Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Sep 4.
Published in final edited form as: Expert Rev Anticancer Ther. 2017 Dec 19;18(2):131–148. doi: 10.1080/14737140.2018.1417844

The evolution into personalized therapies in pancreatic ductal adenocarcinoma: challenges and opportunities

Anteneh A Tesfaye 1,2,*, Mandana Kamgar 1,2, Asfar Azmi 1,2, Philip A Philip 1,2,3
PMCID: PMC6121777  NIHMSID: NIHMS1500871  PMID: 29254387

Abstract

Introduction:

Pancreatic ductal adenocarcinoma (PDAC) is projected to be the second leading cause of cancer related mortality in the United States in 2030, with a 5-year overall survival of less than 10% despite decades of extensive research. Pancreatic cancer is marked by the accumulation of complex molecular changes, complex tumor-stroma interaction, and an immunosuppressive tumor microenvironment. PDAC has proven to be resistant to many cytotoxic, targeted and immunologic treatment approaches.

Areas covered:

In this paper, we review the major areas of research in PDAC, with highlights on the challenges and areas of opportunity for personalized treatment approaches.

Expert commentary:

The focus of research in pancreatic cancer has moved away from developing conventional cytotoxic combinations. The marked advances in understanding the molecular biology of this disease especially in the areas of the microenvironment, metabolism, and DNA repair have opened new opportunities for developing novel treatment strategies. Improved understanding of molecular abnormalities allows the development of personalized treatment approaches.

Keywords: targeted therapy, personalized therapy, pancreas cancer, tumor microenvironment, DNA repair pathways

1. Introduction

It is estimated that 53,670 people would be diagnosed with pancreatic ductal adenocarcinoma (PDAC) in 2017, while approximately 43,090 people would die because of it. PDAC is the third leading cause of cancer related mortality and is projected to become the second leading cause of cancer dead by 2030. [1] Progress in the treatment of PDAC has been painfully slow over the past several decades. In the period 1975 – 1977, the 5-year relative survival rate of patients diagnosed with pancreas cancer in the United States was 3%, while in the period 2006–2012 the 5-year survival rate was at 9%. [2]

At this time, there is no proven method for screening and early detection of PDAC which largely explains why most patients are diagnosed at an advanced stage of the disease. [3] The relative anatomic inaccessibility of the pancreas [4] challenges the development of early detection strategies. Only 10–15% of patients present at an early stage that is amenable to a curative surgical resection. This underscores the need to develop reliable screening methods that will enable detection of the disease at its early and potentially curable stage. Even though surgical resection is the only potentially curative treatment, the median overall survival of patients who undergo radical resection is 11–23 months [5, 6] because most patients who undergo surgical resection will ultimately develop metastatic disease. This readily explains the systemic nature of the disease at presentation even in those with apparent localized stage. A significant number of patients whose disease progresses on front line therapy will not receive further treatment because of rapid clinical deterioration. [7] There is an urgent unmet need in the treatment of this disease, which is often coupled with frustration stemming from the lack of treatments with durable response.

There is also a need for reliable prognostic and predictive biomarkers to appropriately select patients for the available treatment options. The only biomarker approved for clinical use at this time is serum levels of CA19–9, which is used as a prognostic maker and to follow the activity of PDAC. However, this biomarker lacks specificity and sensitivity [8] and the level of CA19–9 doesn’t reliably predict drug response or resistance to treatment. [9].

In this article, we discuss the opportunities and challenges in furthering the development of newer and improved personalized treatment strategies for this deadly cancer.

2. Molecular Biology of Pancreatic ductal adenocarcinoma

A better understanding of the molecular biology of PDAC is the foundation for the development of personalized therapies. The molecular basis of pancreatic carcinogenesis was extensively studied using genetically engineered animal models (GEMM) of pancreatic adenocarcinoma. These models have shown that carcinogenesis is a multi-step process associated with the accumulation of signature mutations along the way. Table 1 summarizes examples of such GEMM models. Frequently encountered mutations and their affected downstream pathways in PDAC are summarized in Table 2.

2.1. Oncogene activation

Activating mutations of the KRAS oncogene are the earliest genetic changes in transformation to pancreatic cancer. [10] These mutations are seen in over 90% of diagnosed pancreatic cancers. [11] Activating KRAS mutations compromise the ability of the RAS protein to hydrolyze GTP to GDP, thus locking the protein in an active conformation. [12] The activation of KRAS in the pancreatic epithelium promotes the formation “pancreatic intraepithelial neoplasia” (PanIN), which is considered a precursor lesion for future pancreatic carcinogenesis. [13] Though these mutations are key steps in PanIN initiation, they are not sufficient for the development of pancreatic adenocarcinoma which needs additional genetic changes including loss of tumor suppressor genes. [14]

2.2. Tumor suppressor gene inactivation

Tumor suppressor genes code for key cellular proteins that prevent malignant transformation of normal cells. As such, tumor suppressor genes inhibit cellular proliferation. They also induce apoptosis when there is a critical damage that cannot be readily repaired by the DNA repair machinery. Consequently, inactivating mutations of tumor suppressor genes lead to the loss of their regulatory function in cell division, senescence and detection of DNA damage. In pancreatic cancer, frequently inactivated tumor suppressor genes include Ink4A (pl6/CDKN2A), TP53, SMAD4 (DPC4) and BRCA2. Inactivating mutations of the Ink4A (p16/CDKN2A) tumor suppressor gene are seen in up to 95% of pancreatic adenocarcinoma. [15] It is shown that KRAS mutations may induce the expression of Ink4A and the induction of cellular death. Ink4A loss therefore may be required to override the cellular senescence induced by constitutively activated KRAS. [16] The TP53 tumor-suppressor gene is mutated in 50–75% of pancreatic adenocarcinomas. [17, 18] TP53 mutations arise in later-stage PanINs that have acquired significant features of dysplasia, reflecting the possible function of TP53 in preventing malignant progression. [10] Following p53 loss, the rate of genomic aberrations increases drastically. This possibly contributes to the immense heterogeneity of pancreatic tumors and their resistance to multiple chemotherapeutic agents. [18] SMAD4 (DPC) mutations are seen in approximately 50% of all pancreatic adenocarcinomas. Similar to p53 mutation, loss of SMAD4 occurs late in the process of PanIN progression to carcinoma. SMAD4 loss is likely to contribute to tumor progression by influencing tumor-stroma interactions. [19]

2.3. DNA repair pathways

DNA damage repair (DDR) pathways are critical to the survival of the cell. To maintain the integrity of the genome, the cell relies on a significant functional overlap in the base excision repair (BER), nucleotide excision repair (NER), mismatch repair (MMR), homologous recombinant (HR) and non-homologous end joining (NHEJ) pathways. [20, 21, 22, 23, 24] The presence of seemingly redundant repair pathways ensure genomic stability in that if one pathway is lost, the cell becomes increasingly dependent on the others. [25]

There has been an increasing interest in targeting the DNA damage repair pathways because of the recognition of synthetic lethality. In a cell with redundant pathways, the inactivation of one function may be compensated for by another. Inactivating the alternative pathway(s) can be lethal to the cell, hence the phenomenon of synthetic lethality. There is an interest in the development of drugs that target the single strand DDR pathways in cancers with a defective double strand DDR pathway.

Mutations in BRCA gene account for approximately 5% of all pancreatic adenocarcinomas. However, BRCA2 gene mutation account for up to 20% of all cases of familial PDAC. [26] Somatic BRCA2 mutations are also seen in sporadic cases of pancreatic adenocarcioma. Similar in function to BRCA1, BRCA2 is involved in the repair of double stranded breaks in DNA. Loss of BRCA2 leads to the rapid accumulation of double strand DNA breaks and chromosomal aberrations. [27] Mutation in BRCA2 is typically seen late in tumorigenesis, probably reflecting the requisite inactivation of mediators of DNA-damage-response pathways, like TP53. [10] Heterozygous germline mutation of PALB2 has been shown to cause a defect in DNA replication and damage response. [28] Anecdotal experience showed inactivating mutation of PALB2 gene lead to a favorable tumor response to mitomycin C treatment, as the mutation leads to disturbed BRCA1 and BRCA2 interactions and result in defective DNA double strand break repair. [29] Ataxia telangiectasia mutated (ATM) is another gene involved in the DNA double strand break repair. [30] In a study of 166 familial pancreatic cancer probands, 2.4% (4/166) carried deleterious ATM mutations. [31] In an integrated genomic analysis of 456 cases of pancreatic adenocarcinoma, the frequency of DNA repair genes (BRCA1, BRCA2, ATM, PALB2) mutation was 17% (5% germline, and 12% somatic). [32]

2.4. Tumor Microenvironment

The role of tumor microenvironment in carcinogenesis, disease progression, metastasis, drug resistance, and immunosuppression has become increasingly recognized in the past decade. [33] It has been reported that up to 80% of the tumor mass in pancreatic adenocarcinoma may be contributed by the dense desmoplastic stroma with various cellular components. [34] The stroma consists of pancreatic stellate cells (PSCs), extracellular matrix (ECM), fibroblasts, macrophages, blood vessels, pericytes, lymphatic vessels, bone marrow-derived cells (BMDCs), stem cell-like cells and inflammatory cells. [35, 36] Studies have shown that the microenvironment-tumor cross talk is complex, dynamic and rapidly changing.

Pancreatic stellate cells, whose exact origin is unknown, play an important role in this network. In the normal pancreas, they make up 4% of all parenchymal cells. [37] These cells are activated by paracrine signaling from the surrounding cancer cells, immune or endothelial cells in the context of benign inflammation or malignancy. [38] Upon activation, these cells undergo transformation into myofibroblast-like cells expressing the cytoskeletal protein alpha smooth muscle actin (aSMA). In this conformation, the stellate cells increase their proliferation, migration and extracellular matrix protein synthesis. [39] Activated pancreatic stellate cells can induce cancer cell proliferation, while decreasing cancer cell apoptosis. They can also stimulate cancer cell migration, through increased epithelial mesenchymal transition (EMT) of cancer cells. [40] Pancreatic stellate cells have chemotactic effect on CD8+ T cells through secretion of CXCL12 [41] and on myeloid derived suppressor cells (MDSCs) through the secretion of IL 6. [42] MDSCs are known to suppress immune response to the tumor, and hence lead to immune evasion by cancer cells. Pancreatic stellate cells also produce VEGF and the hepatocyte growth factor (HGF) and activate cMET pathway hence promoting angiogenesis. [43, 44] It has been proposed that pancreatic stellate cells can also travel to metastatic site and facilitate seeding, survival and proliferation of the metastatic cancer cells at their new sites. [43] It is important to note that these findings are derived from in vitro studies and in vivo murine models and that there are species as well as donor-dependent variances between murine and human pancreatic stellate cells

2.5. Tumor infiltrating immune cells

An immunosuppressive tumor microenvironment is one of the hallmarks of pancreatic cancer. This is characterized by the presence of activated immunosuppressive and tumor supportive immune cells like myeloid derived suppressive cells (MDSCs), regulatory T cells (Tregs), tumor associated macrophages (TAM), and the presence of tumor-supportive immune cells with lack of effector immune cells.[45, 46] MDSCs can suppress effector CD8+ T-lymphocytes and promote initial tumor growth. [47] T-regs can secrete suppressive cytokines such as interleukin 10 (IL-10) and tumor growth factor β (TGFβ) and function through immune check-point inhibitors. [48] The M2 subtype of TAMs has been shown to be immunosuppressive in pancreatic carcinoma. [49] Effector cells such as CD4+, CD8+ T cells and NK cells are found to be minimal or nonfunctional in the pancreatic carcinoma stroma. This phenotype of pancreatic carcinoma tumor microenvironment is considered to be induced by the tumor cells, pancreatic stellate cells and their interaction with each other and with other cells of the stroma. [50, 51]

2.6. Angiogenesis

Angiogenesis is complex process of forming new blood vessel formation to support the growing needs of tumors and is mediated by many cell types and growth factors. [52] Angiogenesis can be independent of vascular endothelial growth factor (VEGF). Angiopoietin-Tie and Delta-Notch family are examples of other systems mediating angiogenesis in tumors. [53] Considering the pancreatic cancer molecular heterogeneity and complexity of stroma, it is not surprising that VEGF-inhibitors have not shown therapeutic benefits in pancreas cancer. [54, 55] The inhibition of angiogenesis has been speculated to increase the aggressiveness of the tumor through the hypoxia induced transcription factor pathway, in which the active HIF transcription factor binds to its promoter, and turns on the cMET gene. The cMET gene has been shown to be one of the major promoters of the epigenetically driven epithelial-mesenchymal transitions that convert closely packed epithelial cancer cells into chemotherapy resistant mesenchymal type cells. [56, 57, 58] Inhibiting cMET was shown to abrogate oncogenic signaling in vitro and impair tumor growth in vivo. [59, 60]

3. Challenges in the treatment of pancreatic ductal adenocarcinoma

3.1. Disease biology

Finding an effective systemic treatment has been extremely challenging largely because of de novo or rapidly acquired resistance to cytotoxic therapy. The lack of a driver genetic mutation that can be inhibited and the molecular heterogeneity of the disease make it difficult to effectively target molecular pathways. [61, 62] Abundant and complex tumor stroma functions more than a physical barrier to therapeutic interventions. This tumor microenvironment has been characterized to be immunosuppressive thereby enabling the cancer to evade the immune system and be unresponsive to immunotherapy. [63, 64]

As the KRAS mutation plays a key role in the oncogenesis in more than 90% of pancreatic cancer cases, it is a rational target for drug development. Unfortunately, this has met with little success because of RAS’s molecular structure. [65] As there is no therapeutic intervention that can target the activated KRAS mutation, blocking the pathways downstream to KRAS has been attempted, but with little success, if any. [66]

3.2. Tissue acquisition for research

Obtaining tissue from pancreatic masses for diagnostic and research purposes may be challenging due to the location of the organ. Percutaneous image guided needle biopsy of pancreas masses has been safely used in humans as early as 1975. [67] Core needle biopsy was introduced in the late 1980s. [68] Both methods have comparable accuracy in establishing the diagnosis. [69, 70, 71] Endoscopic ultrasound (EUS) has changed the way we obtain diagnostic tissue specimens. With increasing focus on personalized therapy and search for predictive biomarkers there has been an increased need for bigger sized core biopsies in the last few years [72] and modified needles are being evaluated for acquisition of such specimens. [73]

