, have certainly led to valuable biological insights in the past years. However, those model systems suffer from several limitations that have contributed to the lack of concordance between preclinical and clinical studies for PDA. Pancreatic ductal organoids have recently emerged as a reliable culture system to establish models from both normal and neoplastic pancreatic tissues. Pancreatic organoid cultures can be efficiently generated from small tissue biopsies, which opens up the possibility of longitudinal studies in individual patients. A proof-of-concept study has demonstrated that patient-derived PDA organoids are able to predict responses to conventional chemotherapy. The use of this three-dimensional culture system has already improved our understanding of PDA biology and promises to implement precision oncology by enabling the alignment of preclinical and clinical platforms to guide therapeutic intervention in PDA.
Collapse
|
1880
|
Overexpression of PODXL/ITGB1 and BCL7B/ITGB1 accurately predicts unfavorable prognosis compared to the TNM staging system in postoperative pancreatic cancer patients. PLoS One 2019; 14:e0217920. [PMID: 31166991 PMCID: PMC6550449 DOI: 10.1371/journal.pone.0217920] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/21/2019] [Indexed: 12/26/2022] Open
Abstract
We previously reported that overexpression of PODXL, BCL7B, and ARHGEF4 in pancreatic cancer tissue is correlated with pancreatic cancer-related survival. The aim of this study was to investigate the use of PODXL, BCL7B, ARHGEF4, and the integrin family member ITGB1 as useful markers for the prognosis of postoperative pancreatic cancer patients in comparison with tumor size and the tumor node metastasis (TNM) staging system. Immunohistochemistry was performed using an anti-ITGB1 antibody on 102 samples of pancreatic cancer tissue surgically resected at the University of Kochi Medical School Hospital and the Matsuyama Shimin Hospital. Univariate Cox proportional hazards regression analysis showed that TNM stage and overexpression of PODXL, BCL7B, and ITGB1 were correlated with postoperative survival. However, tumor size was not significantly associated with postoperative prognosis of pancreatic cancer compared to these features. Multivariate Cox proportional hazards regression analysis showed that the overexpression of both PODXL and ITGB1 and overexpression of both BCL7B and ITGB1 increased the hazard ratio (6.27, 95% confidence interval [CI] 2.58-15.21; and 3.93, 95% CI 1.74-8.91, respectively) compared to that of TNM stage (IIA and IIB vs. III and IV; 3.05, 95% CI 1.25-7.42). These results imply that the combination of PODXL with ITGB1 and the combination of BCL7B with ITGB1 accurately predicted the postoperative outcomes of pancreatic cancer patients, and they were superior compared to the TNM staging system. The combination of PODXL with ITGB1 would be particularly useful, as it was the most highly correlated with postoperative outcomes. Importantly, the present results are useful to determine which adjuvant therapy should be selected.
Collapse
|
1881
|
Borazanci E, Sckolnik S, Amini A. Neo-adjuvant therapy for pancreatic cancer: hope for the future. Expert Rev Gastroenterol Hepatol 2019; 13:579-589. [PMID: 30979348 DOI: 10.1080/17474124.2019.1607294] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In 2018, pancreatic ductal adenocarcinoma (PDAC) was the 3rd highest cause cancer related death in the United States. Worldwide estimates in 2018 indicate 458,918 cases diagnosed with 432,242 deaths. Standard therapy for decades for localized PDAC has been to pursue surgical resection for localized disease. For the individuals who are diagnosed with localized PDAC and undergo surgical resection, historical survival has been reported to be around 24 months. While recent advancements in the use of multiagent systemic therapy has allowed for greater survival benefit, adjuvant therapy does have limitations. Recently, neo-adjuvant therapy for PDAC has become more accepted in practice. Areas covered: In this review, we will discuss the current guidelines for treatment of localized PDAC, the pros and cons of neo-adjuvant versus adjuvant therapy for PDAC, the utilization of available biomarkers for the management of PDAC, and future possibilities for clinical trials. Expert commentary: Neo-adjuvant therapy for localized PDAC has tremendous promise in leading to greater survival by treating for micro-metastatic disease along with selecting for patients for better outcomes. Further work based upon molecular insights will lead to better biomarkers for treatment assessment along with improvements in treatment.
