Abstract
Biomarkers play an essential role in the management of patients with invasive cancers. Pancreatic ductal adenocarcinoma (PDC) associated with poor prognosis due to advanced presentation and limited therapeutic options. This is further complicated by absence of validated screening and predictive biomarkers for early diagnosis and precision treatments respectively. There is emerging data on biomarkers in pancreatic cancer in past two decades. So far, the CA 19-9 remains the only approved biomarker for diagnosis and response assessment but limited by low sensitivity and specificity. In this article, we aim to review current and future biomarkers that has potential serve as critical tools for early diagnostic, predictive and prognostic indications in pancreatic cancer.
Key words: Pancreatic cancer, biomarkers
Introduction
Pancreatic ductal adenocarcinoma (PDC) is the most common subtype of pancreatic cancer, estimated around 85%.1 Age-standardized incidence rate of PDC is 7.2 to 2.8 per 100,000 in developed region versus less developed2 countries. Northern America considered having highest incidence rate globally while Africa has the lowest rate 7.4 and 2 per 100,000 respectively. Globally the mortality rate coincides with the incidence rate, emphasizing the poor prognosis. In United States, pancreatic cancer is the 4th leading cause of cancer-related mortality.3 Recent epidemiology studies show that the incidence of new pancreatic cancer has been gradually increased over time4 and within a decade, it is expected to rise to the second leading cause of cancer-related mortality behind lung cancer. In recent times, five years survival rate is minimally improved and reaches only 7% among all stages of pancreatic cancer.5 The screening programs for PDC remains challenge compared with other tumors-lung, breast, colon and cervix. The barriers to develop screening test to detect pancreatic cancer include specificity of the chosen test and the relatively low incidence of the disease. This can lead to multiple false positive cases and further challenged by the cost and morbidity associated with invasive confirmatory testing. To overcome this in unselected patient population, a high performing screening test with sensitivity and specificity close to 100% is required. Current attempts to discover screening tests in PDC for early diagnosis have focused mainly on serum biomarkers. According to national cancer institute, the biomarker has been defined as any substance, structure, or process that can be measured in the body or its products and influence or predict the incidence of outcome or disease. The carbohydrate antigen (CA) 19-9, the only biomarker approved by US Food and Drug Administration (FDA) is not considered as a screening tool due to its low sensitivity and specificity.6,7 It was reported previously that the median sensitivity for CA19-9 was 79% while median specificity was 82%.8 Therefore, the screening efforts were directed on the high-risk groups with familial risk and chronic pancreatitis; however, these represent the minority of affected individuals. Applying screening strategies to patients with one or more of the risk factors could enhance the performance of a putative screening test. Biomarkers role is crucial in diagnostic and therapeutic approach in cancer treatment and are key assets in identification of a sub-group of population to target preventive interventions.9 In the sporadic pancreatic cancer group, no biomarkers so far with high enough accuracy are currently available for use in screening and therefore an urgent unmet need for identification of right biomarker.9 The aim of this article was to review the novel biological and molecular biomarkers with diagnostic, predictive and prognostic potential in PDC patients.
Diagnostic markers
Most patients with early-stage PDC are asymptomatic, however commonly diagnosed at advanced stages, where the treatment options are limited and associated with worse clinical outcomes. The poor prognosis of PDC attributed to late diagnosis with advanced presentation, where curative therapeutic options are lacking. Identifying robust biomarkers for earlier detection could enable management of these cancers with curative intent and thus reducing the PDC mortality. To date, there is no biomarker approved for early diagnosis. This underscores the unmet need for development of early detection biomarkers.
Carbohydrate antigens and carcinoembryonic antigen
Serum carbohydrate antigen (CA) 19-9 is the most common and validated diagnostic tumor marker with sensitivity and specificity of 79-81% and 82-90% respectively; but have poor predictive value of 0.5-0.9% in asymptomatic patient.10 CA 19-9 can be elevated in other medical conditions such as acute cholangitis, pancreatitis, obstructive jaundice and liver cirrhosis. Additionally, Lewis-negative blood type patient, which makes 5-10% Caucasian, do not produce CA 19-9 levels,10 thus contributing to false negativity. Currently, CA 19-9 is being applied in clinical practice for prediction of treatment response and prognostication. Few other carbohydrate antigens have also been studied extensively including CA-242, CA 50, CA 195, CA 72-4, CEA and CA-125, and found to be overall less sensitive than CA19-9.11,12
MicroRNAs
MicroRNA (miRNAs) belongs to a class of non-coding RNA that involve in expression of post-transcriptional regulatory mechanisms. Use of miRNAs expression profiling has gained importance as a biomarker for early detection of cancer.13,14 In pancreatic cancer, miRNAs dysregulation has been profiled in pancreatic tissue, blood, stool and saliva.15 Among several different miRNAs, miR-21, miR-155 and mi-R 196 have been demonstrated to be upregulated in PDC and can differentiate from pre-cancerous lesions as weli.16-19 Since specimen acquisition from pancreatic juice and pancreatic tissue, requires invasive approach, non-invasive techniques such as fecal and urinary specimen has been studied for diagnostic purposes. Three miRNAs (miR-143, miR-223 and miR-30e) as assessed in urine samples were over expressed in stage I cancer as compared to healthy individuals. Additionally, miR-223 and miR-204 could distinguish early stage cancer from chronic pancreatitis. Furthermore, combination use of miR-143 and miR-30e achieved sensitivity of 83.3% and specificity of 96.2%.20 Similarly, higher levels of miR-21 and miR-155 levels in PDC compared to normal controls was reported in stool specimen.21
Macrophage inhibitory cytokine 1
Macrophage inhibitory cytokine 1 (MIC-1) is an autocrine regulatory molecule, which distantly belong to transformer growth factor beta (TGF-β) superfamily. Serum MIC-1 levels may serve as a novel diagnostic biomarker for early detection of pancreatic cancers. A study by Koopman et al. demonstrated that serum MIC-1 outperform all serum markers including CA 19-9 levels in distinguishing resectable pancreatic cancer from healthy controls.22 Recent studies including meta-analysis showed, serum MIC-1 levels were higher in pancreatic cancer patients as compared to controls. 23,24
PAM 4
PAM4 is a murine monoclonal antibody (mAb) is reactive to Mucin 5 AC, a secretory mucin. The expression of PAM4 is highly restricted to early stages of neoplastic development in pancreas, including pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN).25 PAM4 antibodies were found to be absent in normal pancreatic tissues and solely expressed in pancreatic cancers or those with early neoplastic changes. Gold et.al reported higher specificity of 85% for PAM4 alone in comparison CA 19-9 with 68%.26 Interestingly, combined PAM4/CA19-9 assay reported to have improved sensitivity (84%) for early detection of PDC along-with improved specificity (82%). Additionally, PAM4 has been radiolabeled to enhance diagnostic accuracy27 by radio-immunodetection.