Circulating tumor cells (CTCs) in the peripheral blood is being studied as means of noninvasive tissue acquisition. However, isolation of cancer cells from the blood may be challenging. [74] Circulating cells are likely to be detected with higher disease burden, although there are reports of CTC detection in early disease stage. [75] The presence of CTC in the blood has generally been linked to worse prognosis. [75, 76, 77] The detection techniques and procedures are rapidly developing. The sensitivity of these tests are currently relatively low, and their place in the clinic or research remains to be established. [78] It is believed that with further development, CTCs may provide valuable diagnostic, prognostic or predictive information in the treatment of pancreatic adenocarcinoma. Furthermore, mutational analysis of CTCs and serial monitoring of CTCs and CTC burden might serve as means of monitoring treatment response/emergence of resistance. [75] Circulating cell free DNA (cfDNA) is another resource under investigation. Its utility as a biomarker for pancreatic cancer is similarly being determined. [79]

3.3. Genomic profiling

The use of targeted Next Generation Sequencing (NGS) has been proposed to improve the diagnostic value of fine needle aspiration derived material by detecting key somatic and germline genetic alterations that are unique to pancreatic cancer. [80, 81] The findings of germline mutations may also have implication for unaffected family members. It can also help identify patients at risk of developing pancreatic adenocarcinoma and help define future therapeutic options. [82] NGS in exceptional responders may identify subset of patients that might benefit from specific treatments. Furthermore, NGS may identify potential targets that may help tailor treatments to a specific mutation. However, next generation sequencing has not been prospectively validated for clinical use in patients with pancreatic cancer and remains experimental at this point. There is currently no available drug that can directly target the four most common mutations, KRAS, CDKN2A, TP53 and SMAD4 in pancreatic cancer. All the mutations that can be therapeutically exploited in pancreatic cancer have an exceedingly low prevalence [83] and as such are not interrogated in clinical practice unless enrolled in a clinical trial that is enriched for a particular molecular abnormality. [84] NGS results can also be used to risk stratify patients. In a recent study, a composite of 25-gene signature from NGS in pancreas cancer patients has been shown to identify patients with short and long-term survival benefits after surgical resection of the primary tumor. [85]

3.4. Preclinical models

Preclinical models of pancreatic cancer are important for the investigation of carcinogenesis, progression and therapy. These include in vitro models (pancreatic cancer cell lines, spheroids and organoid) and in vivo models including cancer cellular models subcutaneously or orthotopically transplanted into mice, genetically engineered mouse models (GEMMs) and patient-derived xenografts (PDXs). Among them, the in vivo models especially PDXs are more important for testing the efficacy of novel therapeutics because of assumed similarity with the pathology of the disease in humans.

3.4.1. In vitro Models

Pancreatic cancer cells lines have been widely used in basic research for pancreatic cancer biology and therapy. So far, more than 25 pancreatic cancer cell lines have been established for in vitro studies (http://www.creative-bioarray.com/Products/Pancreatic-Tumor-Cells-list-106.htm) [86]. Among them, MiaPaCa-2, PANC-1, AsPC-1, HPAC and BxPC-3 cell lines have been most frequently used in the studies of signaling transduction pathway, biomarkers, novel drug screen, and targeting drug resistance in pancreatic cancer. Pancreatic cancer cell lines are cultured in 2-D monolayer pattern and provide a relatively easy tool to conduct any in vitro experiments. However, cells in 2-D structure behave differently than cells in 3-D structure which better mimic cells grown in physiological condition in vivo. [87]

To overcome the limitation of 2-D cell culture, scientists have created 3-D spheroid culture models. [88] This model was originally developed and used for testing novel agents and their combinations. Building further on these the field has recently focused on the development of organoids using pancreatic cancer cells grown in a semi-solid environment. [89, 90] Studies found that organoids maintained same genetic and phenotypic features of source cells. [89] Moreover, organoids have the similar structure and function compared to the in vivo counterparts, suggesting that organoids have great potential for preclinical investigations. [90] The development of pancreatic organoid preclinical models to study the molecular basis of pancreatic cancer and discover targeted therapies supports the enthusiasm for improved drug development.

3.4.2. In -vivo Models

Before the emergence of GEMMs and PDXs, human pancreatic cancer cell lines have been subcutaneously or orthotopically transplanted into mice for preclinical research. However, these models lack the interaction between human cancerous cells and human stromal cells. Also, the cancer cells grow in the environment without human immunity. Therefore, the models do not physiologically mimic the human pancreatic cancer. [91] Nevertheless, with the advantage of gene engineering and xenograft techniques, GEMMs and PDXs mouse models (Table 1), which are more suitable for preclinical research, have been developed.

As discussed earlier, multiple gene mutations contribute to the development and progression of PDAC. Based on this information, GEMMs were developed that cover the most commonly found mutations in pancreatic cancer. KRAS mutation is the most frequent genetic alteration in pancreatic cancer. Therefore, transgenic mice have been designed to carry PDX-1-Cre/Lox-Stop-Lox (LSL)-KRAS. [13] PDX-1 (Pancreatic duodenal homeobox 1, also known as insulin promoter factor 1) is a critical transcription factors in the developmental program of the pancreas. The Cre/Lox is site-specific recombination system. PDX-l-Cre/Lox-Stop-Lox (LSL)-KRAS ensures KRAS mutation and activation in pancreatic cells in the mouse model. [13] The pancreas of this transgenic mouse develops ductal lesions identical to all three stages of human PanINs. [13] However, KRAS mutation is not sufficient to induce progression to the invasive pancreatic adenocarcinoma. Therefore, more GEMMs have been designed to have activated KRAS with one or more other gene mutations such as p53, Ink4a, Smad4 and TGF-β mutations commonly found in pancreatic cancer. [92, 93, 94, 95, 96, 97, 98] Recent study shows that LSL-KrasG12D; LSL-Trp53R172H/+; Ink4flox/+; Ptf1/p48-Cre mouse model with multiple mutations is a more applicable model for preclinical investigation of locally invasive and metastatic pancreatic cancer. [99] The activated KRAS together with other gene mutations in pancreatic cells promotes PanINs to progress to invasive pancreatic cancers, making the models more relative to clinical pancreatic cancer. However, the GEMMs harbor same mutations in all pancreatic cells and they are homogeneous. Nonetheless, pancreatic cancer is highly heterogeneous and significantly affected by the tumor microenvironment. Thus, patient-derived xenografts (PDXs) have been developed to overcome the disadvantages of GEMMs,

Pancreatic PDXs are increasingly used in preclinical investigation of efficacy of novel therapeutics. So far, more than 22 PDXs have been developed (Mouse Tumor Biology Database). [100] PDXs are created by the transplantation of a human tumor mass into an immunocompromised mice such as athymic nude mice. PDXs can maintain the original genetic profiles of original cancers from patients even after passaging in vitro and in vivo. Cancer cells in PDX models grow in their original human stroma. These advantages allow testing for efficacy of novel therapeutics in an in vivo setting which mimics the original tumor and environment. The in vivo experiments using these models provide valuable information that can help to design more effective human clinical trials to further test the novel therapeutics for treatment of pancreatic cancer. Recently, a gemcitabine-resistant pancreatic cancer patient-derived orthotopic xenograft (PDOX) has been used to demonstrate that MEK inhibitors more significantly inhibit pancreatic cancer growth compared to other therapies. [101] A limitation of PDX models is that PDXs are created in immunocompromised mice, which are not suitable for testing immunotherapy. Recently, humanized NOD SCID gamma mouse models engrafted with human hematopoietic stem cells have been developed and used for investigation of immunotherapy. [102] We believe that by development of conditional control of human immune system engrafted in the mice, the humanized NOD SCID mouse models could be utilized for creating pancreatic cancer PDXs models which could be used for preclinical investigation of immunotherapy.

3.5. Prioritization of molecular targets

It is a disappointment to many that we still don’t have a cutting-edge target for effective therapy despite decades of research in pancreatic cancer. The very modest advance in patient outcome from the systemic therapy of pancreatic cancer during the era of targeted therapies was still with conventional cytotoxic drugs. Success of personalized therapy in other malignancies was a result of discovering “actionable” molecular targets that can be used in patient selection. This illusive actionable treatment target may be discovered with the use of newer molecular technologies, and advanced computing power that can sift through the extensive databases generated from extensive work in genomics, proteomics and metabolomics. [103, 104]

There is a need for more powerful computing power, and even artificial intelligence, to sift through the plethora of information being generated constantly. Although most models still lack clinical validity, different computational models are being used to identify and validate anticancer drug targets on a genome-wide scale. [105] Before large human studies can be performed on the proper selection of targets and drugs that can affect them, a thorough preclinical evaluation along with clinical validation is needed. Establishing efficacy of new targeted therapies in multiple models would increase the level of confidence for possible eventual clinical benefit. With better design of early stage clinical trials and focus on meaningful clinical benefit, rather than a mere statistical significance of findings, it may be possible to avoid conducting a costly phase III study that winds up being un-necessary. [106] The use of predictive algorithms to incorporate existing and future knowledge to best match therapeutic intervention to the molecular signatures of the tumor needs to be implemented to bring about a meaningful breakthrough treatments for this deadly disease. [107]

3.6. Clinical trial design

The design of trials based on rare molecular abnormalities remains challenging due to the difficulty of patient accrual. Such challenges have led to development of innovative designs of clinical trials. Umbrella study typically enrolls patients with a single tumor type. Patients are directed towards different therapeutic arms of the study based on the molecular characteristic of their tumor. Basket study, on the other hand, enrolls patients with different tumor types. Tumors sharing common biomarkers are stratified to receive certain drugs regardless of their tissue of origin. In other words, in the context of basket trial what matters is the tumor’s molecular signature, rather than tissue of origin. Master or tent protocol uses a single multiplex diagnostic assay to assign participants to different arms of a trial within the same trial or network of trials. In this approach after the initial molecular profiling patients and clinicians are presented with different options for standard therapy versus all possible options of available clinical trials. This type of study would ideally need to be conducted in collaboration of a network of organizations. [84] Adaptive trials use information obtained while trial is ongoing (interim analyses) to modify the course of trial. Depending on the emerging results, a study can be extended or stopped. If no benefit is seen within a specific arm of study, that arm can be dropped. In addition, it allows for change of randomization proportion, and rate of accrual. [84, 107]

Considering, the failure of many of our traditional approach to clinical trials in finding an effective therapy in this disease and the complexity of the above measures, a more comprehensive coordination of the available trials is needed to guide patients to the best available clinical trials.

4. Opportunities for personalized therapy for the near future

Although there has been a great interest in personalizing pancreatic cancer therapy, this has been a very elusive goal to achieve. There is also a significant amount of interpatient variability seen in treatment outcomes of patients receiving the current standard of care in the clinic that is also in need of predictive biomarkers. Consequently, clinical trials continue to be largely based on empiric drug combinations and are minimally dependent on predictive biomarkers. Recent work identified several potential areas of developing targeted therapy strategies that may be coupled with predictive biomarkers. Table 3 illustrates select clinical trials exploring the therapeutic utility of such biomarkers.

4.1. Targeting DNA Repair: BRCA and beyond

DNA damage may re ult from ba se modifications, double-strand DNA breaks, single strand DNA breaks, intrastrand and interstrand DNA cross links. DNA is repaired by distinct pathways to maintain genetic stability and integrity. Two main double strand break repair pathways are prevalent: the more efficient homologous recombination and the error prone non-homologous end joining pathway. [108] The BRCA genes encode proteins essential for the homologous recombination pathway of double stand DNA break repair. They also play a key role as a key cell cycle checkpoint in response to DNA damage. [27, 108] BRCA1/2-deficient cells that lack homologous recombination activity accumulate inefficiently repaired double-strand DNA breaks, resulting in genomic instability and an increased predisposition to malignant transformation and progression. [109] Although the risk of pancreatic cancer in carriers of BRCA1 gene mutation is not well quantified, it is estimated to be a 2 to 3 fold increased over the general population. [110, 111] BRCA1 gene mutation was not seen in a cohort of familial pancreatic cancer patients, raising the question of its significance in familial pancreatic cancer syndromes. On the contrary, BRCA2 gene mutation has been seen in about 6% of patients with familial pancreatic cancer syndromes. [112] The risk of pancreatic cancer in BRCA2 gene mutation carriers is reported to be 3 to 6 fold increased over the general population. [113, 114] Patients with impaired DNA repair pathways have been shown to respond better than those with intact DNA repair pathways to therapeutic interventions that induce DNA damage, such as platinum-based chemotherapy drugs and radiation therapy. [115, 116] In a retrospective study of 58 BRCA-associated advanced pancreatic cancer patients, there was a significant overall survival advantage in patients treated with platinum when compared to those treated with non-platinum chemotherapy agents (22 vs 9 months; P: 0.039). [117] Small sized studies in patients with BRCA1/2 mutation related pancreatic cancer showed improved response rates and survival outcome after treatment with platinum-based chemotherapy. [116, 118, 119]

Single strand breaks are by far the most common DNA damages in a cell. If single strand damages are not repaired efficiently, they evolve into a double strand break. [108, 120] Base excision repair (BER) pathway is an important repair mechanism for single strand DNA breaks. Poly-ADP ribose polymerase (PARP-1) is a critical component of the BER pathway and plays a key role in sensing and binding to single strand DNA damages and results in the activation of catalytic proteins including topoisomerases, histones, and PARP-1 itself for the repair of the damage. [108, 121, 122] When a cell that has a defective homologous recombination pathway loses ability to repair single strand DNA damages, the combination could be lethal to the cell hence the phenomenon of synthetic lethality. To exploit this vulnerability, different studies are looking into using PARP inhibitors in patients with BRCA gene mutation related pancreatic cancers.

The PARP Inhibitor rucaparib was studied in a phase II single arm treatment of 19 patients with known germline or somatic BRCA mutation and relapsed pancreatic cancer, with more than 1 prior systemic treatment. There were 2 partial responses and 1 complete response of the 19 patients, response rate of 15%. [123] There are ongoing clinical trials evaluating the benefit of PARP inhibitors in treatment of pancreas cancer in patients with BRCA gene mutations (NCT01078662, NCT01989546, NCT02042378, NCT01489865). Ataxia telangiectasia mutated (ATM) deficiency is associated with responsiveness to the PARP inhibitor olaparib in gastric cancer. [123]

With increased understanding of the disease and the involved genetic alterations, the PARP inhibitors need to be evaluated further in patients with aberrant genes involved in the DNA repair pathway, beyond the BRCA genes.

