Review
Molecular classification as prognostic factor and guide for treatment decision of pancreatic cancer

https://doi.org/10.1016/j.bbcan.2018.02.001Get rights and content

Abstract

Clinico-pathological factors fail to consistently predict the outcome after pancreatic resection for pancreatic ductal adenocarcinoma (PDAC). PDACs show a high level of inter- and intra- tumor genetic heterogeneity. A molecular classification should help sort patients into less heterogeneous and more appropriate groups regarding the metastatic risk and the therapeutic response, with the consequences of better predicting evolution and better orienting the treatment. PDAC can be classified based on mutational subtypes and 18gene alterations. Whole-genome sequencing identified mutational signatures, mutational burden and hyper-mutated tumors with specific DNA repair defects. Their overlap/similarities allow the definition of molecular subtypes. DNA and RNA classifications can be used in prognosis assessment. They are useful in therapeutic choice for they allow the design of approaches that can predict the respective drug sensitivity of each molecular subtype. This review provides a comprehensive analysis of available molecular classifications in PDAC and how this can help guide clinical decisions.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive human cancers [1] with a 5-year survival inferior to 10% and an increasing incidence [2]. Complete surgical removal of the tumor followed by adjuvant chemotherapy is the only curative treatment. However, <20% of patients are candidate to surgery. Recently, adjuvant combination of gemcitabine and capecitabine following pancreatic resection for PDAC (ESPAC-4) showed a median overall survival of 28 months (95% CI 23·5–31·5) for patients who received gemcitabine plus capecitabine compared to 25·5 months (22·7–27·9) for gemcitabine alone [3]. Patients with borderline resectable or locally-advanced tumor are often treated with neoadjuvant chemotherapy (FOLFIRINOX) to obtain a margin-negative resection with a disease-free survival and overall survival better than upfront resected patients (DFS: 29.1 vs 13.7, P < 0.001; OS: 37.7 vs 25.1 months from diagnosis, P = 0.01) [4].

Only few chemotherapy agents (gemcitabine with or without nab-paclitaxel, FOLFIRINOX regimen combining 5-FU, leucovorin, oxaliplatin and irinotecan) and one targeted therapy (erlotinib, a tyrosine kinase inhibitor, notably for the epidermal growth factor receptor (EGFR)) show some degree of efficacy in unselected patients but with a limited clinical impact. Only 31.6%, 23% and <10% of patients respond to FOLFIRINOX, gemcitabine/nab-paclitaxel and gemcitabine, respectively [5,6]. Thus, it is crucial to both develop novel drugs and specifically identify patients most likely to benefit from one or another treatment.

Currently, the AJCC TNM staging is the only prognostic factor used in clinical practice to assess the survival of a resected PDAC and guide treatment decision. However, this clinicopathological factor fails to consistently predict the outcome after pancreatic resection. Characterization of molecular alterations may help develop techniques for early detection, identify new molecular targets and design novel targeted therapies. In breast and lung cancers, targeting drugs to tumor molecular subtypes improves treatment response and outcome [7,8]. Like these tumors, PDAC is a heterogeneous molecular disease. To solve this inter-tumor heterogeneity, high-throughput molecular studies have been applied to PDAC samples. They revealed molecular subtypes associated with different genetic alterations and prognosis, as well as gene expression signatures associated with survival [[9], [10], [11], [12]].

Section snippets

Molecular classification according to genomic alterations

Early studies had shown that the most frequently altered genes in PDACs are KRAS, SMAD4, TP53 and CDKN2A/B. Many genes such as ARID1A, GATA6, SF3B1, and TGFBR2 were later found altered by using comprehensive genomic approaches including array-comparative genomic hybridization [[13], [14], [15], [16]] and large-scale sequencing [[17], [18], [19], [20]]. Since then, many studies have been published and confirmed the high heterogeneity of genomic alterations found in PDAC. In 2012, exome

Molecular classification according to gene expression

The first profiling of PDAC was published in 2011 based on 27 microdissected surgical samples. Three different PDAC subtypes (“classical”, “quasi-mesenchymal”, and “exocrine-like”) with different clinical outcomes and therapeutic responses were defined [10]. These subtypes were validated in three additional public datasets (n = 102 samples). After surgical resection, the classical tumor subtype had a better survival than the quasi-mesenchymal subtype and showed an overexpression of genes

Molecular classification and prognosis

A major challenge is to improve the imperfect current prognostic factors to aid in therapeutic decision-making. Due to variation in survival within AJCC clinical stages and to the large genomic heterogeneity within PDAC tumors, prognostic gene expression signatures may be used to help predict outcome. Indeed, gene expression profiling remains today the most promising and successful high-throughput molecular approach for identifying new prognostic tools in early-stage cancers. Multigene

Molecular classification and treatment

The recently identified molecular PDAC classifications increased our understanding and are important to establish treatment strategies and therapeutic development. Now that genomic and transcriptomic classifications are established, we should move to in vitro and in vivo experiments to validate the best treatment strategy to use in each of the defined subtypes. What treatments could be proposed based on molecular classifications?