Collapse
Affiliation(s)
| | | | - Albert Amini
- c Arizona Premier Surgery , Scottsdale , AZ , USA
| |
Collapse
|
1882
|
Skau Rasmussen L, Vittrup B, Ladekarl M, Pfeiffer P, Karen Yilmaz M, Østergaard Poulsen L, Østerlind K, Palnæs Hansen C, Bau Mortensen M, Viborg Mortensen F, Sall M, Detlefsen S, Bøgsted M, Wilki Fristrup C. The effect of postoperative gemcitabine on overall survival in patients with resected pancreatic cancer: A nationwide population-based Danish register study. Acta Oncol 2019; 58:864-871. [PMID: 30905248 DOI: 10.1080/0284186x.2019.1581374] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background: Adjuvant chemotherapy following curative resection is the standard treatment for pancreatic adenocarcinoma (PC). Randomized clinical trials using gemcitabine have shown a median overall survival (mOS) of 2 years and a 5-year survival rate of 15-20%. However, the effect of gemcitabine outside these trials is less clear. We examined the effect of postoperative gemcitabine on survival in an unselected cohort of patients receiving curative resection for PC in Denmark during a five-year period. Material and methods: From 1 May 2011 to 30 April 2016, 731 patients treated with curative resection were identified in the Danish Pancreatic Cancer Database (DPCD). Thirty patients died within 10 weeks postoperatively; 78 received other regimens or preoperative chemotherapy and were excluded. Of the remaining 623 patients, the chemotherapy (CT) group (n = 409, 66%) received gemcitabine within 10 weeks after resection, whereas the non-chemotherapy (NCT) group (n = 214, 34%) did not receive CT within 10 weeks. Results: CT patients were slightly younger than NCT patients but did not otherwise differ in baseline characteristics. The CT group showed a mOS of 24 months (95% CI; 21-27) and a 5-year survival rate of 22% (95% CI; 17-27); the NCT group had a mOS of 22 months (95% CI; 16-26, p = .27) and a 5-year survival rate of 26% (95% CI; 19-34, p = .66). Most patients (415/623) had lymph node metastases. Of these patients, those in the CT group (n = 280) had significantly longer mOS [20 months (95% CI; 18-24)] than those in the NCT group (n = 135) [14 months (95% CI; 11-17)]. Conclusions: In this national Danish cohort of PC patients undergoing resection between 2011 and 2016, the survival after postoperative gemcitabine was similar to that reported in previous clinical trials. However, the survival advantage of postoperative gemcitabine was limited to patients with lymph node metastases.
Collapse
Affiliation(s)
- Louise Skau Rasmussen
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Benny Vittrup
- Department of Oncology, University Hospital Herlev, Herlev, Denmark
| | - Morten Ladekarl
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Mette Karen Yilmaz
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Laurids Østergaard Poulsen
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Kell Østerlind
- Department of Oncology, North Sealand Hospital, Hillerød, Denmark
| | - Carsten Palnæs Hansen
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michael Bau Mortensen
- Odense Pancreas Center (OPAC), Department of Surgery, Odense University Hospital, Odense, Denmark
| | | | - Mogens Sall
- Department of Surgical Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
| | - Sönke Detlefsen
- Odense Pancreas Center (OPAC), Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Martin Bøgsted
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Claus Wilki Fristrup
- Danish Pancreatic Cancer Database, Denmark
- Department of Surgery, Odense University Hospital, Odense, Denmark
| |
Collapse
|
1883
|
Hajjar AH, Eid R, Haddad FG, Kourie HR. FOLFIRINOX: a new standard of care in the adjuvant setting of resectable pancreatic adenocarcinomas. Future Oncol 2019; 15:1947-1950. [DOI: 10.2217/fon-2019-0136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Abdel Hadi Hajjar
- Hematology & Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Roland Eid
- Hematology & Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Fady Gh Haddad
- Hematology & Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Hampig Raphael Kourie
- Hematology & Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| |
Collapse
|
1884
|
Daamen LA, Groot VP, Intven MPW, Besselink MG, Busch OR, Koerkamp BG, Mohammad NH, Hermans JJ, van Laarhoven HWM, Nuyttens JJ, Wilmink JW, van Santvoort HC, Molenaar IQ, Stommel MWJ. Postoperative surveillance of pancreatic cancer patients. Eur J Surg Oncol 2019; 45:1770-1777. [PMID: 31204168 DOI: 10.1016/j.ejso.2019.05.