4.2. Hyaluronan in the tumor stroma

Hyaluronic acid (HA) is a highly abundant mucopolysaccharide in the body synthesized by HA synthases (HAS1–3) and degraded by hyaluronidases (HYAL1–4, HYALP1 and PH20). [124] There is high level of hyaluronic acid in up to 40% of patients with pancreas cancer, due to tumor-stroma interaction, and resulting upregulation of HAS. [125, 126] The accumulation of hyaluronic acid in the stroma leads to increased interstitial fluid pressure due to its water binding capacity leading to collapse of intra-tumoral vasculature, which is considered as a barrier to the delivery of drugs. [127, 128] Hyaluronic acid has also been shown to bind surface receptors RHAMM and CD44, possibly promoting tumor proliferation, adhesion, migration, invasion and immune resistance. [129, 130, 131] Hence, hyaluronic acid has been an interesting therapeutic target in pancreas cancer. Contrary to inhibiting synthesis, blocking receptor signaling or depleting the stromal content of hyaluronic acid is the most studied strategy so far.

Recombinant human hyaluronidase (PH20) has a very short half-life in vivo, which is prolonged to weeks by conjugating it with polyethylene glycol (PEGPH20). [13 1] In preclinical models, systemic administration of PEGPH20 lead to intratumoral depletion of hyaluronic acid, reduction of interstitial fluid pressure, and improved penetration of chemotherapeutic drugs to the tumor cells. [127, 132, 133] Furthermore, PEGPH20 was shown beneficial effect in improving delivery of monoclonal antibodies into the tumor microenvironment in vitro. [134] A recently completed phase II study investigated whether the addition of PEGPH20 to standard gemcitabine and nab-paclitaxel improved outcomes in untreated metastatic pancreatic adenocarcinoma. Though the final study outcomes are yet to be published, interim analysis of 146 patients showed improved median PFS in patients with HA-high tumors treated with PEGPH20 (9.2 vs 4.3 months; P =.05). [135] PEGPH20 is currently being tested in a Phase III clinical trial in combination with gemcitabine and nab-paclitaxel that is the current standard of care. [NCT02715804]

4.3. DNA mismatch repair protein deficiency

Immunotherapy is a rapidly advancing field in cancer treatment. Among the recent immunotherapy modalities, immune check point inhibition has shown significant success in several solid malignancies, but with no meaningful benefit in pancreatic cancer. Checkpoint inhibition has been shown to be promising in mismatch repair deficient colorectal and other cancers. [136, 137] In a more recent publication, Dung et al showed that solid tumors with mismatch repair deficiency are sensitive to immune checkpoint blockade with pembrolizumab. Objective radiographic response was seen in 53% of patients with mismatch repair deficiency and complete response was seen in 21%. The responses were durable and the median PFS and OS was not reached at the time of publication. [138] Pembrolizumab has recently been FDA approved for solid tumor with mismatch repair deficiency, irrespective of tissue of origin. Although small studies have reported a higher incidence rate of mismatch repair enzyme deficiency in pancreatic cancer ranging 11 to 22%, [139, 140] After testing 12019 cancers, the prevalence of microsatellite instability was seen to be around 5% in many solid tumors, while in pancreatic adenocarcinoma it was about 2%. [138] There is an ongoing study (COMPASS study) to improve molecular and genetic characterization of pancreatic cancer and help identify who might benefit from immunotherapy. [NCT02750657]

4.4. Metabolic pathways

Pancreatic adenocarcinoma cells have developed metabolic flexibility to survive given the unique challenges of their microenvironment including hypoxia and nutrient deprivation, through metabolic reprogramming with in the cancer cell and the tumor microenvironment. [141, 142]

Aerobic glycolysis switch is one of the well described adaptation in pancreatic cancer cells, regulated by PI3K, hypoxia inducible factor (HIF), p53, MYC, and AMP-activated protein kinase and liver kinase B1 pathways. [141, 143, 144] CPI-613, a highly selective blocker of pyruvate dehydrogenase (PDH) and α-ketoglutarate dehydrogenase (KGDH), was found to be safe in combination with chemotherapy (FOLFIRINOX), and phase III trial is being developed. [145]

Glutamine is used by pancreatic cancer cells for lipid biosynthesis, as a nitrogen donor for amino acids and nucleotide biosynthesis, as a carbonic substrate for the tricyclic acid (TCA) cycle, and even as fuel for energy production. Other metabolic adaptations seen in pancreatic cancer cells include activation of hexosamine biosynthetic pathway, lipid metabolism, autophagy and pinocytosis. [142] Targeting tumor specific metabolic pathways has gained attention for development of innovative therapies.

4.5. Mesothelin

Mesothelin is a surface glycoprotein, normally expressed by mesothelial cells, but also overexpressed by several solid tumors including pancreatic adenocarcinoma. [146, 147] Although its exact role in normal cells is not well established, mesothelin has been implicated in tumor adhesion and metastasis through its ability to bind to MUC16 (CA125). [148, 149] Mesothelin is implicated in tumor progression and resistance to chemotherapy. [150, 151, 152, 153, 154] Given the overexpression of mesothelin in several human malignancies relative to normal cells, it has been regarded a potential target for antibody based therapies. SS1P (CAT-5001) is a recombinant immunotoxin consisting of anti-mesothelin antibody Fv fragment linked to a truncated Pseudomonas exotoxin that mediates cell killing. [155, 156] Two separate phase I studies have established the safety of SS1P. [157, 158] MORAb-009 is a high affinity monoclonal antibody against mesothelin. This antibody is cytotoxic to mesothelin expressing cell lines via antibody dependent cell toxicity. Clinical safety of MORAb-009 is established in two phase I clinical trials. [159, 160] Further clinical studies would be needed to define the clinical utilities of these mesothelin targeted novel therapies. Mesothelin has also been explored in cancer vaccine development. The vaccine CRS-207 is a live-attenuated Listeria monocytogenes strain expressing mesothelin. Although earlier phase studies showed that it induces mesothelin specific T-cell responses [161], but a phase II vaccine study were negative. [162]

4.6. HER/ErbB family

The human epidermal growth factor (HER)/Erythroblast leukemia viral oncogene homologue (ErbB) family of proteins consists of: HER1 (ErbB1/EGFR), HER2 (ErbB2), HER3 (ErbB3) and HER4 (ErbB4). All of the HER-family receptors are transmembrane tyrosine kinases that activate and regulate diverse processes, including cell survival, proliferation, differentiation and migration. [163, 164] Multiple studies so far have tried to target these pathways with little success in PDAC. The empiric addition of erlotinib to gemcitabine was shown to have a marginal improvement in survival outcomes over single agent gemcitabine that led to the approval of the regimen by FDA. [165] However, this regimen is not considered to offer any clinically meaningful benefit, especially after the introduction of modern chemotherapy regimens, considering its cost and added toxicity. The addition of the either antiEGFR antibody cetuximab or the antiHEr2 antibody trastuzumab to gemcitabine did not improve outcomes over single agent gemcitabine. [166, 167, 168] The addition of the tyrosine kinase Her2 inhibitor lapatinib to gemcitabine also did not add any benefit compared to gemcitabine alone and the trial was stopped early for futility. [169]

Combining antiEGFR and AntiHEr2 antibodies have shown synergism in preclinical studies pancreatic cancer models. [170]. But phase II clinical trials of combined trastuzumab, erlotinib and gemcitabine did not show survival benefit over single agent gemcitabine. [171] A phase II study of pertuzumab and erlotinib for refractory adenocarcinoma of the pancreas was also terminated early due to extreme toxicity in combination therapy. [NCT01108458]

The phase I/II study of combination trastuzumab and cetuximab in patients with advanced pancreatic cancer after failure of gemcitabine based chemotherapy showed stable disease in 9 of 39 patients. Treatment was discontinued due to toxicity (skin rash). OS and PFS were positively correlated with skin toxicity severity. [172] Afatinib is an irreversible EGFR, HER2 and HER4 inhibitor. The combination of afatinib and gemcitabine versus gemcitabine alone is being studied in a phase II clinical trial for untreated metastatic pancreatic cancer. [NCT01728818] The combination of afatinib plus capecitabine is being evaluated study in patients with advanced refractory pancreatic or biliary cancer. [NCT02451553]

A common shortcoming in all the above targeted therapies is the lack of predictive marker of response. In the absence of a predictive biomarker to select these treatments, it might be difficult to see benefits from treatment selected on pure empiricism.

4.7. Adoptive cell therapy

Immunogenicity of Her-2 derived peptides has been demonstrated in In vitro studies. [173, 174] Chimeric antigen receptor-modified T cell (CART) therapy represents a novel approach to treatment of many malignancies. There is an ongoing phase I/II clinical trial is studying effectiveness of CAR T cells targeting HER-2 antigen in patients with different HER-2 positive solid cancers including pancreas cancer. [NCT02713984] Feasibility targeting the EGFR molecule for the development of CAR T cells has been previously shown in vitro and in animals. [175] There is also an ongoing Phase I/II study of genetically engineered lymphocyte therapy in treating patients with EGFR positive advanced/unrespectable solid tumors including pancreatic cancer tumors. [NCT01869166] Regardless of their outcomes, these studies represent a novel treatment approach and there is an important lesson that can be gained out of them.

4.8. Role of Natural Compounds

It is estimated that approximately 60% of anti-cancer drugs are either purely natural or are inspired by natural products. [176]. While some natural products are accepted as alternative medicine in western countries, these products are commonly combined with standard chemotherapy in countries like China. It is estimated that around 90% of patients with PDAC in China at some point receive Chinese herbal medicine as an adjunct to their cancer treatment. Therapeutic role of multiple natural compounds is currently under investigation in vitro, in vivo and in the context of clinical trials. [177, 178]. There are several natural compounds under investigation for their potential role against PDAC pre-clinically and in Phase I/II clinical studies. BMS-247550, a novel epothilone derivative, is being developed as an anticancer agent for the treatment of patients with malignant tumors especially pancreatic cancer. [179] BMS-247550 is a semisynthetic analogue of the natural product epothilone B and its mode of action is analogous to that of paclitaxel (i.e., microtubule stabilization). BMS-247 550 was tested in a Phase II study [NCT00016965]. Although the study was completed the outcome of this trial is still awaited. Another natural product triptolide, a diterpenoid, that was shown to be effective against pancreatic cancer cells in vitro as well as in vivo. [180] A water-soluble analog of triptolide, named minnelide had similar potency in preclinical models of pancreatic cancer. Minnelide was shown to reduce the growth and spread of tumors and improving survival in xenograft models of human PDAC. [181] Based on these data, minnelide is currently under Phase II evaluation for pancreatic cancer [NCT03117920].

4.9. Biomarkers of drug effect and toxicity

4.9.1. UGT1A1

Irinotecan is currently being used for treatment of metastatic pancreatic cancer, both in the front-line (as part of the FOLFIRONOX regimen) and in the second line (in the form of Nano liposomal irinotecan in combination with 5FU) settings. [182, 183] A subset of patients receiving irinotecan experience severe hematologic and/or gastrointestinal toxicities. These are patients who may be deficient in the UDP-glucuronosyltransferase 1A1 (UGT1A1) enzyme that catalyzes the glucuronidation of the active irinotecan metabolite, SN-38. Different studies have shown that UGT1A1 gene polymorphism, resulting in the deficient activity of the UDP-lucuronosyltransferase 1A1 enzyme, is a predictor of severe irinotecan toxicity. [184, 185] Different levels of dose reduction have been recommended by different authors. [123] There are different ongoing studies that aim to determine the safety profile of irinotecan in patients with advanced gastrointestinal malignancies based on their UGT1A1 gene status. [NCT01963182, NCT02256800, NCT01643499] As most patients with advanced pancreatic cancer have limited physiologic reserve, avoiding a drug with potentially severe toxicity will be of paramount importance.

4.9.2. Gemcitabine transport molecule: hENT1

Gemcitabine (2, 2 -difluorodeoxycytidine) remains a cornerstone in the systemic treatment of patients with PDAC, especially in the adjuvant setting. It is a nucleoside analogue that inhibits ribonucleotide reductase and DNA polymerase. [186] Gemcitabine is a polar molecule and therefore does not readily diffuse across the cell membrane. The human equilibrative nucleoside transporter (hENT) 1 is the main transporter protein for cellular uptake of gemcitabine in pancreatic cancer cells. [187, 188]

Retrospective studies demonstrated that patients with pancreatic cancers that have increased expression of hENT1 had higher response and survival outcomes when treated with gemcitabine relative to those with low hENT1 expression. These included treatments in the adjuvant setting. [189, 190, 191] However, there is discordance in the predictive value of increased hENT1 expression. Some prospective studies have shown increased expression of hENT1, both by immunohistochemistry and gene sequencing, in the patients treated with gemcitabine, was predictive of increased overall and disease-free survival, in patients treated with gemcitabine. [192, 193, 194] On the other hand, the prospective CONKO-001 trial that investigated the role of adjuvant gemcitabine versus observation in pancreatic cancer, showed that hENT1 was not predictive of response to gemcitabine [195]. The difference in the types of the antibody used in the CONKO-001 study was given as a possible explanation for this discordance. [196] There is no real consensus on the cut off level for low and high expression levels. The transport and metabolism of the drug is very likely to be more complex than the use of single transporter. Additionally, the magnitude of therapeutic benefit from gemcitabine is not sufficient to maintain excitement around this biomarker. Hence the hENT1 was not accepted as a predictive biomarker worthy of further development for the standard clinical practice.