Glypican
Glypican 1 (GPC1), a membrane anchoring protein, found to be overexpressed in various cancers. GPC1 is highly expressed as assessed by immunohistochemical assessment, in pancreatic cancer tissue as compared to normal tissue.28 Additionally, GPC1 had an independent prognostic effect on overall survival.28 Similar results were reported for Glypican 3 (GPC3) in pancreatic cancers. A recent study by Yao et.al reported overexpression of GPC3 associated with progression, carcinogenesis and poor progression in PDC.29 In a novel approach GPC1 circulating exosomes (GPC1 crExos) were monitored with flow cytometry in serum of patients and mice with cancer by Melo et al.30 GPC1 crExos demonstrated nearly perfect values when comparing patients with PDAC, chronic pancreatitis and healthy individuals. GPC1+ crExos showed a sensitivity and specificity of 100% in each stage of pancreas cancer. 30 Moreover, GPC1+ crExos demonstrated superior prognostic indicator to CA19-9 and elevated levels prior to MRI detectable lesions in pancreatic cancer. These evidences suggest its utility as novel, non-invasive biomarker in early diagnosis and potential use in pancreatic cancer progression.
KRAS mutation
KRAS mutations occur very frequently in pancreatic cancer and were extensively studied. The diagnostic accuracy of KRAS mutation was not optimal for diagnostic utility due to non-specificity of these mutations.31 The low level of cell-free circulating tumor DNA (ctDNA) in serum limits non- invasive assessments.32
Osteopontin
Osteopontin (OPN), a protein of extra-cellular matrix, has been reported to be upregulated in pancreatic cancers with sensitivity of 80% and specificity of 97% for detection of pancreatic cancers.33 However, the OPN did not diagnostic accuracy over CA 19-9 levels alone, but a diagnostic panel including OPN,TIMP-1 and CA 19-9 achieved better sensitivity and specificity.34,35 These biomarkers require further investigation to determine their role as a diagnostic biomarker in pancreatic cancer.
Epigenetic markers
Epigenetic changes can contribute to both cancer initiation and progression in PDC that evolves through non-invasive precursor pancreatic intraepithelial neoplasias (PanINs).36 The PanINs typically take 10 to 15 years to develop into malignant lesions that can further metastasize,37 thus an ideal context for the early detection. The serum markers, CA19-9 and radiological imaging are not reliable and therefore epigenetically silenced genes such as NTPX2, SARP2, RPRM, and LHX1 are currently under investigation.38 The sources for assessment of methylation markers include pancreatic juice, cell free tumor DNA and brush samples.39
Pancreatic juice with exfoliated cells from diffuse areas of the pancreas, can form a good source for early detection in pancreatic epithelial changes. Emerging data suggests that patients with malignant transformation can be differentiated from benign changes with higher specificity by assessing methylation of the genes: CCND2, TFPI2, PENK, NPTX2, FOXE1, CD1D, KCNK12, CLEC11A, NDRG4, IKZF1, and PKRCB.40,41 Prediction models generated by assessing methylation promoters of BMP3, RASSF1A, BNC1, MESTv2, TFPI2, APC, SFRP1 and SFRP2 in cell free DNA in plasma generated detection probability with >75% sensitivity and >80% specificity for PDC.42 Methylation levels of TFPI2, NPTX2 and CCND2 in endoscopic biliary brush samples from patients with PDC, correlated with detection in 73% of patients.43 Currently, no epigenetic biomarker has been approved for detection, however independent validation in large samples are anticipated in future with increased availability of genome wide analysis.
Predictive biomarkers
The advanced pancreatic cancer patients associated with poor prognosis in spite of available therapeutic options It appears that overall emphasis in identifying predictive biomarkers is relatively low compared to diagnostic markers, likely due to limited therapeutic options.
Gemcitabine markers
Gemcitabine, a nucleoside analog, since its approval in 1996 has been the cornerstone therapy for neo-adjuvant, adjuvant and palliative chemotherapy in pancreatic cancer. It was suggested that the two genes, GSTM1 and ONECU were found to be differentially methylated between responders and the non-responders.44 Cellular uptake mechanisms are the key to develop gemcitabine toxicity and resistance.45 The following nucleoside transporters involved in the uptake of this drug have been evaluated for predicting the gemcitabine response.46
Human equilibrate nucleoside transporter 1 (hENT1)
hENT1 relationship with gemcitabine as a predictive biomarker was initially evaluated by immunostaining in a small study that demonstrated significantly longer survival with gemcitabine chemotherapy as compared to without detectable hENT1 (13 months versus 4 months, P=0.001).47 These findings were validated in a larger cohort and in adjuvant setting validating that hENT1 expression can predict the gemcitabine response correlating with improved survival outcomes.48,49 An ongoing randomized clinical trial, evaluating if hENT1 can predict response to gemcitabine treatment and whether combination therapy of 5-FU, leucovorin and oxaliplatin (FOLFOX) might be a superior treatment instead of gemcitabine in patients with low hENT1 expression (NCT01586611). Locally advanced unresectable and distantly metastatic PDC are treated with multiagent systemic chemotherapy, combination therapy of folinic acid, 5-FU, irinotecan and oxaliplatin (FOLFIRINOX) that showed longer OS and PFS with FOLFIRINOX as compared to single agent gemcitabine regardless of hENT1 expression. Interestingly, in HENT1 positive patients no significant differences were noted in between gemcitabine alone or FOLFIRINOX.50 Additionally, patients positive for hENT1 expression in curative resection specimens had better prognosis compared to hENT1 negative patients.51 Thus, prognostic potential to select subgroup for surgical management.
