1
|
Jain AJ, Maxwell JE, Katz MHG, Snyder RA. Surgical Considerations for Neoadjuvant Therapy for Pancreatic Adenocarcinoma. Cancers (Basel) 2023; 15:4174. [PMID: 37627202 PMCID: PMC10453019 DOI: 10.3390/cancers15164174] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a challenging disease process with a 5-year survival rate of only 11%. Neoadjuvant therapy in patients with localized pancreatic cancer has multiple theoretical benefits, including improved patient selection for surgery, early delivery of systemic therapy, and assessment of response to therapy. Herein, we review key surgical considerations when selecting patients for neoadjuvant therapy and curative-intent resection. Accurate determination of resectability at diagnosis is critical and should be based on not only anatomic criteria but also biologic and clinical criteria to determine optimal treatment sequencing. Borderline resectable or locally advanced pancreatic cancer is best treated with neoadjuvant therapy and resection, including vascular resection and reconstruction when appropriate. Lastly, providing nutritional, prehabilitation, and supportive care interventions to improve patient fitness prior to surgical intervention and adequately address the adverse effects of therapy is critical.
Collapse
Affiliation(s)
| | | | | | - Rebecca A. Snyder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.J.)
| |
Collapse
|
2
|
Piper M, Ross RB, Hu J, Watanabe S, Knitz M, Mehrotra S, Shulick R, Chiaro MD, Karam SD. Vasculitis, CA19-9, and Perineural Invasion Differentially Predict Response and Surgical Outcome in Pancreatic Ductal Adenocarcinoma. Int J Radiat Oncol Biol Phys 2023:S0360-3016(22)03692-6. [PMID: 36599398 DOI: 10.1016/j.ijrobp.2022.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/01/2022] [Accepted: 12/24/2022] [Indexed: 01/02/2023]
Abstract
PURPOSE Curative intent treatment of pancreatic adenocarcinoma (PDAC) relies on surgical resection. Modern treatment protocols focus on optimizing neoadjuvant therapy to increase resectability and improve oncologic outcomes. To elucidate differences in outcomes, we investigated the relationship between neoadjuvant chemotherapy (NAC), either with or without stereotactic body radiation therapy (SBRT), and vascular inflammation, surgical outcomes, and the resultant transcriptomic changes. METHODS AND MATERIALS Clinical data were collected from patients with borderline resectable PDAC (clinical T3-T4N0-1) who underwent NAC or NAC-SBRT followed by curative intent resection between 2014 and 2019. Vascular structures on surgical specimens were histologically evaluated for vasculitis. RNA sequencing was used to evaluate differential gene expression and to generate enrichment maps. Multivariate analysis was used to analyze the relationship between patient characteristics and oncological outcome. RESULTS In total, 46 patients met inclusion criteria (n = 12 NAC, n = 34 NAC-SBRT) with a median follow-up of 20.1 months. All patients underwent curative resection, with 91.3% achieving R0. There was no significant difference in patterns of failure, overall survival, or progression-free survival between NAC and NAC-SBRT groups. Patients with vasculitis had a lower median overall survival compared with those without (14.5 vs 28.3 months; hazard ratio, 12.96; 95% confidence interval, 3.55-47.28; P < .001). There was no significant correlation between inflammation and surgical complications or pathologic response. Neoadjuvant therapy did not have a significant effect on development of vasculitis (odds radio, 1.64 for NAC-SBRT; 95% confidence interval, 0.40-8.43; P = .52). Predictors of poor survival included perineural invasion and high baseline carbohydrate antigen 19-9 (CA19-9) (>191 U/mL). Patients with robust CA19-9 (>20% decrease) responses to neoadjuvant therapy had enrichment in immune response, chemotaxis, and cytotoxic T-cell and natural killer-cell proliferation. CONCLUSIONS Vasculitis predicts for poor survival outcomes in patients with PDAC; NAC-SBRT did not increase the rate of vasculitis compared with NAC. Perineural invasion and CA19-9 remain strong prognosticators. Understanding and optimizing immune interactions remain a crucial hurdle in achieving response in pancreatic cancer.
Collapse
Affiliation(s)
- Miles Piper
- Departments of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Richard Blake Ross
- Departments of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Junxiao Hu
- Departments of Biostatistics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shuichi Watanabe
- Departments of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael Knitz
- Departments of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sanjana Mehrotra
- Departments of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Richard Shulick
- Departments of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Marco Del Chiaro
- Departments of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sana D Karam
- Departments of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
3
|
Sha M, Kunduzi B, Froghi S, Quaglia A, Davidson B, Fusai GK. Role of circulating exosomal biomarkers and their diagnostic accuracy in pancreatic cancer. JGH Open 2022; 7:30-39. [PMID: 36660044 PMCID: PMC9840196 DOI: 10.1002/jgh3.12848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/07/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022]
Abstract
Background and Aim New biomarkers have the potential to facilitate early diagnosis of pancreatic cancer (PC). Circulating exosomes are cell-derived protein complexes containing RNA that can be used as indicators of cancer development. The aim of this review is to evaluate the current literature involving PC patient groups for highly accurate exosomal biomarkers. Methods The literature search followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Eight-hundred and seventy-five studies were identified across various databases (Ovid MEDLINE, Embase, and Cochrane) published between 2009 and 2020. Nine studies fulfilled the inclusion criteria: human PC patients, diagnosis as outcome of interest, serum biomarker of exosomal content, reporting of diagnostic values, and disease progress. Area under the curve (AUC) of the exosomal biomarker was compared against that of CA19-9. Results Nine papers were reviewed for relevant outcomes based on the inclusion criteria. These studies involved 565 participants (331 PC, 234 controls; male/female ratio 1.21; mean age 64.1). Tumor staging was reported in all studies, with 45.6% of PC patients diagnosed with early-stage PC (T1-2). The mRNA panel (ARG1, CD63, CK18, Erbb3, GAPDH, H3F3A, KRAS, ODC1) and GPC 1 reported the highest performing sensitivity and specificity at 100% each. The microRNA panel (miR-10b, miR-21, miR-30c, miR-181a, and miR-let7a), mRNA panel (ARG1, CD63, CK18, Erbb3, GAPDH, H3F3A, KRAS, ODC1), and GPC 1 showed a perfect AUC of 1.0. Five studies compared the AUC of the exosomal biomarker against CA19-9, each being superior to that of CA19-9. Conclusion The potential of exosomal biomarkers remains promising in PC diagnosis. Standardization of future studies will allow for larger comparative analyses and overcoming contrasting findings.
Collapse
Affiliation(s)
- Menazir Sha
- University College London, Medical SchoolLondonUK,Division of Surgery and Interventional Sciences/UCLRoyal Free HospitalLondonUK
| | | | - Saied Froghi
- Division of Surgery and Interventional Sciences/UCLRoyal Free HospitalLondonUK,Department of HPB and Liver TransplantationRoyal Free HospitalLondonUK
| | | | - Brian Davidson
- Division of Surgery and Interventional Sciences/UCLRoyal Free HospitalLondonUK,Department of HPB and Liver TransplantationRoyal Free HospitalLondonUK
| | - Giuseppe K Fusai
- Division of Surgery and Interventional Sciences/UCLRoyal Free HospitalLondonUK,Department of HPB and Liver TransplantationRoyal Free HospitalLondonUK
| |
Collapse
|
4
|
Kamal MA, Siddiqui I, Belgiovine C, Barbagallo M, Paleari V, Pistillo D, Chiabrando C, Schiarea S, Bottazzi B, Leone R, Avigni R, Migliore R, Spaggiari P, Gavazzi F, Capretti G, Marchesi F, Mantovani A, Zerbi A, Allavena P. Oncogenic KRAS-Induced Protein Signature in the Tumor Secretome Identifies Laminin-C2 and Pentraxin-3 as Useful Biomarkers for the Early Diagnosis of Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14112653. [PMID: 35681634 PMCID: PMC9179463 DOI: 10.3390/cancers14112653] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
KRAS mutations characterize pancreatic cell transformation from the earliest stages of carcinogenesis, and are present in >95% of pancreatic ductal adenocarcinoma (PDAC) cases. In search of novel biomarkers for the early diagnosis of PDAC, we identified the proteins secreted by the normal human pancreatic cell line (HPDE) recently transformed by inducing the overexpression of the KRASG12V oncogene. We report a proteomic signature of KRAS-induced secreted proteins, which was confirmed in surgical tumor samples from resected PDAC patients. The putative diagnostic performance of three candidates, Laminin-C2 (LAMC2), Tenascin-C (TNC) and Pentraxin-3 (PTX3), was investigated by ELISA quantification in two cohorts of PDAC patients (n = 200) eligible for surgery. Circulating levels of LAMC2, TNC and PTX3 were significantly higher in PDAC patients compared to the healthy individuals (p < 0.0001). The Receiver Operating Characteristics (ROC) curve showed good sensitivity (1) and specificity (0.63 and 0.85) for LAMC2 and PTX3, respectively, but not for TNC, and patients with high levels of LAMC2 had significantly shorter overall survival (p = 0.0007). High levels of LAMC2 and PTX3 were detected at early stages (I−IIB) and in CA19-9-low PDAC patients. In conclusion, pancreatic tumors release LAMC2 and PTX3, which can be quantified in the systemic circulation, and may be useful in selecting patients for further diagnostic imaging.
Collapse
Affiliation(s)
- Mohammad Azhar Kamal
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
| | - Imran Siddiqui
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
| | - Cristina Belgiovine
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
| | - Marialuisa Barbagallo
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
| | - Valentina Paleari
- Biobank, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (V.P.); (D.P.)
| | - Daniela Pistillo
- Biobank, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (V.P.); (D.P.)
| | - Chiara Chiabrando
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (C.C.); (S.S.)
| | - Silvia Schiarea
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (C.C.); (S.S.)
| | - Barbara Bottazzi
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
| | - Roberto Leone
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
| | - Roberta Avigni
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
| | - Roberta Migliore
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
| | - Paola Spaggiari
- Department of Pathology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy;
| | - Francesca Gavazzi
- Pancreatic Surgery Unit, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (F.G.); (G.C.); (A.Z.)
| | - Giovanni Capretti
- Pancreatic Surgery Unit, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (F.G.); (G.C.); (A.Z.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
| | - Federica Marchesi
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Alberto Mantovani
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
- The William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Alessandro Zerbi
- Pancreatic Surgery Unit, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (F.G.); (G.C.); (A.Z.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
| | - Paola Allavena
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, 20089 Rozzano, Italy; (M.A.K.); (I.S.); (C.B.); (M.B.); (B.B.); (R.L.); (R.A.); (R.M.); (F.M.); (A.M.)
- Correspondence:
| |
Collapse
|
5
|
Woeste MR, Wilson KD, Kruse EJ, Weiss MJ, Christein JD, White RR, Martin RCG. Optimizing Patient Selection for Irreversible Electroporation of Locally Advanced Pancreatic Cancer: Analyses of Survival. Front Oncol 2022; 11:817220. [PMID: 35096621 PMCID: PMC8793779 DOI: 10.3389/fonc.2021.817220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
Background Irreversible electroporation (IRE) has emerged as a viable consolidative therapy after induction chemotherapy, in which this combination has improved overall survival of locally advanced pancreatic cancer (LAPC). Optimal timing and patient selection for irreversible electroporation remains a clinically unmet need. The aim of this study was to investigate preoperative factors that may assist in predicting progression-free and overall survival following IRE. Methods A multi-institutional, prospectively maintained database was reviewed for patients with LAPC treated with induction chemotherapy followed by open-technique irreversible electroporation from 7/2015-5/2019. RECIST 1.1 criteria were used to assess tumor response and radiological progression. Overall survival (OS) and progression-free survival (PFS) were recorded. Survival analyses were performed using Kaplan Meier and Cox multivariable regression analyses. Results 187 LAPC patients (median age 62 years range, 21 – 91, 65% men, 35% women) were treated with IRE. Median PFS was 21.7 months and median OS from diagnosis was 25.5 months. On multivariable analysis, age ≤ 61 (HR 0.41, 95%CI 0.21-0.78, p<0.008) and no prior radiation (HR 0.49, 95%CI 0.26-0.94, p=0.03) were positive predictors of OS after IRE. Age ≤ 61(HR 0.53, 95%CI, 0.28-.99, p=0.046) and FOLFIRINOX followed by gemcitabine/abraxane induction chemotherapy (HR 0.37,95%CI 0.15-0.89, p=0.027) predicted prolonged PFS after IRE. Abnormal CA19-9 values at the time of surgery negatively impacted both OS (HR 2.46, 95%CI 1.28-4.72, p<0.007) and PFS (HR 2.192, 95%CI 1.143-4.201, p=0.018) following IRE. Conclusions Age, CA 19-9 response, avoidance of pre-IRE radiation, and FOLFIRINOX plus gemcitabine/abraxane induction chemotherapy are prominent factors to consider when referring or selecting LAPC patients to undergo IRE.