4.9.3. SMAD4

SMAD4/DPC4 (deleted in pancreatic carcinoma, locus 4) encodes a key intracellular messenger in the transforming growth factor (TGF) signaling cascade. As TGF is a potent inhibitor of growth of epithelial cells, loss of SMAD4 releases this inhibition and accelerate KRAS mediated pancreatic neoplastic transformation. [197] SMAD4 is also considered to have anti-angiogenesis activity, decreasing expression of VEGF and increasing expression of thrombosponding-1. [198] In a retrospective study of 76 patients with pancreatic cancer, 30% were found to have died with locoregional disease progression, while 70% with widespread metastasis. Only 22% of patients with locally advanced carcinomas at autopsy showed loss of SMAD4, compared to 78% of patients with metastatic disease with loss of SMAD4. [199] Unlike pancreatic cancers with intact SMAD4 who have more local pattern of disease progression, tumor with SMAD4 loss commonly have distant disease progression. [200] The combination of RUNX3 and SMAD4 status was proposed as a better predictor or disease progression compared to SMAD4 alone. Loss of SMAD4/DPC4 by immunohistochemistry correlated with metastatic pattern of disease progression. On the other hand, SMAD4/DPC4 intact tumors with high level of RUNX3 are seen to have high metastatic potential. [201] The roles of SMAD4 or SMAD4/RUNX3 as a biomarker in the care of patients with PDAC should be further tested in prospective studies before it can be widely applied in the clinic, especially in situations where radiation therapy is being considered.

4.9.4. MicroRNAs

MicroRNAs (miRNAs) are small non-coding RNAs consisting of 18–24 nucleotides which regulate the stability and translation of their target mRNAs by binding to their 30UTR region. [202, 203] They are part of the cellular regulatory network that control cell functions such as cell growth, proliferation, differentiation, development and apoptosis. [203] Studies have shown that several miRNAs are aberrantly expressed in pancreatic tumor, contrary to their adjacent normal tissue. [204, 205] It has been shown that miRNAs could elicit oncogenic or tumor-suppressive functions by directly targeting oncogenes or tumor suppressor genes. [206] The inhibition of oncogenic miRNAs and restoration of expression of tumor suppressor miRNAs are therefore potential therapeutic strategies. MicroRNAs are also linked to resistance and sensitivity to chemotherapeutic agents and targeted therapies. [207, 208] Most studies of miRNAs in pancreatic cancer are still in the pre-clinical phase. The better identification, validation and selective delivery of miRNAs could help in their clinical application.

5. Prospects of Molecular Classification of Pancreatic Adenocarcinoma

Recent biotechnological advances give us the ability to perform comprehensive genomic, transcriptomic, proteomic and metabolomic studies at a faster rate and for a much cheaper cost. However, these tests are not ready for wide clinical application. A large amount of data is generated from these new tests, which with the application of advanced bioinformatics can be used to better understand the disease process and possibly be used to design better treatment strategies.

Hoadley et al used five genome-wide platforms and one proteomic platform on 3257 specimens from 12 cancer types to examine how much is shared across tissues. [209]This study revealed that tissues can be classified into common subtypes regardless of their tissue of origin and stage. As an example, subset of bladder cancer, lung squamous and head and neck could be classified as a subtype with TP53 alterations, TP63 amplifications and high expression of immune and proliferation pathway. This innovative system of reclassifying tumors could guide novel treatment decisions and open the door for studying regimens that are considered nonstandard on the current disease classification based on tissue of origin. While such studies can find shared features among different cancers, there is also a great deal of interest in understanding the differences between different cancer types and even among different tumors of the same tissue of origin.

Bailey et al performed integrated genomic analysis of 456 cases of pancreatic adenocarcinoma and identified 32 recurrently mutated genes that aggregate into 10 pathways: KRAS, TGF-β, WNT, NOTCH, ROBO/SLIT signaling, G1/S transition, SWI-SNF, chromatin modification, DNA repair and RNA processing. [32] The authors categorized pancreatic adenocarcinoma into four molecular subtypes based on the genetic mutations observed: squamous, pancreatic progenitor, immunogenic; and aberrantly differentiated endocrine exocrine. Within each subtype, tumors share certain pathways that might have therapeutic implications. Clinical utility of such new classifications is yet to be evaluated in prospective studies. Other authors also have proposed different molecular classification system for pancreatic cancer based genomic analysis of tumor specimens. [210, 211] Collisson et al proposed classification of PDACs in to three subtypes of Classical, quasimesenchymal (QM) and exocrine like. Exploring the predictive role of this classification in PDAC cell lines, they proposed differential response to Gemcitabine and erlotinib in the classical and QM subtype. Mofitt et al expanded molecular profiling from primary tumor to metastatic lesions and normal tumors. They classified PDAC tumors in to 2 subtypes of classical and basal like. Stroma was also classified to normal and activated. Wadell et al has proposed a classification mechanism with reliance on structural variation events. [116] They classified tumors to 4 subtypes. Unstable subtype (subtype 4) which accounted for 14% of samples in this study is possibly what is currently known as pancreatic cancer with BRCAness. These classifications need to be prospectively evaluated and be used in the design of scientifically rational treatment strategies.

6. Conclusion

Pancreatic cancer remains a very challenging disease. Despite decades of research, there is no good treatment strategy that offers meaningful and durable outcomes at this time. Several areas of research are showing some promising results such as targeting the stroma, exploiting DNA repair deficiencies and immune modulation. However, it is very unlikely that a single treatment option will be effective in the treatment of this disease, given the cellular and microenvironment complexity of the tumor that has endowed it with resistance to multiple treatment strategies. A better understanding and characterization of the molecular composition and cellular cross talk in the tumor microenvironment is needed to design scientifically rational novel treatment strategies for tumors that have specific molecular profiles. Combination of targeted therapies guided by predictive biomarkers will be the ultimate therapeutic goal.

7. Expert commentary

Pancreatic ductal adenocarcinoma remains a disease with very poor outcome because of late stage disease in the majority of newly diagnosed patients. A fifth or less of the patients are diagnosed with localized disease that may be amenable to radical and potentially curative surgical resection. However, only a fifth of the resected patients survive long term making the long-term survival of the entire population of pancreatic cancer patients less than 10 percent. There is major concern with the rising incidence of this disease and the projected mortality from pancreas cancer being second to lung cancer by 2030. Improvements in systemic therapy are essential to prolong survival of patients with this disease. However, thus far, active drugs have been very limited to a handful of conventional cytotoxic drugs. The benefit of therapy is very much limited by drug resistance. Successful developments of targeted and other novel therapies was limited by the poor understanding of the disease biology. However, in the last decade there has been a rapid increase in our knowledge of the molecular aspects of this disease with the development of better preclinical models that mimic human disease. New opportunities for targeted and personalized therapies are now being actively pursued along several fronts including exploitation of DNA repair abnormalities, disrupting the hyaluronan, targeting mesothelin, and blocking certain enzymes involved in metabolism unique to the cancer cell. Immunotherapy is still at its infancy in this disease. Unlike some other cancers, driver gene mutations are very scarce in pancreatic cancer. So far there are no molecular biomarkers to help direct, choose therapy or select patients but several may be emerging and include those involved in DNA repair, hyaouronan and mesothelin.

8. Five-year view

Pancreatic ductal adenocarcinoma is a drug resistant disease. Only few conventional cytotoxic agents have shown activity in this cancer. There is a continued work being done to optimize the delivery of these drugs, especially how they should be sequenced to improve the outcome of patients. The cytotoxic chemotherapy platforms of either gemcitabine/nab-paclitaxel or FOLFIRINOX will continue to be used especially in frontline setting to which will be added any newly developed agent. A number of targeted and novel agents are currently in phase I-III clinical trials. Within the next 2–3 years we may be able to see the clinical activities of these agents. High interest agents include PEGPH20 targeting hyaluronan, and platinum compounds with or without PARP inhibitors in patients with defective DNA repair pathways. PARP inhibitors may be used as maintenance therapy following the use of combination cytotoxic therapy in select patients with BRCA mutations. Biomarkers that may be of high interest in the next few years and be part of the everyday clinical practice include BRCA and other DNA repair gene mutations, hyaluronan expression and mesothelin expression. Innovative immunotherapy approaches are currently being actively pursued. No major immunotherapeutic breakthrough is anticipated in the near future for this disease. Molecular profiling of pancreatic cancers will expand in clinical practice and to guide clinical trial eligibility. Microsatellite testing, BRCA and related gene testing (e.g., PALB2, ATM) are examples of tests that are increasingly adopted by oncologists. Improvements in tissue collection and capture of circulating tumor cells and cell free DNA are to be expected.

Key issues.

  • Pancreas adenocarcinoma has proven resistant to various treatment strategies and is projected to become the second leading cause of cancer related mortality in few years. There is a strong unmet need to advance systemic therapy for this disease. More effective treatments are tightly linked to better understanding of the disease biology.

  • Gene mutations that can be effectively targeted by single agent therapy are extremely low in this disease. Advances in systemic therapy have been limited to conventional cytotoxic drugs at this time resulting in a very modest benefit.

  • Defective DNA repair pathways is seen in minority of patients but has a potential for becoming an effective treatment for those patients using platinum compounds with or without PARP inhibitors. Examples include patients with BRCA, PALB2, or ATM mutations. Multiple trials are addressing this area of research.

  • The disease is also characterized by complex interaction between the cancer cells and their microenvironment posing major challenges to design effective therapies. Targeting hyaluronic acid in the tumor stroma is a promising strategy when combined with gemcitabine and nab-paclitaxel. There is ongoing phase III trial in patients with tumors that overexpress hyalouronan. Other approaches include targeting the cellular component of the microenvironment responsible for disease progression and resistance to therapy

  • Immunotherapy with checkpoint inhibitors has not been effective in pancreas cancer in general. In very few patients with defective mismatch repair enzymes, this treatment is a very good treatment option.

  • Personalizing cancer treatment may be achieved by the use of biomarkers to guide the selection of therapeutic agents. Better molecular characterization of the disease coupled with increasing capabilities in molecular techniques may help develop more personalized treatment options.

Acknowledgments

Funding

The authors are grateful for support from the SKY Foundation, James H Thie Foundation and Perri Family Foundation and the National Institute of Health R21 grant 1R21CA188818–01A1

8. Appendix: Tables and figures

Table 1.

Pancreatic cancer GEMMs and PDXs

Mouse Models Mutation Ref
Genetically engineered mouse models (GEMMS)
Genetically engineered mouse models (GEMMS)
PDX-1-Cre/Lox-Stop-Lox (LSL)-Kras or p48/LSL-Kras
K-ras mutation in pancreatic cells [13]
PDX-1-Cre, LSL-KrasG12D, LSL-Trp53R172H/- K-ras and p53 mutations [92]
PDX-1-Cre, Brca2F11, LSL-KrasG12D, Trp53 F2–10 K-ras, BRCA2, and p53 mutations [93]
Mist1 KrasG12D/+ transgenic model KrasG12D into ORF of Mist1 [95]
PDX1-Cre, KrasG12D, Ink4a/Arfflox/flox transgenic model K-ras and Ink4a mutations [96]
PDX1-Cre, KrasG12D, Smad4flox/flox transgenic model K-ras and Smad4 mutations [97]
Ptf1acre/+, LSL-KrasG12D/+, Tgfbr2flox/flox transgenic model K-ras and TGF-β mutations [98]
LSL-KrasG12D; LSL-Trp53R172H/+; Ink4flox/+; K-ras, p53, and Ink4 [99]
Ptf1/p48-Cre mutations

Patient derived Xenografts (PDXs)
TM00310 K-ras and p53 mutations [100]
TM00312 p53 mutation [100]
TM00314 K-ras mutation [100]
More PDX models can been found in Mouse Tumor Biology Database [100]

Table 2.

Frequency of altered intracellular molecular pathways in pancreatic cancer [32]

Altered Pathways Mutated Genes Reported frequency
KRAS Activation KRAS, MAPK4 92%
Cell Cycle, GI/S Checkpoint Disruption TP53, CDKN2A, TP53BP2 78%
TGF Beta Signaling SMAD3, SMAD4, TGF, TGPBR1, TGFBR2, ACVR1B andACVR2A 47%
Histone Modification KDM6A, SETD2, ASCOM Complex members MLL2 and MLL3 24%
DNA Repair BRCA1, BRCA2, ATM, PALB2, SATF2 17% (5% germline, 12% somatic)
RNA Processing SF3B1, U2AF1, RBM10 16%
SWI/SNF Complex ARID1A, ARID1B, PBRM1 and SMARCA4 14%
WNT Signaling RNF43, MAPK2, TLE4 5%

Table 3:

Select clinical trials with targets of interest

Mechanism Target Agent Combined with Phase N NCT
Stroma Hyaloronan PEGPH20 mFOLFIRINOX 1/2 <170 01959139
Gemcitabine/nabpaclitaxel 3 420 01839487
Tumor infiltrating monocytes or macrophages CCR2 PF-04136309 Gemcitabine/nabpaclitaxel 1b/2 02732938
FOLFORINOX 1b 47 01413022
CCX872-B FOLFIRINOX 1b 54 02345408
CCR2/CCR5 BMS-813160 Nivolumab 1/2 260* 03184870
CSF-1R AMG 820 pembrolizumab 1/2 197* 02713529
Pexidartinib Durvalumab 1 58* 02777710
DNA repair PARP inhibitor Veliparib Gemcitabin/Cisplatin 2 107 01585805
FOLFOX 1/2 79 01489865
FOLFIRI 2 143 02890355
Olaparib none 3 145 02184195
Olaparib Cediranib 2 126* 02498613
Tumor metabolism PDH/KGDH CPI-613 Low dose FOLFIRINOX 1b 21 01835041
Mesothelin targeted Mesothelin anetumab ravtansine 1 348* 03102320
ravtansine 2 30 03023722
CAR T cells none 1 30 02706782
CAR T cells none 1/2 136 01583686
Antibody/LMB-100 none 1/2 100 02810418
Sternness STAT3 Napabucasin Gemcitabine/nabpaclitaxel 1b/2 71 02231723
Gemcitabine/nabpaclitaxel 3 1132 02231723
Cell death NQ01 ARQ-761 Gemcitabine/nabpaclitaxel 1/1b 20 02514031
Immune modulation Check point inhibitors/vaccines Nivolumab GVAX/CRS-207 2 108 02243371
Pembrolizumab ACP-196 2 76 02362048
Epacadostat/CRS-207 2 70 03006302
Durvalumab T remelimumab/SBRT 1 60 02311361
Pexidartinib 1 58 02777710
Gemcitabine/Nabpaclitaxel/Teremlimumab 2 180 02879318