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/21/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The aim of this study is to collect the best available evidence for diagnostic modalities, frequency, and duration of surveillance after resection for pancreatic ductal adenocarcinoma (PDAC). METHODS PDAC guidelines published after 2015 were collected. Furthermore, a systematic search of the literature on postoperative surveillance was performed in PubMed and Embase from 2000 to 2019. Articles comparing different diagnostic modalities and frequencies of postoperative surveillance in PDAC patients with regard to survival, quality of life, morbidity and cost-effectiveness were selected. RESULTS The literature search resulted in 570 articles. A total of seven guidelines and twelve original clinical studies were eventually evaluated. PDAC guidelines increasingly recommend a combination of tumor marker testing and computed tomography (CT) imaging every three to six months during the first two years after resection. These guidelines are, however, based on expert opinion and other low-level evidence. Prospective studies comparing different surveillance strategies are lacking. According to recent studies, surveillance with tumor markers and imaging at regular intervals results in the detection of PDAC recurrence before the onset of symptoms and more frequent administration of further therapy, such as chemotherapy or radiotherapy. CONCLUSION Current evidence for recurrence-focused surveillance after PDAC resection is limited and contradictory. Consequently, recommendations on surveillance are conflicting. To define the clinical merit of recurrence-focused surveillance, patients who are most likely to benefit from early detection and treatment of PDAC recurrence need to be identified. To this purpose, well-designed prospective studies are needed, accounting for both economical and psychosocial implications of surveillance.
Collapse
Affiliation(s)
- L A Daamen
- Dept. of Surgery, UMC Utrecht Cancer Center, Utrecht, the Netherlands; Dept. of Radiation Oncology, UMC Utrecht Cancer Center, Utrecht, the Netherlands.
| | - V P Groot
- Dept. of Surgery, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - M P W Intven
- Dept. of Radiation Oncology, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - M G Besselink
- Dept. of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - O R Busch
- Dept. of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | | | - N Haj Mohammad
- Dept. of Medical Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - J J Hermans
- Dept. of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - H W M van Laarhoven
- Dept. of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - J J Nuyttens
- Dept. of Radiation Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - J W Wilmink
- Dept. of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - H C van Santvoort
- Dept. of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands
| | - I Q Molenaar
- Dept. of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, the Netherlands
| | - M W J Stommel
- Dept. of Surgery, Radboud University Medical Center, Nijmegen, the Netherlands.
| | | |
Collapse
|
1885
|
FOLFIRINOX nach Resektion von Pankreaskarzinomen setzt neue Maßstäbe – Systemtherapie als Impulsgeber für eine Lokaltherapie beim resektablen Pankreaskarzinom. Strahlenther Onkol 2019; 195:570-572. [DOI: 10.1007/s00066-019-01449-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
1886
|
Groot VP, Mosier S, Javed AA, Teinor JA, Gemenetzis G, Ding D, Haley LM, Yu J, Burkhart RA, Hasanain A, Debeljak M, Kamiyama H, Narang A, Laheru DA, Zheng L, Lin MT, Gocke CD, Fishman EK, Hruban RH, Goggins MG, Molenaar IQ, Cameron JL, Weiss MJ, Velculescu VE, He J, Wolfgang CL, Eshleman JR. Circulating Tumor DNA as a Clinical Test in Resected Pancreatic Cancer. Clin Cancer Res 2019; 25:4973-4984. [PMID: 31142500 DOI: 10.1158/1078-0432.ccr-19-0197] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/15/2019] [Accepted: 05/23/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE In research settings, circulating tumor DNA (ctDNA) shows promise as a tumor-specific biomarker for pancreatic ductal adenocarcinoma (PDAC). This study aims to perform analytical and clinical validation of a KRAS ctDNA assay in a Clinical Laboratory Improvement Amendments (CLIA) and College of American Pathology-certified clinical laboratory. EXPERIMENTAL DESIGN Digital-droplet PCR was used to detect the major PDAC-associated somatic KRAS mutations (G12D, G12V, G12R, and Q61H) in liquid biopsies. For clinical validation, 290 preoperative and longitudinal postoperative plasma samples were collected from 59 patients with PDAC. The utility of ctDNA status to predict PDAC recurrence during follow-up was assessed. RESULTS ctDNA was detected preoperatively in 29 (49%) patients and was an independent predictor of decreased recurrence-free survival (RFS) and overall survival (OS). Patients who had neoadjuvant chemotherapy were less likely to have preoperative ctDNA than were chemo-naïve patients (21% vs. 69%; P < 0.001). ctDNA levels dropped significantly after tumor resection. Persistence of ctDNA in the immediate postoperative period was associated with a high rate of recurrence and poor median RFS (5 months). ctDNA detected during follow-up predicted clinical recurrence [sensitivity 90% (95% confidence interval (CI), 74%-98%), specificity 88% (95% CI, 62%-98%)] with a median lead time of 84 days (interquartile range, 25-146). Detection of ctDNA during postpancreatectomy follow-up was associated with a median OS of 17 months, while median OS was not yet reached at 30 months for patients without ctDNA (P = 0.011). CONCLUSIONS Measurement of KRAS ctDNA in a CLIA laboratory setting can be used to predict recurrence and survival in patients with PDAC.