Human equilibrate nucleoside transporter 1 (hENT1)
hCNT1 and hCNT3 are the major gemcitabine transporters in hCNT group.52 Higher hCNT3 expression on tumor blocks from patients treated with adjuvant gemcitabine based chemoradiation associated with higher survival rate at 3-years, 54.6% vs 26.1% (P=0.028).52 Additionally, in a combined analysis, patients with two favorable prognostic factors (hENT1(high)/hCNT3(high) expression) had significantly longer survival than those having one or no favorable prognostic factor.52 With limited data and lack of prospective trial, further studies are warranted to assess use of hCNT as treatment predictive biomarker.
FOLFIRINOX markers
In metastatic pancreatic cancer, FOLFIRINOX (combination of folinic acid, 5-FU, irinotecan and oxaliplatin) reported to have survival advantage as compared to gemcitabine alone.53 Predictive biomarkers are essential for FOLFIRINOX therapy to avoid unfavorable side-effect profile. Higher tissue CES2 expression was correlated with longer OS and PFS who received neoadjuvant FOLFIRINOX treatment.54
Nab-paclitaxel markers
In metastatic pancreatic cancers, combination therapy with albumin based nab-paclitaxel and gemcitabine reported significant improvement in OS and PFS compared to Gemcitabine alone.55 Glycoprotein osteonectin, also known as secreted protein acidic and rich in cysteine (SPARC), identified as a frequent site for aberrant methylation in pancreatic cancer.56 Several studies described the role of SPARC overexpression in pancreatic cancer and suggested its role in enhancement of paclitaxel delivery into the tumor as well. This was further clinically evaluated in phase I/II trial, that demonstrated high-SPARC group compared to low-SPARC group, was associated with improved median OS (17.8 months vs 8.1 months respectively).57,58 However, these findings were not validated in phase III study in metastatic pancreatic cancer treated with nab-paclitaxel and gemcitabine.59 To date, no other reliable marker is reported for nab-paclitaxel therapy in pancreatic cancer patients.
Stromal markers
PDC is quite unique because of extensive fibrosis that surrounds cancer cells, this fibrosis along with a poor blood supply has been found to limit delivery of drugs into cancer cells. A dense desmoplastic stroma surrounding the PDC can cause physical barrier to the delivery of chemotherapy60 and develop hypoxic tumor microenvironment that is immunosuppressive in nature. This is one mechanism by which pancreatic cancer is resistant to our current standard treatment. Hyaluronan is a major component of the extracellular matrix that comprises the stromal components of PDC and recently emerged as novel therapeutic target. Hyaluronidase is an enzyme that degrades this hyaluronan. The recombinant pegylated form of hyaluronidase (PEGPH 20) has been shown to improve clinical outcomes by stromal depletion leading to tumor vasculature expansion and improvement in drug delivery. This was investigated in the phase 2 HALO-202 study and reported improved median progression- free survival in the PDC patients receiving PEGPH20 with gemcitabine and nab-paclitaxel versus gemcitabine and nab-paclitaxel alone.61 In the secondary endpoint analysis of patients with high levels of hyaluronan, the median PFS increased to 9.2 months from 5.2 months in favor of combination PEGPH20 and chemotherapy. Therefore, hyaluronan-high status has been considered as a potential predictive biomarker of benefit of PEGPH20 in PDC.61 The combination of hyaluronidase with immunotherapies is currently under validation in a phase 3 trial.62
BRCA mutated tumors
About 4-10% of pancreatic cancer patient are believed to have hereditary predisposition. Patients with familial history of pancreatic cancer, BRCA mutation prevalence can be up to 17%.63 Inactivation of BRCA1 and BRCA2, PALB2 a subset of tumors may predict response to platinum-based treatments (oxaliplatin, cisplatin and carboplatin).64 BRCA mutations are a potential predictive biomarker of response to PARP inhibitors and platinumbased chemotherapies. Superior overall survival was reported in stage III-IV pancreatic cancer patients having BRCA mutations treated with platinum (22 month vs non-platinum (9 months) chemotherapies.65 PARP inhibitors are pharmacologic inhibitor of poly(ADP-ribose) polymerase enzymes, studied as potential chemotherapeutic agents in breast and ovarian cancers. Tumors with germ line mutations in DNA repair genes are susceptible to PARP inhibitors. Olaparib, an oral PARP inhibitor was given in a phase II multi centered trial in advanced cancers harboring germline BRCA1/2 mutations. A total of 298 patients received treatments, 23 of whom had pancreatic cancer. Tumor response rate was 21.7% and stable disease >8 weeks was reported in 35% cases of pancreatic cancer.66 There are several trials currently evaluating PARP inhibitor effectiveness in patients with pancreatic cancers and BRCA mutations.67
Microsatellite instability
The reported incidence rate of microsatellite instability (MSI) in PDCs has been variable ranging from <5% to 13–17% of PDC patients.68,69 While outcomes from single agent immunotherapy trials in PC was disappointing, results from the pivotal KEYNOTE study revealed that pembrolizumab demonstrated 83% objective response rate (ORR) in the six evaluable pancreatic cancer patients70 suggesting that MSI status can predict the benefit from anti-PD-1/PD-L1 blockade C. It is recommended now to test all PCs for MSI status.
