Next Article in Journal
From Prostate to Bone: Key Players in Prostate Cancer Bone Metastasis
Next Article in Special Issue
Experimental Animal Models of Pancreatic Carcinogenesis for Prevention Studies and Their Relevance to Human Disease
Previous Article in Journal
Progress in Nanotechnology Based Approaches to Enhance the Potential of Chemopreventive Agents
Previous Article in Special Issue
Pancreatic Cancer Gene Therapy: From Molecular Targets to Delivery Systems
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabolic Disorder, Inflammation, and Deregulated Molecular Pathways Converging in Pancreatic Cancer Development: Implications for New Therapeutic Strategies

1
Department of Medical Oncology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa 920-0293, Japan
2
Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa 920-0293, Japan
3
Division of Translational & Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
*
Author to whom correspondence should be addressed.
Cancers 2011, 3(1), 446-460; https://doi.org/10.3390/cancers3010446
Submission received: 17 December 2010 / Revised: 18 January 2011 / Accepted: 21 January 2011 / Published: 24 January 2011
(This article belongs to the Special Issue Pancreatic Cancer)

Abstract

: Pancreatic cancer develops and progresses through complex, cumulative biological processes involving metabolic disorder, local inflammation, and deregulated molecular pathways. The resulting tumor aggressiveness hampers surgical intervention and renders pancreatic cancer resistant to standard chemotherapy and radiation therapy. Based on these pathologic properties, several therapeutic strategies are being developed to reverse refractory pancreatic cancer. Here, we outline molecular targeting therapies, which are primarily directed against growth factor receptor-type tyrosine kinases deregulated in tumors, but have failed to improve the survival of pancreatic cancer patients. Glycogen synthase kinase-3β (GSK3β) is a member of a serine/threonine protein kinase family that plays a critical role in various cellular pathways. GSK3β has also emerged as a mediator of pathological states, including glucose intolerance, inflammation, and various cancers (e.g., pancreatic cancer). We review recent studies that demonstrate the anti-tumor effects of GSK3β inhibition alone or in combination with chemotherapy and radiation. GSK3β inhibition may exert indirect anti-tumor actions in pancreatic cancer by modulating metabolic disorder and inflammation.

Graphical Abstract

1. Introduction

Pancreatic cancer is a devastating disease and a major unresolved health problem due to its late clinical diagnosis and predisposition towards metastasis [1,2]. Pancreatic cancer is characterized by highly proliferative and invasive tumor cells [3]. Despite progress in approaches to treatment [4], such aggressive biological behavior thwarts early diagnosis and curative surgical intervention and renders tumors resistant to conventional chemotherapy, radiation therapy, and their combination [5-7], leading to a poor prognosis [8]. Therefore, understanding the detailed molecular and biological basis of pancreatic cancer pathogenesis facilitates advances in the diagnosis, treatment, and prevention of this disease.

Pancreatic cancer develops through a series of complex, cumulative biological processes involving metabolic disorder and chronic local inflammation in association with stromal changes and deregulated molecular pathways (Figure 1). Based on these pathologic properties, new therapeutic strategies are being developed to reverse the refractory stage of pancreatic cancer. Here, we review the multidimensional processes leading to pancreatic cancer development and progression, and discuss biology-based therapeutic alternatives to conventional cancer treatments. In addition, we highlight an emerging strategy for cancer treatment that targets glycogen synthase kinase 3β (GSK3β), focusing on the effect of its deregulation on pancreatic cancer.

2. Metabolic Disorder Coincides with Pancreatic Cancer

Although systemic metabolic disorders such as obesity and type II diabetes mellitus (DM) influence the risk of developing pancreatic cancer and clinical outcomes (reviewed in [9,10]), the abnormal metabolic profile of cancer cells dictates their survival, proliferation, and invasion, as well as susceptibility to chemotherapy and radiation [11,12]. Thus, metabolic disorder and altered tumor cell metabolism are potential targets for cancer treatment and (chemo) prevention [13,14].