Collapse
Affiliation(s)
- Matthew R Woeste
- Division of Surgical Oncology, Department of Surgery, University of Louisville School of Medicine, Louisville, KY, United States
| | - Khaleel D Wilson
- Division of Surgical Oncology, Department of Surgery, University of Louisville School of Medicine, Louisville, KY, United States
| | - Edward J Kruse
- Department of Surgery, Section of Surgical Oncology, Augusta University Medical Center, Augusta, GA, United States
| | - Matthew J Weiss
- Department of Surgery, Division of Surgical Oncology, Johns Hopkins University, Baltimore, MD, United States
| | - John D Christein
- Department of Surgery, Division of Gastrointestinal Surgery, University of Alabama, Birmingham, AL, United States
| | - Rebekah R White
- Gastrointestinal Cancer Unit, University of California San Diego Moores Cancer Center, San Diego, CA, United States
| | - Robert C G Martin
- Division of Surgical Oncology, Department of Surgery, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
6
|
Vanek P, Eid M, Psar R, Zoundjiekpon V, Urban O, Kunovský L. Current trends in the diagnosis of pancreatic cancer. VNITRNI LEKARSTVI 2022; 68:363-370. [PMID: 36316197 DOI: 10.36290/vnl.2022.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a dreaded malignancy with a dismal 5-year survival rate despite maximal efforts on optimizing treatment strategies. Currently, early detection is considered to be the most effective way to improve survival as radical resection is the only potential cure. PDAC is often divided into four categories based on the extent of disease: resectable, borderline resectable, locally advanced, and metastatic. Unfortunately, the majority of patients are diagnosed with locally advanced or metastatic disease, which renders them ineligible for curative resection. This is mainly due to the lack of or vague symptoms while the disease is still localized, although appropriate utilization and prompt availability of adequate diagnostic tools is also critical given the aggressive nature of the disease. A cost-effective biomarker with high specificity and sensitivity allowing early detection of PDAC without the need for advanced or invasive methods is still not available. This leaves the diagnosis dependent on radiodiagnostic methods or endoscopic ultrasound. Here we summarize the latest epidemiological data, risk factors, clinical manifestation, and current diagnostic trends and implications of PDAC focusing on serum biomarkers and imaging modalities. Additionally, up-to-date management and therapeutic algorithms are outlined.
Collapse
|
7
|
Clinical Characteristics of Resected Acinar Cell Carcinoma of the Pancreas: A Korean Multi-Institutional Study. Cancers (Basel) 2021; 13:cancers13205095. [PMID: 34680244 PMCID: PMC8534044 DOI: 10.3390/cancers13205095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Pancreatic acinar cell carcinoma accounts for less than 1% of primary pancreatic neoplasms. Because of its rarity, its characteristics and clinical outcomes remain unclear. Treatment strategies for pancreatic acinar cell carcinoma have relied on those of pancreatic ductal adenocarcinoma. In previous studies, it has been difficult to identify its characteristics due to the lack of cohort numbers in single institutional studies with detailed data and the lack of detailed data in large cohort multi-institutional studies. This retrospective multicenter cohort used a database founded in 2015 by the Korean Association of Hepato-Biliary Pancreatic Surgery. This database has collected nationwide patient data with details. In the present study, we aimed to better understand clinical outcomes of resected pancreatic acinar cell carcinoma and to lay the groundwork for establishing proper treatment strategies. Abstract Given the rare incidence of pancreatic acinar cell carcinoma (PACC), its post-resection clinical outcomes remain unclear. Treatment strategies for PACC have relied on those of pancreatic ductal adenocarcinoma (PDAC). The present study retrospectively investigated clinicopathologic characteristics of resected PACC registered in the Korea Tumor Registry System Biliary Pancreas database. Among 59 patients with a mean age of 59.2 years and a male predominance (83.1%), 43, 5, 7, and 4 had pure PACC, ductal differentiations, mixed neuroendocrine carcinomas, and intraductal and papillary variants, respectively. The mean tumor size was 4.6 cm, consisting of eight at T1, 26 at T2, and 25 at T3 stages. Metastasis to regional lymph node was identified in 15 (25.4%) patients. Thirty-one (52.5%) patients received adjuvant therapy. Five-year survival rate was 57.4%. The median survival was 78.8 months. In survival comparison according to the stage with AJCC system, N stage (lymph node metastasis), but not T stage, showed significant differences (p = 0.027). Resected PACC appeared to have clinical outcomes distinct from those of PDAC in this nationwide study. Therefore, large-scale multinational studies are needed to overcome the rarity of PACC and to establish an appropriate treatment strategies and staging system.
Collapse
|
8
|
Kang YM, Wang H, Li R, Pan G. Prognostic Role of Carbohydrate Antigen 19 to 9 in Predicting Survival of Patients With Pancreatic Cancer: A Meta-Analysis. Technol Cancer Res Treat 2021; 20:15330338211043030. [PMID: 34617852 PMCID: PMC8642114 DOI: 10.1177/15330338211043030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
This study evaluates the prognostic role of carbohydrate antigen 19 to 9 (CA19-9) in predicting survival of pancreatic cancer patients. Literature search was conducted in electronic databases (Google Scholar, Ovid, PubMed, and Science Direct) and study selection was based on precise eligibility criteria. Random-effects meta-analyses were performed to achieve overall estimates of median survival and hazard ratios (HRs) of survival with cutoff defined lower and higher CA19-9 levels before and after surgery or chemotherapy (CT)/radiotherapy (RT) and the changes in CA19-9 levels after any treatment. A total of 41 studies (6519 patients; 42% females; age 63.3 years [95% confidence interval [CI]: 62.2, 64.4]) were included. A pooled HR of 1.79 with a narrow 95% CI (1.58, 2.01) showed that higher CA19-9 levels or less decrease in CA19-9 levels after treatment predicted shorter survival. Median survival in patients with lower and higher preoperative CA19-9 levels was 23.2 months [95% CI: 17.2, 29.2] and 14.0 months [95% CI: 10.9, 17.2], respectively, whereas median survival with lower and higher postoperative CA19-9 levels was 25.0 months [95% CI: 21.9, 28.0] and 13.0 months [95% CI: 10.9, 15.0] respectively. Median survival with lower and higher pre-CT/RT CA19-9 levels was 11.9 months [95% CI: 10.2, 13.6] and 7.7 months [95% CI: 6.2, 9.2], respectively, whereas median survival with lower and higher post-CT/RT CA19-9 levels was 15.1 months [95% CI: 13.2, 17.0] and 10.7 months [95% CI: 7.3, 14.0] respectively. A decrease in CA19-9 levels after treatment was also associated with longer survival. Thus, both pretreatment and posttreatment CA19-9 levels or their changes after treatment have good prognostic value in determining the survival of pancreatic cancer patients.
Collapse
Affiliation(s)
- Yong-Ming Kang
- 159365Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Hao Wang
- Heilongjiang Province Land Reclamation Headquarter General Hospital, Harbin, Heilongjiang, China
| | - Ran Li
- Harbin Red Cross Central Hospital, Harbin, Heilongjiang, China
| | - Gu Pan
- 159365Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
9
|
Reames BN, Blair AB, Krell RW, Groot VP, Gemenetzis G, Padussis JC, Thayer SP, Falconi M, Wolfgang CL, Weiss MJ, Are C, He J. Management of Locally Advanced Pancreatic Cancer: Results of an International Survey of Current Practice. Ann Surg 2021; 273:1173-1181. [PMID: 31449138 DOI: 10.1097/sla.0000000000003568] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The aim of this study was to investigate surgeon preferences for the management of patients with locally advanced pancreatic cancer (LAPC). BACKGROUND Select patients with LAPC may become candidates for curative resection following neoadjuvant therapy, and recent reports of survival are encouraging. Yet the optimal management approach remains unclear. METHODS An extensive electronic survey was systematically distributed by email to an international cohort of pancreas surgeons. Data collected included practice characteristics, management preferences, attitudes regarding contraindications to surgery, and 6 clinical vignettes of patients that ultimately received a margin negative resection (with detailed videos of post-neoadjuvant imaging) to assess propensity for surgical exploration if resection status is not known. RESULTS A total of 153 eligible responses were received from 4 continents. Median duration of practice is 12 years (interquartile range 6-20) and 77% work in a university setting. Most surgeons (86%) are considered high volume (>10 resections/yr), 33% offer a minimally-invasive approach, and 50% offer arterial resections in select patients. Most (72%) always recommend neoadjuvant chemotherapy, and 65% prefer FOLFIRINOX. Preferences for the duration of chemotherapy varied widely: 39% prefer ≥2 months, 43% prefer ≥4 months, and 11% prefer ≥6 months. Forty-one percent frequently recommend neoadjuvant radiotherapy, and 53% prefer 5 to 6 weeks of chemoradiation. The proportion of surgeons favoring exploration following neoadjuvant varied extensively across 5 vignettes of LAPC, from 14% to 53%. In a vignette of oligometastatic liver metastases, 31% would offer exploration if a favorable therapy response is observed. CONCLUSIONS In an international cohort of pancreas surgeons, there is substantial variation in management preferences, perceived contraindications to surgery, and the propensity to consider exploration in LAPC. These results emphasize the importance of a robust and nuanced multidisciplinary discussion for each patient, and suggest an evolving concept of "resectability."
Collapse
Affiliation(s)
- Bradley N Reames
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE
| | - Alex B Blair
- Department of Surgery, Johns Hopkins Hospital, Baltimore, MD
| | - Robert W Krell
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Vincent P Groot
- Department of Surgery, UMC Utrecht Cancer Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Georgios Gemenetzis
- Department of Surgery, University of Glasgow School of Medicine, Glasgow, UK
| | - James C Padussis
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE
| | - Sarah P Thayer
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE
| | - Massimo Falconi
- Department of Surgery, Università Vita-Salute, San Raffaele Hospital IRCCS, Milano, Italy
| | | | | | - Chandrakanth Are
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE
| | - Jin He
- Department of Surgery, Johns Hopkins Hospital, Baltimore, MD
| |
Collapse
|
10
|
Luo G, Jin K, Deng S, Cheng H, Fan Z, Gong Y, Qian Y, Huang Q, Ni Q, Liu C, Yu X. Roles of CA19-9 in pancreatic cancer: Biomarker, predictor and promoter. Biochim Biophys Acta Rev Cancer 2021; 1875:188409. [PMID: 32827580 DOI: 10.1016/j.bbcan.2020.188409] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/29/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023]
Abstract
Carbohydrate antigen 19-9 (CA19-9) is the best validated biomarker and an indicator of aberrant glycosylation in pancreatic cancer. CA19-9 functions as a biomarker, predictor, and promoter in pancreatic cancer. As a biomarker, the sensitivity is approximately 80%, and the major challenges involve false positives in conditions of inflammation and nonpancreatic cancers and false negatives in Lewis-negative Individuals. Lewis antigen status should be determined when using CA19-9 as a biomarker. CA19-9 has screening potential when combined with symptoms and/or risk factors. As a predictor, CA19-9 could be used to assess stage, prognosis, resectability, recurrence, and therapeutic efficacy. Normal baseline levels of CA19-9 are associated with long-term survival. As a promoter, CA19-9 could be used to evaluate the biology of pancreatic cancer. CA19-9 can accelerate pancreatic cancer progression by glycosylating proteins, binding to E-selectin, strengthening angiogenesis, and mediating the immunological response. CA19-9 is an attractive therapeutic target for cancer, and strategies include therapeutic antibodies and vaccines, CA19-9-guided nanoparticles, and inhibition of CA19-9 biosynthesis.