PD-1/PD-L1
Pancreatic cancers are typically deficient in T-cell infiltration which may explain the poor response to single agent immunotherapeutic. 71 PD-L1 overexpression is associated with worse prognosis in a range of solid tumors, including PDAC.72 PD-L1 expression has been evaluated as a predictive biomarker for response to PD-1 inhibitors in other tumor types and was found to be correlated with better outcomes with PD-1/PD-L1 blockade;73 however, the relative lack of response to single agent checkpoint inhibitors in PC precludes assessment of PD-L1 as a predictive marker. Several novel vaccine therapies are under investigation to induce T-cell responses and overcome the immune-resistance in PC. IL17 expression in the tumor microenvironment is currently being evaluated as a biomarker for vaccine induced anti-tumor response (NCT02451982).
Prognostic markers
CA 19-9
Ca19-9 has also been studied for its prognostic value. Berardi et.al, reported high levels of Ca 19-9 independent unfavorable prognostic factor. Median overall survival if CA19-9 ≤37 U/mL vs >37 U/mL was 18.49 vs. 9.21 months respectively.74 These findings were validated in other study and reported post-operative decrease in CA 19-9 and post-op value less than 200 U/mL of CA 19-9 predicts improved survival outcomes.75
SMAD4
SMAD4 signal transformer from transforming growth factorbeta (TGF-β), involves in pancreatic cells proliferation, apoptosis and serve as tumor suppressor gene. It was reported to be inactivated in more than 50% pancreatic cancer cases.76 Loss of SMAD4 expression was correlated with distant metastasis. The prognostic role is conflicting with few reports on worse prognosis with loss of SMAD3 expression while few other studies could not confirm those findings.77,78 To clarify further, Shugang et.al reported prognostic value of SMAD4 in a recent meta-analysis with 14 studies demonstrated the worse prognosis with loss of SMAD4 expression. 79 It is hypothesized that patients with intact SMAD 4 expression associated with relatively less distant progression and therefore local treatment with radiation could improve clinical outcomes. Its value as prognostic biomarkers is currently being validated in the ongoing Radiation Therapy Oncology Trial (RTOG) 1201 trial will further evaluate response of therapy to SMAD 4 status in locally advanced pancreatic cancer patients.80
Angiogenesis markers
Stromal cells in pancreatic cancer contribute in tumor progression by releasing angiogenesis factors such as platelet-derived growth factors (PDGF), matrix metalloproteinases (MMPs) and vascular endothelial growth factors (VEGF). Higher tissue and serum levels of these angiogenic markers were correlated strongly with better survival.81
Inflammatory markers
Cancer cells activate systemic inflammation pathways which anticipate tumor progression via complicated route involving cancer cell proliferation, inducing angiogenesis, evading growth suppressors and activation of metastasis. Several inflammatory biomarkers have been proposed to predict prognosis in various cancer such as C-reactive protein, platelet to lymphocyte ratio, neutrophil to lymphocyte ratio (NLR)82 and modified Glasgow prognostic score.83 Among these, neutrophil to lymphocyte (NLR) has been shown to most valuable in predicting prognosis. NLR >5 appears to indicate shorter OS and poor prognosis in pancreatic cancer.84
Immune markers
Several immune markers were investigated immunohistochemically (IHC) markers and correlated with prognosis. IHC markers associated with a worse prognosis include FOXP3, CD68, CD163, CD204, and CD66b;85 and the markers associated with an improved prognosis include CD3, CD8, CD4, CD20.86 High CD4+/CD8+ tumor infiltrating lymphocytes following curative resection was found to be an independent favorable prognostic factor for overall survival.87 The presence of intra-tumoral tertiary lymphoid organs (lymphoid follicles), was associated with longer survival.88 However the data is conflicting as Hart et al. scored intra-tumoral lymphocytes as high or low and found no survival difference.89 This needs to be investigated in the context of a larger clinical trial. Emerging data also suggests that neoadjuvant therapy may selectively modulate immunosuppressive cells. In a retrospective analysis, significantly lower numbers of Tregs (T-regulatory cells) were identified in resected PDC specimens following neoadjuvant therapy compared with resected tumor specimens from untreated patients.90 This is likely due to adaption of the immune infiltrate, or by the immune reactivation by neoadjuvant therapy. Whether this activation could be further harnessed by concurrent or sequential administration of immunotherapies is currently under investigation.
Micro RNA’s
Beside its role as diagnostic biomarker, miRNAs, have been evaluated as potential prognostic marker. In a recent meta-analysis by Frampton et.al, demonstrated decreased OS and disease-free survival (DFS) in patients expressing high miR-21, miR-155 and miR-203; and low miR-34a levels.91 Other studies demonstrated that lower expression of miR-494 and miR-218 and high miR-221 and miR-744 levels predict poor prognosis in pancreatic cancer.92-95
SPARC
SPARC as discussed above as predictive biomarker for nabpaclitaxel, was evaluated for prognostication. SPARC overexpression in pancreatic cancer indicate poor outcome.96 Interestingly, SPARC overexpression in pancreatic cancer stromal cell demonstrated poor prognosis but its expression in tumor cells was not associated with prognosis.97 Comparable results were reported for SAPRC mRNA expression in tissues of PDC patients.98
Challenges in biomarkers studies
One of the major challenges in biomarker development is the collection of tumor tissue of adequate quality for analysis. Early diagnosis of PDC is usually performed with fine needle aspiration and therefore adequate tissue procurement is difficult to obtain. Pre-chemotherapy tumor biopsies frequently contain limited tumor cells (15%) or did not have ≥50% tumor content for high-quality tissue assessments.99 The failure in finding high-sensitive and high-specific biomarkers may also be attributed to the availability of relative fewer samples and lack of proper matching with cases and controls. It is also challenged by inadequate standard operating procedures in terms of sample collection, storage, analysis and interpretation of results. The antibody-based technologies for biomarker discovery have been challenged by lack of robustness, and relatively low throughput requiring multiplexing and complex assay validations.100 These in general also associated with high costs and requirement of large volume of samples
Conclusions
A diverse array of novel biomarkers in terms of their diagnostic, predictive and prognostic potentials are currently being studied with the hope of finding effective management for this challenging cancer. Studies on innovative molecular markers such Glypican-1 and micro-RNA’s, yielded encouraging results. The emerging immunomodulatory treatments for PDC present an opportunity for predictive biomarker development. Various combinations of these biomarkers demonstrated their potential use. However, the biomarker studies have been challenged by relatively low case numbers, absence of feasibility studies only, selection of early stage samples and non-specificity of molecular markers. Nevertheless, large studies with novel study designs are warranted to validate these biomarkers for clinical application. The optimal therapeutic management should be guided by the molecular composition of their tumor and these biomarkers play a crucial role in defining the way for precision treatment.
References
- 1.Ryan DP, Hong TS, Bardeesy N. Pancreatic Adenocarcinoma. N Engl J Med 2014;371:1039-49. [DOI] [PubMed] [Google Scholar]
- 2.Simoes PK, Olson SH, Saldia A, Kurtz RC. Epidemiology of pancreatic adenocarcinoma. Chin Clin Oncol 2017;6:24. [DOI] [PubMed] [Google Scholar]
- 3.Jemal A, Siegel R, Ward E, et al. Cancer Statistics, 2009. Cancer J Clin 2009;59:225-49. [DOI] [PubMed] [Google Scholar]
- 4.American Cancer Society. What is pancreatic cancer? Available from: https://www.cancer.org/cancer/pancreaticcancer/about/what-is-pancreatic-cancer.html [Google Scholar]
- 5.Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2014;136:E359-86. [DOI] [PubMed] [Google Scholar]
- 6.Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol (EJSO) 2007;33:266-70. [DOI] [PubMed] [Google Scholar]
- 7.Gui J-C, Yan W-L, Liu X-D. CA19-9 and CA242 as tumor markers for the diagnosis of pancreatic cancer: a metaanalysis. Clin Exp Med 2013;14:225-33. [DOI] [PubMed] [Google Scholar]
- 8.Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol 2007; 33:266-70. [DOI] [PubMed] [Google Scholar]
- 9.Poruk KE, Firpo MA, Adler DG, Mulvihill SJ. Screening for Pancreatic Cancer: Why, How, and Who? Ann Surg 2013;257:17-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ballehaninna UK, Chamberlain RS. The clinical utility of serum CA 19-9 in the diagnosis, prognosis and management of pancreatic adenocarcinoma: An evidence based appraisal. J Gastrointest Oncol 2012;3:105-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhang Y, Yang J, Li H, et al. Tumor markers CA19-9, CA242 and CEA in the diagnosis of pancreatic cancer: a meta-analysis. Int J Clin Exp Med 2015;8:11683-91. [PMC free article] [PubMed] [Google Scholar]
- 12.Bunger S, Laubert T, Roblick UJ, Habermann JK. Serum biomarkers for improved diagnostic of pancreatic cancer: a current overview. J Cancer Res Clin Oncol 2011;137:375-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Lu J, Getz G, Miska EA, et al. MicroRNA expression profiles classify human cancers. Nature 2005;435:834-8. [DOI] [PubMed] [Google Scholar]
- 14.Rosenfeld N, Aharonov R, Meiri E, et al. MicroRNAs accurately identify cancer tissue origin. Nat Biotechnol 2008;26:462-9. [DOI] [PubMed] [Google Scholar]
- 15.Hernandez YG, Lucas AL. MicroRNA in pancreatic ductal adenocarcinoma and its precursor lesions. World J Gastrointest Oncol 2016;8:18-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bloomston M, Frankel WL, Petrocca F, et al. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA 2007;297:1901-8. [DOI] [PubMed] [Google Scholar]
- 17.Caponi S, Funel N, Frampton AE, et al. The good, the bad and the ugly: a tale of miR-101, miR-21 and miR-155 in pancreatic intraductal papillary mucinous neoplasms. Ann Oncol 2013;24:734-41. [DOI] [PubMed] [Google Scholar]
- 18.Szafranska AE, Davison TS, John J, et al. MicroRNA expression alterations are linked to tumorigenesis and nonneoplastic processes in pancreatic ductal adenocarcinoma. Oncogene 2007;26:4442-52. [DOI] [PubMed] [Google Scholar]
- 19.Tang S, Bonaroti J, Unlu S, et al. Sweating the small stuff: microRNAs and genetic changes define pancreatic cancer. Pancreas 2013;42:740-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Debernardi S, Massat NJ, Radon TP, et al. Noninvasive urinary miRNA biomarkers for early detection of pancreatic adenocarcinoma. Am J Cancer Res 2015;5:3455-66. [PMC free article] [PubMed] [Google Scholar]
- 21.Yang JY, Sun YW, Liu DJ, et al. MicroRNAs in stool samples as potential screening biomarkers for pancreatic ductal adenocarcinoma cancer. Am J Cancer Res 2014;4:663-73. [PMC free article] [PubMed] [Google Scholar]
- 22.Koopmann J, Rosenzweig CN, Zhang Z, et al. Serum markers in patients with resectable pancreatic adenocarcinoma: macrophage inhibitory cytokine 1 versus CA19-9. Clin Cancer Res 2006;12:442-6. [DOI] [PubMed] [Google Scholar]
- 23.Chen YZ, Liu D, Zhao YX, et al. Diagnostic performance of serum macrophage inhibitory cytokine-1 in pancreatic cancer: a meta-analysis and meta-regression analysis. DNA Cell Biol 2014;33:370-7. [DOI] [PubMed] [Google Scholar]
- 24.Wang X, Li Y, Tian H, et al. Macrophage inhibitory cytokine 1 (MIC-1/GDF15) as a novel diagnostic serum biomarker in pancreatic ductal adenocarcinoma. BMC Cancer. 2014;14: 578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Liu D, Chang C-H, Gold DV, Goldenberg DM. Identification of PAM4 (clivatuzumab)-reactive epitope on MUC5AC: A promising biomarker and therapeutic target for pancreatic cancer. Oncotarget 2015;6:4274-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gold DV, Gaedcke J, Ghadimi BM, et al. PAM4 Immunoassay Alone and in Combination with CA19-9 for the Detection of Pancreatic Adenocarcinoma. Cancer 2013;119:522-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Han S, Jin G, Wang L, et al. The role of PAM4 in the management of pancreatic cancer: diagnosis, radioimmunodetection, and radioimmunotherapy. J Immunol Res 2014;2014:268479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Duan L, Hu XQ, Feng DY, et al. GPC-1 may serve as a predictor of perineural invasion and a prognosticator of survival in pancreatic cancer. Asian J Surg 2013;36:7-12. [DOI] [PubMed] [Google Scholar]