Gemcitabine is generally used as the first-line therapy but

Authors' contributions

DJB, EM and DB was involved in the conception and the design of study, analysis and interpretation of data, and draft of the manuscript. PF was involved in the acquisition and analysis of all data. FB was involved in the analysis and interpretation of some data. All authors read critically and approved the final manuscript.

Competing interests

The authors declare that they have no conflict of interest.

Funding

Our work was supported by Institut Paoli-Calmettes, Institut National de la Santé et de la Recherche Médicale, Institut National du Cancer, and Site de Recherche Intégrée sur le Cancer Marseille (INCa-DGOS-Inserm 6038 grant). None of them had any role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Transparency document

Transparency document

Acknowledgments

Not applicable.

References (73)

  • L. Rahib et al.

    Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States

    Cancer Res.

    (2014 Jun 1)
  • S. Gourgou-Bourgade et al.

    Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: results from the PRODIGE 4/ACCORD 11 randomized trial

    J. Clin. Oncol.

    (2013 Jan 1)
  • D.D. Von Hoff et al.

    Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine

    N. Engl. J. Med.

    (2013 Oct 31)
  • T.J. Lynch et al.

    Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib

    N. Engl. J. Med.

    (2004 May 20)
  • D.J. Slamon et al.

    Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2

    N. Engl. J. Med.

    (2001 Mar 15)
  • P. Bailey et al.

    Genomic analyses identify molecular subtypes of pancreatic cancer

    Nature

    (2016 Mar 3)
  • E.A. Collisson et al.

    Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy

    Nat. Med.

    (2011 Apr)
  • R.A. Moffitt et al.

    Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma

    Nat. Genet.

    (2015 Oct)
  • N. Waddell et al.

    Whole genomes redefine the mutational landscape of pancreatic cancer

    Nature

    (2015 Feb 26)
  • D.J. Birnbaum et al.

    Genome profiling of pancreatic adenocarcinoma

    Genes Chromosom. Cancer

    (2011 Jun)
  • D.J. Birnbaum et al.

    Endometriosis-associated ovarian carcinomas

    N. Engl. J. Med.

    (2011 Feb 3)
  • T. Harada et al.

    Interglandular cytogenetic heterogeneity detected by comparative genomic hybridization in pancreatic cancer

    Cancer Res.

    (2002 Feb 1)
  • T. Harada et al.

    Evaluation of the reliability of chromosomal imbalances detected by combined use of universal DNA amplification and comparative genomic hybridization

    Jpn. J. Cancer Res.

    (2000 Nov)
  • S.A. Hahn et al.

    DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1

    Science

    (1996 Jan 19)
  • S. Jones et al.

    Core signaling pathways in human pancreatic cancers revealed by global genomic analyses

    Science

    (2008 Sep 26)
  • K.A. Kwei et al.

    Genomic profiling identifies GATA6 as a candidate oncogene amplified in pancreatobiliary cancer

    PLoS Genet.

    (2008 May 23)
  • A.V. Biankin et al.

    Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes

    Nature

    (2012 Nov 15)
  • A.K. Witkiewicz et al.

    Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets

    Nat. Commun.

    (2015 Apr 9)
  • L.B. Alexandrov et al.

    Signatures of mutational processes in human cancer

    Nature

    (2013 Aug 22)
  • J.L. Humphris et al.

    Hypermutation in pancreatic cancer

    Gastroenterology

    (2017 Jan)
  • M. Carrasco et al.

    GATA4 and GATA6 control mouse pancreas organogenesis

    J. Clin. Invest.

    (2012 Oct)
  • S. Xuan et al.

    Pancreas-specific deletion of mouse Gata4 and Gata6 causes pancreatic agenesis

    J. Clin. Invest.

    (2012 Oct)
  • R. Janky et al.

    Prognostic relevance of molecular subtypes and master regulators in pancreatic ductal adenocarcinoma

    BMC Cancer

    (2016 Aug 12)
  • A. Maitra et al.

    Pancreatic cancer

    Annu. Rev. Pathol.

    (2008)
  • E.S. Knudsen et al.

    Stratification of pancreatic ductal adenocarcinoma: combinatorial genetic, stromal, and immunologic markers

    Clin. Cancer Res.

    (2017 Aug 1)
  • D.J. Birnbaum et al.

    Prognostic value of PDL1 expression in pancreatic cancer

    Oncotarget

    (2016 Nov 1)
  • Cited by (25)

    • The dynamic interactions between the stroma, pancreatic stellate cells and pancreatic tumor development: Novel therapeutic targets

      2019, Cytokine and Growth Factor Reviews
      Citation Excerpt :

      The classical subtype displayed better survival rates and was characterized by the upregulation of adhesion and epithelial genes and GATA6, which codes for a TF mediating pancreatic development [22]. In contrast, quasi-mesenchymal tumors correlated with the poorest survival rates of all three classes and exhibited increased levels of mesenchymal genes, including CAV1, HK2 and TWIST1 [6,7]. Finally, the exocrine-like tumors were characterized by the overexpression of genes coding for digestive enzymes and linked to exocrine pancreas function [6,7].

    View all citing articles on Scopus
    View full text