Collapse
Affiliation(s)
- Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China
| | - Shengming Deng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China
| | - Zhiyao Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China
| | - Yitao Gong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China
| | - Yunzhen Qian
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China
| | - Qiuyi Huang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China; Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, China.
| |
Collapse
|
11
|
Yu S, Zhang C, Xie KP. Therapeutic resistance of pancreatic cancer: Roadmap to its reversal. Biochim Biophys Acta Rev Cancer 2020; 1875:188461. [PMID: 33157162 DOI: 10.1016/j.bbcan.2020.188461] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is a lethal disease with limited opportunity for resectable surgery as the first choice for cure due to its late diagnosis and early metastasis. The desmoplastic stroma and cellular genetic or epigenetic alterations of pancreatic cancer impose physical and biological barriers to effective therapies, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy. Here, we review the current therapeutic options for pancreatic cancer, and underlying mechanisms and potential reversal of therapeutic resistance, a hallmark of this deadly disease.
Collapse
Affiliation(s)
- Sen Yu
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital Affiliated to the South China University of Technology, School of Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Chunyu Zhang
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital Affiliated to the South China University of Technology, School of Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Ke-Ping Xie
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital Affiliated to the South China University of Technology, School of Medicine, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
12
|
Colloca GA, Venturino A, Guarneri D. Tumor growth kinetics by CA 19-9 in patients with unresectable pancreatic cancer receiving chemotherapy: A retrospective analysis. Pancreatology 2020; 20:1189-1194. [PMID: 32747196 DOI: 10.1016/j.pan.2020.07.397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Recently, measures of tumor growth kinetics calculated by carbohydrate antigen 19-9 (CA 19-9) determinations after cytotoxic chemotherapy (CHT) have been reported as effective prognostic indicators in locally-advanced unresectable and metastatic pancreatic adenocarcinoma (mPDAC). The study aims to evaluate the prognostic role of tumor kinetics measured by CA 19-9 in patients with mPDAC, measuring it by three different ways. METHODS Patients with mPDAC receiving a first-line CHT between 2009 and 2017 were identified, and those for whom CA 19-9 data were available were enrolled. Three CA 19-9-related variables were calculated: CA 19-9 related reduction rate (RR) and tumor growth rate (G), after 8 weeks of CHT, tumor growth and inflammation index (TGII), after 90 days of CHT. The relationships with the outcome were analysed, and a Cox model has been build with each of the three variables. RESULTS Of 118 patients only 48 were eligible for the analysis. RR, G, or TGII appear as significant prognostic factors, and, after multivariate analysis, a reduction rate of 20% the baseline or more was associated with good survival (HR 0.321; CIs 0.156-0.661) as well as a G > -0.4%/day (HR 2.114; CIs 1.034-4.321), whereas TGII >190 was not correlated with the outcome (HR 1.788; CIs 0.789-4.055). CONCLUSIONS In patients with mPDAC, after 8 weeks of first-line CHT, CA 19-9-related tumor reduction or growth rate appear as valuable prognostic factors.
Collapse
|
13
|
Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. Lancet 2020; 395:2008-2020. [PMID: 32593337 DOI: 10.1016/s0140-6736(20)30974-0] [Citation(s) in RCA: 1414] [Impact Index Per Article: 353.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/06/2020] [Accepted: 04/17/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is a highly fatal disease with a 5-year survival rate of approximately 10% in the USA, and it is becoming an increasingly common cause of cancer mortality. Risk factors for developing pancreatic cancer include family history, obesity, type 2 diabetes, and tobacco use. Patients typically present with advanced disease due to lack of or vague symptoms when the cancer is still localised. High quality computed tomography with intravenous contrast using a dual phase pancreatic protocol is typically the best method to detect a pancreatic tumour and to determine surgical resectability. Endoscopic ultrasound is an increasingly used complementary staging modality which also allows for diagnostic confirmation when combined with fine needle aspiration. Patients with pancreatic cancer are often divided into one of four categories based on extent of disease: resectable, borderline resectable, locally advanced, and metastatic; patient condition is also an important consideration. Surgical resection represents the only chance for cure, and advancements in adjuvant chemotherapy have improved long-term outcomes in these patients. Systemic chemotherapy combinations including FOLFIRINOX (5-fluorouracil, folinic acid [leucovorin], irinotecan, and oxaliplatin) and gemcitabine plus nab-paclitaxel remain the mainstay of treatment for patients with advanced disease. Data on the benefit of PARP inhibition as maintenance therapy in patients with germline BRCA1 or BRACA2 mutations might prove to be a harbinger of advancement in targeted therapy. Additional research efforts are focusing on modulating the pancreatic tumour microenvironment to enhance the efficacy of the immunotherapeutic strategies.
Collapse
Affiliation(s)
- Jonathan D Mizrahi
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rishi Surana
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Juan W Valle
- Division of Cancer Sciences, University of Manchester, Department of Medical Oncology, Christie NHS Foundation Trust, Manchester, UK
| | - Rachna T Shroff
- Division of Hematology and Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ, USA.
| |
Collapse
|
14
|
Importance of Normalization of CA19-9 Levels Following Neoadjuvant Therapy in Patients With Localized Pancreatic Cancer. Ann Surg 2020; 271:740-747. [PMID: 30312198 DOI: 10.1097/sla.0000000000003049] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Carbohydrate antigen 19-9 (CA19-9) is a prognostic marker for patients with pancreatic cancer (PC), but its value as a treatment biomarker is unclear. SUMMARY BACKGROUND DATA Although CA19-9 is an established prognostic marker for patients with PC, it is unclear how CA19-9 monitoring should be used to guide multimodality treatment and what level of change in CA19-9 constitutes a meaningful treatment response. METHODS CA19-9 measurements at diagnosis (pretx), after completion of all planned neoadjuvant therapy (preop), and after surgery (postop) were analyzed in patients with localized PC who had an elevated CA19-9 (≥35 U/dL) at diagnosis. Patients were classified by: 1) quartiles of pretx CA19-9 (Q1-4); 2) proportional changes in CA19-9 (ΔCA19-9) after the completion of neoadjuvant therapy; 3) normalization (CA19-9 <35 U/dL) of preop CA19-9; and 4) normalization of postop CA19-9. RESULTS Among 131 patients, the median overall survival (OS) was 30 months; 68 months for the 33 patients in Q1 of pretx CA19-9 (<80 U/dL) compared with 25 months for the 98 patients in Q2-4 (P = 0.03). For the 98 patients in Q2-4, preop CA19-9 declined (from pretx) in 86 (88%), but there was no association between the magnitude of ΔCA19-9 and OS (P = 0.77). Median OS of the 98 patients who did (n = 29) or did not (n = 69) normalize their preop CA19-9 were 46 and 23 months, respectively (P = 0.02). Of the 69 patients with an elevated preop CA19-9, 32 (46%) normalized their postop CA19-9. Failure to normalize preop or postop CA19-9 was associated with a 2.77-fold and 4.03-fold increased risk of death, respectively (P < 0.003) as compared with patients with normal preop CA19-9. CONCLUSIONS Following neoadjuvant therapy, normalization of CA19-9, rather than the magnitude of change, is the strongest prognostic marker for long-term survival.
Collapse
|
15
|
|
16
|
Hua J, Tian L. A comprehensive and comparative review of optimal cut-points selection methods for diseases with multiple ordinal stages. J Biopharm Stat 2019; 30:46-68. [PMID: 31250693 DOI: 10.1080/10543406.2019.1632876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cut-points selection is a key topic in the field of diagnostic studies. For binary classification, there exist several well-developed methods, some of which have been extended to three-class settings and beyond. This paper focuses on optimal cut-points selection methods for diseases with multiple ordinal stages. The purpose of this paper is two-fold: 1) to propose three new cut-points selection methods; and 2) to present a comprehensive simulation study to assess and compare the performance of all the available methods. Two real data sets, one from ovarian cancer and the other from pancreatic cancer, are analyzed.
Collapse
Affiliation(s)
- Jia Hua
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Lili Tian
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
17
|
Mathew MP, Donaldson JG. Glycosylation and glycan interactions can serve as extracellular machinery facilitating clathrin-independent endocytosis. Traffic 2019; 20:295-300. [PMID: 30706592 DOI: 10.1111/tra.12636] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 01/04/2023]
Abstract
In contrast to clathrin-mediated endocytosis (CME) which is well characterized and understood, little is known about the regulation and machinery underlying clathrin-independent endocytosis (CIE). There is also a wide variation in the requirements each individual CIE cargo has for its internalization. Recent studies have shown that CIE is affected by glycosylation and glycan interactions. We briefly review these studies and explore how these studies mesh with one another. We then discuss what this sensitivity to glycan interactions could indicate for the regulation of CIE. We address the spectrum of responses CIE has been shown to have with respect to changes in glycan interactions and attempt to reconcile disparate observations onto a shared conceptual landscape. We focus on the mechanisms by which cells can alter the glycan interactions at the plasma membrane and propose that glycosylation and glycan interactions could provide cells with a tool box with which cells can manipulate CIE. Altered glycosylation is often associated with a number of diseases and we discuss how under different disease settings, glycosylation-based modulation of CIE could play a role in disease progression.
Collapse
Affiliation(s)
- Mohit P Mathew
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Julie G Donaldson
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
18
|
Consensus statement on mandatory measurements in pancreatic cancer trials (COMM-PACT) for systemic treatment of unresectable disease. Lancet Oncol 2019; 19:e151-e160. [PMID: 29508762 DOI: 10.1016/s1470-2045(18)30098-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/19/2017] [Accepted: 10/04/2017] [Indexed: 12/17/2022]
Abstract
Variations in the reporting of potentially confounding variables in studies investigating systemic treatments for unresectable pancreatic cancer pose challenges in drawing accurate comparisons between findings. In this Review, we establish the first international consensus on mandatory baseline and prognostic characteristics in future trials for the treatment of unresectable pancreatic cancer. We did a systematic literature search to find phase 3 trials investigating first-line systemic treatment for locally advanced or metastatic pancreatic cancer to identify baseline characteristics and prognostic variables. We created a structured overview showing the reporting frequencies of baseline characteristics and the prognostic relevance of identified variables. We used a modified Delphi panel of two rounds involving an international panel of 23 leading medical oncologists in the field of pancreatic cancer to develop a consensus on the various variables identified. In total, 39 randomised controlled trials that had data on 15 863 patients were included, of which 32 baseline characteristics and 26 prognostic characteristics were identified. After two consensus rounds, 23 baseline characteristics and 12 prognostic characteristics were designated as mandatory for future pancreatic cancer trials. The COnsensus statement on Mandatory Measurements in unresectable PAncreatic Cancer Trials (COMM-PACT) identifies a mandatory set of baseline and prognostic characteristics to allow adequate comparison of outcomes between pancreatic cancer studies.