*Includes other cancers

Footnotes

Declaration of interest

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

References

Reference annotations

* Of interest

** Of considerable interest

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA: A Cancer Journal for Clinicians. 2017;67(1):7–30. doi: 10.3322/caac.21387. [DOI] [PubMed] [Google Scholar]
  • 2.American Cancer Society. Cancer Facts and Figures 2017. Atlanta, GA: American Cancer Society; 2017. [Google Scholar]
  • 3.Heinemann V, Boeck S. Perioperative management of pancreatic cancer. Ann Oncol. 2008. September;19 Suppl 7:vii273–8. doi: 10.1093/annonc/mdn450. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 4.Sjoquist KM, Chin VT, Chantrill LA, et al. Personalising pancreas cancer treatment: When tissue is the issue. World J Gastroenterol. 2014. June 28;20(24):7849–63. doi: 10.3748/wjg.v20.i24.7849. PubMed PMID: ; PubMed Central PMCID: PMCPMC4069313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cress RD, Yin D, Clarke L, et al. Survival among patients with adenocarcinoma of the pancreas: a population-based study (United States). Cancer Causes Control. 2006. May;17(4):403–9. doi: 10.1007/s10552-005-0539-4. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 6.Alexakis N, Halloran C, Raraty M, et al. Current standards of surgery for pancreatic cancer. Br J Surg. 2004. November;91(11):1410–27. doi: 10.1002/bjs.4794. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 7.Schrag D, Archer L, Wang X, et al. A patterns-of-care study of post-progression treatment (Rx) among patients (pts) with advanced pancreas cancer (APC) after gemcitabine therapy on Cancer and Leukemia Group B (CALGB) study #80303. Journal of Clinical Oncology. 2007;25(18_suppl):4524–4524. doi: 10.1200/jco.2007.25.18_suppl.4524.17925547 [DOI] [Google Scholar]
  • 8.Kim HJ, Kim MH, Myung SJ, et al. A new strategy for the application of CA19–9 in the differentiation of pancreaticobiliary cancer: analysis using a receiver operating characteristic curve. The American journal of gastroenterology. 1999. July;94(7): 1941–6. doi: 10.1111/j.1572-0241.1999.01234.x. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 9.Steinberg W The clinical utility of the CA 19–9 tumor-associated antigen. The American journal of gastroenterology. 1990. April;85(4):350–5. PubMed PMID: [PubMed] [Google Scholar]
  • 10.Bardeesy N, DePinho RA. Pancreatic cancer biology and genetics. Nat Rev Cancer. 2002. December;2(12):897–909. doi: 10.1038/nrc949. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 11.Fang Y, Yao Q, Chen Z, et al. Genetic and molecular alterations in pancreatic cancer: implications for personalized medicine. Med Sci Monit. 2013. October 31;19:916–26. doi: 10.12659/MSM.889636. PubMed PMID: ; PubMed Central PMCID: PMCPMC3 818103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS oncogenes: weaving a tumorigenic web. Nat Rev Cancer. 2011. October 13;11(11):761–74. doi: 10.1038/nrc3106. PubMed PMID: ; PubMed Central PMCID: PMCPMC3632399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hingorani SR, Petricoin EF, Maitra A, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell. 2003. December;4(6):437–50. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 14.Rhim AD, Stanger BZ. Molecular biology of pancreatic ductal adenocarcinoma progression: aberrant activation of developmental pathways. Prog Mol Biol Transl Sci. 2010;97:41–78. doi: 10.1016/B978-0-12-385233-5.00002-7. PubMed PMID: ; PubMed Central PMCID: PMCPMC3117430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hustinx SR, Leoni LM, Yeo CJ, et al. Concordant loss of MTAP and p16/CDKN2A expression in pancreatic intraepithelial neoplasia: evidence of homozygous deletion in a noninvasive precursor lesion. Mod Pathol. 2005. July;18(7):959–63. doi: 10.1038/modpathol.3800377. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 16.Sharpless NE, DePinho RA. Cancer: crime and punishment. Nature. 2005. August 04;436(7051):636–7. doi: 10.1038/436636a. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 17.Redston MS, Caldas C, Seymour AB, et al. p53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Res. 1994. June 01;54(11):3025–33. PubMed PMID: [PubMed] [Google Scholar]
  • 18.Rozenblum E, Schutte M, Goggins M, et al. Tumor-suppressive pathways in pancreatic carcinoma. Cancer Res. 1997. May 01;57(9):1731–4. PubMed PMID: . [PubMed] [Google Scholar]
  • 19.Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996. January 19;271(5247):350–3. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 20.Cleaver JE, Lam ET, Revet I. Disorders of nucleotide excision repair: the genetic and molecular basis of heterogeneity. Nat Rev Genet. 2009. November;10(11):756–68. doi: 10.1038/nrg2663. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 21.Lieber MR. The mechanism of human nonhomologous DNA end joining. J Biol Chem. 2008. January 04;283(1):1–5. doi: 10.1074/jbc.R700039200. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 22.Caldecott KW. Single-strand break repair and genetic disease. Nat Rev Genet. 2008. August;9(8):619–31. doi: 10.1038/nrg2380. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 23.Jiricny J The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol. 2006. May;7(5):335–46. doi: 10.1038/nrm1907. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 24.Moynahan ME, Jasin M. Mitotic homologous recombination maintains genomic stability and suppresses tumorigenesis. Nat Rev Mol Cell Biol. 2010. March;11(3):196–207. doi: 10.1038/nrm2851. PubMed PMID: ; PubMed Central PMCID: PMCPMC3261768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Maginn EN, de Sousa CH, Wasan HS, et al. Opportunities for translation: targeting DNA repair pathways in pancreatic cancer. Biochim Biophys Acta. 2014. August;1846(1):45–54. doi: 10.1016/j.bbcan.2014.04.002. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 26.Murphy KM, Brune KA, Griffin C, et al. Evaluation of candidate genes MAP2K4, MADH4, ACVR1B, and BRCA2 in familial pancreatic cancer: deleterious BRCA2 mutations in 17%. Cancer Res. 2002. July 01;62(13):3789–93. PubMed PMID: . [PubMed] [Google Scholar]
  • 27.Venkitaraman AR. Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell. 2002. January 25;108(2):171–82. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 28.Nikkila J, Parplys AC, Pylkas K, et al. Heterozygous mutations in PALB2 cause DNA replication and damage response defects. Nat Commun. 2013;4:2578. doi: 10.1038/ncomms3578. PubMed PMID: ; PubMed Central PMCID: PMCPMC3 826652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Villarroel MC, Rajeshkumar NV, Garrido-Laguna I, et al. Personalizing cancer treatment in the age of global genomic analyses: PALB2 gene mutations and the response to DNA damaging agents in pancreatic cancer. Mol Cancer Ther. 2011. January;10(1):3–8. doi: 10.1158/1535-7163.MCT-10-0893. PubMed PMID: ; PubMed Central PMCID: PMCPMC3307340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Peng G, Lin S-YY. Exploiting the homologous recombination DNA repair network for targeted cancer therapy. World journal of clinical oncology. 2011;2(2):73–79. doi: 10.5306/wjco.v2.i2.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Roberts NJ, Jiao Y, Yu J, et al. ATM mutations in patients with hereditary pancreatic cancer. Cancer Discov. 2012. January;2(1):41–6. doi: 10.n58/2159-8290.CD-11-0194. PubMed PMID: ; PubMed Central PMCID: PMCPMC3676748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.**.Bailey P, Chang DK, Nones K, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531(7592):47–52. doi: 10.1038/nature16965. This paper classifies pancreatic cancer based on frequently seen mutations that have prognostic and therapeutic implications. [DOI] [PubMed] [Google Scholar]
  • 33.Erkan M, Hausmann S, Michalski CW, et al. The role of stroma in pancreatic cancer: diagnostic and therapeutic implications. Nat Rev Gastroenterol Hepatol. 2012. August;9(8):454–67. doi: 10.1038/nrgastro.2012.115. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 34.Erkan M, Michalski CW, Rieder S, et al. The activated stroma index is a novel and independent prognostic marker in pancreatic ductal adenocarcinoma. Clin Gastroenterol Hepatol. 2008. October;6(10):1155–61. doi: 10.1016/j.cgh.2008.05.006. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 35.Hidalgo M Pancreatic cancer. N Engl J Med. 2010. April 29;362(17):1605–17. doi: 10.1056/NEJMra0901557. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 36.Korc M Pancreatic cancer-associated stroma production. Am J Surg. 2007. October;194(4 Suppl):S84–6. doi: 10.1016/j.amjsurg.2007.05.004. PubMed PMID: ; PubMed Central PMCID: PMCPMC2094116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Apte MV, Haber PS, Applegate TL, et al. Periacinar stellate shaped cells in rat pancreas: identification, isolation, and culture. Gut. 1998. July;43(1):128–33. PubMed PMID: ; PubMed Central PMCID: PMCPMC1727174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Masamune A, Shimosegawa T. Signal transduction in pancreatic stellate cells. J Gastroenterol. 2009;44(4):249–60. doi: 10.1007/s00535-009-0013-2. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 39.Apte MV, Xu Z, Pothula S, et al. Pancreatic cancer: The microenvironment needs attention too! Pancreatology. 2015. July;15(4 Suppl):S32–8. doi: 10.1016/j.pan.2015.02.013. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 40.Apte MV, Wilson JS, Lugea A, et al. A starring role for stellate cells in the pancreatic cancer microenvironment. Gastroenterology. 2013. June;144(6):1210–9. doi: 10.1053/j.gastro.2012.11.037. PubMed PMID: ; PubMed Central PMCID: PMCPMC3729446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ene-Obong A, Clear AJ, Watt J, et al. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology. 2013. November;145(5):1121–32. doi: 10.1053/j.gastro.2013.07.025. PubMed PMID: ; PubMed Central PMCID: PMCPMC3896919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mace TA, Bloomston M, Lesinski GB. Pancreatic cancer-associated stellate cells: A viable target for reducing immunosuppression in the tumor microenvironment. Oncoimmunology. 2013. July 01;2(7):e24891. doi: 10.4161/onci.24891. PubMed PMID: ; PubMed Central PMCID: PMCPMC3782129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Xu Z, Vonlaufen A, Phillips PA, et al. Role of pancreatic stellate cells in pancreatic cancer metastasis. Am J Pathol. 2010. November;177(5):2585–96. doi: 10.2353/ajpath.2010.090899. PubMed PMID: ; PubMed Central PMCID: PMCPMC2966814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Patel MB, Pothula SP, Xu Z, et al. The role of the hepatocyte growth factor/c-MET pathway in pancreatic stellate cell-endothelial cell interactions: antiangiogenic implications in pancreatic cancer. Carcinogenesis. 2014. August;35(8):1891–900. doi: 10.1093/carcin/bgu122. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 45.Rucki AA, Zheng L. Pancreatic cancer stroma: understanding biology leads to new therapeutic strategies. World J Gastroenterol. 2014. March 07;20(9):2237–46. doi: 10.3748/wjg.v20.i9.2237. PubMed PMID: ; PubMed Central PMCID: PMCPMC3 942829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zheng L, Xue J, Jaffee EM, et al. Role of immune cells and immune-based therapies in pancreatitis and pancreatic ductal adenocarcinoma. Gastroenterology. 2013. June;144(6):1230–40. doi: 10.1053/j.gastro.2012.12.042. PubMed PMID: ; PubMed Central PMCID: PMCPMC3641650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Porembka MR, Mitchem JB, Belt BA, et al. Pancreatic adenocarcinoma induces bone marrow mobilization of myeloid-derived suppressor cells which promote primary tumor growth. Cancer Immunol Immunother. 2012. September;61(9):1373–85. doi: 10.1007/s00262-011-1178-0. PubMed PMID: ; PubMed Central PMCID: PMCPMC3697836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Beyer M, Schultze JL. Regulatory T cells in cancer. Blood. 2006. August 01;108(3):804–11. doi: 10.1182/blood-2006-02-002774. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 49.Sica A, Larghi P, Mancino A, et al. Macrophage polarization in tumour progression. Semin Cancer Biol. 2008. October;18(5):349–55. doi: 10.1016/j.semcancer.2008.03.004. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 50.Loos M, Giese NA, Kleeff J, et al. Clinical significance and regulation of the costimulatory molecule B7-H1 in pancreatic cancer. Cancer Lett. 2008. September 08;268(1):98–109. doi: 10.1016/j.canlet.2008.03.056. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 51.Nomi T, Sho M, Akahori T, et al. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2007. April 01;13(7):2151–7. doi: 10.1158/1078-0432.CCR-06-2746. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 52.Loges S, Schmidt T, Carmeliet P. Mechanisms of resistance to anti-angiogenic therapy and development of third-generation anti-angiogenic drug candidates. Genes Cancer. 2010. January;1(1):12–25. doi: 10.1177/1947601909356574. PubMed PMID: ; PubMed Central PMCID: PMCPMC3092176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Shibuya M Vascular endothelial growth factor-dependent and -independent regulation of angiogenesis. BMB Rep. 2008. April 30;41(4):278–86. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 54.Kindler HL, Niedzwiecki D, Hollis D, et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303). Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010. August 01;28(22):3617–22. doi: 10.1200/jco.2010.28.1386. PubMed PMID: ; PubMed Central PMCID: PMCPMC2917317. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Van Cutsem E, Vervenne WL, Bennouna J, et al. Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009. May 01;27(13):2231–7. doi: 10.1200/JCO.2008.20.0238. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 56.Pennacchietti S, Michieli P, Galluzzo M, et al. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell. 2003. April;3(4):347–61. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 57.Tsarfaty I, Rong S, Resau JH, et al. The Met proto-oncogene mesenchymal to epithelial cell conversion. Science. 1994. January 07;263(5143):98–101. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 58.Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009. June;119(6):1420–8. doi: 10.1172/JCI39104. PubMed PMID: ; PubMed Central PMCID: PMCPMC2689101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Avan A, Caretti V, Funel N, et al. Crizotinib inhibits metabolic inactivation of gemcitabine in c-Met-driven pancreatic carcinoma. Cancer research. 