Collapse
Affiliation(s)
- Vincent P Groot
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Surgery, UMC Utrecht Cancer Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stacy Mosier
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Molecular Pathology Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ammar A Javed
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan A Teinor
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Georgios Gemenetzis
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ding Ding
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,The Pancreatic Cancer Precision Medicine Center of Excellence Program, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lisa M Haley
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Molecular Pathology Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jun Yu
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Richard A Burkhart
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alina Hasanain
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marija Debeljak
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Molecular Pathology Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hirohiko Kamiyama
- Department of Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Amol Narang
- Department of Radiation Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel A Laheru
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lei Zheng
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,The Pancreatic Cancer Precision Medicine Center of Excellence Program, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ming-Tseh Lin
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Molecular Pathology Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher D Gocke
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Molecular Pathology Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elliot K Fishman
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael G Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - I Quintus Molenaar
- Department of Surgery, UMC Utrecht Cancer Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - John L Cameron
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Matthew J Weiss
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Victor E Velculescu
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jin He
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher L Wolfgang
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - James R Eshleman
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland. .,Molecular Pathology Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
1887
|
Kaissis G, Braren R. Pancreatic cancer detection and characterization-state of the art cross-sectional imaging and imaging data analysis. Transl Gastroenterol Hepatol 2019; 4:35. [PMID: 31231702 DOI: 10.21037/tgh.2019.05.04] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents a deadly disease, prognosticated to become the 2nd most common cause of cancer related death in the western world by 2030. State of the art radiologic high-resolution cross-sectional imaging by computed tomography (CT) and magnetic resonance imaging (MRI) represent advanced techniques for early lesion detection, pre-therapeutic patient staging and therapy response monitoring. In light of molecular taxonomies currently under development, the implementation of advanced imaging data post-processing pipelines and the integration of imaging and clinical data for the development of risk assessment and clinical decision support tools are required. This review will present the current state of cross-sectional radiologic imaging and image post-processing related to PDAC.
Collapse
Affiliation(s)
- Georgios Kaissis
- Institute of Diagnostic and Interventional Radiology, Faculty of Medicine, Technical University of Munich, Translational Oncology and Quantitative Imaging/Data Science Laboratory, Munich, Germany
| | - Rickmer Braren
- Institute of Diagnostic and Interventional Radiology, Faculty of Medicine, Technical University of Munich, Translational Oncology and Quantitative Imaging/Data Science Laboratory, Munich, Germany
| |
Collapse
|
1888
|
Chiorean EG, Cheung WY, Giordano G, Kim G, Al-Batran SE. Real-world comparative effectiveness of nab-paclitaxel plus gemcitabine versus FOLFIRINOX in advanced pancreatic cancer: a systematic review. Ther Adv Med Oncol 2019; 11:1758835919850367. [PMID: 31205510 PMCID: PMC6535755 DOI: 10.1177/1758835919850367] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/09/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND No clinical trial has directly compared nab-paclitaxel/gemcitabine (nab-P/G) with FOLFIRINOX (fluorouracil/leucovorin/oxaliplatin/irinotecan) in metastatic or advanced pancreatic cancer (mPC or aPC). We conducted a systematic review of real-world studies comparing these regimens in the first-line setting. METHODS Embase and MEDLINE databases through 22 January 2019, and Gastrointestinal Cancers Symposium 2019 abstracts were searched for real-world, retrospective studies comparing first-line nab-P/G versus FOLFIRINOX in mPC or aPC that met specific parameters. Studies with radiotherapy were excluded. Study quality was assessed using the Newcastle-Ottawa Scale. RESULTS Of 818 records initially identified, 35 were duplicates and 749 did not meet the eligibility criteria, mostly because they were either not comparative (n = 356) or not first line (n = 245). The remaining 34 studies (21 mPC; 13 aPC) assessed >6915 patients who received nab-P/G or FOLFIRINOX. In the studies identified, the median overall survival (OS) reached 14.4 and 15.9 months with nab-P/G and FOLFIRINOX, respectively, and median progression-free survival reached 8.5 and 11.7 months, respectively. Safety data were reported in 14 studies (2205 patients), including 8 single-institutional studies. In most single-institutional studies that reported safety data, rates were higher with FOLFIRINOX versus nab-P/G for grade 3/4 neutropenia (five of six studies) and febrile neutropenia (all three studies), while rates of grade 3/4 peripheral neuropathy were higher with nab-P/G in four of seven studies. CONCLUSIONS Although FOLFIRINOX was associated with slightly longer median OS in more studies, the differences, when available, were not statistically significant. Therefore, a randomized, controlled trial is warranted. Toxicity profile differences represent key considerations for treatment decisions.
Collapse
Affiliation(s)
- Elena Gabriela Chiorean
- Fred Hutchinson Cancer Research Center,
University of Washington School of Medicine, 825 Eastlake Ave East, Seattle,
WA 98109, USA
| | | | - Guido Giordano
- IRCCS Casa Sollievo della Sofferenza, San
Giovanni Rotondo, Italy
| | - George Kim
- 21st Century Oncology, Jacksonville, FL,
USA
| | - Salah-Eddin Al-Batran
- Institute of Clinical Cancer Research,
Krankenhaus Nordwest, University Cancer Center, Frankfurt, Germany
| |
Collapse
|
1889
|
Abstract
PURPOSE OF REVIEW Despite all efforts, pancreatic ductal adenocarcinoma (PDAC) remains a disease that causes substantial morbidity and mortality, with a 5-year survival rate of 7%. Innovative paradigms for treating PDAC are urgently needed. RECENT FINDINGS We discuss the advances and difficulties in using immunotherapy and developing immunotherapeutic vaccines for PDAC. Current excitement about antigen-specific immunotherapy has been propelled by advances in multiple areas, such as next-generation sequencing to identify neoantigens and manufacturing to produce immunotherapeutic vaccines. Antigen-specific immunotherapy is being actively explored in clinical trials. As the field of immunotherapy matures and as our understanding of the complex interactions between tumor and host develops, we hope to identify new methods for treating and managing PDAC.
Collapse
Affiliation(s)
- Annie A Wu
- Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 488, Baltimore, MD, 21287, USA
| | - Elizabeth Jaffee
- Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 488, Baltimore, MD, 21287, USA
| | - Valerie Lee
- Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 488, Baltimore, MD, 21287, USA.
| |
Collapse
|
1890
|
Djanani A, Schmiderer A, Niederreiter L, Niederreiter M, Tilg H. Management of ductal pancreatic cancer. Eur Surg 2019. [DOI: 10.1007/s10353-019-0583-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
1891
|
Immunomodulatory Effect after Irreversible Electroporation in Patients with Locally Advanced Pancreatic Cancer. JOURNAL OF ONCOLOGY 2019; 2019:9346017. [PMID: 31214261 PMCID: PMC6535893 DOI: 10.1155/2019/9346017] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/24/2022]
Abstract
Purpose Irreversible electroporation (IRE) has been demonstrated to be a safe and effective method for locally advanced pancreatic cancer (LAPC). The aim of this study was to evaluate the immunomodulatory effect after IRE and to evaluate the prognostic value of variations of the immune parameters in LAPC patients after IRE. Methods Peripheral blood samples of 34 patients were obtained preoperatively and on the third day (D3) and seventh day (D7) after IRE, respectively. The phenotypes of lymphocytes were analyzed by flow cytometry, and dynamic changes of serum levels of cytokines, complement, and immunoglobulin were assayed by enzyme-linked immunosorbent assay. Receiver operating characteristic (ROC) curve and concordance index (C-index) were used to compare the survival predictive ability. Results There was a transitory decrease followed by a steady increase for CD4+ T cell, CD8+ T cell, NK cell, IL-2, C3, C4, and IgG while a reverse trend was detected for Treg cell, IL-6, and IL10 after IRE. The alteration of CD8+ T cell between D3 and D7 was identified as a prognostic factor for both overall survival (OS) and progression-free survival (PFS). The values of ROC curve (AUC) and C-indexes of the alteration of CD8+ T cell for OS and PFS were 0.816 and 0.773 and 0.816 and 0.639, respectively, which were larger than those of other immune or inflammation-based indexes. Conclusions This study presented the first evidence of IRE-based immunomodulatory in patients with LAPC. The alteration of CD8+ T cell between D3 and D7 showed relatively good performance and could be used as an effective tool for prognostic evaluation for LAPC patients after IRE.
Collapse
|
1892
|
Primavesi F, Stättner S, Schlick K, Kiesslich T, Mayr C, Klieser E, Urbas R, Neureiter D. Pancreatic cancer in young adults: changes, challenges, and solutions. Onco Targets Ther 2019; 12:3387-3400. [PMID: 31118690 PMCID: PMC6508149 DOI: 10.2147/ott.s176700] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Despite improvements in multidisciplinary treatments, survival of pancreatic cancer (PC) patients remains dismal. Studies dealing with early onset pancreatic cancer (EOPC) patients are scarce. In this review, we discuss differences between EOPC and late-onset pancreatic cancer based on findings in original papers and reviews with a focus on morphology, genetics, clinical outcomes and therapy. In conclusion, families with a positive history of PC and patients with BRCA 1 or 2 mutations should be monitored. Patients with EOPC usually present with better overall fitness compared to the average PC population, however often with even more aggressive cancer behaviour. Therefore, potent state-of-the-art multi-modal systemic therapies should be applied whenever possible. Large-scale registries and randomized clinical trials dealing with EOPC in regard to distinct biology and outcome are warranted.
Collapse
Affiliation(s)
- Florian Primavesi
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Stättner
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Konstantin Schlick
- Department of Internal Medicine III - Division of Hematology, Medical Oncology, Hemostaseology, Rheumatology, Infectiology and Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Tobias Kiesslich
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria.,Institute of Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria
| | - Christian Mayr
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria.,Institute of Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria
| | - Eckhard Klieser
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria, .,Cancer Cluster Salzburg, Salzburg, Austria,
| | - Romana Urbas
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria, .,Cancer Cluster Salzburg, Salzburg, Austria,
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria, .,Cancer Cluster Salzburg, Salzburg, Austria,
| |
Collapse
|
1893
|
Affiliation(s)
- Natalia Khalaf
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas.
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
1894
|
Hu Q, Wang X, Chen Y, Li X, Cao P, Cao D. Which is the optimal adjuvant chemotherapy for resected pancreatic ductal adenocarcinoma?: A protocol for a network meta-analysis of randomized controlled trials. Medicine (Baltimore) 2019; 98:e15761. [PMID: 31124963 PMCID: PMC6571414 DOI: 10.1097/md.0000000000015761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 04/29/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Although adjuvant chemotherapy has been shown to reduce relapse and prolong survival after surgery, it is still unclear which adjuvant chemotherapy regimen will be favorable over the all adjuvant treatments evaluated for patients with resected pancreatic ductal adenocarcinoma. METHODS PubMed, Embase (Ovid version), Cochrane Library, the American Society of Clinical Oncology, and ClinicalTrials.gov database will be searched from their inception to January 19, 2019. We will include studies that contain adjuvant chemotherapy following surgery in patients with pancreatic ductal adenocarcinoma. The outcomes are overall survival, disease-free survival, and grade 3-4 hematological and nonhematological toxicity. The risk of bias for each randomized controlled trial will be assessed as low, moderate, or high using Cochrane Collaboration's tool independently. Pairwise and network meta-analysis will be performed using STATA 13.0, GeMTC, and WinBUGS, respectively. The competing adjuvant chemotherapy regimens will be ranked by an advantage index. RESULTS The study is ongoing and the results will be submitted to a peer-reviewed journal for publication. CONCLUSION This network meta-analysis will systematically provide suggestions to select optimum adjuvant treatment for clinical practice in the future.PROSPERO registration number: CRD42019123907 (https://www.crd.york.ac.uk/PROSPERO/#searchadvanced).
Collapse
|
1895
|
Klaiber U, Hackert T, Neoptolemos JP. Adjuvant treatment for pancreatic cancer. Transl Gastroenterol Hepatol 2019; 4:27. [PMID: 31143848 PMCID: PMC6509427 DOI: 10.21037/tgh.2019.04.04] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is the third leading cause of cancer-associated mortality in Western countries. Upfront resection with adjuvant chemotherapy is the treatment of choice in resectable tumors, offering the chance for cure. Until the 1990s, adjuvant therapy was not routinely used after resection for pancreatic cancer. During the last three decades however, enormous progress has been made in evidence-based onco-surgical management of resectable pancreatic cancer. Based on the results from multicenter randomized controlled trials, primarily initiated by the European Study Group of Pancreatic Cancer (ESPAC), adjuvant chemotherapy has become the gold standard after upfront resection, while adjuvant chemoradiotherapy is not recommended. Combination chemotherapy with gemcitabine and capecitabine was shown to significantly prolong median overall survival after resection compared to monotherapy with either gemcitabine or 5-fluorouracil/folinic acid. Recent data from the French-Canadian Uni-Cancer GI PRODIGE 24/CCTG PA.6 trial showed that adjuvant poly-agent chemotherapy with modified FOLFIRINOX achieved median survival times of 54.4 months in selected patients. Despite improved survival times after resection followed by adjuvant chemotherapy, however, recurrence occurs still in more than 75% of patients within the first 2 years after resection. Further efforts are therefore to be made in early detection tools, the evaluation of neoadjuvant strategies, the development of new drug targets, and stratification strategies to better select patients for the available therapies. This review article summarizes the body of evidence on adjuvant treatment for pancreatic cancer, identifies evidence gaps and provides future perspectives.
Collapse
Affiliation(s)
- Ulla Klaiber
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - John P Neoptolemos
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
1896
|
He C, Wang J, Sun S, Zhang Y, Lin X, Lao X, Cui B, Li S. Irreversible electroporation versus radiotherapy after induction chemotherapy on survival in patients with locally advanced pancreatic cancer: a propensity score analysis. BMC Cancer 2019; 19:394. [PMID: 31029111 PMCID: PMC6486960 DOI: 10.1186/s12885-019-5607-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Locally advanced pancreatic cancer (LAPC) represents more than one third of pancreatic cancers and owns poor survival after the standard chemotherapy. Irreversible electroporation (IRE) is a novel method and has been recently used in LAPC. The aim of this study was to compare the efficacy of IRE and radiotherapy after induction chemotherapy for patients with LAPC. METHODS From August 2015 to August 2017, a total of 76 patients with biopsy proven LAPC and who had received IRE or radiotherapy after chemotherapy were included. Thirty-two pairs of patients were selected through propensity score matching (PSM) analysis and the efficacy of two treatments was compared. RESULTS Before PSM analysis, after induction chemotherapy, patients with LAPC benefited more in terms of overall survival (OS) and progression free survival (PFS) from IRE, compared with radiotherapy (2-year OS rates, 53.5% vs 26.9%, p = 0.039; 2-year PFS rates, 28.4% vs 13.3%, p = 0.045). After PSM analysis, the survival benefits of OS and PFS of patients after induction chemotherapy followed by IRE were more obvious than those of patients treated with radiotherapy (2-year OS rates, 53.5% vs 20.7%, p = 0.011; 2-year PFS rates, 28.4% vs 5.6%, p = 0.004). Multivariate Cox regression analysis indicated that IRE after induction chemotherapy was identified as a significant favourable factor for both OS and PFS in both the whole and matched cohort. CONCLUSIONS Induction chemotherapy followed by IRE is superior to induction chemotherapy followed by radiotherapy for treating LAPC. A randomized clinical trial comparing the efficacy of IRE and radiotherapy after the induction chemotherapy is therefore considerable.
Collapse
Affiliation(s)
- Chaobin He
- Department of Hepatobiliary and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jun Wang
- Department of Hepatobiliary and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Ultrasonics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Shuxin Sun
- Department of Hepatobiliary and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Xiaojun Lin
- Department of Hepatobiliary and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiangming Lao
- Department of Hepatobiliary and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Bokang Cui
- Department of Hepatobiliary and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Shengping Li
- Department of Hepatobiliary and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
1897
|
Bakadlag R, Jandaghi P, Hoheisel JD, Riazalhosseini Y. The potential of dopamine receptor D2 (DRD2) as a therapeutic target for tackling pancreatic cancer. Expert Opin Ther Targets 2019; 23:365-367. [DOI: 10.1080/14728222.2019.1606904] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Rowa Bakadlag
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Pouria Jandaghi
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Jörg D. Hoheisel
- Functional Genome Analysis, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Yasser Riazalhosseini
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| |
Collapse
|
1898
|
Unveiling the mechanisms of immune evasion in pancreatic cancer: may it be a systemic inflammation responsible for dismal survival? Clin Transl Oncol 2019; 22:81-90. [DOI: 10.1007/s12094-019-02113-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/09/2019] [Indexed: 02/07/2023]
|
1899
|
Petrelli F, Zaniboni A, Ghidini A, Ghidini M, Turati L, Pizzo C, Ratti M, Libertini M, Tomasello G. Timing of Adjuvant Chemotherapy and Survival in Colorectal, Gastric, and Pancreatic Cancer. A Systematic Review and Meta-Analysis. Cancers (Basel) 2019; 11:cancers11040550. [PMID: 30999653 PMCID: PMC6520704 DOI: 10.3390/cancers11040550] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/13/2019] [Accepted: 04/15/2019] [Indexed: 12/18/2022] Open
Abstract
(1) Background: The optimal timing of adjuvant chemotherapy (CT) in gastrointestinal malignancies is still a matter of debate. For colorectal cancer, it is recommended to start post-operative treatment within eight weeks. The objective of this study was to assess the clinical effects of starting adjuvant CT within or after 6–8 weeks post-surgery in colorectal, gastric, and pancreatic cancer. (2) Methods: MEDLINE, EMBASE, and the Cochrane Library were searched in December 2018. Publications comparing the outcomes of patients treated with adjuvant CT administered before (early) or after (delayed) 6–8 weeks post-surgery for colorectal, gastric, and pancreatic cancer were identified. The primary endpoint was overall survival (OS). (3) Results: Out of 8752 publications identified, 34 comparative studies assessing a total of 141,853 patients were included. Meta-analysis indicated a statistically significant increased risk of death with delayed CT (>6–8 weeks post-surgery) in colorectal cancer (hazard ratio (HR) = 1.27, 95% confidence interval (CI) 1.21–1.33; p <0.001). Similarly, for gastric cancer, delaying adjuvant CT was associated with inferior overall survival (HR = 1.2, 95% CI 1.04–1.38; p = 0.01). Conversely, the benefit of earlier CT was not evident in pancreatic cancer (HR = 1, 95% CI 1–1.01; p = 0.37). Conclusions: Starting adjuvant CT within 6–8 weeks post-surgery is associated with a significant survival benefit for colorectal and gastric cancer, but not for pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | | | - Luca Turati
- Surgical Oncology Unit, ASST of Bergamo, 24100 Bergamo Ovest, Italy.
| | | | | | | | | |
Collapse
|
1900
|
Hruban RH, Gaida MM, Thompson E, Hong SM, Noë M, Brosens LA, Jongepier M, Offerhaus GJA, Wood LD. Why is pancreatic cancer so deadly? The pathologist's view. J Pathol 2019; 248:131-141. [PMID: 30838636 DOI: 10.1002/path.5260] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/19/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022]
Abstract
The remarkable aggressiveness of pancreatic cancer has never been fully explained. Although clearly multifactorial, we postulate that venous invasion, a finding seen in most pancreatic cancers but not in most cancers of other organs, may be a significant, underappreciated contributor to the aggressiveness of this disease. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ralph H Hruban
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthias M Gaida
- Department of General Pathology, The University Hospital of Heidelberg, Heidelberg, Germany
| | - Elizabeth Thompson
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seung-Mo Hong
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Michaël Noë
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lodewijk Aa Brosens
- Department of Pathology, The University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martine Jongepier
- Department of Pathology, The University Medical Center Utrecht, Utrecht, The Netherlands
| | - G Johan A Offerhaus
- Department of Pathology, The University Medical Center Utrecht, Utrecht, The Netherlands
| | - Laura D Wood
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|