Collapse
|
19
|
van der Sijde F, Vietsch EE, Mustafa DAM, Besselink MG, Groot Koerkamp B, van Eijck CHJ. Circulating Biomarkers for Prediction of Objective Response to Chemotherapy in Pancreatic Cancer Patients. Cancers (Basel) 2019; 11:cancers11010093. [PMID: 30650521 PMCID: PMC6356815 DOI: 10.3390/cancers11010093] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/03/2019] [Accepted: 01/11/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a lethal disease with increasing incidence. Most patients present with advanced disease, for which palliative systemic chemotherapy is the only therapeutic option. Despite improved median survival rates with FOLFIRINOX or gemcitabine chemotherapy compared to the best supportive care, many individual patients may not benefit from chemotherapy. Biomarkers are needed to predict who will benefit from chemotherapy and to monitor a patient’s response to chemotherapy. This review summarizes current research and future perspectives on circulating biomarkers for systemic chemotherapy response.
Collapse
Affiliation(s)
- Fleur van der Sijde
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands.
| | - Eveline E Vietsch
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands.
| | - Dana A M Mustafa
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands.
| | - Marc G Besselink
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands.
| | - Casper H J van Eijck
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
20
|
Sanh N, Fadul H, Hussein N, Lyn-Cook BD, Hammons G, Ramos-Cardona XE, Mohamed K, Mohammed SI. Proteomics Profiling of Pancreatic Cancer and Pancreatitis for Biomarkers Discovery. ACTA ACUST UNITED AC 2018; 9. [PMID: 31032145 DOI: 10.4172/2157-7013.1000287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pancreatic cancer is one of the most aggressive malignancies with an increase in incidence predicted, particularly in African Americans. Pancreatic cancer is considered a silent disease with poor prognosis and a lack of early biomarkers for detection. Proteomics has been applied in many diseases for identifying or discovering biomarkers. It has long been suggested that chronic pancreatitis may be a risk factor for developing pancreatic cancer. This study identified proteins that are altered in expression in pancreatic cancer and pancreatitis compared to normal using proteomic technology. Proteins were extracted from laser captured micro-dissected tissues and separated in 2-DPAGE and imaged. The protein profiles of pancreatic cancer and pancreatitis are similar but differed with the protein profile of normal adjacent tissues. Representative proteins, overexpressed in tumor and pancreatitis but not normal tissues, were excised from gels, subjected to in-gel digestion, and analyzed by MALDI-TOF mass spectrometry. Proteins identified included transferrin, ER-60 protein, proapolipoprotein, tropomyosin 1, alpha 1 actin precursor, ACTB protein, and gamma 2 propeptide, aldehyde dehydrogenase 1A1, pancreatic lipase and annexin A1. Several proteins, which were shown in pancreatic cancer, were also observed in pancreatitis samples. Understanding the role of these specific proteins and their mechanistic action will give insights into their involvement in pancreatic cancers.
Collapse
Affiliation(s)
- N Sanh
- Department of Comparative Pathobiology and Purdue University Center for Cancer Research, Purdue University, West Lafayette, USA
| | - H Fadul
- Department of Comparative Pathobiology and Purdue University Center for Cancer Research, Purdue University, West Lafayette, USA
| | - N Hussein
- Franklin College, IUPUI- Indiana University Purdue University Indianapolis, Indianapolis, USA
| | - B D Lyn-Cook
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, USA
| | - G Hammons
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, USA
| | - X E Ramos-Cardona
- Department of Comparative Pathobiology and Purdue University Center for Cancer Research, Purdue University, West Lafayette, USA
| | - K Mohamed
- Radiation and Isotopes Center Khartoum (RICK), Sudan
| | - S I Mohammed
- Department of Comparative Pathobiology and Purdue University Center for Cancer Research, Purdue University, West Lafayette, USA
| |
Collapse
|
21
|
Rieser CJ, Zenati M, Hamad A, Al Abbas AI, Bahary N, Zureikat AH, Zeh HJ, Hogg ME. CA19-9 on Postoperative Surveillance in Pancreatic Ductal Adenocarcinoma: Predicting Recurrence and Changing Prognosis over Time. Ann Surg Oncol 2018; 25:3483-3491. [PMID: 29786131 DOI: 10.1245/s10434-018-6521-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Serum carbohydrate antigen 19-9 (CA19-9) correlates with response to therapy and overall survival (OS) for patients with pancreatic ductal adenocarcinoma (PDAC). This study aimed to define the chronologic relationship between CA19-9 elevation and radiographic recurrence to develop a model that can predict the risk of recurrence (RFS) and prognosis during interval surveillance for patients with resected PDAC. METHODS A retrospective review examined patients undergoing surgery for pancreatic adenocarcinoma from January 2010 to May 2016. Their CA19-9 levels were classified at diagnosis, after surgery, and at 6-month surveillance intervals. Recurrence was defined by radiographic evidence. The CA19-9 levels were correlated with RFS and OS at every time point using multivariate analysis. RESULTS The study examined 525 patients. Five patterns of CA19-9 were identified: normal ("nonsecretors," 18.5%), always elevated, and high at diagnosis but normal after resection involving three patterns with varied behavior during surveillance. These five patterns had implications for RFS and OS. When elevation of CA19-9, as assessed at 6-month intervals, was analyzed relative to detection of radiographic disease, CA19-9 had poor positive predictive value (average, 35%) but high negative predictive value (average, 92%) for radiographic recurrence. Conditional RFS showed that CA19-9 elevation did not equal radiographic recurrence but predicted subsequent RFS. Additionally, conditional OS showed that CA19-9 elevation alone was predictive at each time point. CONCLUSION This study showed that CA19-9 patterns beyond the post-resection period predict RFS and OS. High CA19-9 frequently is discordant with recurrence on imaging and may precede it by more than 6 months. At each surveillance interval, CA19-9 is predictive of prognosis, which may help in counseling patients and could be used to direct protocols of salvage chemotherapy.
Collapse
Affiliation(s)
- Caroline J Rieser
- Department of Surgical Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mazen Zenati
- Department of Surgical Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ahmad Hamad
- Department of Surgical Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amr I Al Abbas
- Department of Surgical Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nathan Bahary
- Department of Medical Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amer H Zureikat
- Department of Surgical Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Herbert J Zeh
- Department of Surgical Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melissa E Hogg
- Department of Surgical Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
22
|
Chen I, Raymond VM, Geis JA, Collisson EA, Jensen BV, Hermann KL, Erlander MG, Tempero M, Johansen JS. Ultrasensitive plasma ctDNA KRAS assay for detection, prognosis, and assessment of therapeutic response in patients with unresectable pancreatic ductal adenocarcinoma. Oncotarget 2017; 8:97769-97786. [PMID: 29228650 PMCID: PMC5716690 DOI: 10.18632/oncotarget.22080] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 10/11/2017] [Indexed: 12/18/2022] Open
Abstract
Precision oncology requires sensitive and specific clinical biomarkers. Carbohydrate Antigen 19-9 (CA19-9) is widely used in pancreatic ductal adenocarcinoma (PDA) but lacks sensitivity and specificity. Nearly all PDAs harbor somatic KRAS mutations, nominating circulating tumor DNA (ctDNA) KRAS as an alternative disease biomarker, however, variable clinical performance has limited its clinical utility. We applied an ultrasensitive, PCR mutation enrichment, next generation sequencing ctDNA KRAS assay in a large cohort of patients with unresectable PDA (N = 189) recruited to the BIOPAC study between 2008-2015. Baseline and longitudinal serum CA19-9 and plasma ctDNA KRAS were correlated with time to progression (TTP) and overall survival (OS). Baseline ctDNA KRAS detection rate was 93.7% (86.4% in patients with non-elevated CA19-9). ctDNA KRAS and CA19-9 were positively correlated yet independently associated with TTP and OS (ctDNA KRAS p = 0.0018 and 0.0014; CA19-9 p = 0.0294 and 0.0007, respectively). A generated model quantitating longitudinal ctDNA KRAS correctly assessed greater than 80% of patient responses. Quantitative detection of KRAS ctDNA is an informative prognostic biomarker, complementary to CA19-9 in patients with unresectable PDA. Longitudinal ctDNA KRAS may inform therapeutic decision making and provides a kinetically dynamic and quantitative metric of patient response.
Collapse
Affiliation(s)
- Inna Chen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Eric A Collisson
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Benny V Jensen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kirstine L Hermann
- Department of Radiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Margaret Tempero
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Julia S Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Zhao YP, Zhou PT, Ji WP, Wang H, Fang M, Wang MM, Yin YP, Jin G, Gao CF. Validation of N-glycan markers that improve the performance of CA19-9 in pancreatic cancer. Clin Exp Med 2017; 17:9-18. [PMID: 26714469 DOI: 10.1007/s10238-015-0401-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 11/02/2015] [Indexed: 01/05/2023]
Abstract
Pancreatic cancer (PC) has a high mortality rate because it is usually diagnosed late. Glycosylation of proteins is known to change in tumor cells during the development of PC. The objectives of this study were to identify and validate the diagnostic value of novel biomarkers based on N-glycomic profiling for PC. In total, 217 individuals including subjects with PC, pancreatitis, and healthy controls were divided randomly into a training group (n = 164) and validation groups (n = 53). Serum N-glycomic profiling was analyzed by DSA-FACE. The diagnostic model was constructed based on N-glycan markers with logistic stepwise regression. The diagnostic performance of the model was assessed further in validation cohort. The level of total core fucose residues was increased significantly in PC. Two diagnostic models designated GlycoPCtest and PCmodel (combining GlycoPCtest and CA19-9) were constructed to differentiate PC from normal. The area under the receiver operating characteristic curve (AUC) of PCmodel was higher than that of CA19-9 (0.925 vs. 0.878). The diagnostic models based on N-glycans are new, valuable, noninvasive alternatives for identifying PC. The diagnostic efficacy is improved by combined GlycoPCtest and CA19-9 for the discrimination of patients with PC from healthy controls.
Collapse
Affiliation(s)
- Yun-Peng Zhao
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Rd, Shanghai, 200438, China
| | - Ping-Ting Zhou
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Rd, Shanghai, 200438, China
| | - Wei-Ping Ji
- Department of Surgery, Changhai Hospital, Second Military Medical University, 116 Changhai Rd, Shanghai, 200438, China
| | - Hao Wang
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Meng Fang
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Rd, Shanghai, 200438, China
| | - Meng-Meng Wang
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Rd, Shanghai, 200438, China
| | - Yue-Peng Yin
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Rd, Shanghai, 200438, China
| | - Gang Jin
- Department of Surgery, Changhai Hospital, Second Military Medical University, 116 Changhai Rd, Shanghai, 200438, China.
| | - Chun-Fang Gao
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Rd, Shanghai, 200438, China.
| |
Collapse
|
24
|
Herreros-Villanueva M, Bujanda L. Non-invasive biomarkers in pancreatic cancer diagnosis: what we need versus what we have. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:134. [PMID: 27162784 DOI: 10.21037/atm.2016.03.44] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pancreatic cancer (PC) is probably the most lethal tumor being forecast as the second most fatal cancer by 2020 in developed countries. Only the earliest forms of the disease are a curable disease but it has to be diagnosed before symptoms starts. Detection at curable phase demands screening intervention for early detection and differential diagnosis. Unfortunately, no successful strategy or image technique has been concluded as effective approach and currently non-invasive biomarkers are the hope. Multiple translational research studies have explored minimally or non-invasive biomarkers in biofluids-blood, urine, stool, saliva or pancreatic juice, but diagnostic performance has not been validated yet. Nowadays no biomarker, alone or in combination, has been superior to carbohydrate antigen 19-9 (CA19-9) in sensitivity and specificity. Although the number of novel biomarkers for early diagnosis of PC has been increasing during the last couple of years, no molecular signature is ready to be implemented in clinical routine. Under the uncertain future, miRNAs profiling and methylation status seem to be the most promising biomarkers. However, good results in larger validations are urgently needed before application. Industry efforts through biotech and pharmaceutical companies are urgently required to demonstrate accuracy and validate promising results from basic and translational results.
Collapse
Affiliation(s)
- Marta Herreros-Villanueva
- Department of Gastroenterology, Hospital Donostia/Biodonostia Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco UPV/EHU, San Sebastian, Spain
| | - Luis Bujanda
- Department of Gastroenterology, Hospital Donostia/Biodonostia Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco UPV/EHU, San Sebastian, Spain
| |
Collapse
|
25
|
Jenkinson C, Elliott VL, Evans A, Oldfield L, Jenkins RE, O’Brien DP, Apostolidou S, Gentry-Maharaj A, Fourkala EO, Jacobs IJ, Menon U, Cox T, Campbell F, Pereira SP, Tuveson DA, Park BK, Greenhalf W, Sutton R, Timms JF, Neoptolemos JP, Costello E. Decreased Serum Thrombospondin-1 Levels in Pancreatic Cancer Patients Up to 24 Months Prior to Clinical Diagnosis: Association with Diabetes Mellitus. Clin Cancer Res 2016; 22:1734-1743. [PMID: 26573598 PMCID: PMC4820087 DOI: 10.1158/1078-0432.ccr-15-0879] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 09/19/2015] [Indexed: 12/24/2022]
Abstract
PURPOSE Identification of serum biomarkers enabling earlier diagnosis of pancreatic ductal adenocarcinoma (PDAC) could improve outcome. Serum protein profiles in patients with preclinical disease and at diagnosis were investigated. EXPERIMENTAL DESIGN Serum from cases up to 4 years prior to PDAC diagnosis and controls (UKCTOCS,n= 174) were studied, alongside samples from patients diagnosed with PDAC, chronic pancreatitis, benign biliary disease, type 2 diabetes mellitus, and healthy subjects (n= 298). Isobaric tags for relative and absolute quantification (iTRAQ) enabled comparisons of pooled serum from a test set (n= 150). Validation was undertaken using multiple reaction monitoring (MRM) and/or Western blotting in all 472 human samples and samples from a KPC mouse model. RESULTS iTRAQ identified thrombospondin-1 (TSP-1) as reduced preclinically and in diagnosed samples. MRM confirmed significant reduction in levels of TSP-1 up to 24 months prior to diagnosis. A combination of TSP-1 and CA19-9 gave an AUC of 0.86, significantly outperforming both markers alone (0.69 and 0.77, respectively;P< 0.01). TSP-1 was also decreased in PDAC patients compared with healthy controls (P< 0.05) and patients with benign biliary obstruction (P< 0.01). Low levels of TSP-1 correlated with poorer survival, preclinically (P< 0.05) and at clinical diagnosis (P< 0.02). In PDAC patients, reduced TSP-1 levels were more frequently observed in those with confirmed diabetes mellitus (P< 0.01). Significantly lower levels were also observed in PDAC patients with diabetes compared with individuals with type 2 diabetes mellitus (P= 0.01). CONCLUSIONS Circulating TSP-1 levels decrease up to 24 months prior to diagnosis of PDAC and significantly enhance the diagnostic performance of CA19-9. The influence of diabetes mellitus on biomarker behavior should be considered in future studies.
Collapse
Affiliation(s)
- Claire Jenkinson
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
- National Institute for Health Research Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, UK
| | - Victoria L. Elliott
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
- National Institute for Health Research Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, UK
| | - Anthony Evans
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
- National Institute for Health Research Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, UK
| | - Lucy Oldfield
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
- National Institute for Health Research Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, UK
| | - Rosalind E. Jenkins
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, UK
| | - Darragh P. O’Brien
- Department of Women’s Cancer, Institute for Women’s Health, University College London, UK
| | - Sophia Apostolidou
- Department of Women’s Cancer, Institute for Women’s Health, University College London, UK
| | | | - Evangelia-O Fourkala
- Department of Women’s Cancer, Institute for Women’s Health, University College London, UK
| | - Ian J. Jacobs
- Department of Women’s Cancer, Institute for Women’s Health, University College London, UK
- Faculty of Medical & Human Sciences, 1.018 Core Technology Facility, University of Manchester, UK
| | - Usha Menon
- Department of Women’s Cancer, Institute for Women’s Health, University College London, UK
| | - Trevor Cox
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
| | | | | | - David A. Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - B. Kevin Park
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, UK
| | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
- National Institute for Health Research Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, UK
| | - Robert Sutton
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
- National Institute for Health Research Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, UK
| | - John F. Timms
- Department of Women’s Cancer, Institute for Women’s Health, University College London, UK
| | - John P. Neoptolemos
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
- National Institute for Health Research Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, UK
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
- National Institute for Health Research Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, UK
| |
Collapse
|
26
|
Herreros-Villanueva M, Bujanda L. Glypican-1 in exosomes as biomarker for early detection of pancreatic cancer. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:64. [PMID: 27004211 DOI: 10.3978/j.issn.2305-5839.2015.10.39] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
On June 24, 2015 Nature published an article entitle "Glypican-1 identifies cancer exosomes and detects early pancreatic cancer'', which demonstrates that exosomes positives for the proteoglycan glypican-1 (GPC1) are expressed in serum of patients with pancreatic cancer since very early stages but not in benign pancreatic disease. Additionally, these GPC1(+) circulating exosomes correlate with tumor burden and could be used as prognostic biomarker in pre and post-surgical patients. The study is pioneer since GPC1 biomarker in exosomes offers better sensitivity and specificity than any other under evaluation or used in clinical practice. However, methodology for exosomes isolation still remains at investigational phase. Further studies are need to translate this technology to a practicable clinical method. Further research is also required to validate this biomarker in larger prospective cohort including more cases of premalignant lesions and then replicated results could possible guide changes in clinical practice.
Collapse
Affiliation(s)
- Marta Herreros-Villanueva
- Department of Gastroenterology, Hospital Donostia/Biodonostia Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco UPV/EHU, San Sebastian, Spain
| | - Luis Bujanda
- Department of Gastroenterology, Hospital Donostia/Biodonostia Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco UPV/EHU, San Sebastian, Spain
| |
Collapse
|
27
|
Chung KH, Ryu JK, Lee BS, Jang DK, Lee SH, Kim YT. Early decrement of serum carbohydrate antigen 19-9 predicts favorable outcome in advanced pancreatic cancer. J Gastroenterol Hepatol 2016; 31:506-12. [PMID: 26250642 DOI: 10.1111/jgh.13075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 07/14/2015] [Accepted: 07/23/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIM The role of carbohydrate antigen 19-9 (CA 19-9) for predicting treatment outcome in pancreatic ductal adenocarcinoma (PDAC) remains to be elucidated. This study was aimed to determine the correlation between early decrement in CA 19-9 concentration and prognosis of advanced PDAC after chemotherapy. METHODS All patients confirmed with locally advanced or metastatic PDAC who received initial systemic chemotherapy for at least two cycles in our institution between January 2012 and December 2013 were included. Serum CA 19-9 concentrations at baseline and 8 weeks after the initiation of chemotherapy were obtained. Correlation between CA 19-9 decrement and survival outcomes (time to progression [TTP] and overall survival [OS]) were evaluated. RESULTS A total of 183 patients with initially elevated CA 19-9 were included. OS and TTP was significantly longer for patients whose serum CA 19-9 concentration decreased more than 10% from baseline (n = 103), than that for patients whose serum CA 19-9 was not decreased (n = 80) (423 vs 155 days, P < 0.001 for OS and 222 vs 75 days, P < 0.001 for TTP). In multivariate analysis, CA 19-9 decrement more than 10% from baseline was still a significant factor for longer OS (hazard ratio for progression 0.275 [0.184-0.412], P < 0.001) and TTP (0.322 [0.219-0.473], P < 0.001) in both stage III and IV. CONCLUSIONS The early decrement of CA 19-9 after the initiation of chemotherapy was an independent factor related with better survival outcomes in unresectable PDAC.
Collapse
Affiliation(s)
- Kwang Hyun Chung
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Kon Ryu
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ban Seok Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Kee Jang
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sang Hyub Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong-Tae Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
28
|
Aldakkak M, Christians KK, Krepline AN, George B, Ritch PS, Erickson BA, Johnston FM, Evans DB, Tsai S. Pre-treatment carbohydrate antigen 19-9 does not predict the response to neoadjuvant therapy in patients with localized pancreatic cancer. HPB (Oxford) 2015; 17:942-52. [PMID: 26255895 PMCID: PMC4571763 DOI: 10.1111/hpb.12448] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 04/28/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND The prognostic value of CA19-9 in patients with pancreatic cancer (PC) treated with neoadjuvant therapy has not been well described. METHODS Pre-treatment CA19-9 levels (with concomitant normal bilirubin level) in patients with localized PC were categorized as normal (≤35), low (36-200), moderate (201-1000), or high (>1000). Post-treatment CA19-9 was measured after neoadjuvant therapy, prior to surgery. RESULTS Pre-treatment CA19-9 levels were evaluable in 235 patients, levels were normal in 60 (25%) patients, low in 78 (33%) patients, moderate in 69 (29%) and high in 28 (12%). After neoadjuvant therapy, post-treatment CA19-9 normalized (≤ 35) in 40 (51%) of the patients in the low group, 14 (21%) of the moderate and 5 (19%) of the high group (P < 0.001). Of the 235 patients, 168 (71%) completed all intended therapy including a pancreatectomy; 44 (73%), 62 (79%), 46 (67%) and 16 (57%) of the normal, low, moderate and high groups (P = 0.10). Among these 168 patients, the median overall survival was 38.4, 43.6, 44.7, 27.2 and 26.4 months for normal, low, moderate and high CA19-9 groups (log rank P = 0.72). Among resected patients, an elevated pre-treatment CA19-9 was of little prognostic value; instead, it was the CA19-9 response to neoadjuvant therapy that was prognostic [hazard ratio (HR): 1.80, P = 0.02]. CONCLUSIONS Among patients who completed neoadjuvant therapy and surgery, pre-treatment CA19-9 obtained at the time of diagnosis was not predictive of overall survival, but normalization of post-treatment CA19-9 in response to neoadjuvant therapy was highly prognostic.
Collapse
Affiliation(s)
- Mohammed Aldakkak
- Departments of Surgery, Pancreatic Cancer Program, The Medical College of WisconsinMilwaukee, WI, USA
| | - Kathleen K Christians
- Departments of Surgery, Pancreatic Cancer Program, The Medical College of WisconsinMilwaukee, WI, USA
| | - Ashley N Krepline
- Departments of Surgery, Pancreatic Cancer Program, The Medical College of WisconsinMilwaukee, WI, USA
| | - Ben George
- Departments of Medicine, Pancreatic Cancer Program, The Medical College of WisconsinMilwaukee, WI, USA
| | - Paul S Ritch
- Departments of Medicine, Pancreatic Cancer Program, The Medical College of WisconsinMilwaukee, WI, USA
| | - Beth A Erickson
- Departments of Radiation Oncology, Pancreatic Cancer Program, The Medical College of WisconsinMilwaukee, WI, USA
| | - Fabian M Johnston
- Departments of Surgery, Pancreatic Cancer Program, The Medical College of WisconsinMilwaukee, WI, USA
| | - Douglas B Evans
- Departments of Surgery, Pancreatic Cancer Program, The Medical College of WisconsinMilwaukee, WI, USA
| | - Susan Tsai
- Departments of Surgery, Pancreatic Cancer Program, The Medical College of WisconsinMilwaukee, WI, USA
| |
Collapse
|
29
|
Jenkinson C, Earl J, Ghaneh P, Halloran C, Carrato A, Greenhalf W, Neoptolemos J, Costello E. Biomarkers for early diagnosis of pancreatic cancer. Expert Rev Gastroenterol Hepatol 2015; 9:305-15. [PMID: 25373768 DOI: 10.1586/17474124.2015.965145] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma is an aggressive malignancy with a 5-year survival rate of approximately 5%. The lack of established strategies for early detection contributes to this poor prognosis. Although several novel candidate biomarkers have been proposed for earlier diagnosis, none have been adopted into routine clinical use. In this review, the authors examine the challenges associated with finding new pancreatic cancer diagnostic biomarkers and explore why translation of biomarker research for patient benefit has thus far failed. The authors also review recent progress and highlight advances in the understanding of the biology of pancreatic cancer that may lead to improvements in biomarker detection and implementation.
Collapse
Affiliation(s)
- Claire Jenkinson
- Department of Molecular and Clinical Cancer Medicine, National Institute for Health Research Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Daulby Street, Liverpool L69 3GA, UK
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Jin K, Luo G, Xiao Z, Liu Z, Liu C, Ji S, Xu J, Liu L, Long J, Ni Q, Yu X. Noncoding RNAs as potential biomarkers to predict the outcome in pancreatic cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:1247-55. [PMID: 25750521 PMCID: PMC4348055 DOI: 10.2147/dddt.s77597] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a common digestive system cancer, is highly malignant and has a poor disease outcome. Currently, all available examination and detection methods cannot accurately predict the clinical outcome. Therefore, it is extremely important to identify novel molecular biomarkers for personalized medication and to significantly improve the overall outcome. The “noncoding RNAs” (ncRNAs) are a group of RNAs that do not code for proteins, and they are categorized as structural RNAs and regulatory RNAs. It has been shown that microRNAs and long ncRNAs function as regulatory RNAs to affect the progression of various diseases. Many studies have confirmed a role for ncRNAs in the progression of PDAC during the last few years. Because of the significant role of ncRNAs in PDAC, ncRNA profiling may be used to predict PDAC outcome with high accuracy. This review comprehensively analyzes the value of ncRNAs as potential biomarkers to predict the outcome in PDAC and the possible mechanisms thereof.
Collapse
Affiliation(s)
- Kaizhou Jin
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Guopei Luo
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Zhiwen Xiao
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Zuqiang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Chen Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Shunrong Ji
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Jin Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Liang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Jiang Long
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Quanxing Ni
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China ; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China ; Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
31
|
Rossi ML, Rehman AA, Gondi CS. Therapeutic options for the management of pancreatic cancer. World J Gastroenterol 2014; 20:11142-11159. [PMID: 25170201 PMCID: PMC4145755 DOI: 10.3748/wjg.v20.i32.11142] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/11/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Since its initial characterization, pancreatic ductal adenocarcinoma has remained one of the most devastating and difficult cancers to treat. Pancreatic cancer is the fourth leading cause of death in the United States, resulting in an estimated 38460 deaths annually. With few screening tools available to detect this disease at an early stage, 94% of patients will die within five years of diagnosis. Despite decades of research that have led to a better understanding of the molecular and cellular signaling pathways in pancreatic cancer cells, few effective therapies have been developed to target these pathways. Other treatment options have included more sophisticated pancreatic cancer surgeries and combination therapies. While outcomes have improved modestly for these patients, more effective treatments are desperately needed. One of the greatest challenges in the future of treating this malignancy will be to develop therapies that target the tumor microenvironment and surrounding pancreatic cancer stem cells in addition to pancreatic cancer cells. Recent advances in targeting pancreatic stellate cells and the stroma have encouraged researchers to shift their focus to the role of desmoplasia in pancreatic cancer pathobiology in the hopes of developing newer-generation therapies. By combining novel agents with current cytotoxic chemotherapies and radiation therapy and personalizing them to each patient based on specific biomarkers, the goal of prolonging a patient’s life could be achieved. Here we review the most effective therapies that have been used for the treatment of pancreatic cancer and discuss the future potential of therapeutic options.
Collapse
|
32
|
Shaw VE, Lane B, Jenkinson C, Cox T, Greenhalf W, Halloran CM, Tang J, Sutton R, Neoptolemos JP, Costello E. Serum cytokine biomarker panels for discriminating pancreatic cancer from benign pancreatic disease. Mol Cancer 2014; 13:114. [PMID: 24884871 PMCID: PMC4032456 DOI: 10.1186/1476-4598-13-114] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/23/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND We investigated whether combinations of serum cytokines, used with logistic disease predictor models, could facilitate the detection of pancreatic ductal adenocarcinoma (PDAC). METHODS The serum levels of 27 cytokines were measured in 241 subjects, 127 with PDAC, 49 with chronic pancreatitis, 20 with benign biliary obstruction and 45 healthy controls. Samples were split randomly into independent training and test sets. Cytokine biomarker panels were selected by identifying the top performing cytokines in best fit logistic regression models during multiple rounds of resampling from the training dataset. Disease prediction by logistic models, built using the resulting cytokine panels, was evaluated with training and test sets and further examined using resampled performance evaluation. RESULTS For the discrimination of PDAC patients from patients with benign disease, a panel of IP-10, IL-6, PDGF plus CA19-9 offered improved diagnostic performance over CA19-9 alone in the training (AUC 0.838 vs. 0.678) and independent test set (AUC 0.884 vs. 0.798). For the discrimination of PDAC from CP, a panel of IL-8, CA19-9, IL-6 and IP-10 offered improved diagnostic performance over CA19-9 alone with the training (AUC 0.880 vs. 0.758) and test set (AUC 0.912 vs. 0.848). Finally, for the discrimination of PDAC in the presence of jaundice from benign controls with jaundice, a panel of IP-10, IL-8, IL-1b and PDGF demonstrated improvement over CA19-9 in the training (AUC 0.810 vs. 0.614) and test set (AUC 0.857 vs. 0.659). CONCLUSIONS These findings support the potential role for cytokine panels in the discrimination of PDAC from patients with benign pancreatic diseases and warrant additional study.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, Tumor-Associated, Carbohydrate/blood
- Antigens, Tumor-Associated, Carbohydrate/genetics
- Biomarkers/blood
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Case-Control Studies
- Cholestasis/blood
- Cholestasis/diagnosis
- Cholestasis/genetics
- Cholestasis/pathology
- Cytokines/blood
- Cytokines/genetics
- Diagnosis, Differential
- Female
- Gene Expression
- Humans
- Logistic Models
- Male
- Middle Aged
- Pancreas/metabolism
- Pancreas/pathology
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Pancreatitis, Chronic/blood
- Pancreatitis, Chronic/diagnosis
- Pancreatitis, Chronic/genetics
- Pancreatitis, Chronic/pathology
- Platelet-Derived Growth Factor/genetics
- Platelet-Derived Growth Factor/metabolism
Collapse
Affiliation(s)
- Victoria E Shaw
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| | - Brian Lane
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| | - Claire Jenkinson
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| | - Trevor Cox
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| | - William Greenhalf
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| | - Christopher M Halloran
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| | - Joseph Tang
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| | - Robert Sutton
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| | - John P Neoptolemos
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| | - Eithne Costello
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool and Broadgreen University Hospital NHS Trust, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
| |
Collapse
|
33
|
Jazieh KA, Foote MB, Diaz LA. The clinical utility of biomarkers in the management of pancreatic adenocarcinoma. Semin Radiat Oncol 2014; 24:67-76. [PMID: 24635863 DOI: 10.1016/j.semradonc.2013.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related deaths in the United States and survival rates have seen minimal improvement over the past few decades. Although results are poor, surgical resection is considered the only curative therapeutic intervention for pancreatic cancer, thereby emphasizing the significance of effective diagnostic and prognostic tools to improve outcomes. As such, biomarkers play a promising role in the development of personalized treatments for patients with pancreatic cancer. Prognostic biomarkers, such as serum carbohydrate antigen 19-9 in particular, as well as cancer stem cell markers, provide valuable insight into the biological processes of an individual and their likely course of disease. This, consequently, allows for the assessment of optimal therapeutic intervention. Furthermore, current efforts target putative predictive biomarkers such as BRCA2, PALB2, and SPARC so as to determine their influence on tumor response on targeted therapies. As research progresses, more evidence will provide clinicians with guidelines on the utilization of biomarkers to accurately stage and tailor personalized treatment to the needs of specific patients with pancreatic cancer.
Collapse
Affiliation(s)
- Khalid A Jazieh
- The Swim Across America Laboratory, The Ludwig Center for Cancer Genetics and Therapeutics, Baltimore, MD; The Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Michael B Foote
- The Swim Across America Laboratory, The Ludwig Center for Cancer Genetics and Therapeutics, Baltimore, MD; The Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Luis A Diaz
- The Swim Across America Laboratory, The Ludwig Center for Cancer Genetics and Therapeutics, Baltimore, MD; The Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD.
| |
Collapse
|
34
|
S-1 plus CIK as second-line treatment for advanced pancreatic cancer. Med Oncol 2013; 30:747. [PMID: 24122257 DOI: 10.1007/s12032-013-0747-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/02/2013] [Indexed: 01/26/2023]
Abstract
This study aimed to evaluate the efficacy and tolerability of S-1 (Tegafur, Gimeracil, and Oteracil Potassium Capsules) plus CIK (Cytokine-induced killer cells) in patients with advanced pancreatic cancer who had previously received gemcitabine-based therapy. In this prospective study, fifty-eight patients were randomly divided into two groups. One group (CT group) was given S-1 alone, and the other group (immuno-CT group) was given S-1 plus CIK. S-1 was administered orally twice a day at 80 mg/m(2)/day on days 1-21 of a 28-day cycle till disease progression or unacceptable toxicity occurred. CIK was given for one cycle of 28 days. The disease control rate for S-1 and CIK was 40.0 and 53.6%, respectively (p = 0.621). The serum CA19-9 level decreased for more than 25% was significantly different (33.3 and 60.7 % in CT group and immuno-CT group, respectively, p = 0.037). The median time to progression was 2.5 (95% CI 2.3-2.8) and 2.9 (95% CI 2.6-3.2) months (p = 0.037) for CT group and immuno-CT group, respectively. The median overall survival was 6.1 (95% CI 5.7-6.5) and 6.6 (95% CI 6.1-7.1) months (p = 0.09) for CT group and immuno-CT group, respectively. The difference in hematological toxicity, including leukocytopenia, anemia, and neutropenia, was insignificant between the two groups. In contrast, the differences in non-hematological toxicity, fatigue, and non-infective fever were significantly different between the two groups (p < 0.05). The S-1 plus CIK regimen was well tolerated in a second-line setting in patients with gemcitabine-refractory and advanced pancreatic cancer.
Collapse
|
35
|
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related death in the United States. There has been minimal progress with regard to cancer-specific outcomes in recent decades. Although effective therapies will undoubtedly change the natural history of the disease, effective biomarkers are a promising tool that will likely have a positive impact and will undoubtedly have an important role in the management of patients with pancreatic ductal adenocarcinoma (PDA) in the future. At present, serum CA-19-9 (carbohydrate antigen 19-9) is the only Food and Drug Administration-approved biomarker for PDA, and it has utility as a prognostic marker and as a marker of disease recurrence. There has been a recent explosion in the pancreatic cancer biomarker field with more than 2000 biomarker studies implicating thousands of informative genes as candidate biomarkers. In this review, we summarize the literature on CA-19-9 in PDA and highlight the most promising investigational biomarkers. Distinctions are made between diagnostic biomarkers (detection of disease), prognostic biomarkers (provide information on prognosis and recurrence pattern), and predictive biomarkers (predict treatment response).