2013. November 15;73(22):6745–56. doi: 10.1158/0008-5472.CAN-13-0837. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 60.Brandes F, Schmidt K, Wagner C, et al. Targeting cMET with INC280 impairs tumour growth and improves efficacy of gemcitabine in a pancreatic cancer model. BMC Cancer. 2015. February 19;15:71. doi: 10.1186/s12885-015-1064-9. PubMed PMID: ; PubMed Central PMCID: PMCPMC4340491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lili LN, Matyunina LV, Walker LD, et al. Evidence for the importance of personalized molecular profiling in pancreatic cancer. Pancreas. 2014. March;43(2):198–211. doi: 10.1097/MPA.0000000000000020. PubMed PMID: ; PubMed Central PMCID: PMCPMC42063 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gutierrez ML, Sayagues JM, Abad Mdel M, et al. Cytogenetic heterogeneity of pancreatic ductal adenocarcinomas: identification of intratumoral pathways of clonal evolution. Histopathology. 2011. February;58(3):486–97. doi: 10.1111/j.1365-2559.2011.03771.x. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 63.Mukherjee P, Ginardi AR, Madsen CS, et al. MUC1-specific CTLs are non-functional within a pancreatic tumor microenvironment. Glycoconj J. 2001. Nov-Dec;18(11–12):931–42. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 64.Mei L, Du W, Ma WW. Targeting stromal microenvironment in pancreatic ductal adenocarcinoma: controversies and promises. J Gastrointest Oncol. 2016. June;7(3):487–94. doi: 10.21037/jgo.2016.03.03. PubMed PMID: ; PubMed Central PMCID: PMCPMC4880763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.di Magliano MP, Logsdon CD. Roles for KRAS in pancreatic tumor development and progression. Gastroenterology. 2013. June;144(6):1220–9. doi: 10.1053/j.gastro.2013.01.071. PubMed PMID: ; PubMed Central PMCID: PMCPMC3902845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chung V, McDonough S, Philip PA, et al. Effect of Selumetinib and MK-2206 vs Oxaliplatin and Fluorouracil in Patients With Metastatic Pancreatic Cancer After Prior Therapy: SWOG S1115 Study Randomized Clinical Trial. JAMA Oncol. 2016. December 15. doi: 10.1001/jamaoncol.2016.5383. PubMed PMID: . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hancke S, Holm HH, Koch F. Ultrasonically guided percutaneous fine needle biopsy of the pancreas. Surg Gynecol Obstet. 1975. March;140(3):361–4. PubMed PMID: . [PubMed] [Google Scholar]
  • 68.Livraghi T, Sangalli G, Giordano F, et al. Fine aspiration versus fine cutting needle, and comparison between smear cytology, inclusion cytology and microhistology in abdominal lesions. Tumori. 1988. June 30;74(3):361–4. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 69.Li L, Liu LZ, Wu QL, et al. CT-guided core needle biopsy in the diagnosis of pancreatic diseases with an automated biopsy gun. J Vasc Interv Radiol. 2008. January;19(1): 89–94. doi: 10.1016/j.jvir.2007.09.001. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 70.Lee YN, Moon JH, Kim HK, et al. Core biopsy needle versus standard aspiration needle for endoscopic ultrasound-guided sampling of solid pancreatic masses: a randomized parallel-group study. Endoscopy. 2014. December;46(12):1056–62. doi: 10.1055/s-0034-1377558. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 71.Bhutani M, Koduru P, Lanke G, et al. The emerging role of endoscopic ultrasound-guided core biopsy for the evaluation of solid pancreatic masses. Minerva Gastroenterol Dietol. 2015. June;61(2):51–9. PubMed PMID: [PubMed] [Google Scholar]
  • 72.Kalogeraki A, Papadakis GZ, Tamiolakis D, et al. EUS - Fine-Needle Aspiration Biopsy (FNAB) in the Diagnosis of Pancreatic Adenocarcinoma: A Review. Rom J Intern Med. 2016. Jan-Mar;54(1):24–30. doi: 10.1515/rjim-2016-0002. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 73.Iglesias-Garcia J, Poley JW, Larghi A, et al. Feasibility and yield of a new EUS histology needle: results from a multicenter, pooled, cohort study. Gastrointest Endosc. 2011. June;73(6):1189–96. doi: 10.1016/j.gie.2011.01.053. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 74.Allard WJ, Matera J, Miller MC, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clinical cancer research : an official journal of the American Association for Cancer Research. 2004. October 15;10(20):6897–904. doi: 10.1158/1078-0432.CCR-04-0378. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 75.Hiraiwa K, Takeuchi H, Hasegawa H, et al. Clinical significance of circulating tumor cells in blood from patients with gastrointestinal cancers. Ann Surg Oncol. 2008. November;15(11):3092–100. doi: 10.1245/s10434-008-0122-9. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 76.Tjensvoll K, Nordgard O, Smaaland R. Circulating tumor cells in pancreatic cancer patients: methods of detection and clinical implications. International journal of cancer Journal international du cancer. 2014. January 01;134(1):1–8. doi: 10.1002/ijc.28134. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 77.Bidard FC, Huguet F, Louvet C, et al. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: the ancillary CirCe 07 study to the LAP 07 trial. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO. 2013. August;24(8):2057–61. doi: 10.1093/annonc/mdt176. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 78.Okubo K, Uenosono Y, Arigami T, et al. Clinical impact of circulating tumor cells and therapy response in pancreatic cancer. Eur J Surg Oncol. 2017. February 12. doi: 10.1016/j.ejso.2017.01.241. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 79.Takai E, Totoki Y, Nakamura H, et al. Clinical Utility of Circulating Tumor DNA for Molecular Assessment and Precision Medicine in Pancreatic Cancer. Adv Exp Med Biol. 2016;924:13–17. doi: 10.1007/978-3-319-42044-8_3. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 80.Sibinga Mulder BG, Mieog JS, Handgraaf HJ, et al. Targeted next-generation sequencing of FNA-derived DNA in pancreatic cancer. J Clin Pathol. 2017. February;70(2):174–178. doi: 10.1136/jclinpath-2016-203928. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 81.Young G, Wang K, He J, et al. Clinical next-generation sequencing successfully applied to fine-needle aspirations of pulmonary and pancreatic neoplasms. Cancer Cytopathol. 2013. December;121(12):688–94. doi: 10.1002/cncy.21338. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 82.Amato E, Molin MD, Mafficini A, et al. Targeted next-generation sequencing of cancer genes dissects the molecular profiles of intraductal papillary neoplasms of the pancreas. J Pathol. 2014. July;233(3):217–27. doi: 10.1002/path.4344. PubMed PMID: ; PubMed Central PMCID: PMCPMC4057302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kleeff J, Korc M, Apte M, et al. Pancreatic cancer. Nat Rev Dis Primers. 2016. April 21;2:16022. doi: 10.1038/nrdp.2016.22. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 84.Biankin AV, Piantadosi S, Hollingsworth SJ. Patient-centric trials for therapeutic development in precision oncology. Nature. 2015. October 15;526(7573):361–70. doi: 10.1038/nature15819. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 85.Birnbaum DJ, Finetti P, Lopresti A, et al. A 25-gene classifier predicts overall survival in resectable pancreatic cancer. BMC Med. 2017. September 20;15(1):170. doi: 10.1186/s12916-017-0936-z. PubMed PMID: ; PubMed Central PMCID: PMCPMC5606023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Creative Bioarray. Pancreatic Tumor Cells 2017. [cited 2017 June 3] Available from: http://www.creative-bioarray.com/Products/Pancreatic-Tumor-Cells-list-106.htm
  • 87.Masters JR. Human cancer cell lines: fact and fantasy. Nat Rev Mol Cell Biol. 2000. December;1(3):233–6. doi: 10.1038/35043102. PubMed PMID: ; eng. [DOI] [PubMed] [Google Scholar]
  • 88.Zanoni M, Piccinini F, Arienti C, et al. 3D tumor spheroid models for in vitro therapeutic screening: a systematic approach to enhance the biological relevance of data obtained. Sci Rep. 2016. January 11;6:19103. doi: 10.1038/srep19103. PubMed PMID: ; PubMed Central PMCID: PMCPMC4707510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Boj SF, Hwang CI, Baker LA, et al. Model organoids provide new research opportunities for ductal pancreatic cancer. Molecular & cellular oncology. 2016. January;3(1):e1014757. doi: 10.1080/23723556.2015.1014757. PubMed PMID: ; PubMed Central PMCID: PMCPMC4845167. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Boj SF, Hwang CI, Baker LA, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015. January 15;160(1–2):324–38. doi: 10.1016/j.cell.2014.12.021. PubMed PMID: ; PubMed Central PMCID: PMCPMC4334572. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Logsdon CD, Arumugam T, Ramachandran V. Animal Models of Gastrointestinal and Liver Diseases. The difficulty of animal modeling of pancreatic cancer for preclinical evaluation of therapeutics. American journal of physiology Gastrointestinal and liver physiology. 2015. September 01;309(5):G283–91. doi: 10.1152/ajpgi.00169.2015. PubMed PMID: ; PubMed Central PMCID: PMCPMC4556944. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hingorani SR, Wang L, Multani AS, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005. May;7(5):469–83. doi: 10.1016/j.ccr.2005.04.023. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 93.Rowley M, Ohashi A, Mondal G, et al. Inactivation of Brca2 promotes Trp53-associated but inhibits KrasG12D-dependent pancreatic cancer development in mice. Gastroenterology. 2011. April;140(4):1303–1313.e1–3. doi: 10.1053/j.gastro.2010.12.039. PubMed PMID: ; PubMed Central PMCID: PMCPMC3066280. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Skoulidis F, Cassidy LD, Pisupati V, et al. Germline Brca2 heterozygosity promotes Kras(G12D) -driven carcinogenesis in a murine model of familial pancreatic cancer. Cancer Cell. 2010. November 16;18(5):499–509. doi: 10.1016/j.ccr.2010.10.015. PubMed PMID: ; eng. [DOI] [PubMed] [Google Scholar]
  • 95.Tuveson DA, Zhu L, Gopinathan A, et al. Mist1-KrasG12D knock-in mice develop mixed differentiation metastatic exocrine pancreatic carcinoma and hepatocellular carcinoma. Cancer Res. 2006. January 01;66(1):242–7. doi: 10.1158/0008-5472.can-05-2305. PubMed PMID: ; eng. [DOI] [PubMed] [Google Scholar]
  • 96.Aguirre AJ, Bardeesy N, Sinha M, et al. Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes & development. 2003. December 15;17(24):3112–26. doi: 10.1101/gad.1158703. PubMed PMID: ; PubMed Central PMCID: PMCPMC305262. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Bardeesy N, Cheng KH, Berger JH, et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes & development. 2006. November 15;20(22):3130–46. doi: 10.1101/gad.1478706. PubMed PMID: ; PubMed Central PMCID: PMCPMC1635148. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ijichi H, Chytil A, Gorska AE, et al. Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-beta signaling in cooperation with active Kras expression. Genes & development. 2006. November 15;20(22):3147–60. doi: 10.1101/gad.1475506. PubMed PMID: ; PubMed Central PMCID: PMCPMC1635149. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ma L, Saiyin H. LSL-KrasG12D; LSL-Trp53R172H/+; Ink4flox/+; Ptf1/p48-Cre mice are an applicable model for locally invasive and metastatic pancreatic cancer. PloS one. 2017;12(5):e0176844. doi: 10.1371/journal.pone.0176844. PubMed PMID: ; PubMed Central PMCID: PMCPmc5419507. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Mouse Tumor Biology Database. Pancreatic Cancer Patient Derived Xenograft 2017. [cited 2017 June 3] Available from: http://tumor.informatics.jax.org/mtbwi/pdxSearchResults.do?primarySites=Pancreas&
  • 101.Kawaguchi K, Igarashi K, Murakami T, et al. MEK inhibitors cobimetinib and trametinib, regressed a gemcitabine-resistant pancreatic-cancer patient-derived orthotopic xenograft (PDOX). Oncotarget. 2017. May 07. doi: 10.18632/oncotarget.17667. PubMed PMID: ; eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hasgur S, Aryee KE, Shultz LD, et al. Generation of Immunodeficient Mice Bearing Human Immune Systems by the Engraftment of Hematopoietic Stem Cells. Methods in molecular biology (Clifton, NJ). 2016;1438:67–78. doi: 10.1007/978-1-4939-3661-8_4. PubMed PMID: ; PubMed Central PMCID: PMCPMC5268072. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Long J, Liu Z, Wu X, et al. Screening for genes and subnetworks associated with pancreatic cancer based on the gene expression profile. Mol Med Rep. 2016. May;13(5):3779–86. doi: 10.3892/mmr.2016.5007. PubMed PMID: ; PubMed Central PMCID: PMCPMC4838159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Long J, Liu Z, Wu X, et al. Gene expression profile analysis of pancreatic cancer based on microarray data. Mol Med Rep. 2016. May;13(5):3913–9. doi: 10.3892/mmr.2016.5021. PubMed PMID: ; PubMed Central PMCID: PMCPMC4838162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Jeon J, Nim S, Teyra J, et al. A systematic approach to identify novel cancer drug targets using machine learning, inhibitor design and high-throughput screening. Genome Med. 2014;6(7):57. doi: 10.1186/s13073-014-0057-7. PubMed PMID: PubMed Central PMCID: PMCPMC4143549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Tempero MA, Berlin J, Ducreux M, et al. Pancreatic cancer treatment and research: an international expert panel discussion. Ann Oncol. 2011. July;22(7):1500–6. doi: 10.1093/annonc/mdq545. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 107.Dienstmann R, Rodon J, Tabernero J. Optimal design of trials to demonstrate the utility of genomically-guided therapy: Putting Precision Cancer Medicine to the test. Mol Oncol. 2015. May;9(5):940–50. doi: 10.1016/j.molonc.2014.06.014. PubMed PMID: . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Ashworth A A synthetic lethal therapeutic approach: poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2008. August 01;26(22):3785–90. doi: 10.1200/JCO.2008.16.0812. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 109.Boyerinas B, Jochems C, Fantini M, et al. Antibody-Dependent Cellular Cytotoxicity Activity of a Novel Anti-PD-L1 Antibody Avelumab (MSB0010718C) on Human Tumor Cells. Cancer Immunol Res. 2015. October;3(10):1148–57. doi: 10.1158/2326-6066.CIR-15-0059. PubMed PMID: . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Thompson D, Easton DF, Breast Cancer Linkage C. Cancer Incidence in BRCA1 mutation carriers. J Natl Cancer Inst. 2002. September 18;94(18):1358–65. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 111.Brose MS, Rebbeck TR, Calzone KA, et al. Cancer risk estimates for BRCA1 mutation carriers identified in a risk evaluation program. J Natl Cancer Inst. 2002. September 18;94(18):1365–72. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 112.Couch FJ, Johnson MR, Rabe KG, et al. The prevalence of BRCA2 mutations in familial pancreatic cancer. Cancer Epidemiol Biomarkers Prev. 2007. February;16(2):342–6. doi: 10.1158/1055-9965.EPI-06-0783. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 113.Breast Cancer Linkage C. Cancer risks in BRCA2 mutation carriers. J Natl Cancer Inst. 1999. August 04;91(15):1310–6. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 114.van Asperen CJ, Brohet RM, Meijers-Heijboer EJ, et al. Cancer risks in BRCA2 families: estimates for sites other than breast and ovary. J Med Genet. 2005. September;42(9):711–9. doi: 10.1136/jmg.2004.028829. PubMed PMID: ; PubMed Central PMCID: PMCPMC1736136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Vyas O, Leung K, Ledbetter L, et al. Clinical outcomes in pancreatic adenocarcinoma associated with BRCA-2 mutation. Anticancer Drugs. 2015. February;26(2):224–6. doi: 10.1097/CAD.0000000000000178. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 116.*.Waddell N, Pajic M, Patch AM, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015. February 26;518(7540):495–501. doi: 10.1038/nature14169. PubMed PMID: ; PubMed Central PMCID: PMCPMC4523082.This paper subclassifies pancreatic cancer based on whole genome sequencing to uncover potential molecular vulnerabilities. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Golan T, Kanji ZS, Epelbaum R, et al. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. Br J Cancer. 2014. September 09; 111 (6): 1132–8. doi: 10.1038/bjc.2014.418. PubMed PMID: ; PubMed Central PMCID: PMCPMC4453851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Tran B, Moore S, Zogopoulos G, et al. Platinum-based chemotherapy (Pt-chemo) in pancreatic adenocarcinoma (PC) associated with BRCA mutations: A translational case series. Journal of Clinical Oncology. 2012;30(4_suppl):217–217. doi: 10.1200/jco.2012.30.4_suppl.217. PubMed PMID: . [DOI] [Google Scholar]
  • 119.Golan T, Sella T, O’Reilly EM, et al. Overall survival and clinical characteristics of BRCA mutation carriers with stage I/II pancreatic cancer. British journal of cancer. 2017. March 14;116(6):697–702. doi: 10.1038/bjc.2017.19. PubMed PMID: ; PubMed Central PMCID: PMCPMC5355924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Haber JE. DNA recombination: the replication connection. Trends Biochem Sci. 1999. July;24(7):271–5. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 121.Lee JM, Ledermann JA, Kohn EC. PARP Inhibitors for BRCA1/2 mutation-associated and BRCA-like malignancies. Ann Oncol. 2014. January;25(1):32–40. doi: 10.1093/annonc/mdt384. PubMed PMID: ; PubMed Central PMCID: PMCPMC3868320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Hay T, Matthews JR, Pietzka L, et al. Poly(ADP-ribose) polymerase-1 inhibitor treatment regresses autochthonous Brca2/p53-mutant mammary tumors in vivo and delays tumor relapse in combination with carboplatin. Cancer Res. 2009. May 01;69(9):3850–5. doi: 10.1158/0008-5472.CAN-08-2388. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 123.Domchek SM, Hendifar AE, McWilliams RR, et al. RUCAPANC: An open-label, phase 2 trial of the PARP inhibitor rucaparib in patients (pts) with pancreatic cancer (PC) and a known deleterious germline or somatic BRCA mutation. J Clin Oncol (Meeting Abstracts). 2016 January, 2016;34(suppl):abstr 4110. [Google Scholar]
  • 124.Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev Cancer. 2004. July;4(7):528–39. doi: 10.1038/nrc1391. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 125.Knudson W, Biswas C, Toole BP. Interactions between human tumor cells and fibroblasts stimulate hyaluronate synthesis. Proc Natl Acad Sci U S A. 1984. November;81(21):6767–71. PubMed PMID: ; PubMed Central PMCID: PMCPMC3 92012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Sato N, Kohi S, Hirata K, et al. Role of hyaluronan in pancreatic cancer biology and therapy: Once again in the spotlight. Cancer Sci. 2016. May;107(5):569–75. doi: 10.1111/cas.12913. PubMed PMID: ; PubMed Central PMCID: PMCPMC4970823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Provenzano PP, Cuevas C, Chang AE, et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012. March 20;21(3):418–29. doi: 10.1016/j.ccr.2012.01.007. PubMed PMID: ; PubMed Central PMCID: PMCPMC3371414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Provenzano PP, Hingorani SR. Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. Br J Cancer. 2013. January 15;108(1): 1–8. doi: 10.1038/bjc.2012.569. PubMed PMID: ; PubMed Central PMCID: PMCPMC3553539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Misra S, Hascall VC, Markwald RR, et al. Interactions between Hyaluronan and Its Receptors (CD44, RHAMM) Regulate the Activities of Inflammation and Cancer. Frontiers in immunology. 2015;6:201. doi: 10.3389/fimmu.2015.00201. PubMed PMID: ; PubMed Central PMCID: PMCPMC4422082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Cheng XB, Kohi S, Koga A, et al. Hyaluronan stimulates pancreatic cancer cell motility. Oncotarget. 2016. January 26;7(4):4829–40. doi: 10.18632/oncotarget.6617. PubMed PMID: ; PubMed Central PMCID: PMCPMC4826246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Kultti A, Zhao C, Singha NC, et al. Accumulation of extracellular hyaluronan by hyaluronan synthase 3 promotes tumor growth and modulates the pancreatic cancer microenvironment. Biomed Res Int. 2014;2014:817613. doi: 10.1155/2014/817613. PubMed PMID: ; PubMed Central PMCID: PMCPMC4131462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Thompson CB, Shepard HM, O’Connor PM, et al. Enzymatic depletion of tumor hyaluronan induces antitumor responses in preclinical animal models. Molecular cancer therapeutics. 2010. November;9(11):3052–64. doi: 10.1158/1535-7163.MCT-10-0470. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 133.Jacobetz MA, Chan DS, Neesse A, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013. January;62(1):112–20. doi: 10.1136/gutjnl-2012-302529. PubMed PMID: ; PubMed Central PMCID: PMCPMC3551211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Singha NC, Nekoroski T, Zhao C, et al. Tumor-associated hyaluronan limits efficacy of monoclonal antibody therapy. Molecular cancer therapeutics. 2015. February;14(2):523–32. doi: 10.1158/1535-7163.MCT-14-0580. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 135.**.Hingorani SR, Harris WP, Hendifar AE, et al. High response rate and PFS with PEGPH20 added to nab-paclitaxel/gemcitabine in stage IV previously untreated pancreatic cancer patients with high-HA tumors: Interim results of a randomized phase II study. J Clin Oncol (Meeting Abstracts). 2015 January, 2015;33(Suppl):abstract 4006. This paper shows benefit of adding PEGPH20 to chemotherapy was shown in pancreatic cancers with high hayluronan content. [Google Scholar]
  • 136.Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015. June 25;372(26):2509–20. doi: 10.1056/NEJMoa1500596. PubMed PMID: ; PubMed Central PMCID: PMCPMC4481136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Overman MJ, Lonardi S, Leone F, et al. Nivolumab in patients with DNA mismatch repair deficient/microsatellite instability high metastatic colorectal cancer: Update from CheckMate 142. J Clin Oncol (Meeting Abstracts). 2017 January, 2017;35(suppl 4s):abstr 519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.*.Le DT, Durham JN, Smith KN, et al. Mismatch-repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017. June 08. doi: 10.1126/science.aan6733. PubMed PMID: This paper showed the effectiveness of antiPD-1 therapy in mismatch deficient tumors including pancreatic cancer. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Salo-Mullen EE, O’Reilly EM, Kelsen DP, et al. Identification of germline genetic mutations in patients with pancreatic cancer. Cancer. 2015;121(24):4382–4388. doi: 10.1002/cncr.29664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Eatrides JM, Coppola D, Diffalha SA, et al. Microsatellite instability in pancreatic cancer. J Clin Oncol (Meeting Abstracts). 2016 June, 2016;34(suppl):abstr e15753. [Google Scholar]
  • 141.Cairns RA, Harris IS, Mak TW. Regulation of cancer cell metabolism. Nat Rev Cancer. 2011. February;11(2):85–95. doi: 10.1038/nrc2981. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 142.*.Cohen R, Neuzillet C, Tijeras-Raballand A, et al. Targeting cancer cell metabolism in pancreatic adenocarcinoma. Oncotarget. 2015. July 10;6(19):16832–47. doi: 10.18632/oncotarget.4160. PubMed PMID: ; PubMed Central PMCID: PMCPMC4627277. This paper presents the potential role of targeting metabolism in pancreas cancer. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Zhou W, Capello M, Fredolini C, et al. Proteomic analysis of pancreatic ductal adenocarcinoma cells reveals metabolic alterations. Journal of proteome research. 2011. April 01;10(4):1944–52. doi: 10.1021/pr101179t. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 144.Vander Heiden MG, Plas DR, Rathmell JC, et al. Growth factors can influence cell growth and survival through effects on glucose metabolism. Mol Cell Biol. 2001. September;21(17):5899–912. PubMed PMID: ; PubMed Central PMCID: PMCPMC87309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Alistar A, Morris BB, Desnoyer R, et al. Safety and tolerability of the first-in-class agent CPI-613 in combination with modified FOLFIRINOX in patients with metastatic pancreatic cancer: a single-centre, open-label, dose-escalation, phase 1 trial. The Lancet Oncology. 2017. June;18(6):770–778. doi: 10.1016/S1470-2045(17)30314-5. PubMed PMID: . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Hassan R, Laszik ZG, Lerner M, et al. Mesothelin is overexpressed in pancreaticobiliary adenocarcinomas but not in normal pancreas and chronic pancreatitis. Am J Clin Pathol. 2005. December;124(6):838–45. PubMed PMID: . [PubMed] [Google Scholar]
  • 147.Argani P, Iacobuzio-Donahue C, Ryu B, et al. Mesothelin is overexpressed in the vast majority of ductal adenocarcinomas of the pancreas: identification of a new pancreatic cancer marker by serial analysis of gene expression (SAGE). Clin Cancer Res. 2001. December;7(12):3862–8. PubMed PMID: [PubMed] [Google Scholar]
  • 148.Rump A, Morikawa Y, Tanaka M, et al. Binding of ovarian cancer antigen CA125/MUC16 to mesothelin mediates cell adhesion. J Biol Chem. 2004. March 05;279(10):9190–8. doi: 10.1074/jbc.M312372200. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 149.Chen SH, Hung WC, Wang P, et al. Mesothelin binding to CA125/MUC16 promotes pancreatic cancer cell motility and invasion via MMP-7 activation. Sci Rep. 2013;3:1870. doi: 10.1038/srep01870. PubMed PMID: ; PubMed Central PMCID: PMCPMC3660778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Li M, Bharadwaj U, Zhang R, et al. Mesothelin is a malignant factor and therapeutic vaccine target for pancreatic cancer. Molecular cancer therapeutics. 2008. February;7(2):286–96. doi: 10.1158/1535-7163.MCT-07-0483. PubMed PMID: ; PubMed Central PMCID: PMCPMC2929838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Bharadwaj U, Marin-Muller C, Li M, et al. Mesothelin overexpression promotes autocrine IL-6/sIL-6R trans-signaling to stimulate pancreatic cancer cell proliferation. Carcinogenesis. 2011. July;32(7):1013–24. doi: 10.1093/carcin/bgr075. PubMed PMID: ; PubMed Central PMCID: PMCPMC3128561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Bharadwaj U, Li M, Chen C, et al. Mesothelin-induced pancreatic cancer cell proliferation involves alteration of cyclin E via activation of signal transducer and activator of transcription protein 3. Mol Cancer Res. 2008. November;6(11):1755–65. doi: 10.1158/1541-7786.MCR-08-0095. PubMed PMID: ; PubMed Central PMCID: PMCPMC2929833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Chang MC, Chen CA, Hsieh CY, et al. Mesothelin inhibits paclitaxel-induced apoptosis through the PI3K pathway. Biochem J. 2009. December 10;424(3):449–58. doi: 10.1042/BJ20082196. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 154.Cheng WF, Huang CY, Chang MC, et al. High mesothelin correlates with chemoresistance and poor survival in epithelial ovarian carcinoma. Br J Cancer. 2009. April 07;100(7):1144–53. doi: 10.1038/sj.bjc.6604964. PubMed PMID: ; PubMed Central PMCID: PMCPMC2669998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Chowdhury PS, Viner JL, Beers R, et al. Isolation of a high-affinity stable single-chain Fv specific for mesothelin from DNA-immunized mice by phage display and construction of a recombinant immunotoxin with anti-tumor activity. Proc Natl Acad Sci U S A. 1998. January 20;95(2):669–74. PubMed PMID: ; PubMed Central PMCID: PMCPMC18478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Chowdhury PS, Pastan I. Improving antibody affinity by mimicking somatic hypermutation in vitro. Nat Biotechnol. 1999. June;17(6):568–72. doi: 10.1038/9872. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 157.Hassan R, Bullock S, Premkumar A, et al. Phase I study of SS1P, a recombinant anti-mesothelin immunotoxin given as a bolus I.V. infusion to patients with mesothelin-expressing mesothelioma, ovarian, and pancreatic cancers. Clin Cancer Res. 2007. September 01;13(17):5144–9. doi: 10.1158/1078-0432.CCR-07-0869. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 158.Uchida T, Muramoto M, Kyunou H, et al. Clinical outcome of high-intensity focused ultrasound for treating benign prostatic hyperplasia: preliminary report. Urology. 1998. July;52(1):66–71. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 159.Hassan R, Cohen SJ, Phillips M, et al. Phase I clinical trial of the chimeric anti-mesothelin monoclonal antibody MORAb-009 in patients with mesothelin-expressing cancers. Clin Cancer Res. 2010. December 15;16(24):6132–8. doi: 10.1158/1078-0432.CCR-10-2275. PubMed PMID: ; PubMed Central PMCID: PMCPMC3057907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Fujisaka Y, Kurata T, Tanaka K, et al. Phase I study of amatuximab, a novel monoclonal antibody to mesothelin, in Japanese patients with advanced solid tumors. Investigational new drugs 2015. April;33(2):380–8. doi: 10.1007/s10637-014-0196-0. PubMed PMID: ; PubMed Central PMCID: PMCPMC4387254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Brockstedt DG, Giedlin MA, Leong ML, et al. Listeria-based cancer vaccines that segregate immunogenicity from toxicity. Proc Natl Acad Sci U S A. 2004. September 21;101(38):13832–7. doi: 10.1073/pnas.0406035101. PubMed PMID: ; PubMed Central PMCID: PMCPMC518841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Le DT, Ko AH, Wainberg ZA, et al. Results from a phase 2b, randomized, multicenter study of GVAX pancreas and CRS-207 compared to chemotherapy in adults with previously-treated metastatic pancreatic adenocarcinoma (ECLIPSE Study). J Clin Oncol (Meeting Abstracts). 2017 January 20, 2017;35(Suppl 4S):abstract 345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Yarden Y The EGFR family and its ligands in human cancer. signalling mechanisms and therapeutic opportunities. Eur J Cancer. 2001. September;37 Suppl 4:S3–8. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 164.Perez-Soler R HER1/EGFR targeting: refining the strategy. Oncologist. 2004;9(1):58–67. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 165.Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007. May 20;25(15):1960–6. doi: 10.1200/jco.2006.07.9525. PubMed PMID: 17452677; eng. [DOI] [PubMed] [Google Scholar]