- 29.Yao H, Yang Z, Liu Z, et al. Glypican-3 and KRT19 are markers associating with metastasis and poor prognosis of pancreatic ductal adenocarcinoma. Cancer Biomark 2016;17:397-404. [DOI] [PubMed] [Google Scholar]
- 30.Melo SA, Luecke LB, Kahlert C, et al. Glypican1 identifies cancer exosomes and facilitates early detection of cancer. Nature 2015;523:177-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fuccio L, Hassan C, Laterza L, et al. The role of K-ras gene mutation analysis in EUS-guided FNA cytology specimens for the differential diagnosis of pancreatic solid masses: a meta-analysis of prospective studies. Gastrointest Endosc 2013;78:596-608. [DOI] [PubMed] [Google Scholar]
- 32.Kinugasa H, Nouso K, Miyahara K, et al. Detection of K-ras gene mutation by liquid biopsy in patients with pancreatic cancer. Cancer 2015;121:2271-80. [DOI] [PubMed] [Google Scholar]
- 33.Koopmann J, Fedarko NS, Jain A, et al. Evaluation of osteopontin as biomarker for pancreatic adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2004;13:487-91. [PubMed] [Google Scholar]
- 34.Poruk KE, Firpo MA, Scaife CL, et al. Serum Osteopontin andTIMP-1 as Diagnostic and Prognostic Biomarkers for Pancreatic Adenocarcinoma. Pancreas 2013;42:193-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Brand RE, Nolen BM, Zeh HJ, et al. Serum biomarker panels for the detection of pancreatic cancer. Clin Cancer Res 2011;17:805-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Vincent A, Herman J, Schulick R, et al. Pancreatic cancer. Lancet 2011;378:607-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Yachida S, Jones S, Bozic I, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 2010;467:1114-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sato N, Fukushima N, Hruban RH, Goggins M. CpG island methylation profile of pancreatic intraepithelial neoplasia. Modern Pathol 2007;21:238-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Vedeld HM, Goel A, Lind GE. Epigenetic biomarkers in gastrointestinal cancers: The current state and clinical perspectives. Semin Cancer Biol 2017. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kisiel JB, Raimondo M, Taylor WR, et al. New DNA Methylation Markers for Pancreatic Cancer: Discovery, Tissue Validation, and Pilot Testing in Pancreatic Juice. Clin Cancer Res 2015;21:4473-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Matsubayashi H, Canto M, Sato N, et al. DNA Methylation Alterations in the Pancreatic Juice of Patients with Suspected Pancreatic Disease. Cancer Res 2006;66:1208-17. [DOI] [PubMed] [Google Scholar]
- 42.Dam Henriksen S, Henning Madsen P, Christian Larsen A, et al. Cell-free DNA promoter hypermethylation in plasma as a predictive marker for survival of patients with pancreatic adenocarcinoma. Oncotarget 2017. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Parsi MA, Li A, Li CP, Goggins M. DNA Methylation Alterations In Endoscopic Retrograde Cholangio - pancreatography Brush Samples of Patients With Suspected Pancreaticobiliary Disease. Clin Gastroenterol Hepatol 2008;6:1270-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Tan AC, Jimeno A, Lin SH, et al. Characterizing DNA methylation patterns in pancreatic cancer genome. Mol Oncol 2009;3:425-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Binenbaum Y, Na’ara S, Gil Z. Gemcitabine resistance in pancreatic ductal adenocarcinoma. Drug Resist Updat 2015;23:55-68. [DOI] [PubMed] [Google Scholar]
- 46.Mini E, Nobili S, Caciagli B, et al. Cellular pharmacology of gemcitabine. Ann Oncol 2006;17:v7-12. [DOI] [PubMed] [Google Scholar]
- 47.Spratlin J, Sangha R, Glubrecht D, et al. The absence of human equilibrative nucleoside transporter 1 is associated with reduced survival in patients with gemcitabine-treated pancreas adenocarcinoma. Clin Cancer Res 2004;10:6956-61. [DOI] [PubMed] [Google Scholar]
- 48.Farrell JJ, Elsaleh H, Garcia M, et al. Human equilibrative nucleoside transporter 1 levels predict response to gemcitabine in patients with pancreatic cancer. Gastroenterology 2009;136:187-95. [DOI] [PubMed] [Google Scholar]
- 49.Bird NT, Elmasry M, Jones R, et al. Immunohistochemical hENT1 expression as a prognostic biomarker in patients with resected pancreatic ductal adenocarcinoma undergoing adjuvant gemcitabine-based chemotherapy. Br J Surg 2017;104:328-36. [DOI] [PubMed] [Google Scholar]
- 50.Orlandi A, Calegari MA, Martini M, et al. Gemcitabine versus FOLFIRINOX in patients with advanced pancreatic adenocarcinoma hENT1-positive: everything was not too bad back when everything seemed worse. Clin Transl Oncol 2016;18:988-95. [DOI] [PubMed] [Google Scholar]
- 51.Yamada R, Mizuno S, Uchida K, et al. Human Equilibrative Nucleoside Transporter 1 Expression in Endoscopic Ultrasonography-Guided Fine-Needle Aspiration Biopsy Samples Is a Strong Predictor of Clinical Response and Survival in the Patients With Pancreatic Ductal Adenocarcinoma Undergoing Gemcitabine-Based Chemoradiotherapy. Pancreas 2016;45:761-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Marechal R, Mackey JR, Lai R, et al. Human equilibrative nucleoside transporter 1 and human concentrative nucleoside transporter 3 predict survival after adjuvant gemcitabine therapy in resected pancreatic adenocarcinoma. Clin Cancer Res 2009;15:2913-9. [DOI] [PubMed] [Google Scholar]
- 53.Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011;364:1817-25. [DOI] [PubMed] [Google Scholar]
- 54.Capello M, Lee M, Wang H, et al. Carboxylesterase 2 as a Determinant of Response to Irinotecan and Neoadjuvant FOLFIRINOX Therapy in Pancreatic Ductal Adenocarcinoma. J Natl Cancer Inst 2015;107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Von Hoff DD, Ervin T, Arena FP, et al. Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus Gemcitabine. N Engl J Med 2013;369:1691-703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sato N, Fukushima N, Maehara N, et al. SPARC//osteonectin is a frequent target for aberrant methylation in pancreatic adenocarcinoma and a mediator of tumor-stromal interactions. Oncogene 2000;22:5021-30. [DOI] [PubMed] [Google Scholar]
- 57.Hoff DDV, Ramanathan R, Borad M, et al. SPARC correlation with response to gemcitabine (G) plus nab-paclitaxel (nab-P) in patients with advanced metastatic pancreatic cancer: A phase I/II study. J Clin Oncol 2009;27:4525-5. [Google Scholar]
- 58.Von Hoff DD, Ramanathan RK, Borad MJ, et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol 2011;29:4548-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Hidalgo M, Plaza C, Musteanu M, et al. SPARC Expression Did Not Predict Efficacy of nab-Paclitaxel plus Gemcitabine or Gemcitabine Alone for Metastatic Pancreatic Cancer in an Exploratory Analysis of the Phase III MPACT Trial. Clin Cancer Res 2015;21:4811-8. [DOI] [PubMed] [Google Scholar]
- 60.Provenzano P, Cuevas C, Chang AE, et al. Enzymatic Targeting of the Stroma Ablates Physical Barriers to Treatment of Pancreatic Ductal Adenocarcinoma. Cancer Cell 2012;21:418-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hingorani SR, Bullock A, Seery T, et al. 763PRandomized phase 2 study of PEGPH20 Plus nab-paclitaxel/gemcitabine (PAG) vs AG in patients (Pts) with untreated, metastatic pancreatic ductal adenocarcinoma (mPDA). Ann Oncol 2017;28. [Google Scholar]
- 62.Wong KM, Horton KJ, Coveler AL, et al. Targeting the Tumor Stroma: the Biology and Clinical Development of Pegylated Recombinant Human Hyaluronidase (PEGPH20). Curr Oncol Rep 2017;19. [DOI] [PubMed] [Google Scholar]
- 63.Couch FJ, Johnson MR, Rabe KG, et al. The Prevalence of BRCA2 Mutations in Familial Pancreatic Cancer. Cancer Epidemiol Biomark Prevent 2007;16:342-46. [DOI] [PubMed] [Google Scholar]
- 64.Waddell N, Pajic M, Patch A-M, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015;518:495-501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Golan T, Kanji ZS, Epelbaum R, et al. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. Br J Cancer 2014;111:1132-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol 2015;33:244-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Douillard JY, Ostoros G, Cobo M, et al. Gefitinib treatment in EGFR mutated caucasian NSCLC: circulating-free tumor DNA as a surrogate for determination of EGFR status. J Thorac Oncol 2014;9:1345-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Nakata B, Yashiro M, Nishioka N, et al. Very low incidence of microsatellite instability in intraductal papillary-mucinous neoplasm of the pancreas. Int J Cancer 2002;102:655-59. [DOI] [PubMed] [Google Scholar]
- 69.Yamamoto H, Perezpiteira J, Yoshida T, et al. Gastric cancers of the microsatellite mutator phenotype display characteristic genetic and clinical features. Gastroenterol 1999;116:1348-57. [DOI] [PubMed] [Google Scholar]
- 70.Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Royal RE, Levy C, Turner K, et al. Phase 2 Trial of Single Agent Ipilimumab (Anti-CTLA-4) for Locally Advanced or Metastatic Pancreatic Adenocarcinoma. J Immunother 2010;33:828-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Birnbaum DJ, Finetti P, Lopresti A, et al. Prognostic value of PDL1 expression in pancreatic cancer. Oncotarget 2016;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti-PD-1 Therapy. Clin Cancer Res 2014;20:5064-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Berardi RMA, Pellei C, Maccaroni E, et al. Prognostic Factors in Pancreatic Cancer: The Role of Perineural, Vascular and Lymphatic Invasion and of Ca19-9. J Gastrointest Dig Syst 2013;3. [Google Scholar]
- 75.Ferrone CR, Finkelstein DM, Thayer SP, et al. Perioperative CA19-9 Levels Can Predict Stage and Survival in Patients With Resectable Pancreatic Adenocarcinoma. J Clin Oncol 2006;24:2897-902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 1996;271:350-3. [DOI] [PubMed] [Google Scholar]
- 77.Oshima M, Okano K, Muraki S, et al. Immunohistochemically detected expression of 3 major genes (CDKN2A/p16, TP53, and SMAD4/DPC4) strongly predicts survival in patients with resectable pancreatic cancer. Ann Surg 2013;258:336-46. [DOI] [PubMed] [Google Scholar]
- 78.Ottenhof NA, Morsink FH, Ten Kate F, et al. Multivariate analysis of immunohistochemical evaluation of protein expression in pancreatic ductal adenocarcinoma reveals prognostic significance for persistent Smad4 expression only. Cell Oncol (Dordr) 2012;35:119-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Shugang X, Hongfa Y, Jianpeng L, et al. Prognostic Value of SMAD4 in Pancreatic Cancer: A Meta-Analysis. Transl Oncol 2016;9:1-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kim ES, Hirsh V, Mok T, et al. Gefitinib versus docetaxel in previously treated non-small-cell lung cancer (INTEREST): a randomised phase III trial. Lancet 2008;372:1809-18. [DOI] [PubMed] [Google Scholar]
- 81.Kahlert C, Fiala M, Musso G, et al. Prognostic impact of a compartment-specific angiogenic marker profile in patients with pancreatic cancer. Oncotarget 2014;5:12978-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wei Y, Jiang YZ, Qian WH. Prognostic role of NLR in urinary cancers: a meta-analysis. PLoS One 2014;9:e92079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Nakagawa K, Tanaka K, Nojiri K, et al. The modified Glasgow prognostic score as a predictor of survival after hepatectomy for colorectal liver metastases. Ann Surg Oncol 2014;21: 1711-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Xue P, Kanai M, Mori Y, et al. Neutrophil-to-lymphocyte ratio for predicting palliative chemotherapy outcomes in advanced pancreatic cancer patients. Cancer Med 2014;3:406-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Tang Y, Xu X, Guo S, et al. An Increased Abundance of Tumor-Infiltrating Regulatory T Cells Is Correlated with the Progression and Prognosis of Pancreatic Ductal Adenocarcinoma. PLoS One 2014;9:e91551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ino Y, Yamazaki-Itoh R, Shimada K, et al. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. Br J Cancer 2013;108:914-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Shibuya KC, Goel VK, Xiong W, et al. Pancreatic Ductal Adenocarcinoma Contains an Effector and Regulatory Immune Cell Infiltrate that Is Altered by Multimodal Neoadjuvant Treatment. PLoS One 2014;9:e96565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hiraoka N, Ino Y, Yamazaki-Itoh R, et al. Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer. Br J Cancer 2015;112:1782-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Hart PA, Smyrk TC, Bamlet WR, Chari ST. Impact of Intratumoral Inflammation on Survival After Pancreatic Cancer Resection. Pancreas 2016;45:123-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Fukunaga A, Miyamoto M, Cho Y, et al. CD8+ Tumor- Infiltrating Lymphocytes Together with CD4+ Tumor- Infiltrating Lymphocytes and Dendritic Cells Improve the Prognosis of Patients with Pancreatic Adenocarcinoma. Pancreas 2004;28:e26-e31. [DOI] [PubMed] [Google Scholar]
- 91.Frampton AE, Krell J, Jamieson NB, et al. microRNAs with prognostic significance in pancreatic ductal adenocarcinoma: A meta-analysis. Eur J Cancer 2015;51:1389-404. [DOI] [PubMed] [Google Scholar]
- 92.Duan F, Wang K, Dai L, et al. Prognostic significance of low microRNA-218 expression in patients with different types of cancer: Evidence from published studies. Medicine (Baltimore) 2016;95:e4773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Ma YB, Li GX, Hu JX, et al. Correlation of miR-494 expression with tumor progression and patient survival in pancreatic cancer. Genet Mol Res 2015;14:18153-9. [DOI] [PubMed] [Google Scholar]
- 94.Miyamae M, Komatsu S, Ichikawa D, et al. Plasma microRNA profiles: identification of miR-744 as a novel diagnostic and prognostic biomarker in pancreatic cancer. Br J Cancer 2015;113:1467-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Passadouro M, Pedroso de Lima MC, Faneca H. MicroRNA modulation combined with sunitinib as a novel therapeutic strategy for pancreatic cancer. Int J Nanomed 2014;9:3203-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Han W, Cao F, Chen M-B, et al. Prognostic Value of SPARC in Patients with Pancreatic Cancer: A Systematic Review and Meta-Analysis. PLoS One 2016;11:e0145803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Infante JR, Matsubayashi H, Sato N, et al. Peritumoral fibroblast SPARC expression and patient outcome with resectable pancreatic adenocarcinoma. J Clin Oncol 2007;25:319-25. [DOI] [PubMed] [Google Scholar]
- 98.Miyoshi K, Sato N, Ohuchida K, et al. SPARC mRNA expression as a prognostic marker for pancreatic adenocarcinoma patients. Anticancer Res 2010;30:867-71. [PubMed] [Google Scholar]
- 99.Aguilar-Mahecha A, Lafleur J, Pelmus M, et al. The identification of challenges in tissue collection for biomarker studies: the Q-CROC-03 neoadjuvant breast cancer translational trial experience. Modern Pathol 2017;30:1567. [DOI] [PubMed] [Google Scholar]
- 100.Yoneyama T, Ohtsuki S, Honda K, et al. Identification of IGFBP2 and IGFBP3 As Compensatory Biomarkers for CA19-9 in Early-Stage Pancreatic Cancer Using a Combination of Antibody-Based and LC-MS/MS-Based Proteomics. PLoS One 2016;11:e0161009. [DOI] [PMC free article] [PubMed] [Google Scholar]