Collapse
|
36
|
Tonack S, Jenkinson C, Cox T, Elliott V, Jenkins RE, Kitteringham NR, Greenhalf W, Shaw V, Michalski CW, Friess H, Neoptolemos JP, Costello E. iTRAQ reveals candidate pancreatic cancer serum biomarkers: influence of obstructive jaundice on their performance. Br J Cancer 2013; 108:1846-53. [PMID: 23579209 PMCID: PMC3658525 DOI: 10.1038/bjc.2013.150] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 01/09/2013] [Accepted: 03/14/2013] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The aims of our study were to identify serum biomarkers that distinguish pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC) patients from benign pancreatic disease patients and healthy subjects, and to assess the effects of jaundice on biomarker performance. METHODS Isobaric tags for relative and absolute quantification were used to compare pooled serum and pancreatic juice samples from a test set of 59 and 25 subjects, respectively. Validation was undertaken in 113 independent subjects. RESULTS Candidate proteins Complement C5, inter-α-trypsin inhibitor heavy chain H3, α1-β glycoprotein and polymeric immunoglobulin receptor were elevated in cancer, as were the reference markers CA19-9 and Reg3A. Biliary obstruction had a significant effect on the performance of the markers, in particular within the PDAC group where the presence of jaundice was associated with a significant increase in the levels of all six proteins (P<0.01). Consequently, in the absence of jaundice, proteins showed reduced sensitivity for PDAC patients over benign subjects and healthy controls (HCs). Similarly, in the presence of jaundice, markers showed reduced specificity for PDAC patients over benign subjects with jaundice. Combining markers enabled improved sensitivity for non-jaundiced PDAC patients over HCs and improved specificity for jaundiced PDAC patients over jaundiced benign disease subjects. CONCLUSIONS The presence-absence of jaundice in the clinical scenario severely impacts the performance of biomarkers for PDAC diagnosis and has implications for their clinical translation.
Collapse
Affiliation(s)
- S Tonack
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - C Jenkinson
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - T Cox
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - V Elliott
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
- National Institute for Health Research Liverpool Pancreatic Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - R E Jenkins
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Liverpool, UK
| | - N R Kitteringham
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Liverpool, UK
| | - W Greenhalf
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
- National Institute for Health Research Liverpool Pancreatic Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - V Shaw
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - C W Michalski
- Department of General Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - H Friess
- Department of General Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - J P Neoptolemos
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
- National Institute for Health Research Liverpool Pancreatic Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - E Costello
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
- National Institute for Health Research Liverpool Pancreatic Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
37
|
Vainshtein JM, Schipper M, Zalupski MM, Lawrence TS, Abrams R, Francis IR, Khan G, Leslie W, Ben-Josef E. Prognostic significance of carbohydrate antigen 19-9 in unresectable locally advanced pancreatic cancer treated with dose-escalated intensity modulated radiation therapy and concurrent full-dose gemcitabine: analysis of a prospective phase 1/2 dose escalation study. Int J Radiat Oncol Biol Phys 2012; 86:96-101. [PMID: 23265573 DOI: 10.1016/j.ijrobp.2012.11.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 11/06/2012] [Accepted: 11/11/2012] [Indexed: 12/19/2022]
Abstract
PURPOSE Although established in the postresection setting, the prognostic value of carbohydrate antigen 19-9 (CA19-9) in unresectable locally advanced pancreatic cancer (LAPC) is less clear. We examined the prognostic utility of CA19-9 in patients with unresectable LAPC treated on a prospective trial of intensity modulated radiation therapy (IMRT) dose escalation with concurrent gemcitabine. METHODS AND MATERIALS Forty-six patients with unresectable LAPC were treated at the University of Michigan on a phase 1/2 trial of IMRT dose escalation with concurrent gemcitabine. CA19-9 was obtained at baseline and during routine follow-up. Cox models were used to assess the effect of baseline factors on freedom from local progression (FFLP), distant progression (FFDP), progression-free survival (PFS), and overall survival (OS). Stepwise forward regression was used to build multivariate predictive models for each endpoint. RESULTS Thirty-eight patients were eligible for the present analysis. On univariate analysis, baseline CA19-9 and age predicted OS, CA19-9 at baseline and 3 months predicted PFS, gross tumor volume (GTV) and black race predicted FFLP, and CA19-9 at 3 months predicted FFDP. On stepwise multivariate regression modeling, baseline CA19-9, age, and female sex predicted OS; baseline CA19-9 and female sex predicted both PFS and FFDP; and GTV predicted FFLP. Patients with baseline CA19-9 ≤ 90 U/mL had improved OS (median 23.0 vs 11.1 months, HR 2.88, P<.01) and PFS (14.4 vs 7.0 months, HR 3.61, P=.001). CA19-9 progression over 90 U/mL was prognostic for both OS (HR 3.65, P=.001) and PFS (HR 3.04, P=.001), and it was a stronger predictor of death than either local progression (HR 1.46, P=.42) or distant progression (HR 3.31, P=.004). CONCLUSIONS In patients with unresectable LAPC undergoing definitive chemoradiation therapy, baseline CA19-9 was independently prognostic even after established prognostic factors were controlled for, whereas CA19-9 progression strongly predicted disease progression and death. Future trials should stratify by baseline CA19-9 and incorporate CA19-9 progression as a criterion for progressive disease.
Collapse
Affiliation(s)
- Jeffrey M Vainshtein
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Martin LK, Li X, Kleiber B, Ellison EC, Bloomston M, Zalupski M, Bekaii-Saab TS. VEGF remains an interesting target in advanced pancreas cancer (APCA): results of a multi-institutional phase II study of bevacizumab, gemcitabine, and infusional 5-fluorouracil in patients with APCA. Ann Oncol 2012; 23:2812-2820. [PMID: 22767582 PMCID: PMC3841413 DOI: 10.1093/annonc/mds134] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 04/06/2012] [Accepted: 04/10/2012] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND We investigated the safety and efficacy of bevacizumab combined with gemcitabine followed by infusional 5-fluorouracil (5-FU) in patients with advanced pancreas cancer (APCA). DESIGN Patients with untreated APCA received bevacizumab 10 mg/kg, gemcitabine 1000 mg/m(2) over 100 min, and 5-FU 2400 mg/m(2) over 48 h on days 1 and 15 of each 28-day cycle. The primary end point was the proportion of patients with progression-free survival (PFS) at 6 months from initiation of therapy. If PFS at 6 months was ≥41%, the regimen would be considered promising. RESULTS Forty-two patients were enrolled in the study; of which, 39 were evaluable for primary end point. PFS at 6 months was 49% (95% CI 34% to 64%). Median PFS was 5.9 months (95% CI 3.5 to 8.1) and median overall survival (OS) was 7.4 months (95% CI 4.7 to 11.2). Partial response and stable disease occurred in 30% and 45% of patients, respectively. Treatment-related hypertension and normal baseline albumin correlated with an improved response rate, PFS and OS. Grade 3 to 4 toxicities included fatigue (14%), hypertension (5%), and venous thrombosis (5%). CONCLUSIONS The study met its primary end point. Further investigation of anti-VEGF therapy in combination with fluoropyrimidine-based therapy is warranted in APCA. Treatment-related hypertension and normal baseline albumin may predict for the efficacy of bevacizumab and should be investigated in prospective studies.
Collapse
Affiliation(s)
- L K Martin
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus
| | - X Li
- Center for Biostatistics, The Ohio State University, Columbus
| | - B Kleiber
- Comprehensive Cancer Center, The Ohio State University, Columbus
| | - E C Ellison
- Department of Surgery, Division of General Surgery, The Ohio State University Medical Center, Columbus
| | - M Bloomston
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus
| | - M Zalupski
- Department of Internal Medicine, Division of Hematology-Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, USA
| | - T S Bekaii-Saab
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus.
| |
Collapse
|
40
|
Ko AH, Truong TG, Kantoff E, Jones KA, Dito E, Ong A, Tempero MA. A phase I trial of nab-paclitaxel, gemcitabine, and capecitabine for metastatic pancreatic cancer. Cancer Chemother Pharmacol 2012; 70:875-81. [PMID: 23053263 DOI: 10.1007/s00280-012-1979-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 09/13/2012] [Indexed: 02/05/2023]
Abstract
BACKGROUND Substantial antitumor activity has previously been demonstrated with the addition of nab-paclitaxel (Abraxane [Celgene, Summit, NJ]), an albumin-bound formulation of paclitaxel, to gemcitabine in patients with advanced pancreatic cancer. Given preclinical evidence of synergy when a fluoropyrimidine is added to gemcitabine plus a taxane in a sequence-specific schedule, we conducted a phase I study to evaluate the combination of nab-paclitaxel, gemcitabine, and capecitabine administered biweekly in patients with metastatic pancreatic adenocarcinoma. MATERIALS AND METHODS Patients with previously untreated metastatic pancreatic cancer and an ECOG performance status of 0-1 were eligible to participate. Study design utilized a 3 + 3 dose-escalation schema, with expanded cohort at maximum-tolerated dose (MTD). Treatment was administered in 14-day cycles, with capecitabine given on days 1-7 and both gemcitabine (at fixed-dose rate infusion) and nab-paclitaxel on day 4 of each cycle. Dose-limiting toxicity (DLT) definitions included grade 3-4 hematologic toxicities and grade 2-4 hand-foot syndrome, neuropathy, or diarrhea. RESULTS Fifteen patients were enrolled across two dose levels. Final MTD was established at nab-paclitaxel 100 mg/m(2), gemcitabine 750 mg/m(2), and capecitabine 750 mg/m(2) twice daily. Patients received a median of four treatment cycles (range 1-16). The most frequent adverse events (any grade) for the entire study cohort included fatigue, rash/hand-foot syndrome, nausea/vomiting, diarrhea, neuropathy, and elevated liver function tests. Ten patients (66.7 %) experienced at least one grade 3-4 adverse event. Grade 3-4 hematologic toxicities were uncommon. Two of 14 evaluable patients (14.3 %) exhibited a partial response, and 6 of 12 patients (50 %) with elevated CA19-9 at baseline had a ≥50 % biomarker decline. CONCLUSION While well tolerated overall, this regimen demonstrated only modest antitumor activity in patients with metastatic pancreatic cancer. Recognizing the limits of cross-study comparisons and small sample size, these results do not match those reported at MTD in the phase I/II trial of gemcitabine/nab-paclitaxel. The lower doses used in the current study suggest that dose intensity may be a critical aspect to optimize multidrug regimens.
Collapse
Affiliation(s)
- Andrew H Ko
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 94115, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Bauer TM, El-Rayes BF, Li X, Hammad N, Philip PA, Shields AF, Zalupski MM, Bekaii-Saab T. Carbohydrate antigen 19-9 is a prognostic and predictive biomarker in patients with advanced pancreatic cancer who receive gemcitabine-containing chemotherapy: a pooled analysis of 6 prospective trials. Cancer 2012; 119:285-92. [PMID: 22786786 DOI: 10.1002/cncr.27734] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 05/17/2012] [Accepted: 06/07/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND Carbohydrate antigen 19-9 (CA19-9) is a widely used biomarker in pancreatic cancer. There is no consensus on the interpretation of the change in CA19-9 serum levels and its role in the clinical management of patients with pancreatic cancer. METHODS Individual patient data from 6 prospective trials evaluating gemcitabine-containing regimens from 3 different institutions were pooled. CA19-9 values were obtained at baseline and after successive cycles of treatment. The objective of this study was to correlate a decline in CA19-9 with outcomes while undergoing treatment. RESULTS A total of 212 patients with locally advanced (n = 50) or metastatic (n = 162) adenocarcinoma of the pancreas were included. Median baseline CA19-9 level was 1077 ng/mL (range, 15-492,241 ng/mL). Groups were divided into those levels below (low) or above (high) the median. Median overall survival (mOS) was 8.7 versus 5.2 months (P = .0018) and median time to progression (mTTP) was 5.8 versus 3.7 months (P = .082) in the low versus high groups, respectively. After 2 cycles of chemotherapy, up to a 5% increase versus ≥ 5% increase in CA19-9 levels conferred an improved mOS (10.3 vs 5.1 months, P = .0022) and mTTP (7.5 vs 3.5 months, P = 0.0005). CONCLUSIONS In patients who have advanced pancreatic cancer treated with gemcitabine-containing regimens baseline CA19-9 is prognostic for outcome. A decline in CA19-9 after the second cycle of chemotherapy is not predictive of improved mOS or mTTP; thus, CA19-9 decline is not a useful surrogate endpoint in clinical trials. Clinically, a ≥ 5% rise in CA19-9 after 2 cycles of chemotherapy serves as a negative predictive marker.
Collapse
Affiliation(s)
- Todd M Bauer
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
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: 1.7] [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
|
43
|
Winter JM, Yeo CJ, Brody JR. Diagnostic, prognostic, and predictive biomarkers in pancreatic cancer. J Surg Oncol 2012; 107:15-22. [PMID: 22729569 DOI: 10.1002/jso.23192] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 05/18/2012] [Indexed: 12/13/2022]
Abstract
Serum CA 19-9 is the only FDA approved biomarker recommended for use in the routine management of pancreatic ductal adenocarcinoma (PDA). Over 2,000 biomarker studies related to pancreatic cancer appear in the literature, highlighting the need to discover and develop improved tests. Diagnostic biomarkers have implications for early detection of PDA, prognostic markers predict patient survival and recurrence patterns, and predictive markers can help personalize treatment regimens.
Collapse
Affiliation(s)
- Jordan M Winter
- Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | | | | |
Collapse
|
44
|
Hecht JR, Farrell JJ, Senzer N, Nemunaitis J, Rosemurgy A, Chung T, Hanna N, Chang KJ, Javle M, Posner M, Waxman I, Reid A, Erickson R, Canto M, Chak A, Blatner G, Kovacevic M, Thornton M. EUS or percutaneously guided intratumoral TNFerade biologic with 5-fluorouracil and radiotherapy for first-line treatment of locally advanced pancreatic cancer: a phase I/II study. Gastrointest Endosc 2012; 75:332-8. [PMID: 22248601 PMCID: PMC4380192 DOI: 10.1016/j.gie.2011.10.007] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 10/04/2011] [Indexed: 02/06/2023]
Abstract
BACKGROUND TNFeradeBiologic (AdGVEGR.TNF.11D) is a replication-deficient adenoviral vector that expresses tumor necrosis factor-α (TNF-α) under the control of the Egr-1 promoter, which is inducible by chemotherapy and radiation. OBJECTIVE This study was conducted to determine the maximal tolerated dose of TNFeradeBiologic with standard chemoradiotherapy and preliminary activity and safety of the combination in the treatment of locally advanced pancreatic cancer (LAPC). DESIGN TNFeradeBiologic was injected into locally advanced pancreatic carcinomas by using EUS or percutaneous administration once a week for 5 weeks together with 50.4 Gy radiation and 5-fluorouracil (5-FU) 200 mg/m(2) daily over 5.5 weeks. Dose levels from 4 × 10(9) to 1 × 10(12) particle units (PU) were studied. SETTING Multicentered, academic institutions. PATIENTS Fifty patients with LAPC were treated. INTERVENTIONS Doses of TNFerade Biologic were administered to patients. MAIN OUTCOME MEASUREMENTS Toleration of TNFerade Biologic was measured through toxicity and tumor response, by using the criteria of the Response Evaluation Criteria in Solid Tumors and the World Health Organization, and was reviewed by a central radiology facility. Overall survival and progression-free survival were also measured. RESULTS Dose-limiting toxicities of pancreatitis and cholangitis were observed in 3 patients at the 1 × 10(12) PU dose, making 4 × 10(11) PU the maximum tolerated dose. One complete response, 3 partial responses, and 12 patients with stable disease were noted. Seven patients eventually went to surgery, 6 had clear margins, and 3 survived >24 months. LIMITATIONS This is a Phase 1/2 non-randomized study. CONCLUSIONS Intratumoral delivery of TNFerade Biologic by EUS with fine-needle viral injection or percutaneously, combined with chemoradiation, shows promise in the treatment of LAPC. There appeared to be better clinical outcome at the maximal tolerated dose than at lower doses. The dose of 4 ×10(11) PU TNFerade Biologic was generally well tolerated, with encouraging indications of activity, and will be tested in the randomized phase of this study. Delivery of TNFerade Biologic did not interfere with subsequent surgical resection.
Collapse
Affiliation(s)
- J Randolph Hecht
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
A phase I/II study of gemcitabine-concurrent proton radiotherapy for locally advanced pancreatic cancer without distant metastasis. Radiother Oncol 2012; 103:25-31. [PMID: 22300608 DOI: 10.1016/j.radonc.2011.12.029] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 12/15/2011] [Accepted: 12/20/2011] [Indexed: 01/12/2023]
Abstract
PURPOSE We conducted the study to assess the feasibility and efficacy of gemcitabine-concurrent proton radiotherapy (GPT) for locally advanced pancreatic cancer (LAPC). MATERIALS AND METHODS Of all 50 patients who participated in the study, 5 patients with gastrointestinal (GI)-adjacent LAPC were enrolled in P-1 (50 Gy equivalent [GyE] in 25 fractions) and 5 patients with non-GI-adjacent LAPC in P-2 (70.2 GyE in 26 fractions), and 40 patients with LAPC regardless of GI-adjacency in P-3 (67.5 GyE in 25 fractions using the field-within-a-field technique). In every protocol, gemcitabine (800 mg/m(2)/week for 3 weeks) was administered concurrently. Every patient received adjuvant chemotherapy including gemcitabine after GPT within the tolerable limit. RESULTS The median follow-up period was 12.5 months. The scheduled GPT was feasible for all except 6 patients (12%) due to acute hematologic or GI toxicities. Grade 3 or greater late gastric ulcer and hemorrhage were seen in 5 patients (10%) in P-2 and P-3. The one-year freedom from local-progression, progression-free, and overall survival rates were 81.7%, 64.3%, and 76.8%, respectively. CONCLUSION GPT was feasible and showed high efficacy. Although the number of patients and the follow-up periods are insufficient, the clinical results seem very encouraging.
Collapse
|
46
|
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: 202] [Impact Index Per Article: 16.8] [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
|
47
|
Heinemann V, Haas M, Boeck S. Systemic treatment of advanced pancreatic cancer. Cancer Treat Rev 2012; 38:843-53. [PMID: 22226241 DOI: 10.1016/j.ctrv.2011.12.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer belongs to the most malignant gastrointestinal cancers and, in its advanced stage, remains a deadly disease for nearly all affected patients. Treatment of metastatic adenocarcinoma of the pancreas not only involves chemotherapy and targeted therapy, but also requires attention to accompanying comorbidities as well as frequently intensive supportive treatment and psychosocial support. Gemcitabine-based combinations with fluoropyrimidines and platin analogs have essentially failed to provide a substantial prolongation of survival and may constitute a treatment option only in patients with a good performance status. Among targeted therapies, only the EGFR tyrosine kinase inhibitor erlotinib has shown activity which is marginal in the overall population, but clinically relevant in patients developing skin rash. New avenues of polychemotherapy are presently explored since the gemcitabine-free FOLFIRINOX-regimen (infusional 5-fluorouracil/folinic acid plus irinotecan and oxaliplatin) was shown to be markedly superior to gemcitabine in selected good-performance patients. Pancreatic cancer is notably characterized as a hypovascular tumor rich in desmoplastic stromal tissue. An innovative approach to treatment therefore focuses on peritumoral fibroblasts and aims to induce a depletion of the stroma either by inhibition of the hedgehog pathway or by targeting SPARC (secreted protein acidic and rich in cysteine) via application of albumin-bound paclitaxel.
Collapse
Affiliation(s)
- Volker Heinemann
- Klinikum Grosshadern, Department of Oncology and Comprehensive Cancer Center, Ludwig-Maximilians-Universität München, Germany.
| | | | | |
Collapse
|
48
|
Goldstein D, Spry N, Cummins MM, Brown C, van Hazel GA, Carroll S, Selva-Nayagam S, Borg M, Ackland SP, Wratten C, Shapiro J, Porter IWT, Hruby G, Horvath L, Bydder S, Underhill C, Harvey J, Gebski VJ. The GOFURTGO Study: AGITG phase II study of fixed dose rate gemcitabine-oxaliplatin integrated with concomitant 5FU and 3-D conformal radiotherapy for the treatment of localised pancreatic cancer. Br J Cancer 2011; 106:61-9. [PMID: 22134511 PMCID: PMC3251866 DOI: 10.1038/bjc.2011.526] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background: Locally advanced inoperable pancreatic cancer (LAPC) has a poor prognosis. By increasing intensity of systemic therapy combined with an established safe chemoradiation technique, our intention was to enhance the outcomes of LAPC. In preparation for phase III evaluation, the feasibility and efficacy of our candidate regimen gemcitabine–oxaliplatin chemotherapy with sandwich 5-fluorouracil (5FU) and three-dimensional conformal radiotherapy (3DCRT) needs to be established. Methods: A total of 48 patients with inoperable LAPC without metastases were given gemcitabine (1000 mg m−2 d1 + d15 q28) and oxaliplatin (100 mg m−2 d2 + d16 q28) in induction (one cycle) and consolidation (three cycles), and 5FU 200 mg m−2 per day over 6 weeks during 3DCRT 54 Gy. Results: Median duration of sustained local control (LC) was 15.8 months, progression-free survival (PFS) was 11.0 months, and overall survival was 15.7 months. Survival rates for 1, 2, and 3 years were 70.2%, 21.3%, and 12.8%, respectively. Global quality of life did not significantly decline from baseline during treatment, which was associated with modest treatment-related toxicity. Conclusion: Fixed-dose gemcitabine and oxaliplatin, combined with an effective and safe regimen of 5FU and 3DCRT radiotherapy, was feasible and reasonably tolerated. The observed improved duration of LC and PFS with more intensive therapy over previous trials may be due to patient selection, but suggest that further evaluation in phase III trials is warranted.
Collapse
Affiliation(s)
- D Goldstein
- Department of Medical Oncology, Prince of Wales Hospital, High Street, Randwick, New South Wales 2031, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Brody JR, Witkiewicz AK, Yeo CJ. The past, present, and future of biomarkers: a need for molecular beacons for the clinical management of pancreatic cancer. Adv Surg 2011; 45:301-21. [PMID: 21954696 DOI: 10.1016/j.yasu.2011.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Affiliation(s)
- Jonathan R Brody
- Department of Surgery, Jefferson Pancreas Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
50
|
Maeda S, Motoi F, Onogawa T, Morikawa T, Shigeru O, Sakata N, Takadate T, Naitoh T, Rikiyama T, Katayose Y, Egawa S, Unno M. Paclitaxel as second-line chemotherapy in patients with gemcitabine-refractory pancreatic cancer: a retrospective study. Int J Clin Oncol 2011; 16:539-45. [PMID: 21455624 DOI: 10.1007/s10147-011-0220-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 02/25/2011] [Indexed: 12/13/2022]
Abstract
BACKGROUND We evaluated the efficacy and feasibility of paclitaxel in patients with gemcitabine-refractory pancreatic cancer. We also evaluated the correlation between tumor marker decline and response rate. METHODS Thirty patients histologically diagnosed with pancreatic cancer who had undergone paclitaxel treatment as salvage chemotherapy were retrospectively analyzed. Paclitaxel treatment was performed with 80 mg/(m(2) week) for 3 weeks followed by 1 week rest, and this was continued until failure. Tumor marker response was evaluated by measuring levels of carbohydrate antigen 19-9, carcinoembryonic antigen, and DUPAN-2 monthly. RESULTS In total, 272 weekly paclitaxel treatments were performed. The median number of treatments per patient was 8 (range 1-22). The median overall survival from the start of paclitaxel treatment was 6.7 months (range 1.2-18.8). The response rate was 10% (3/30 patients) and the disease control rate was 46.7% (14/30 patients). Although grade 3 and 4 hematological and non-hematological toxicities were seen in 7 and 6 patients, respectively, adverse events were managed by conservative treatment. We found a significant correlation between the disease control rate and tumor marker decline within 2 months of paclitaxel treatment (P = 0.01). Patients with tumor marker decline tended to survive longer. CONCLUSION Weekly administration of paclitaxel in patients with gemcitabine-refractory pancreatic cancer seems to be well tolerated and can be effective. Paclitaxel treatment should be considered as salvage chemotherapy after gemcitabine failure in patients with good performance status.
Collapse
Affiliation(s)
- Shimpei Maeda
- Division of Hepato-Biliary Pancreatic Surgery, Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|