  • 166.Safran H, Iannitti D, Ramanathan R, et al. Herceptin and gemcitabine for metastatic pancreatic cancers that overexpress HER-2/neu. Cancer Invest. 2004;22(5):706–12. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 167.Philip PA, Benedetti J, Corless CL, et al. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010. August 01;28(22):3605–10. doi: 10.1200/JCO.2009.25.7550. PubMed PMID: ; PubMed Central PMCID: PMCPMC2917315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Harder J, Ihorst G, Heinemann V, et al. Multicentre phase II trial of trastuzumab and capecitabine in patients with HER2 overexpressing metastatic pancreatic cancer. British journal of cancer. 2012. March 13;106(6):1033–8. doi: 10.1038/bjc.2012.18. PubMed PMID: ; PubMed Central PMCID: PMCPMC3304403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Safran H, Miner T, Bahary N, et al. Lapatinib and gemcitabine for metastatic pancreatic cancer. A phase II study. American journal of clinical oncology. 2011. February;34(1):50–2. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 170.Maron R, Schechter B, Mancini M, et al. Inhibition of pancreatic carcinoma by homo- and heterocombinations of antibodies against EGF-receptor and its kin HER2/ErbB-2. Proc Natl Acad Sci U S A. 2013. September 17;110(38):15389–94. doi: 10.1073/pnas.1313857110. PubMed PMID: ; PubMed Central PMCID: PMCPMC3780883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Assenat E, Mineur L, Mollevi C, et al. Phase II study evaluating the association of gemcitabine, trastuzumab, and erlotinib as first-line treatment in patients with metastatic pancreatic adenocarcinoma (GATE 1). J Clin Oncol (Meeting Abstracts). 2015 January, 2015;33(suppl 3):abstr 379. [DOI] [PubMed] [Google Scholar]
  • 172.Assenat E, Azria D, Mollevi C, et al. Dual targeting of HER1/EGFR and HER2 with cetuximab and trastuzumab in patients with metastatic pancreatic cancer after gemcitabine failure: results of the “THERAPY”phase 1–2 trial. Oncotarget. 2015. May 20;6(14):12796–808. doi: 10.18632/oncotarget.3473. PubMed PMID: ; PubMed Central PMCID: PMCPMC4494975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Perez SA, Sotiropoulou PA, Sotiriadou NN, et al. HER-2/neu-derived peptide 884–899 is expressed by human breast, colorectal and pancreatic adenocarcinomas and is recognized by in-vitro-induced specific CD4(+) T cell clones. Cancer Immunol Immunother. 2002. January;50(11):615–24. doi: 10.1007/s002620100225. PubMed PMID: . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Sotiriadou R, Perez SA, Gritzapis AD, et al. Peptide HER2(776–788) represents a naturally processed broad MHC class II-restricted T cell epitope. Br J Cancer. 2001. November 16;85(10):1527–34. doi: 10.1054/bjoc.2001.2089. PubMed PMID: ; PubMed Central PMCID: PMCPMC2363935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Zhou X, Li J, Wang Z, et al. Cellular immunotherapy for carcinoma using genetically modified EGFR-specific T lymphocytes. Neoplasia (New York, NY). 2013. May; 15(5):544–53. PubMed PMID: ; PubMed Central PMCID: PMCPMC3638357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Cragg GM, Grothaus PG, Newman DJ. Impact of natural products on developing new anti-cancer agents. Chem Rev. 2009. July;109(7):3012–43. doi: 10.1021/cr900019j. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 177.Yue Q, Gao G, Zou G, et al. Natural Products as Adjunctive Treatment for Pancreatic Cancer: Recent Trends and Advancements. Biomed Res Int. 2017;2017:8412508. doi: 10.1155/2017/8412508. PubMed PMID: ; PubMed Central PMCID: PMCPMC5292383 authors declared no potential conflict of interests with respect to the research, authorship, and/or publication of this article. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Fairman D, Narwal R, Liang M, et al. Pharmacokinetics of MEDI4736, a fully human anti-PDL1 monoclonal antibody, in patients with advanced solid tumors. J Clin Oncol (Meeting Abstracts). 2014 May 20, 2014;32(15_suppl):2602. [Google Scholar]
  • 179.Lee FY, Borzilleri R, Fairchild CR, et al. BMS-247550: a novel epothilone analog with a mode of action similar to paclitaxel but possessing superior antitumor efficacy. Clin Cancer Res. 2001. May;7(5):1429–37. PubMed PMID: [PubMed] [Google Scholar]
  • 180.Phillips PA, Dudeja V, McCarroll JA, et al. Triptolide induces pancreatic cancer cell death via inhibition of heat shock protein 70. Cancer Res. 2007. October 1;67(19):9407–16. doi: 10.1158/0008-5472.CAN-07-1077. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 181.Chugh R, Sangwan V, Patil SP, et al. A preclinical evaluation of Minnelide as a therapeutic agent against pancreatic cancer. Sci Transl Med. 2012. October 17;4(156):156ra139. doi: 10.1126/scitranslmed.3004334. PubMed PMID: ; PubMed Central PMCID: PMCPMC3656604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. The New England journal of medicine. 2011;364(19):1817–1825. doi: 10.1056/NEJMoa1011923. [DOI] [PubMed] [Google Scholar]
  • 183.Wang-Gillam A, Li C-P, Bodoky G, et al. Updated overall survival analysis of NAPOLI-1: Phase III study of nanoliposomal irinotecan (nal-IRI, MM-398), with or without 5-fluorouracil and leucovorin (5-FU/LV), versus 5-FU/LV in metastatic pancreatic cancer (mPAC) previously treated with gemcitabine-based therapy. ASCO Meeting Abstracts. 2016 January 30, 2016;34(4_suppl):417. [Google Scholar]
  • 184.Ando Y, Saka H, Ando M, et al. Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res. 2000. December 15;60(24):6921–6. PubMed PMID: . [PubMed] [Google Scholar]
  • 185.Marcuello E, Altes A, Menoyo A, et al. UGT1A1 gene variations and irinotecan treatment in patients with metastatic colorectal cancer. Br J Cancer. 2004. August 16;91(4):678–82. doi: 10.1038/sj.bjc.6602042. PubMed PMID: ; PubMed Central PMCID: PMCPMC2364770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Saif MW, Lee Y, Kim R. Harnessing gemcitabine metabolism: a step towards personalized medicine for pancreatic cancer. Ther Adv Med Oncol. 2012. November;4(6):341–6. doi: 10.1177/1758834012453755. PubMed PMID: ; PubMed Central PMCID: PMCPMC3481558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Young JD, Yao SY, Sun L, et al. Human equilibrative nucleoside transporter (ENT) family of nucleoside and nucleobase transporter proteins. Xenobiotica. 2008. July;38(7–8):995–1021. doi: 10.1080/00498250801927427. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 188.Garcia-Manteiga J, Molina-Arcas M, Casado FJ, et al. Nucleoside transporter profiles in human pancreatic cancer cells: role of hCNT1 in 2’,2’-difluorodeoxycytidine-induced cytotoxicity. Clin Cancer Res. 2003. October 15;9(13):5000–8. PubMed PMID: . [PubMed] [Google Scholar]
  • 189.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. October 15;10(20):6956–61. doi: 10.1158/1078-0432.CCR-04-0224. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 190.Morinaga S, Nakamura Y, Watanabe T, et al. Immunohistochemical analysis of human equilibrative nucleoside transporter-1 (hENT1) predicts survival in resected pancreatic cancer patients treated with adjuvant gemcitabine monotherapy. Ann Surg Oncol. 2012. July;19 Suppl 3:S558–64. doi: 10.1245/s10434-011-2054-z. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 191.Marechal R, Bachet JB, Mackey JR, et al. Levels of gemcitabine transport and metabolism proteins predict survival times of patients treated with gemcitabine for pancreatic adenocarcinoma. Gastroenterology. 2012. September;143(3):664–74 e1–6. doi: 10.1053/j.gastro.2012.06.006. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 192.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. January;136(1):187–95. doi: 10.1053/j.gastro.2008.09.067. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 193.Greenhalf W, Ghaneh P, Neoptolemos JP, et al. Pancreatic cancer hENT1 expression and survival from gemcitabine in patients from the ESPAC-3 trial. J Natl Cancer Inst. 2014. January;106(1):djt347. doi: 10.1093/jnci/djt347. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 194.Giovannetti E, Del Tacca M, Mey V, et al. Transcription analysis of human equilibrative nucleoside transporter-1 predicts survival in pancreas cancer patients treated with gemcitabine. Cancer Res. 2006. April 01;66(7):3928–35. doi: 10.1158/0008-5472.CAN-05-4203. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 195.Sinn M, Riess H, Sinn BV, et al. Human equilibrative nucleoside transporter 1 expression analysed by the clone SP 120 rabbit antibody is not predictive in patients with pancreatic cancer treated with adjuvant gemcitabine - Results from the CONKO-001 trial. Eur J Cancer. 2015. August;51(12):1546–54. doi: 10.1016/j.ejca.2015.05.005. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 196.Svrcek M, Cros J, Marechal R, et al. Human equilibrative nucleoside transporter 1 testing in pancreatic ductal adenocarcinoma: a comparison between murine and rabbit antibodies. Histopathology. 2015. February;66(3):457–62. doi: 10.1111/his.12577. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 197.Kojima K, Vickers SM, Adsay NV, et al. Inactivation of Smad4 accelerates Kras(G12D)-mediated pancreatic neoplasia. Cancer Res. 2007. September 01;67(17):8121–30. doi: 10.1158/0008-5472.CAN-06-4167. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 198.Schwarte-Waldhoff I, Volpert OV, Bouck NP, et al. Smad4/DPC4-mediated tumor suppression through suppression of angiogenesis. Proc Natl Acad Sci U S A. 2000. August 15;97(17):9624–9. PubMed PMID: ; PubMed Central PMCID:PMCPMC16915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Iacobuzio-Donahue CA, Fu B, Yachida S, et al. DPC4 gene status of the primary carcinoma correlates with patterns of failure in patients with pancreatic cancer. J Clin Oncol. 2009. April 10;27(11):1806–13. doi: 10.1200/JCO.2008.17.7188. PubMed PMID: ; PubMed Central PMCID: PMCPMC2668706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Crane CH, Varadhachary GR, Yordy JS, et al. Phase II trial of cetuximab, gemcitabine, and oxaliplatin followed by chemoradiation with cetuximab for locally advanced (T4) pancreatic adenocarcinoma: correlation of Smad4(Dpc4) immunostaining with pattern of disease progression. J Clin Oncol. 2011. Aug 01;29(22):3037–43. doi: 10.1200/JCO.2010.33.8038. PubMed PMID: ; PubMed Central PMCID: PMCPMC3157965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Whittle MC, Izeradjene K, Rani PG, et al. RUNX3 Controls a Metastatic Switch in Pancreatic Ductal Adenocarcinoma. Cell. 2015. June 04;161(6):1345–60. doi: 10.1016/j.cell.2015.04.048. PubMed PMID: ; PubMed Central PMCID: PMCPMC4458240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 2009. October;10(10):704–14. doi: 10.1038/nrg2634. PubMed PMID: ; PubMed Central PMCID: PMCPMC3467096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006. April;6(4):259–69. doi: 10.1038/nrc1840. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 204.Lee EJ, Gusev Y, Jiang J, et al. Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer. 2007. March 01;120(5):1046–54. doi: 10.1002/ijc.22394. PubMed PMID: ; PubMed Central PMCID: PMCPMC2680248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Bloomston M, Frankel WL, Petrocca F, et al. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA. 2007. May 02;297(17):1901–8. doi: 10.1001/jama.297.17.1901. PubMed PMID: . [DOI] [PubMed] [Google Scholar]
  • 206.Volinia S, Calin GA, Liu CG, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A. 2006. February 14;103(7):2257–61. doi: 10.1073/pnas.0510565103. PubMed PMID: ; PubMed Central PMCID: PMCPMC1413718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Bryant JL, Britson J, Balko JM, et al. A microRNA gene expression signature predicts response to erlotinib in epithelial cancer cell lines and targets EMT. Br J Cancer. 2012. January 03;106(1):148–56. doi: 10.1038/bjc.2011.465. PubMed PMID: ; PubMed Central PMCID: PMCPMC3251842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Garajova I, Le Large TY, Giovannetti E, et al. The Role of MicroRNAs in Resistance to Current Pancreatic Cancer Treatment: Translational Studies and Basic Protocols for Extraction and PCR Analysis. Methods Mol Biol. 2016;1395:163–87. doi: 10.1007/978-1-4939-3347-1_10. PubMed PMID: [DOI] [PubMed] [Google Scholar]
  • 209.Hoadley KA, Yau C, Wolf DM, et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell. 2014. August 14;158(4):929–44. doi: 10.1016/j.cell.2014.06.049. PubMed PMID: ; PubMed Central PMCID: PMCPMC4152462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Collisson EA, Sadanandam A, Olson P, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nature Medicine. 2011;17(4):500–503. doi: 10.1038/nm.2344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.*.Moffitt RA, Marayati R, Flate EL, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nature Genetics. 2015;47(10):1168–1178. doi: 10.1038/ng.3398.This paper presents molecular subtyping of pancreas cancer based on the gene signatures of the cancer cells and their stromal cells. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES