901
|
Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012; 491:399-405. [PMID: 23103869 DOI: 10.1038/nature11547] [Citation(s) in RCA: 1616] [Impact Index Per Article: 124.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 09/04/2012] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer is a highly lethal malignancy with few effective therapies. We performed exome sequencing and copy number analysis to define genomic aberrations in a prospectively accrued clinical cohort (n = 142) of early (stage I and II) sporadic pancreatic ductal adenocarcinoma. Detailed analysis of 99 informative tumours identified substantial heterogeneity with 2,016 non-silent mutations and 1,628 copy-number variations. We define 16 significantly mutated genes, reaffirming known mutations (KRAS, TP53, CDKN2A, SMAD4, MLL3, TGFBR2, ARID1A and SF3B1), and uncover novel mutated genes including additional genes involved in chromatin modification (EPC1 and ARID2), DNA damage repair (ATM) and other mechanisms (ZIM2, MAP2K4, NALCN, SLC16A4 and MAGEA6). Integrative analysis with in vitro functional data and animal models provided supportive evidence for potential roles for these genetic aberrations in carcinogenesis. Pathway-based analysis of recurrently mutated genes recapitulated clustering in core signalling pathways in pancreatic ductal adenocarcinoma, and identified new mutated genes in each pathway. We also identified frequent and diverse somatic aberrations in genes described traditionally as embryonic regulators of axon guidance, particularly SLIT/ROBO signalling, which was also evident in murine Sleeping Beauty transposon-mediated somatic mutagenesis models of pancreatic cancer, providing further supportive evidence for the potential involvement of axon guidance genes in pancreatic carcinogenesis.
Collapse
|
902
|
Turaga KK, Tsai S, Wiebe LA, Evans DB, Gamblin TC. Novel multimodality treatment sequencing for extrahepatic (mid and distal) cholangiocarcinoma. Ann Surg Oncol 2012; 20:1230-9. [PMID: 23064778 DOI: 10.1245/s10434-012-2648-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Indexed: 12/15/2022]
Abstract
Neoadjuvant chemoradiation has demonstrated significant advantages in the management of pancreatic adenocarcinoma. A similar tumor in a nearby anatomical location is extrahepatic cholangiocarcinoma, which has proven to be largely unresponsive to current forms of therapy. Neoadjuvant therapy for hilar cholangiocarcinoma has been combined with surgical resection and/or liver transplantation with a 25-33 % complete pathological response rate. We propose a wider application of neoadjuvant chemoradiation for patients with distal cholangiocarcinoma and present our rationale for this form of treatment sequencing.
Collapse
Affiliation(s)
- Kiran K Turaga
- Department of Surgery, Dvision of Surgical Oncology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | | | | | | | | |
Collapse
|
903
|
Affiliation(s)
- Saxon Connor
- Department of Surgery, Christchurch Hospital ChristchurchNew Zealand
| | - Magdalena Sakowska
- Department of General Surgery, Christchurch Hospital ChristchurchNew Zealand
| |
Collapse
|
904
|
Zhang Y, Frampton AE, Cohen P, Kyriakides C, Bong JJ, Habib NA, Spalding DRC, Ahmad R, Jiao LR. Tumor infiltration in the medial resection margin predicts survival after pancreaticoduodenectomy for pancreatic ductal adenocarcinoma. J Gastrointest Surg 2012; 16:1875-82. [PMID: 22878786 DOI: 10.1007/s11605-012-1985-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 07/24/2012] [Indexed: 01/31/2023]
Abstract
BACKGROUND Microscopic tumor involvement (R1) in different surgical resection margins after pancreaticoduodenectomy (PD) for pancreatic ductal adenocarcinoma (PDAC) has been debated. METHODS Clinico-pathological data for 258 patients who underwent PD between 2001 and 2010 were retrieved from a prospective database. The rates of R1 resection in the circumferential resection margin (pancreatic transection, medial, posterior, and anterior surfaces) and their prognostic influence on survival were assessed. RESULTS For PDAC, the R1 rate was 57.1% (48/84) for any margin, 31.0% (26/84) for anterior surface, 42.9% (36/84) for posterior surface, 29.8% (25/84) for medial margin, and 7.1% (3/84) for pancreatic transection margin. Overall and disease-free survival for R1 resections were significantly worse than those for R0 resection (17.2 vs. 28.7 months, P = 0.007 and 12.3 vs. 21.0 months, P = 0.019, respectively). For individual margins, only medial positivity had a significant impact on survival (13.8 vs. 28.0 months, P < 0.001), as opposed to involvement in the anterior (19.7 vs. 23.3 months, P = 0.187) or posterior margin (17.5 vs. 24.2 months, P = 0.104). Multivariate analysis demonstrated R0 medial margin was an independent prognostic factor (P = 0.002, HR = 0.381; 95% CI 0.207-0.701). CONCLUSION The medial surgical resection margin is the most important after PD for PDAC, and an R1 resection here predicts poor survival.
Collapse
Affiliation(s)
- Yaojun Zhang
- HPB Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, W12 0HS, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
905
|
Schmidt J, Abel U, Debus J, Harig S, Hoffmann K, Herrmann T, Bartsch D, Klein J, Mansmann U, Jäger D, Capussotti L, Kunz R, Büchler MW. Open-label, multicenter, randomized phase III trial of adjuvant chemoradiation plus interferon Alfa-2b versus fluorouracil and folinic acid for patients with resected pancreatic adenocarcinoma. J Clin Oncol 2012; 30:4077-83. [PMID: 23008325 DOI: 10.1200/jco.2011.38.2960] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Adjuvant chemotherapy prolongs survival in patients with pancreatic cancer, but its benefit is limited. Long-term survival times of up to 44 months after adjuvant chemoradioimmunotherapy in phase II trials motivated the present study. PATIENTS AND METHODS Between 2004 and 2007, 132 R0/R1 resected patients received either fluorouracil (FU), cisplatin, and interferon alfa-2b (IFN α-2b) plus radiotherapy followed by two cycles of FU (arm A, n=64) or six cycles of FU monotherapy (arm B, n=68). One hundred ten patients (arm A, n=53; arm B, n=57) received at least one dose of the study medication, and these patients composed the per-protocol (PP) population. Biomarkers were analyzed longitudinally for their predictive value. RESULTS Median survival for all randomly assigned patients was 26.5 months (95% CI, 21.6 to 39.5 months) in arm A and 28.5 months (95% CI, 20.4 to 38.6 months) in arm B. The hazard ratio was 1.04 (arm A v arm B: 95% CI, 0.66 to 1.53; P=.99). Median survival for the PP population was 32.1 months (95% CI, 22.8 to 42.2 months) in arm A and 28.5 months (95% CI, 19.5 to 38.6 months) in arm B (P=.49). Eighty-five percent of patients in arm A and 16% of patients in arm B experienced grade 3 or 4 toxicity. The quality of life was temporarily negatively affected in arm A. CONCLUSION The FU, cisplatin, and IFN α-2b plus radiotherapy regimen did not improve the survival compared with FU monotherapy. Given the substantial adverse effects, this treatment can currently not be recommended. Nevertheless, the outcome in both arms represents the best survival, to our knowledge, ever reported for patients with resected pancreatic cancer in randomized controlled trials. Future studies will demonstrate whether immune response to IFN α-2b challenge has a predictive value.
Collapse
Affiliation(s)
- Jan Schmidt
- Department of Surgery, Ruprecht-Karls-University, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
906
|
Bachet JB, Maréchal R, Demetter P, Bonnetain F, Couvelard A, Svrcek M, Bardier-Dupas A, Hammel P, Sauvanet A, Louvet C, Paye F, Rougier P, Penna C, Vaillant JC, André T, Closset J, Salmon I, Emile JF, Van Laethem JL. Contribution of CXCR4 and SMAD4 in predicting disease progression pattern and benefit from adjuvant chemotherapy in resected pancreatic adenocarcinoma. Ann Oncol 2012; 23:2327-2335. [PMID: 22377565 DOI: 10.1093/annonc/mdr617] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Prognosis of patients with pancreatic adenocarcinoma is poor. Many prognostic biomarkers have been tested, but most studies included heterogeneous patients. We aimed to investigate the prognostic and/or predictive values of four relevant biomarkers in a multicentric cohort of patients. PATIENTS AND METHODS A total of 471 patients who had resected pancreatic adenocarcinoma were included. Using tissue microarray, we assessed the relationship of biomarker expressions with the overall survival: Smad4, type II TGF-β receptor, CXCR4, and LKB1. RESULTS High CXCR4 expression was found to be the only independent negative prognostic biomarker [hazard ratio (HR) = 1.74; P < 0.0001]. In addition, it was significantly associated with a distant relapse pattern (HR = 2.19; P < 0.0001) and was the strongest prognostic factor compared with clinicopathological factors. In patients who did not received adjuvant treatment, there was a trend toward decrease in the overall survival for negative Smad4 expression. Loss of Smad4 expression was not correlated with recurrence pattern but was shown to be predictive for adjuvant chemotherapy (CT) benefit (HR = 0.59; P = 0.002). CONCLUSIONS CXCR4 is a strong independent prognostic biomarker associated with distant metastatic recurrence and appears as an attractive target to be evaluated in pancreatic adenocarcinoma. Negative SMAD4 expression should be considered as a potential predictor of adjuvant CT benefit.
Collapse
Affiliation(s)
- J B Bachet
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris; EA4340 "Epidémiologie et oncogènes des tumeurs digestives", Versailles Saint-Quentin-en-Yvelines University, Versailles; Department of Hepato-Gastroenterology, Pitié-Salpêtrière Hospital, APHP, Paris, France; Department of Gastroenterology, Gastrointestinal cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Brussels.
| | - R Maréchal
- Department of Gastroenterology, Gastrointestinal cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Brussels
| | - P Demetter
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, and DiaPath, Brussels, Belgium
| | - F Bonnetain
- Department of Biostatistic and Epidemiology (EA 4184), Georges François Leclerc Center, Dijon
| | - A Couvelard
- Department of Pathology, Beaujon Hospital, APHP, Clichy
| | - M Svrcek
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris; Department of Pathology, Saint Antoine Hospital, APHP, Paris
| | - A Bardier-Dupas
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris; Department of Pathology, Pitié-Salpêtrière Hospital, APHP, Paris
| | - P Hammel
- Department of Gastroenterology, Beaujon Hospital, APHP, Clichy
| | - A Sauvanet
- Department of Surgery, Beaujon Hospital, APHP, Clichy
| | - C Louvet
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris; Department of Oncology, Saint Antoine Hospital, APHP, Paris; Department of Oncology, Institut Mutualiste Montsouris, Paris
| | - F Paye
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris; Department of Surgery, Saint Antoine Hospital, APHP, Paris
| | - P Rougier
- EA4340 "Epidémiologie et oncogènes des tumeurs digestives", Versailles Saint-Quentin-en-Yvelines University, Versailles; Department of Digestive Oncology, European Georges Pompidou Hospital, APHP, Paris
| | - C Penna
- EA4340 "Epidémiologie et oncogènes des tumeurs digestives", Versailles Saint-Quentin-en-Yvelines University, Versailles; Department of Surgery, Ambroise Paré Hospital, APHP, Boulogne-Billancourt
| | - J C Vaillant
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris; Department of Surgery, Pitié-Salpêtrière Hospital, APHP, Paris, France
| | - T André
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris; Department of Hepato-Gastroenterology, Pitié-Salpêtrière Hospital, APHP, Paris, France
| | - J Closset
- Department of Surgery, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - I Salmon
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, and DiaPath, Brussels, Belgium
| | - J F Emile
- EA4340 "Epidémiologie et oncogènes des tumeurs digestives", Versailles Saint-Quentin-en-Yvelines University, Versailles; Department of Pathology, Ambroise Paré Hospital, APHP, Boulogne-Billancourt, France
| | - J L Van Laethem
- Department of Gastroenterology, Gastrointestinal cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Brussels
| |
Collapse
|
907
|
Maréchal R, Bachet JB, Mackey JR, Dalban C, Demetter P, Graham K, Couvelard A, Svrcek M, Bardier-Dupas A, Hammel P, Sauvanet A, Louvet C, Paye F, Rougier P, Penna C, André T, Dumontet C, Cass CE, Jordheim LP, Matera EL, Closset J, Salmon I, Devière J, Emile JF, Van Laethem JL. Levels of gemcitabine transport and metabolism proteins predict survival times of patients treated with gemcitabine for pancreatic adenocarcinoma. Gastroenterology 2012; 143:664-674.e6. [PMID: 22705007 DOI: 10.1053/j.gastro.2012.06.006] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 05/11/2012] [Accepted: 06/05/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Patients who undergo surgery for pancreatic ductal adenocarcinoma (PDAC) frequently receive adjuvant gemcitabine chemotherapy. Key determinants of gemcitabine cytotoxicity include the activities of the human equilibrative nucleoside transporter 1 (hENT1), deoxycytidine kinase (dCK), and ribonucleotide reductase subunit 1 (RRM1). We investigated whether tumor levels of these proteins were associated with efficacy of gemcitabine therapy following surgery. METHODS Sequential samples of resected PDACs were retrospectively collected from 434 patients at 5 centers; 142 patients did not receive adjuvant treatment (33%), 243 received adjuvant gemcitabine-based regimens (56%), and 49 received nongemcitabine regimens (11%). We measured protein levels of hENT1, dCK, and RRM1 by semiquantitative immunohistochemistry with tissue microarrays and investigated their relationship with patients' overall survival time. RESULTS The median overall survival time of patients was 32.0 months. Among patients who did not receive adjuvant treatment, levels of hENT1, RRM1, and dCK were not associated with survival time. Among patients who received gemcitabine, high levels of hENT1 and dCK were significantly associated with longer survival time (hazard ratios of 0.34 [P < .0001] and 0.57 [P = .012], respectively). Interaction tests for gemcitabine administration and hENT1 and dCK status were statistically significant (P = .0007 and P = .016, respectively). On multivariate analysis of this population, hENT1 and dCK retained independent predictive values, and those patients with high levels of each protein had the longest survival times following adjuvant therapy with gemcitabine. CONCLUSIONS High levels of hENT1 and dCK in PDAC predict longer survival times in patients treated with adjuvant gemcitabine.
Collapse
Affiliation(s)
- Raphaël Maréchal
- Department of Gastroenterology and Gastrointestinal Cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium.
| | - Jean-Baptiste Bachet
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris, France; EA4340 "Epidémiologie et oncogènes des tumeurs digestives," Versailles Saint-Quentin-en-Yvelines University, Saint-Quentin-en-Yvelines, France; Department of Hepato-Gastroenterology, Pitié Salpêtrière Hospital, APHP, Paris, France
| | - John R Mackey
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Cécile Dalban
- Department of Biostatistics and Epidemiology (EA4184), Georges François Leclerc Center, Dijon, France
| | - Pieter Demetter
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, and DIAPATH, Brussels, Belgium
| | - Kathryn Graham
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Anne Couvelard
- Department of Pathology, Beaujon Hospital, APHP, Clichy, France
| | - Magali Svrcek
- Department of Pathology, Saint Antoine Hospital, APHP, Paris, France
| | - Armelle Bardier-Dupas
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris, France; Department of Pathology, Pitié Salpêtrière Hospital, APHP, Paris, France
| | - Pascal Hammel
- Department of Gastroenterology, Beaujon Hospital, APHP, Clichy, France
| | - Alain Sauvanet
- Department of Surgery, Beaujon Hospital, APHP, Clichy, France
| | - Christophe Louvet
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris, France; Department of Oncology, Saint Antoine Hospital, APHP, Paris, France
| | - François Paye
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris, France; Department of Surgery, Saint Antoine Hospital, APHP, Paris, France
| | - Philippe Rougier
- EA4340 "Epidémiologie et oncogènes des tumeurs digestives," Versailles Saint-Quentin-en-Yvelines University, Saint-Quentin-en-Yvelines, France; Department of Digestive Oncology, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | - Christophe Penna
- EA4340 "Epidémiologie et oncogènes des tumeurs digestives," Versailles Saint-Quentin-en-Yvelines University, Saint-Quentin-en-Yvelines, France; Department of Surgery, Ambroise Paré Hospital, APHP, Boulogne Billancourt, France
| | - Thierry André
- Medical University Pierre et Marie Curie, UFR Paris VI, Paris, France; Department of Hepato-Gastroenterology, Pitié Salpêtrière Hospital, APHP, Paris, France
| | - Charles Dumontet
- Centre de Cancer de Lyon, Lyon, France; Hospices Civils de Lyon, Lyon, France
| | - Carol E Cass
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | | | | | - Jean Closset
- Department of Surgery, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, and DIAPATH, Brussels, Belgium
| | - Jacques Devière
- Department of Gastroenterology and Gastrointestinal Cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-François Emile
- EA4340 "Epidémiologie et oncogènes des tumeurs digestives," Versailles Saint-Quentin-en-Yvelines University, Saint-Quentin-en-Yvelines, France; Department of Pathology, Ambroise Paré Hospital, APHP, Boulogne Billancourt, France
| | - Jean-Luc Van Laethem
- Department of Gastroenterology and Gastrointestinal Cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
908
|
Hsueh CT. Pancreatic cancer: current standards, research updates and future directions. J Gastrointest Oncol 2012; 2:123-5. [PMID: 22811841 DOI: 10.3978/j.issn.2078-6891.2011.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Accepted: 08/15/2011] [Indexed: 01/05/2023] Open
Affiliation(s)
- Chung-Tsen Hsueh
- Division of Medical Oncology and Hematology, Loma Linda University Medical Center, Loma Linda, California, USA
| |
Collapse
|
909
|
Nguyen Kovochich A, Arensman M, Lay AR, Rao NP, Donahue T, Li X, French SW, Dawson DW. HOXB7 promotes invasion and predicts survival in pancreatic adenocarcinoma. Cancer 2012; 119:529-39. [PMID: 22914903 DOI: 10.1002/cncr.27725] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 05/08/2012] [Accepted: 05/24/2012] [Indexed: 12/19/2022]
Abstract
BACKGROUND The homeobox gene HOXB7 is overexpressed across a range of cancers and promotes tumorigenesis through varying effects on proliferation, survival, invasion, and angiogenesis. Although published microarray data suggest HOXB7 is overexpressed in pancreatic ductal adenocarcinoma (PDAC), its function in pancreatic cancer has not been studied. METHODS HOXB7 message and protein levels were examined in PDAC cell lines and patient samples, as well as in normal pancreas. HOXB7 protein expression in patient tumors was determined by immunohistochemistry and correlated with clinicopathologic factors and survival. The impact of HOXB7 on cell proliferation, growth, and invasion was assessed by knockdown and overexpression in PDAC cell lines. Candidate genes whose expression levels were altered following HOXB7 knockdown were determined by microarray analysis. RESULTS HOXB7 message and protein levels were significantly elevated in PDAC cell lines and patient tumor samples relative to normal pancreas. Evaluation of a tissue microarray of 145 resected PDACs found high HOXB7 protein expression was correlated with lymph node metastasis (P = .034) and an independent predictor of worse overall survival in multivariate analysis (hazard ratio = 1.56, 95% confidence interval = 1.02-2.39). HOXB7 knockdown or overexpression in PDAC cell lines resulted in decreased or increased invasion, respectively, without influencing proliferation or cell viability. CONCLUSIONS HOXB7 is frequently overexpressed in PDAC, specifically promotes invasive phenotype, and is associated with lymph node metastasis and worse survival outcome. HOXB7 and its downstream targets may represent novel clinical biomarkers or targets of therapy for inhibiting the invasive and metastatic capacity of PDAC.
Collapse
Affiliation(s)
- Anne Nguyen Kovochich
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1732, USA
| | | | | | | | | | | | | | | |
Collapse
|
910
|
Kimple RJ, Russo S, Monjazeb A, Blackstock AW. The role of chemoradiation for patients with resectable or potentially resectable pancreatic cancer. Expert Rev Anticancer Ther 2012; 12:469-80. [PMID: 22500684 DOI: 10.1586/era.12.18] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Conflicting data and substantial controversy exist regarding optimal adjuvant treatment for those patients with resectable or potentially resectable adenocarcinoma of the pancreas. Despite improvements in short-term surgical outcomes, the use of newer chemotherapeutic agents, development of targeted agents and more precise delivery of radiation, the 5-year survival rates for early-stage patients remains less than 25%. This article critically reviews the existing data for various adjuvant treatment approaches for patients with surgically resectable pancreatic cancer. Our review confirms that despite several randomized clinical trials, the optimal adjuvant treatment approach for these patients remains unclear.
Collapse
Affiliation(s)
- Randall J Kimple
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | | | | | | |
Collapse
|
911
|
Murakami Y, Uemura K, Sudo T, Hashimoto Y, Nakashima A, Kondo N, Nakagawa N, Sueda T. Benefit of portal or superior mesenteric vein resection with adjuvant chemotherapy for patients with pancreatic head carcinoma. J Surg Oncol 2012; 107:414-21. [PMID: 22886567 DOI: 10.1002/jso.23229] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 07/06/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVES The aim of this study was to evaluate the efficacy of portal or superior mesenteric vein (PV/SMV) resection for patients with pancreatic carcinoma who underwent pancreatoduodenectomy. METHODS Medical records of 125 patients with pancreatic head carcinoma who underwent pancreatoduodenectomy were reviewed retrospectively. Sixty-one patients underwent PV/SMV resection and 64 patients did not. Clinicopathological factors were compared between the two groups and the prognostic impact of PV/SMV resection was evaluated using univariate and multivariate survival analysis. RESULTS The frequency of mortality and morbidity did not differ between the two groups. Univariate analysis revealed that a significant difference in overall survival was found between patients who did and did not undergo PV/SMV resection (P = 0.046) as well as between patients with and without pathological PV/SMV invasion (P = 0.012). However, PV/SMV resection and pathological PV/SMV invasion were not independent prognostic factors by multivariate survival analysis. Among patients with PV/SMV resection, the use of adjuvant chemotherapy was the only independent prognostic factor of overall survival (P = 0.003). CONCLUSIONS Resection of the PV/SMV with adjuvant chemotherapy may provide an acceptable survival benefit to patients with pancreatic head carcinoma, which involves the PV/SMV without additional mortality and morbidity.
Collapse
Affiliation(s)
- Yoshiaki Murakami
- Department of Surgery, Division of Clinical Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
912
|
O'Reilly EM. Adjuvant therapy for pancreas adenocarcinoma. J Surg Oncol 2012; 107:78-85. [PMID: 22886586 DOI: 10.1002/jso.23230] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/10/2012] [Indexed: 01/04/2023]
Abstract
Adjuvant therapy for pancreas adenocarcinoma in 2012 includes consideration of systemic therapy based on high level evidence and combined chemoradiotherapy based on less robust data. Current major adjuvant questions are examining the role of the addition of a second agent, either cytotoxic or targeted agent, to gemcitabine and whether or not the utilization of combined chemoradiotherapy improves overall survival. Progress to date has been modest and incremental in the adjuvant setting.
Collapse
Affiliation(s)
- Eileen M O'Reilly
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.
| |
Collapse
|
913
|
Costello E, Greenhalf W, Neoptolemos JP. New biomarkers and targets in pancreatic cancer and their application to treatment. Nat Rev Gastroenterol Hepatol 2012; 9:435-44. [PMID: 22733351 DOI: 10.1038/nrgastro.2012.119] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Late diagnosis of pancreatic ductal adenocarcinoma (pancreatic cancer) and the limited response to current treatments results in an exceptionally poor prognosis. Advances in our understanding of the molecular events underpinning pancreatic cancer development and metastasis offer the hope of tangible benefits for patients. In-depth mutational analyses have shed light on the genetic abnormalities in pancreatic cancer, providing potential treatment targets. New biological studies in patients and in mouse models have advanced our knowledge of the timing of metastasis of pancreatic cancer, highlighting new directions for the way in which patients are treated. Furthermore, our increasing understanding of the molecular events in tumorigenesis is leading to the identification of biomarkers that enable us to predict response to treatment. A major drawback, however, is the general lack of an adequate systematic approach to advancing the use of biomarkers in cancer drug development, highlighted in a Cancer Biomarkers Collaborative consensus report. In this Review, we summarize the latest insights into the biology of pancreatic cancer, and their repercussions for treatment. We provide an overview of current treatments and, finally, we discuss novel therapeutic approaches, including the role of biomarkers in therapy for pancreatic cancer.
Collapse
Affiliation(s)
- Eithne Costello
- National Institute for Health Research Pancreas Biomedical Research Unit and Liverpool Cancer Research UK Centre, Department of Molecular, University of Liverpool, Liverpool, L69 3GA, UK
| | | | | |
Collapse
|
914
|
Auriemma WS, Berger AC, Bar-Ad V, Boland PM, Cohen SJ, Roche-Lima CMS, Morris GJ. Locally Advanced Pancreatic Cancer. Semin Oncol 2012; 39:e9-22. [DOI: 10.1053/j.seminoncol.2012.05.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
915
|
RhoT1 and Smad4 are correlated with lymph node metastasis and overall survival in pancreatic cancer. PLoS One 2012; 7:e42234. [PMID: 22860091 PMCID: PMC3409151 DOI: 10.1371/journal.pone.0042234] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 07/05/2012] [Indexed: 12/22/2022] Open
Abstract
Cancer cell invasion and metastasis are the most important adverse prognostic factors for pancreatic cancer. Identification of biomarkers associated with outcome of pancreatic cancer may provide new approaches and targets for anticancer therapy. The aim of this study is to examine the relationship between the expression of RhoT1, Smad4 and p16 and metastasis and survival in patients with pancreatic cancer. The analysis showed that the high cytoplasmic expression levels of RhoT1, Smad4 and p16 in pancreatic cancer tissues had significantly negative correlation with lymph node metastasis (LNM) (P = 0.017, P = 0.032, P = 0.042, respectively). However, no significant association was observed between perineural invasion (PNI) and the expression of above three proteins (all P>0.05). Additionally, the survival analysis showed that the low expression levels of RhoT1 and Smad4 were significantly associated with worse survival (P = 0.034, P = 0.047, respectively). In conclusion, these results indicated that the low-expression levels of RhoT1 and Smad4 were significantly associated with LNM and shorter survival. RhoT1 may be considered as a potential novel marker for predicting the outcome in patients with pancreatic cancer.
Collapse
|
916
|
Liu QH, Zhao CY, Zhang J, Chen Y, Gao L, Ni CY, Zhu MH. Role of heat shock protein 27 in gemcitabine-resistant human pancreatic cancer: comparative proteomic analyses. Mol Med Rep 2012; 6:767-73. [PMID: 22858734 DOI: 10.3892/mmr.2012.1013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 07/19/2012] [Indexed: 11/06/2022] Open
Abstract
The most notable obstacle hindering the effective treatment of human pancreatic cancer is intrinsic chemoresistance. In order to identify the candidate protein(s) responsible for the intrinsic chemoresistance, the protein expression profiling of human pancreatic adenocarcinoma cell line Capan-1 and its distinct surviving cells following primary treatment with gemcitabine (GEM) were compared by two-dimensional electrophoresis (2-DE) combined with liquid chromatography-mass spectrometry (LC-MS) or mass spectrometry (MS). In total, nine proteins were identified, and heat shock protein B1 (HSP27), one of the differentially expressed proteins, was selected for further validation. Furthermore, the results of western blotting and immunohistochemical staining indicated that HSP27 may be significant in pancreatic intrinsic chemoresistance to GEM. The findings of this study provide a platform for further elucidation of the underlying mechanisms of pancreatic cancer intrinsic chemoresistance and demonstrate that HSP27 may be a valid target for anticancer drug development.
Collapse
Affiliation(s)
- Qing-Hua Liu
- Department of Pathology, Changhai Hospital, Secondary Military Medical University, Shanghai, PR China
| | | | | | | | | | | | | |
Collapse
|
917
|
Wingren C, Sandström A, Segersvärd R, Carlsson A, Andersson R, Löhr M, Borrebaeck CAK. Identification of serum biomarker signatures associated with pancreatic cancer. Cancer Res 2012; 72:2481-90. [PMID: 22589272 DOI: 10.1158/0008-5472.can-11-2883] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is an aggressive disease with poor prognosis, due, in part, to the lack of disease-specific biomarkers that could afford early and accurate diagnosis. With a recombinant antibody microarray platform, targeting mainly immunoregulatory proteins, we screened sera from 148 patients with pancreatic cancer, chronic pancreatitis, autoimmune pancreatitis (AIP), and healthy controls (N). Serum biomarker signatures were derived from training cohorts and the predictive power was evaluated using independent test cohorts. The results identified serum portraits distinguishing pancreatic cancer from N [receiver operating characteristics area under the curve (AUC) of 0.95], chronic pancreatitis (0.86), and AIP (0.99). Importantly, a 25-serum biomarker signature discriminating pancreatic cancer from the combined group of N, chronic pancreatitis, and AIP was determined. This signature exhibited a high diagnostic potential (AUC of 0.88). In summary, we present the first prevalidated, multiplexed serum biomarker signature for diagnosis of pancreatic cancer that may improve diagnosis and prevention in premalignant diseases and in screening of high-risk individuals.
Collapse
|
918
|
Kraft M, Kraft K, Gärtner S, Mayerle J, Simon P, Weber E, Schütte K, Stieler J, Koula-Jenik H, Holzhauer P, Gröber U, Engel G, Müller C, Feng YS, Aghdassi A, Nitsche C, Malfertheiner P, Patrzyk M, Kohlmann T, Lerch MM. L-Carnitine-supplementation in advanced pancreatic cancer (CARPAN)--a randomized multicentre trial. Nutr J 2012; 11:52. [PMID: 22824168 PMCID: PMC3439338 DOI: 10.1186/1475-2891-11-52] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 07/23/2012] [Indexed: 12/29/2022] Open
Abstract
Background Cachexia, a >10% loss of body-weight, is one factor determining the poor prognosis of pancreatic cancer. Deficiency of L-Carnitine has been proposed to cause cancer cachexia. Findings We screened 152 and enrolled 72 patients suffering from advanced pancreatic cancer in a prospective, multi-centre, placebo-controlled, randomized and double-blinded trial to receive oral L-Carnitine (4 g) or placebo for 12 weeks. At entry patients reported a mean weight loss of 12 ± 2,5 (SEM) kg. During treatment body-mass-index increased by 3,4 ± 1,4% under L-Carnitine and decreased (−1,5 ± 1,4%) in controls (p < 0,05). Moreover, nutritional status (body cell mass, body fat) and quality-of-life parameters improved under L-Carnitine. There was a trend towards an increased overall survival in the L-Carnitine group (median 519 ± 50 d versus 399 ± 43 d, not significant) and towards a reduced hospital-stay (36 ± 4d versus 41 ± 9d,n.s.). Conclusion While these data are preliminary and need confirmation they indicate that patients with pancreatic cancer may have a clinically relevant benefit from the inexpensive and well tolerated oral supplementation of L-Carnitine.
Collapse
Affiliation(s)
- Matthias Kraft
- Department of Medicine A, University Medicine Greifswald, Friedrich Löffler Straße 23a, Greifswald 17475, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
919
|
Strobel O, Berens V, Hinz U, Hartwig W, Hackert T, Bergmann F, Debus J, Jäger D, Büchler MW, Werner J. Resection after neoadjuvant therapy for locally advanced, "unresectable" pancreatic cancer. Surgery 2012; 152:S33-42. [PMID: 22770956 DOI: 10.1016/j.surg.2012.05.029] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 12/29/2022]
Abstract
BACKGROUND For pancreatic cancer, complete macroscopic resection in combination with chemotherapy is the only potentially curative treatment. Many patients present with locally advanced cancers deemed unresectable. We sought to assess the results of exploration after neoadjuvant therapy for locally advanced possibly unresectable pancreatic cancer. METHODS From a prospective database, all consecutive patients undergoing operation from October 2001 to December 2009 after neoadjuvant therapy for locally advanced pancreatic cancer were identified. Main criteria for "unresectability" were infiltration of the celiac axis or superior mesenteric artery. Resection rates, perioperative results, and survival were analyzed. RESULTS Of 257 patients, 199 (77.4%) had received neoadjuvant chemoradiation, and 58 (22.6%) chemotherapy only. Of 257 patients, 120 (46.7%) underwent successful resection, whereas 137 patients underwent exploration only; 47 (39.2%) multivisceral and 45 (37.5%) vascular resections (12 arterial reconstructions) were performed. There were 6 (5%) ypT0 neoplasms, 36 (30.0%) R0, 61 (50.8%) R1, and 16 (13.3%) R2 resections. The median follow-up of surviving patients (n = 22) was 22 months. Median postoperative survival was greater after resection (12.7 months) than after exploration alone (8.8 months; P < .0001). Median postoperative survival was 24.6 months after R0, 11.9 months after R1, and 8.9 months after R2 resection. The 3-year survival rate after R0 resection was 24%. To determine survival after start of neoadjuvant therapy, 3.7 months (median) have to be added. CONCLUSION In locally advanced, unresectable pancreatic cancer, R0/R1 resections can be achieved in up to 40% of patients who undergo operation after neoadjuvant therapy. In these cases, survival rates are similar to those observed for initially resectable pancreatic cancer.
Collapse
Affiliation(s)
- Oliver Strobel
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
920
|
Pancreatic ductal adenocarcinoma: long-term survival does not equal cure. Surgery 2012; 152:S43-9. [PMID: 22763261 DOI: 10.1016/j.surg.2012.05.020] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma represents 90% of pancreatic cancers and is an important cause of cancer death in the United States. Operative resection remains as the only treatment providing prolonged survival, but even after a curative resection, 5-year survival rates are low. Our aim was to identify the prognostic factors for long-term survival after resection of pancreatic ductal adenocarcinoma related to patients, treatments, and tumor biology. METHODS Retrospective review identified 959 patients who underwent resection of their pancreatic adenocarcinoma between February 1985 and December 2010, of whom 499 were resected before November 2006 and represent the cohort we describe in this study. Patient, tumor, and treatment-related variables were assessed for their associations with 5- and 10-year overall survival. RESULTS Of the 499 patients, 49% were female and median age was 65 years. The majority of patients had stage IIb disease (60%). Actual 5-year survival after resection of pancreatic adenocarcinoma was 19% (95/499), and actual 10-year survival was 10% (33/329). Significant clinicopathologic factors predicting 5- and 10-year survival were negative margins and negative nodal status. Interestingly, 41% (39/95) of long-term survivors had positive nodes and 24% (23/95) had positive margins. CONCLUSION Pancreatic ductal adenocarcinoma demonstrates a very heterogeneous biology, but patients with negative resection margins and node negative cancers are more likely to survive 5 years after resection. However, our series demonstrates that the biology of the cancer rather than simple pathologic factors determine a patient's prognosis.
Collapse
|
921
|
Crippa S, Partelli S, Zamboni G, Barugola G, Capelli P, Inama M, Bassi C, Pederzoli P, Falconi M. Poorly differentiated resectable pancreatic cancer: is upfront resection worthwhile? Surgery 2012; 152:S112-9. [PMID: 22766365 DOI: 10.1016/j.surg.2012.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Poorly differentiated, resectable pancreatic ductal adenocarcinoma is associated with early recurrence and may benefit from neoadjuvant treatment. The aim of this study was to evaluate clinicopathologic characteristics and survival of patients with resectable pancreatic ductal adenocarcinoma according to histologic grading. METHODS A total of 502 patients who underwent resection for pancreatic ductal adenocarcinoma between 1990 and 2008 were analyzed via the use of different histologic grading. RESULTS Well-differentiated (G1), moderately differentiated (G2), and poorly differentiated (G3) pancreatic ductal adenocarcinomas were found in 23 (4.5%), 310 (62%), and 169 (33.5%) patients. Adjuvant therapy, N status, grading, and R status were independent predictors of disease-specific survival for the entire cohort, with 1- and 5-year disease-specific survival rates of 81% and 21%, respectively. Only the presence of symptoms was a significant clinical predictor of G3 status (P = .035). G3 neoplasms were characterized by a greater rate of lymph node metastases, microvascular/perineural invasion, and R2 resections. Median disease-specific survival was 77, 26, and 20 months for G1, G2, and G3 neoplasms (P < .0001). Median disease-free survival was 63, 14, and 9 months for G1, G2, and G3 pancreatic ductal adenocarcinoma (P < .0001). Adjuvant therapy improved disease-specific survival in G2 (P < .04) and G3 (P < .0001) pancreatic ductal adenocarcinoma, with a greater survival benefit for G3 neoplasms (hazard ratio: 1.334 vs 2.116). CONCLUSION G3 pancreatic ductal adenocarcinoma is associated with a lesser rate of disease-free survival after resection and with the presence of other poor prognostic factors. The benefit of adjuvant therapy is greater in G3 than in G1 and G2 neoplasms. On the basis of these findings, patients with resectable G3 PDAC can be considered as possible targets for neoadjuvant treatment.
Collapse
Affiliation(s)
- Stefano Crippa
- Department of Surgery, University of Verona, Verona, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
922
|
Martin LK, Wei L, Trolli E, Bekaii-Saab T. Elevated baseline CA19-9 levels correlate with adverse prognosis in patients with early- or advanced-stage pancreas cancer. Med Oncol 2012; 29:3101-7. [PMID: 22729400 DOI: 10.1007/s12032-012-0278-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/06/2012] [Indexed: 12/16/2022]
Abstract
CA19-9 is the most specific biomarker for pancreas cancer. We investigated the prognostic significance of normal (≤ 37 U/mL) versus elevated (>37 U/mL) CA19-9 levels in patients with resected and advanced pancreas cancer. Relevant data were obtained from patients treated for early-stage or advanced pancreatic adenocarcinoma at our institution. Log-rank tests were used to evaluate relationship between CA19-9 and clinical outcomes of interest for both early- and advanced-stage patients. A total of 123 patients were included (Group A: N = 30 stage I/II; Group B: N = 93 stage III/IV). In group A, elevated preoperative CA19-9 was significantly associated with lymph node involvement (p = 0.031), tumor ≥ 3 cm (p = 0.011), and lack of tumor differentiation (p = 0.048). Failure of postoperative CA19-9 to normalize predicted significantly worse DFS (p = 0.021). For group B, elevated baseline CA19-9 was associated with shorter OS on chemotherapy (p = 0.0008) and decline in CA19-9 >25 % with treatment was a significant predictor of improved OS (p = 0.0099). Higher than normal CA19-9 level is an adverse prognostic factor in both early and advanced settings and may prove to be useful in the selection of patients for more aggressive therapy in future trials. CA19-9 level decrease of >25 % predicts improved survival in advanced disease on chemotherapy.
Collapse
Affiliation(s)
- Ludmila Katherine Martin
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, M365 Starling Loving Hall, 320 West 10th Avenue, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
923
|
Dovzhanskiy DI, Arnold SM, Hackert T, Oehme I, Witt O, Felix K, Giese N, Werner J. Experimental in vivo and in vitro treatment with a new histone deacetylase inhibitor belinostat inhibits the growth of pancreatic cancer. BMC Cancer 2012; 12:226. [PMID: 22681698 PMCID: PMC3407493 DOI: 10.1186/1471-2407-12-226] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 06/08/2012] [Indexed: 02/07/2023] Open
Abstract
Background Treatment options for pancreatic ductal adenocarcinoma (PDAC) are limited. Histone deacetylase inhibitors are a new and promising drug family with strong anticancer activity. The aim of this study was to examine the efficacy of in vitro and in vivo treatment with the novel pan-HDAC inhibitor belinostat on the growth of human PDAC cells. Methods The proliferation of tumour cell lines (T3M4, AsPC-1 and Panc-1) was determined using an MTT assay. Apoptosis was analysed using flow cytometry. Furthermore, p21Cip1/Waf1 and acetylated histone H4 (acH4) expression were confirmed by immunoblot analysis. The in vivo effect of belinostat was studied in a chimeric mouse model. Antitumoural activity was assessed by immunohistochemistry for Ki-67. Results Treatment with belinostat resulted in significant in vitro and in vivo growth inhibition of PDAC cells. This was associated with a dose-dependent induction of tumour cell apoptosis. The apoptotic effect of gemcitabine was further enhanced by belinostat. Moreover, treatment with belinostat increased expression of the cell cycle regulator p21Cip1/Waf1 in Panc-1, and of acH4 in all cell lines tested. The reductions in xenograft tumour volumes were associated with inhibition of cell proliferation. Conclusion Experimental treatment of human PDAC cells with belinostat is effective in vitro and in vivo and may enhance the efficacy of gemcitabine. A consecutive study of belinostat in pancreatic cancer patients alone, and in combination with gemcitabine, could further clarify these effects in the clinical setting.
Collapse
|
924
|
Nakai Y, Isayama H, Sasaki T, Sasahira N, Tsujino T, Toda N, Kogure H, Matsubara S, Ito Y, Togawa O, Arizumi T, Hirano K, Tada M, Omata M, Koike K. A multicentre randomised phase II trial of gemcitabine alone vs gemcitabine and S-1 combination therapy in advanced pancreatic cancer: GEMSAP study. Br J Cancer 2012; 106:1934-1939. [PMID: 22555398 PMCID: PMC3388559 DOI: 10.1038/bjc.2012.183] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 03/27/2012] [Accepted: 04/03/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND This randomised phase II trial compared gemcitabine alone vs gemcitabine and S-1 combination therapy in advanced pancreatic cancer. METHODS Patients were randomly assigned to 4-week treatment with gemcitabine alone (1000, mg m(-2) gemcitabine by 30-min infusion on days 1, 8, and 15) or gemcitabine and S-1 combination therapy (1000, mg m(-2) gemcitabine by 30-min infusion on days 1 and 15 and 40 mg m(-2) S-1 orally twice daily on days 1-15). The primary end point was progression-free survival (PFS). RESULTS Between July 2006 and February 2009, 106 patients were enrolled. The PFS in gemcitabine and S-1 combination arm was significantly longer than in gemcitabine arm (5.4 vs 3.6 months), with a hazard ratio of 0.64 (P=0.036). Overall survival (OS) for gemcitabine and S-1 combination was longer than that for gemcitabine monotherapy (13.5 vs 8.8 months), with a hazard ratio of 0.72 (P=0.104). Overall, grade 3 or 4 adverse events were similar in both arms. CONCLUSION Gemcitabine and S-1 combination therapy demonstrated longer PFS in advanced pancreatic cancer. Improved OS duration of 4.7 months was found for gemcitabine and S-1 combination therapy, though this was not statistically significant.
Collapse
Affiliation(s)
- Y Nakai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - H Isayama
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - T Sasaki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - N Sasahira
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - T Tsujino
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - N Toda
- Department of Gastroenterology, Mitsui Memorial Hospital, Tokyo, Japan
| | - H Kogure
- Department of Gastroenterology, Kanto Central Hospital, Tokyo, Japan
| | - S Matsubara
- Department of Gastroenterology, Kanto Central Hospital, Tokyo, Japan
| | - Y Ito
- Department of Gastroenterology, Japanese Red Cross Hospital, Tokyo, Japan
| | - O Togawa
- Department of Gastroenterology, JR Tokyo General Hospital, Tokyo, Japan
| | - T Arizumi
- Department of Gastroenterology, Teikyo Chiba Medical Center, Chiba, Japan
| | - K Hirano
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - M Tada
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| | - M Omata
- Yamanashi Prefectural Hospital Organization, Yamanashi, Japan
| | - K Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
925
|
Sykes PD, Neoptolemos JP, Costello E, Halloran CM. Nanotechnology advances in upper gastrointestinal, liver and pancreatic cancer. Expert Rev Gastroenterol Hepatol 2012; 6:343-56. [PMID: 22646256 DOI: 10.1586/egh.12.13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancers of the upper GI tract, liver and pancreas have some of the poorest prognoses of any malignancies. Advances in diagnosis and treatment are sorely needed to improve the outcomes of patients. Nanotechnology offers the potential for constructing tailor-made therapies capable of targeting specific cancers. The particles themselves may be endowed with multifunctional properties that can be exploited for both diagnosis and treatment. Although development of therapies is still in the early stages, the use of nanoparticles (NPs) is widespread in diagnostic applications and will probably involve all areas of medicine in the future. Research into NPs is ongoing for upper gastrointestinal, liver and pancreatic cancers, and their use is becoming increasingly popular as contrast media for radiological investigations. Although more sophisticated technologies capable of active targeting are still in the early stages of assessment for clinical use, a small number of NP-based therapies are in clinical use.
Collapse
Affiliation(s)
- Paul D Sykes
- Liverpool Cancer Research UK Centre, Department of Molecular and Clinical Cancer Studies, University of Liverpool, Daulby Street, Liverpool, L69 3GA, UK
| | | | | | | |
Collapse
|
926
|
Zhang Y, Frampton AE, Kyriakides C, Bong JJ, Habib N, Ahmad R, Jiao LR. Loco-recurrence after resection for ductal adenocarcinoma of the pancreas: predictors and implications for adjuvant chemoradiotherapy. J Cancer Res Clin Oncol 2012; 138:1063-71. [PMID: 22392075 DOI: 10.1007/s00432-012-1165-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 01/25/2012] [Indexed: 12/29/2022]
Abstract
PURPOSE Loco (regional)-recurrence rate after pancreaticoduodenectomy (PD) for pancreatic ductal adenocarcinoma (PDAC) remains high, and the efficiency of adjuvant chemoradiotherapy is still debated. We aimed to assess predictors of loco-recurrence in order to tailor the indications for adjuvant chemoradiotherapy. METHODS Patients who underwent PD for PDAC between January 2001 and December 2010 were retrieved from a prospective database. Tumor recurrence was categorized as either loco-recurrence or distant recurrence. Clinicopathological characteristics and survivals were compared between patients with different recurrence patterns. The predictors for loco-recurrence were assessed. RESULTS Seventy-nine patients were included. Loco-recurrence alone was identified in 22 patients (27.8%), distant recurrence alone in 33 (41.8%), both loco- and distant recurrences in 17 (21.5%) and no recurrence in 7 (8.9%). Median survival after recurrence (SAR) was significantly better in patients with loco-recurrence alone than in those with distant recurrence alone (10.4 vs. 5.0 months, P = 0.002) or in those with both loco- and distant recurrences (10.4 vs. 5.8 months, P = 0.044); the survival for patients with distant recurrence alone and those with both patterns was identical. Patients with early recurrence had a significantly poorer SAR than those with late recurrence (median, 5.5 vs. 9.0 months, P = 0.001). Logistic regression analysis revealed that positive resection margin (P = 0.001, HR = 14.532; 95% CI 7.399-38.466), early T stage (P = 0.018, HR = 0.014; 95% CI 0.000-0.475) and large tumor size (P = 0.030, HR = 4.345; 95% CI 1.152-16.391) were the determinant factors directly related to loco-recurrence alone. CONCLUSIONS Patients with PDAC loco-recurrence alone had a significantly better SAR than those with distant recurrence. Adjuvant chemoradiotherapy should be considered to reduce loco-recurrence further and improve long-term survival.
Collapse
Affiliation(s)
- Yaojun Zhang
- HPB Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London W12 0HS, UK
| | | | | | | | | | | | | |
Collapse
|
927
|
Horgan AM, Amir E, Walter T, Knox JJ. Adjuvant therapy in the treatment of biliary tract cancer: a systematic review and meta-analysis. J Clin Oncol 2012; 30:1934-40. [PMID: 22529261 DOI: 10.1200/jco.2011.40.5381] [Citation(s) in RCA: 502] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The benefit of adjuvant therapy (AT) for biliary tract cancer (BTC) is unclear, with conflicting results from nonrandomized studies. We report a systematic review and meta-analysis to determine the impact of AT on survival. METHODS Studies published between 1960 and November 2010, which evaluated adjuvant chemotherapy (CT), radiotherapy (RT), or both (CRT) compared with curative-intent surgery alone for resected BTC were included. Only tumors of the gallbladder and bile ducts were assessed. Published data were extracted and computed into odds ratios (ORs) for death at 5 years. Subgroup analyses of benefit based on lymph node (LN) or resection margin positivity (R1) were prespecified. Data were weighted by generic inverse variance and pooled using random-effect modeling. RESULTS Twenty studies involving 6,712 patients were analyzed. There was a nonsignificant improvement in overall survival with any AT compared with surgery alone (pooled OR, 0.74; P = .06). There was no difference between gallbladder and bile duct tumors (P = .68). The association was significant when the two registry analyses were excluded. Those receiving CT or CRT derived statistically greater benefit than RT alone (OR, 0.39, 0.61, and 0.98, respectively; P = .02). The greatest benefit for AT was in those with LN-positive disease (OR, 0.49; P = .004) and R1 disease (OR, 0.36; P = .002). CONCLUSION This analysis supports AT for BTC. Prospective randomized trials are needed to provide better rationale for this commonly used strategy. On the basis of our data, such trials could involve two active comparators rather than a no-treatment arm among patients with LN-positive or R1 disease.
Collapse
Affiliation(s)
- Anne M Horgan
- Princess Margaret Hospital, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
928
|
Kircher SM, Krantz SB, Nimeiri HS, Mulcahy MF, Munshi HG, Benson AB. Therapy of locally advanced pancreatic adenocarcinoma: unresectable and borderline patients. Expert Rev Anticancer Ther 2012; 11:1555-65. [PMID: 21999129 DOI: 10.1586/era.11.125] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Systemic chemotherapy for advanced pancreatic cancer is commonly used in practice; however, the optimal strategy for both neoadjuvant and adjuvant therapy in this disease remains controversial. A particular challenge remains in patients who are considered to be locally advanced and either unresectable or borderline resectable. Offering optimal neoadjuvant therapy to this group of patients may give them the opportunity to have a curative surgical approach. This article will discuss the potential role of neoadjuvant therapy in borderline, potentially resectable pancreatic cancer. It will also discuss areas of interest in potential targets as the biology of pancreatic adenocarcinoma is further explored.
Collapse
Affiliation(s)
- Sheetal M Kircher
- Division of Hematology/Oncology, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
929
|
Herreros-Villanueva M, Hijona E, Cosme A, Bujanda L. Adjuvant and neoadjuvant treatment in pancreatic cancer. World J Gastroenterol 2012; 18:1565-72. [PMID: 22529684 PMCID: PMC3325521 DOI: 10.3748/wjg.v18.i14.1565] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Revised: 10/23/2011] [Accepted: 01/22/2012] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma is one of the most aggressive human malignancies, ranking 4th among causes for cancer-related death in the Western world including the United States. Surgical resection offers the only chance of cure, but only 15 to 20 percent of cases are potentially resectable at presentation. Different studies demonstrate and confirm that advanced pancreatic cancer is among the most complex cancers to treat and that these tumors are relatively resistant to chemotherapy and radiotherapy. Currently there is no consensus around the world on what constitutes “standard” adjuvant therapy for pancreatic cancer. This controversy derives from several studies, each fraught with its own limitations. Standards of care also vary somewhat with regard to geography and economy, for instance chemo-radiotherapy followed by chemotherapy or vice versa is considered the optimal therapy in North America while chemotherapy alone is the current standard in Europe. Regardless of the efforts in adjuvant and neoadjuvant improved therapy, the major goal to combat pancreatic cancer is to find diagnostic markers, identifying the disease in a pre-metastatic stage and making a curative treatment accessible to more patients. In this review, authors examined the different therapy options for advanced pancreatic patients in recent years and the future directions in adjuvant and neoadjuvant treatments for these patients.
Collapse
|
930
|
Ansari D, Chen BC, Dong L, Zhou MT, Andersson R. Pancreatic cancer: translational research aspects and clinical implications. World J Gastroenterol 2012; 18:1417-24. [PMID: 22509073 PMCID: PMC3319937 DOI: 10.3748/wjg.v18.i13.1417] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 12/29/2011] [Accepted: 01/18/2012] [Indexed: 02/06/2023] Open
Abstract
Despite improvements in surgical techniques and adjuvant chemotherapy, the overall mortality rates in pancreatic cancer have generally remained relatively unchanged and the 5-year survival rate is actually below 2%. This paper will address the importance of achieving an early diagnosis and identifying markers for prognosis and response to therapy such as genes, proteins, microRNAs or epigenetic modifications. However, there are still major hurdles when translating investigational biomarkers into routine clinical practice. Furthermore, novel ways of secondary screening in high-risk individuals, such as artificial neural networks and modern imaging, will be discussed. Drug resistance is ubiquitous in pancreatic cancer. Several mechanisms of drug resistance have already been revealed, including human equilibrative nucleoside transporter-1 status, multidrug resistance proteins, aberrant signaling pathways, microRNAs, stromal influence, epithelial-mesenchymal transition-type cells and recently the presence of cancer stem cells/cancer-initiating cells. These factors must be considered when developing more customized types of intervention ("personalized medicine"). In the future, multifunctional nanoparticles that combine a specific targeting agent, an imaging probe, a cell-penetrating agent, a biocompatible polymer and an anti-cancer drug may become valuable for the management of patients with pancreatic cancer.
Collapse
|
931
|
Abstract
OBJECTIVES The aims of this study were to study the biological and clinical significance of 3 main proteins of the mitogen-activated protein kinase (MAPK) signaling pathway, ERK1/2, P38, and MKK4, in a series of patients having pancreatic adenocarcinomas treated by surgery. METHODS We examined the immunohistochemical expression of 3 MAPK proteins, ERK1/2, P38, and MKK4 in 99 surgically resected pancreatic ductal adenocarcinomas. Tumor protein expression was studied with regard to pathological characteristics and to postsurgical recurrence-free and overall survivals. RESULTS MKK4 expression was related to tumor cell proliferation, evaluated by the Ki67 index (P < 0.01). ERK1/2 expression was related to a shorter recurrence-free survival on both univariate and multivariate analysis (P < 0.01; odds ratio, 8.39; 95% confidence interval, 2.68-26.26) independently of lymph node metastases and tumor size, and to a shorter overall survival (P = 0.01) on univariate analysis. In patients without postsurgical treatment, both ERK1/2 and P38 tumor expression correlated with a shorter recurrence-free survival (P < 0.01 and P = 0.02, respectively). CONCLUSIONS The results of our study suggest that in pancreatic ductal adenocarcinomas, the MKK4 protein was directly related to high cell proliferation, and that tumor ERK1/2 and P38 expression correlated to shorter postsurgical recurrence-free and overall survivals.
Collapse
|
932
|
Neo-adjuvant chemoradiation therapy using S-1 followed by surgical resection in patients with pancreatic cancer. J Gastrointest Surg 2012; 16:784-92. [PMID: 22160780 DOI: 10.1007/s11605-011-1795-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/23/2011] [Indexed: 01/31/2023]
Abstract
OBJECTIVE The aim of this study was to compare short-term surgical results in pancreatic cancer patients who underwent surgical resection after neo-adjuvant chemoradiation therapy (NACRT) using S-1. METHODS The study population comprised 77 patients with pancreatic cancer between 2006 and 2010. Out of 34 patients who underwent staging laparoscopy between 2008 and 2010, 31 patients without occult distant organ metastasis underwent chemoradiation and of whom 30 underwent pancreatectomy (NACRT group). Of the other 43 patients, 36 underwent surgical resection in 2006-2008, followed by adjuvant therapy (adjuvant group). The primary endpoint was frequency of pathological curative resection (R0). RESULTS The new regimen of NACRT was feasible and safe. Twenty-eight of 30 (93%) patients in the NACRT group had R0 resection, which was significantly higher than in the adjuvant group (21 of 36 patients, 58%, p = 0.005). The number and extent of metastatic lymph nodes in the NACRT group (1 (0-25), N0/1; 18 of 38) was significantly lower than in the adjuvant group (2 (0-19), N0/1; 23 of 30), p = 0.0363). The frequency of intractable ascites in the NACRT group (eight of 30) was significantly higher than in the adjuvant group (two of 36, p = 0.035). CONCLUSION Neo-adjuvant chemoradiation therapy using S-1 followed by pancreatectomy can improve the rate of pathologically curative resection and reduces the number and extent of lymph node metastasis.
Collapse
|
933
|
Safety and Optimal Management of Hepatic Arterial Infusion Chemotherapy After Pancreatectomy for Pancreatobiliary Cancer. AJR Am J Roentgenol 2012; 198:923-30. [DOI: 10.2214/ajr.11.6751] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
934
|
Abrams RA. Radiotherapy in the adjuvant management of pancreatic adenocarcinoma: is it helpful? Expert Rev Gastroenterol Hepatol 2012; 6:149-61. [PMID: 22375521 DOI: 10.1586/egh.12.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Curative-intent management for pancreatic adenocarcinoma requires gross total resection. Only 15-20% of patients are eligible for resection and of these only approximately 20% are long-term survivors. Adjuvant and neoadjuvant approaches have been, and are being, sought to improve upon surgical results. Radiotherapy has a role in the adjuvant management of many gastrointestinal malignancies and historically has also had a role in the adjuvant management of pancreatic adenocarcinoma. However, the role of radiotherapy in the management of pancreatic adenocarcinoma has been called into question. This review examines this controversy and the underlying issues and considers current efforts towards resolving them, as well as some likely future developments.
Collapse
Affiliation(s)
- Ross A Abrams
- Department of Radiation Oncology, Rush University Medical Center, Atrium Bldg, Ground Floor, 500 S. Paulina, Chicago, IL 60612, USA.
| |
Collapse
|
935
|
Baschnagel A, Shah C, Margolis J, Nadeau L, Stein J, Jury R, Robertson JM. Survival after chemoradiation in resected pancreatic cancer: the impact of adjuvant gemcitabine. Int J Radiat Oncol Biol Phys 2012; 83:e331-5. [PMID: 22420967 DOI: 10.1016/j.ijrobp.2012.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 01/03/2012] [Accepted: 01/04/2012] [Indexed: 01/04/2023]
Abstract
PURPOSE To evaluate survival in patients with resected pancreatic cancer treated with concurrent chemoradiation with or without adjuvant gemcitabine (Gem). METHODS AND MATERIALS From 1998 to 2010, 86 patients with pancreatic adenocarcinoma who underwent resection were treated with adjuvant concurrent chemoradiation. Thirty-four patients received concurrent 5-fluorouracil-based chemoradiation (5-FU/RT) with traditional field radiation (range, 45-61.2 Gy; median, 50.4 Gy) without further adjuvant therapy. Thirty patients received traditional field 5-FU/RT (range, 45-60.4 Gy; median, 50.4 Gy) with Gem (1,000 mg/m(2) weekly) either before and after radiotherapy or only after radiotherapy. Twenty-two patients received concurrent full-dose Gem (1,000 mg/m(2) weekly)-based chemoradiation (Gem/RT), consisting of involved-field radiation (range, 27-38 Gy; median, 36 Gy) followed by further adjuvant Gem. RESULTS The median age of the cohort was 65 years (range, 40-80 years). Of the patients, 58 had T3 tumors (67%), 22 had T2 tumors (26%), and 6 had T1 tumors (7%). N1 disease was present in 61 patients (71%), whereas 18 patients (21%) had R1 resections. Performance status, lymph node status, and margin status were all similar among the treatment groups. Median follow-up was 19.0 months. Median overall survival (OS) (19.2 months, 19.0 months, and 21.0 months) and 3-year OS rates (26.5%, 27.2%, and 32.1%) were similar among patients with 5-FU/RT with no adjuvant Gem, those with 5-FU/RT with adjuvant Gem, and those with Gem/RT with adjuvant Gem, respectively (p = 0.88). Patients who received adjuvant Gem had a similar median OS (22.1 months) and 3-year OS rate (29%) compared to patients who did not (19.2 months and 26.5%, respectively) (p = 0.62). There was a trend for improved 3-year OS rates in patients with R0 vs. R1 resections (28.1% vs. 14.2%, p = 0.06) and in patients with T1 and T2 vs. T3 tumors (38% vs. 20%, p = 0.09). Node-negative patients had an improved 3-year OS rate (30.1%) when compared with patients with N1 disease (16.2%) (p = 0.02). CONCLUSION In our cohort of patients with resected pancreatic cancer, Gem chemotherapy did not improve OS after chemoradiotherapy.
Collapse
Affiliation(s)
- Andrew Baschnagel
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan 48072, USA
| | | | | | | | | | | | | |
Collapse
|
936
|
Donahue TR, Tran LM, Hill R, Li Y, Kovochich A, Calvopina JH, Patel SG, Wu N, Hindoyan A, Farrell JJ, Li X, Dawson DW, Wu H. Integrative survival-based molecular profiling of human pancreatic cancer. Clin Cancer Res 2012; 18:1352-63. [PMID: 22261810 PMCID: PMC3816537 DOI: 10.1158/1078-0432.ccr-11-1539] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE To carry out an integrative profile of human pancreatic ductal adenocarcinoma (PDAC) to identify prognosis-significant genes and their related pathways. EXPERIMENTAL DESIGN A concordant survival-based whole genome in silico array analysis of DNA copy number, and mRNA and miRNA expression in 25 early-stage PDAC was carried out. A novel composite score simultaneously integrated gene expression with regulatory mechanisms to identify the signature genes with the most levels of prognosis-significant evidence. The predominant signaling pathways were determined via a pathway-based approach. Independent patient cohorts (n = 148 and 42) were then used as in vitro validation of the array findings. RESULTS The composite score identified 171 genes in which expressions were able to define two prognosis subgroups (P = 3.8e-5). Eighty-eight percent (151 of 171) of the genes were regulated by prognosis-significant miRNAs. The phosphoinositide 3-kinase/AKT pathway and SRC signaling were densely populated by prognosis-significant genes and driven by genomic amplification of SRC and miRNA regulation of p85α and CBL. On tissue microarray validation (n = 148), p85α protein expression was associated with improved survival for all patients (P = 0.02), and activated P-SRC (Y418) was associated shorter survival for patients with low-grade histology tumors (P = 0.04). Interacting P-SRC and p85α revealed that they define two distinct PDAC patient subgroups (P = 0.0066). Furthering the importance of these pathways, CBL protein expression was associated with improved survival (P = 0.03) on a separate cohort (n = 42). CONCLUSIONS These pathways and related genes may represent putative clinical biomarkers and possible targets of individualized therapy in the distinct patient subgroups they define.
Collapse
Affiliation(s)
- Timothy R. Donahue
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Institute for Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Linh M. Tran
- Institute for Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Reginald Hill
- Institute for Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Yunfeng Li
- Institute for Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Anne Kovochich
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Joseph H. Calvopina
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Sanjeet G. Patel
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Nanping Wu
- Institute for Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Antreas Hindoyan
- Institute for Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - James J. Farrell
- Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Xinmin Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - David W. Dawson
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Hong Wu
- Institute for Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
- Eli and Edy the Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| |
Collapse
|
937
|
Trakul N, Koong AC, Maxim PG, Chang DT. Modern radiation therapy techniques for pancreatic cancer. Gastroenterol Clin North Am 2012; 41:223-35. [PMID: 22341260 DOI: 10.1016/j.gtc.2011.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Radiation therapy is a rapidly evolving field, and recent technical advances have spurred an increasing number of new treatments as well as marked improvements in previously existing treatments. Despite a growing body of published evidence demonstrating that radiotherapy for the treatment of pancreatic cancer is improving in efficacy and safety, the ultimate effect on patient outcomes remains to be seen. It is an unfortunate fact that the majority of pancreatic cancer patients will ultimately have metastases and succumb to distant disease. Thus, improvements in local tumor control engendered by these recent advances will have little impact on overall survival without the coincident development of better systemic treatment regimens.
Collapse
Affiliation(s)
- Nicholas Trakul
- Department of Radiation Oncology, Stanford University School of Medicine and Cancer Center, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
938
|
Long-term results of surgical resection after preoperative chemoradiation in patients with pancreatic cancer. Pancreas 2012; 41:333-5. [PMID: 22343984 DOI: 10.1097/mpa.0b013e318229758f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
939
|
Páez D, Labonte MJ, Lenz HJ. Pancreatic cancer: medical management (novel chemotherapeutics). Gastroenterol Clin North Am 2012; 41:189-209. [PMID: 22341258 DOI: 10.1016/j.gtc.2011.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pancreatic adenocarcinoma is the fourth leading cause of cancer death and has an extremely poor prognosis: The 5-year survival probability is less than 5% for all stages. The only chance for cure or longer survival is surgical resection; however, only 10% to 20% of patients have resectable disease. Although surgical techniques have improved, most who undergo complete resection experience a recurrence. Adjuvant systemic therapy reduces the recurrence rate and improves outcomes. There is a potential role for radiation therapy as part of treatment for locally advanced disease, although its use in both the adjuvant and neoadjuvant settings remains controversial. Palliative systemic treatment is the only option for patients with metastatic disease. To date, however, only the gemcitabine plus erlotinib combination, and recently the FOLFIRINOX regimen, have been associated with relatively small but statistically significant improvements in OS when compared directly with gemcitabine alone. Although several meta-analyses have suggested a benefit associated with combination chemotherapy, whether this benefit is clinically meaningful remains unclear, particularly in light of the enhanced toxicity associated with combination regimens. There is growing evidence that the exceptionally poor prognosis in PC is caused by the tumor's characteristic abundant desmoplastic stroma that plays a critical role in tumor cell growth, invasion, metastasis, and chemoresistance. Carefully designed clinical trials that include translational analysis will provide a better understanding of the tumor biology and its relation to the host stromal cells. Future directions will involve testing of new targeted agents, understanding the pharmacodynamics of our current targeted agents, searching for predictive and prognostic biomarkers, and exploring the efficacy of different combinations strategies.
Collapse
Affiliation(s)
- David Páez
- Division of Medical Oncology, University of Southern California/Norris Comprehensive Cancer Center, Keck School of Medicine, Los Angeles, CA 90033, USA
| | | | | |
Collapse
|
940
|
Van Laethem JL, Verslype C, Iovanna JL, Michl P, Conroy T, Louvet C, Hammel P, Mitry E, Ducreux M, Maraculla T, Uhl W, Van Tienhoven G, Bachet JB, Maréchal R, Hendlisz A, Bali M, Demetter P, Ulrich F, Aust D, Luttges J, Peeters M, Mauer M, Roth A, Neoptolemos JP, Lutz M. New strategies and designs in pancreatic cancer research: consensus guidelines report from a European expert panel. Ann Oncol 2012; 23:570-576. [PMID: 21810728 DOI: 10.1093/annonc/mdr351] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although the treatment of pancreatic ductal adenocarcinoma (PDAC) remains a huge challenge, it is entering a new era with the development of new strategies and trial designs. Because there is an increasing number of novel therapeutic agents and potential combinations available to test in patients with PDAC, the identification of robust prognostic and predictive markers and of new targets and relevant pathways is a top priority as well as the design of adequate trials incorporating molecular-driven hypothesis. We presently report a consensus strategy for research in pancreatic cancer that was developed by a multidisciplinary panel of experts from different European institutions and collaborative groups involved in pancreatic cancer. The expert panel embraces the concept of exploratory early proof of concept studies, based on the prediction of response to novel agents and combinations, and randomised phase II studies permitting the selection of the best therapeutic approach to go forward into phase III, where the recommended primary end point remains overall survival. Trials should contain as many translational components as possible, relying on standardised tissue and blood processing and robust biobanking, and including dynamic imaging. Attention should not only be paid to the pancreatic cancer cells but also to microenvironmental factors and stem/stellate cells.
Collapse
Affiliation(s)
- J-L Van Laethem
- Gastrointestinal Cancer Unit, Hôpital Erasme, Université Libre de Bruxelles, Brussels.
| | - C Verslype
- Department of Hepatology, Universitair Ziekenhuis Gasthuisberg, Leuven, Belgium
| | - J L Iovanna
- Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - P Michl
- Department of Gastroenterology and Endocrinology, University of Marburg, Marburg, Germany
| | - T Conroy
- Nancy University and Department of Medical Oncology, Centre Alexis Vautrin, Nancy
| | - C Louvet
- Digestive Surgery Department, Institut Mutualiste Montsouris, Paris
| | - P Hammel
- Gastroenterology Department, Hôpital Beaujon, Clichy
| | - E Mitry
- Medical Oncology Department, Institut Curie, Hôpital René-Huguenin, Saint-Cloud
| | - M Ducreux
- Digestive Oncology Department, Institut G. Roussy, Villejuif, France
| | - T Maraculla
- Medical Oncology Department, Hospital Vall d'Hebron, Barcelona, Spain
| | - W Uhl
- Department of Surgery, St Josef-Hospital, Ruhr-University, Bochum, Germany
| | - G Van Tienhoven
- Department of Radiation Oncology, Academic Medical Centre, Amsterdam, The Netherlands
| | - J B Bachet
- Department of Gastroenterology, Hôpital Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - R Maréchal
- Department of Gastroenterology and Hepato-Pancreatology, Gastrointestinal Cancer Unit, Hôpital Universitaire Erasme, Brussels
| | - A Hendlisz
- Department of Gastroenterology, Institut J. Bordet, Brussels
| | - M Bali
- Department of Medical Imaging, Hôpital Erasme, Brussels, Belgium
| | - P Demetter
- Gastrointestinal Cancer Unit, Hôpital Erasme, Université Libre de Bruxelles, Brussels
| | - F Ulrich
- Department of General and Visceral Surgery, J. W. Goethe University Medical Center, Frankfurt
| | - D Aust
- Institute of Pathology, University Hospital Carl Gustav Carus, Dresden
| | - J Luttges
- Caritasklinik St Theresia, Saarbrücken, Germany
| | - M Peeters
- Department of Oncology, Universitair Ziekenhuis Antwerpen, Edegem
| | - M Mauer
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - A Roth
- Department of Surgery, Clinic for Visceral and Transplantation Surgery, Hôpital Universitaire de Genève, Geneva, Switzerland
| | - J P Neoptolemos
- Department of Surgery, University of Liverpool, Liverpool, UK
| | - M Lutz
- Caritasklinik St Theresia, Saarbrücken, Germany
| |
Collapse
|
941
|
Murakami Y, Uemura K, Sudo T, Hashimoto Y, Nakashima A, Kondo N, Nakagawa N, Sueda T. Long-term results of adjuvant gemcitabine plus S-1 chemotherapy after surgical resection for pancreatic carcinoma. J Surg Oncol 2012; 106:174-80. [DOI: 10.1002/jso.23068] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Accepted: 01/24/2012] [Indexed: 01/11/2023]
|
942
|
Ren F, Xu YC, Wang HX, Tang L, Ma Y. Adjuvant chemotherapy, with or without postoperative radiotherapy, for resectable advanced pancreatic adenocarcinoma: continue or stop? Pancreatology 2012; 12:162-9. [PMID: 22487527 DOI: 10.1016/j.pan.2012.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIMS The aim of the article is to perform a focused review of adjuvant chemotherapy with or without radiotherapy for the treatment of resectable pancreatic adenocarcinoma (PAC). METHODS We performed a Medline database search from 1965 to 2010 using the terms "adjuvant," "trial" and "pancreatic cancer". RESULTS Adding adjuvant chemotherapy to patients with resectable PAC was associated with significantly increased median overall survival (OS) (odds ratio[OR]: 1.98, p < 0.001), disease-free survival (DFS) (OR: 2.12, p < 0.001), two-year survival (OR: 1.38, p = 0.04) and five-year survival (OR: 2.16, p = 0.007) compared to surgery alone. There was no statistically significant difference observed with regard to OS (OR:0.99, p = 0.93), DFS (OR:0.99, p = 0.95), and two-year survival (OR: 0.90, p = 0.57) between adjuvant chemoradiotherapy and surgery alone. The further analysis showed that single agent gemcitabine was as active as combined chemotherapy or chemoradiation, which was reflected by an OR of 1.13 (p = 0.26) for OS and1.08 (p = 0.47) for DFS. CONCLUSIONS A significant benefit with regard to DFS and median OS for adjuvant chemotherapy after PAC resection was demonstrated by this analysis. These results do not support the use of adjuvant radiotherapy for PAC.
Collapse
Affiliation(s)
- Fang Ren
- Department of Oncology, Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | | | | | | | | |
Collapse
|
943
|
Shrikhande SV, Barreto SG. Surgery for pancreatic carcinoma: state of the art. Indian J Surg 2012; 74:79-86. [PMID: 23372311 PMCID: PMC3259176 DOI: 10.1007/s12262-011-0373-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 11/10/2011] [Indexed: 02/06/2023] Open
Abstract
Carcinoma of the pancreas remains a malignancy with a generally dismal outcome owing to the delayed presentation of the disease. To date, surgery affords the best outcomes when a complete resection can be achieved. Improvements in imaging, surgical techniques and adjuvant therapies are perceived advancements in the management of this cancer. This article reviews the latest evidence in terms of the diagnosis and management of pancreatic cancer.
Collapse
Affiliation(s)
- Shailesh V. Shrikhande
- Department of GI and HPB Surgical Oncology, Tata Memorial Centre, Ernest Borges Marg, Parel, Mumbai, 400012 India
| | | |
Collapse
|
944
|
Lim KH, Chung E, Khan A, Cao D, Linehan D, Ben-Josef E, Wang-Gillam A. Neoadjuvant therapy of pancreatic cancer: the emerging paradigm? Oncologist 2012; 17:192-200. [PMID: 22250057 PMCID: PMC3286168 DOI: 10.1634/theoncologist.2011-0268] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 11/07/2011] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer remains one of the deadliest cancers due to difficulty in early diagnosis and its high resistance to chemotherapy and radiation. It is now clear that even patients with potentially resectable disease require multimodality treatment including chemotherapy and/or radiation to improve resectability and reduce recurrence. Tremendous efforts are currently being invested in refining preoperative staging to identify optimal surgical candidates, and also in developing various neoadjuvant or adjuvant regimens to improve surgical outcome. Although at present no studies have been done to directly compare the benefit of neoadjuvant versus adjuvant approaches, accumulating evidence suggests that the neoadjuvant approach is probably beneficial for a subset of the patient population, particularly those with borderline resectable disease in which complete surgical resection is almost certainly unachievable. In this article, we review the literature and rationales of neoadjuvant chemotherapy and chemoradiation, as well as their potential limitations and caveats. We also review the pathological findings following neoadjuvant therapies, and potential surgical complications that may be associated with neoadjuvant therapies.
Collapse
Affiliation(s)
- Kian-Huat Lim
- Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Eugene Chung
- Department of Radiation Oncology, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Adeel Khan
- Department of Surgery, Bay Area Medical Center, Aurora Health Care, Marinette, Wisconsin, USA
| | | | | | - Edgar Ben-Josef
- Department of Radiation Oncology, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Andrea Wang-Gillam
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
945
|
Reni M, Balzano G, Aprile G, Cereda S, Passoni P, Zerbi A, Tronconi MC, Milandri C, Saletti P, Rognone A, Fugazza C, Magli A, Muzio ND, Carlo VD, Villa E. Adjuvant PEFG (Cisplatin, Epirubicin, 5-Fluorouracil, Gemcitabine) or Gemcitabine Followed by Chemoradiation in Pancreatic Cancer: A Randomized Phase II Trial. Ann Surg Oncol 2012; 19:2256-63. [DOI: 10.1245/s10434-011-2205-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Indexed: 01/11/2023]
|
946
|
Humphris JL, Chang DK, Johns AL, Scarlett CJ, Pajic M, Jones MD, Colvin EK, Nagrial A, Chin VT, Chantrill LA, Samra JS, Gill AJ, Kench JG, Merrett ND, Das A, Musgrove EA, Sutherland RL, Biankin AV. The prognostic and predictive value of serum CA19.9 in pancreatic cancer. Ann Oncol 2012; 23:1713-22. [PMID: 22241899 PMCID: PMC3387824 DOI: 10.1093/annonc/mdr561] [Citation(s) in RCA: 217] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Current staging methods for pancreatic cancer (PC) are inadequate, and biomarkers to
aid clinical decision making are lacking. Despite the availability of the serum marker
carbohydrate antigen 19.9 (CA19.9) for over two decades, its precise role in the
management of PC is yet to be defined, and as a consequence, it is not widely used. Methods We assessed the relationship between perioperative serum CA19.9 levels, survival and
adjuvant chemotherapeutic responsiveness in a cohort of 260 patients who underwent
operative resection for PC. Results By specifically assessing the subgroup of patients with detectable CA19.9, we
identified potential utility at key clinical decision points. Low postoperative CA19.9
at 3 months (median survival 25.6 vs 14.8 months,
P = 0.0052) and before adjuvant chemotherapy were
independent prognostic factors. Patients with postoperative CA 19.9 levels >90 U/ml
did not benefit from adjuvant chemotherapy
(P = 0.7194) compared with those with a CA19.9 of
≤90 U/ml (median 26.0 vs 16.7 months, P = 0.0108).
Normalization of CA19.9 within 6 months of resection was also an independent favorable
prognostic factor (median 29.9 vs 14.8 months,
P = 0.0004) and normal perioperative CA19.9 levels
identified a good prognostic group, which was associated with a 5-year survival of
42%. Conclusions Perioperative serum CA19.9 measurements are informative in patients with detectable
CA19.9 (defined by serum levels of >5 U/ml) and have potential clinical utility in
predicting outcome and response to adjuvant chemotherapy. Future clinical trials should
prioritize incorporation of CA19.9 measurement at key decision points to prospectively
validate these findings and facilitate implementation.
Collapse
Affiliation(s)
- J L Humphris
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
947
|
Asuthkar S, Rao JS, Gondi CS. Drugs in preclinical and early-stage clinical development for pancreatic cancer. Expert Opin Investig Drugs 2012; 21:143-52. [PMID: 22217246 DOI: 10.1517/13543784.2012.651124] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Pancreatic cancer (PC) is the fourth leading cause of cancer-related deaths in the US and Europe, and the lethality of this cancer is demonstrated by the fact that the annual incidences are approximately equal to the annual deaths. Current therapy for PC is multimodal, involving surgery and chemotherapy. Clinical symptoms are unspecific, and consequently about 85% of patients with PC are diagnosed at advanced tumor stages without any surgical therapy options. Since the therapeutic rates for PC are so dismal, it is essential to review the clinical targets for diagnosis and treatment of this lethal cancer. AREAS COVERED In this review, we discuss potential treatment options for PC by identifying molecular targets including those involved in cell proliferation, survival, migration, invasion and angiogenesis. Targeting these molecules in combination with surgery could improve the clinical outcome for PC patients. EXPERT OPINION For a decade, gemcitabine has remained the single first-line chemotherapeutic agent for advanced adenocarcinoma of the pancreas; however, less than 25% of patients benefit from gemcitabine. The reason for frequent reoccurrence of PC after conventional methods such as surgery, radiation and/or chemotherapy is due to the lack of understanding of the basic underlying metabolic cause of the cancer and thus consequently remains uncorrected. Our understanding of drug resistance in PC is still not clear and may be answered by focusing on new useful biomarkers and their role in chemo- and radioresistance.
Collapse
Affiliation(s)
- Swapna Asuthkar
- University of Illinois College of Medicine, Cancer Biology and Pharmacology, One Illini Drive, Peoria, 61605, USA
| | | | | |
Collapse
|
948
|
Surviving cells after treatment with gemcitabine or 5-fluorouracil for the study of de novo resistance of pancreatic cancer. Cancer Lett 2012; 314:119-25. [DOI: 10.1016/j.canlet.2011.09.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Revised: 09/06/2011] [Accepted: 09/18/2011] [Indexed: 01/01/2023]
|
949
|
Fischer R, Breidert M, Keck T, Makowiec F, Lohrmann C, Harder J. Early recurrence of pancreatic cancer after resection and during adjuvant chemotherapy. Saudi J Gastroenterol 2012; 18:118-21. [PMID: 22421717 PMCID: PMC3326972 DOI: 10.4103/1319-3767.93815] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND/AIM Adjuvant chemotherapy for 6 months is the current standard of care after potentially curative resection of pancreatic cancer and yields an overall survival of 15-20 months. Early tumor recurrence before or during adjuvant chemotherapy has not been evaluated so far. These patients may not benefit from adjuvant treatment. PATIENTS AND METHODS Thirty-five patients with resection of ductal pancreatic carcinoma and adjuvant chemotherapy with gemcitabine were analyzed between 2005 and 2007. All patients had a computed tomography (CT) scan before and during adjuvant chemotherapy after 2-3 months, 12/35 patients had a histologically confirmed R1 resection. Recurrence of pancreatic cancer was determined by CT scan and the clinical course. RESULTS Median survival of 35 patients with resected pancreatic cancer was 19.7 months, and the 2-year survival was 44%. Thirteen (37%) of the 35 patients analyzed with a CT scan showed tumor recurrence during adjuvant chemotherapy. Overall survival of patients with tumor recurrence was 9.3 months with a 2-year survival rate of 13%, whereas median overall survival of patients without early relapse was 26.3 months (P<0.001). Local recurrence of pancreatic cancer occurred in 38% (5/13); 46% (6/13) of patients developed distant metastasis, and 38% (5/13) developed lymph node metastasis. Early tumor recurrence during or adjuvant chemotherapy did not correlate with R status (R1 vs R0, P=0.69), whereas histologically confirmed lymph node invasion (pN0 vs pN1) and grading showed a statistically significant correlation with early relapse (P<0.05). CONCLUSION A significant fraction of patients with resected pancreatic cancer have early relapse during adjuvant chemotherapy, especially those with lymph node metastasis. Radiologic examinations prior to and during adjuvant chemotherapy will help to identify patients with tumor recurrence who are unlikely to benefit from adjuvant treatment and will need individualized palliative chemotherapy.
Collapse
Affiliation(s)
- Richard Fischer
- Department of Medicine II, University Medical Centre Freiburg, Germany
| | - Matthias Breidert
- Department of Internal Medicine I, Klinik Kösching, Germany,Address for correspondence: Dr. Matthias Breidert, Department of Internal Medicine I, District Hospital Clinic Altmühltal, Krankenhausstrasse 19, D-85092 Kösching. E-mail:
| | - Tobias Keck
- Department of Visceral Surgery, University Medical Centre Freiburg, Germany
| | - Frank Makowiec
- Department of Visceral Surgery, University Medical Centre Freiburg, Germany
| | - Christian Lohrmann
- Department of Diagnostic Radiology, University Medical Centre Freiburg, Germany
| | - Jan Harder
- Department of Medicine II, Hegau-Bodensee-Klinikum Singen, Germany
| |
Collapse
|
950
|
Abstract
Pancreatic cancer is a challenging malignancy to treat, as less than one-fifth of diagnosed cases are resectable, surgery is complex and postoperative recovery slow, treated patients tend to relapse and overall survival rates are low. It is one of the leading causes of cancer-related mortality. Adjuvant therapy has been employed in resectable disease, to target micrometastases and improve prognosis. Chemotherapy, chemoradiotherapy (chemoRT) and chemoradiotherapy (chemoRT) followed on by chemotherapy have been evaluated in randomised controlled trials. The European Study Group for Pancreatic Cancer (ESPAC)-1 and CONKO-001 trials clearly established the survival advantage of adjuvant chemotherapy with 5 fluorouracil (5FU) plus folinic acid and gemcitabine respectively over no chemotherapy. The ESPAC-3 (version 2) trial demonstrated equivalence between 5FU plus folinic acid and gemcitabine in terms of survival parameters, though gemcitabine had a better toxicity profile. The results of these key studies, together with smaller ones have been subjected to meta-analyses, with confirmation of improved survival with adjuvant systemic chemotherapy. The EORTC-40891 and ESPAC-1 trials found no survival advantage with adjuvant chemoRT compared to observation, and this has been reflected in a subsequent meta-analysis. The popularisation of chemoRT, with follow on chemotherapy (versus observation) was based on the small underpowered GITSG trial. The ESPAC-1 trial was unable to find a survival benefit for chemoRT, with follow on chemotherapy compared to observation. The RTOG-9704 trial assessed chemoRT with follow on chemotherapy in both arms and found no difference between survival in the gemcitabine and 5FU arms. There has never been a published head-to-head randomised comparison of adjuvant chemotherapy to chemoRT, with follow on chemotherapy. Ongoing randomised trials are looking into adjuvant combination chemotherapy, chemotherapy with follow on chemoRT, and neoadjuvant therapy. Novel agents continue to be assessed in early phase trials with a major emphasis on predictive and prognostic biomarkers. Based on the available evidence, adjuvant chemotherapy with gemcitabine or 5FU/folinic acid is the current recommended gold standard in the management of resected pancreatic cancer.
Collapse
Affiliation(s)
- Asma Sultana
- Department of Molecular and Clinical Cancer Medicine Centre, University of Liverpool, Liverpool, L69 3GA, UK
| | | | | | | |
Collapse
|