2.1. Obesity, Glucose Intolerance, and Pancreatic Cancer

The association between obesity/DM and pancreatic cancer risk has long been controversial. However, recent reviews and meta-analyses of prospective observational studies have demonstrated that obesity, defined by an increased body mass index, is significantly associated with a risk of pancreatic cancer development [15,16]. DM is also a clinical manifestation of pancreatic cancer, and case-control and prospective studies have demonstrated an increased risk of pancreatic cancer in patients with long-term DM [17-19]. Obesity is associated with the early manifestations of pancreatic cancer and lower overall patient survival [20], although the influence of DM on pancreatic cancer progression or morbidity is not clear [10]. Possible mechanisms behind the association between obesity and worse clinical outcomes may include increased risk of DM, thrombosis, or other comorbidities; impaired immune response leading to aggressive tumor behavior; and poor response to conventional anticancer therapies [10]. The effect of obesity and DM on the development and progression of pancreatic cancer appears to be mediated by adipokines, reactive oxygen species (ROS), inflammatory cytokines, and insulin resistance, which result in activation of insulin-like growth factor-1 (IGF-1) and mammalian target of rapamycin (mTOR) pathways (reviewed in [10]).

A number of studies have reported the potential role of glucose-lowering therapies in reducing the risk of pancreatic cancer (reviewed in [21]). Metformin is a biguanide that is most frequently prescribed for diabetes [22]. Recently, a comprehensive review and meta-analysis of epidemiologic studies demonstrated an inverse correlation between the use of metformin and incidence of pancreatic cancer and overall survival of patients with diabetes [23]. Increasing evidence also suggests that metformin exerts a therapeutic effect against cancer [24,25]. Metformin decreases insulin resistance and indirectly reduces levels of insulin and IGF-1, which promote cancer cell proliferation [26]. Further, metformin activates the tumor suppressor pathway mediated by liver kinase B1 (LKB1) and 5′ AMP-activated protein kinase (AMPK), an important sensor of cellular energy status [27], thereby inactivating mTOR signaling [14]. IGF-1 receptor and G protein-coupled receptor signaling is implicated in the autocrine-paracrine stimulation of a variety of malignant tumors, including exocrine pancreatic cancer. Recent studies demonstrated that metformin-induced activation of AMPK disrupts the crosstalk between insulin/IGF-1 receptor and G protein-coupled receptor signaling pathways in pancreatic cancer cells and inhibits proliferation of these cells in xenograft models, suggesting this crosstalk as a target for treatment of pancreatic cancer by metformin [28].

2.2. Distinct Metabolic Properties of Cancer Cells

The fundamental metabolic characteristics of cancer cells include increased glucose uptake, aerobic glycolysis even under normoxic condition (Warburg effect) [29], and impaired oxidative phosphorylation in the tricarboxylic acid (TCA) cycle, which results in mitochondrial uncoupling [30]. These properties could explain the ability of cancer cells to survive, invade host tissues, and resist the induction of apoptosis by chemotherapeutic agents and ionizing radiation [30,31]. However, the glycolytic phenotype of cancer cells is a potential target for cancer diagnosis and treatment [32]. For example, enhanced glucose uptake by cancer cells can be used to visualize cancer by positron emission tomography (PET) using the radioisotope-labeled glucose analogue 2-[18F]-fluoro-2-deoxy-D-glucose (FDG). FDG-PET in combination with computed tomography (PET-CT) enables detection of metastatic lesions of most cancers with both sensitivity and specificity greater than 90% [33]. Pharmacologic agents targeting the glycolytic phenotype of cancer cells include 2-deoxy-D-glucose (2-DG) and dichloroacetate (DCA).

A glucose analogue, 2-DG, is the most attractive agent for targeting aberrant glucose metabolism in cancer cells [34]. 2-DG inhibits glucose transport by competing with glucose transporters and is subsequently phosphorylated by hexokinase to form 2-DG-6-phospate. Phosphorylated 2-DG is not further metabolized by inhibiting glucose-6-phosphate isomerase, thereby reducing the production of adenosine triphosphate (ATP) and nicotinamide adenine dinucleotide phosphate (NADPH) from glycolysis coupled with the pentose phosphate pathway. Thus, 2-DG exerts antitumor effects by starving cancer cells [34].

The association between the glycolytic phenotype (i.e., TCA cycle defects) and resistance to apoptosis is attributed to decreased mitochondrial hydrogen peroxide production and cytochrome C release [30,31]. Pyruvate dehydrogenase (PDH) plays a crucial role in triggering the TCA cycle by converting pyruvate to citric acid. PDH kinase 1 (PDK1), which phosphorylates and inactivates PDH, is frequently overactivated in cancer cells, resulting in an impaired TCA cycle and mitochondrial hyperpolarization. Thus, inhibiting PDK1 would re-activate PDH and restore mitochondrial membrane polarity, thereby facilitating cancer cell apoptosis in response to chemotherapeutic agents and radiation. DCA, an orally bio-available small molecule, is a well characterized PDK1 inhibitor. The ability of DCA to inhibit lactate production (by stimulating PDH and the TCA cycle) has been long used to treat lactic acidosis, which complicates inherited mitochondrial disorders [35,36]. A recent study demonstrated that DCA induces cancer cell apoptosis by selectively inhibiting PDK1 in cancer cells, leading to metabolic remodeling from glycolysis to glucose oxidation and normalization of mitochondrial function [37]. A recent clinical trial of oral DCA in children with congenital lactic acidosis reported that DCA was well tolerated and safe [36]. Thus, orally available DCA is a promising selective anticancer agent.

3. Inflammation and Stromal Reactions in Pancreatic Cancer

The well recognized link between chronic inflammation and tumor development in many organs [38] is consistent with the reported causative association and interaction between chronic pancreatitis and pancreatic cancer [39-41]. In addition to the etiologic role of inflammation in carcinogenesis, systemic and local inflammation are frequent manifestations of pancreatic cancer and have been implicated in tumor progression and clinical outcomes [39].

3.1. Chronic Pancreatitis and Pancreatic Cancer

Chronic pancreatitis is a risk factor for developing pancreatic cancer [39-41]. This association is supported by a recent meta-analysis of 22 well-performed epidemiologic studies [42]. The risk of developing pancreatic cancer for patients with hereditary pancreatitis is much higher than for patients with sporadic chronic pancreatitis [43-45]. The incidence of chronic pancreatitis in the general population is only about 5 to 10 per 100,000 persons a year, as estimated from hospitalization data. In particular, hereditary pancreatitis caused by germline mutations in the cationic trypsinogen gene [46] accounts for less than 1% of all chronic pancreatitis cases. Therefore, chronic pancreatitis does not constitute a main precursor of pancreatic cancer [9,42].

Despite this low incidence, both forms of chronic pancreatitis have provided substantial evidence for putative inflammatory mechanisms contributing to pancreatic cancer development and progression, including proinflammatory cytokines, nuclear factor-κB (NF-κB), cyclooxygenase-2 (COX-2), peroxisome proliferator-activated receptor-γ (PPARγ), nitric oxide (NO) synthesized by inducible NO synthase (iNOS), DNA damage caused by release of proteolytic enzymes and ROS, and somatic mutations in oncogenes (e.g., K-ras) and tumor suppressor genes (e.g., p53, p16, DPC4/Smad) [47-49]. Pancreatic cancer progression shares these molecular alterations, which are promising targets for early molecular diagnosis, treatment, and prevention of the disease [47-49].

Histopathologic findings of chronic pancreatitis include marked fibrosis, in which pancreatic stellate cells (PSCs) play a crucial role [50,51]. In the normal pancreas, quiescent PSCs produce vitamin A in the periacinar and interlobular space. In response to pancreatic inflammation, PSCs are activated and transformed to a myofibroblast-like phenotype; they proliferate, migrate and produce extracellular matrix components (e.g., collagens, laminin, fibronectin), matrix metalloproteinases, and tissue inhibitors of matrix metalloproteinases. This phenotypic change is induced by inflammatory cytokines (e.g., tumor necrosis factor, interleukin-1, and interleukin-6), growth factors such as transforming growth factor (TGF)-β1, TGF-α, platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF)-2; and ROS [52]. In addition to their roles in the pathogenesis of chronic pancreatitis and cancer, these factors also promote pancreatic cancer progression [50,51].

3.2. Cancer-Stromal Interaction and Tumor Microenvironment

Chronic inflammation, desmoplastic stromal reaction, and neovascularization associated with pancreatic cancer combine to produce a distinct tissue microenvironment where cancer cell proliferation and invasion are facilitated by cancer-stromal interactions [53,54]. Activated PSCs are primarily responsible for the desmoplastic reaction and tumor angiogenesis in response to various growth factors such as TGF-β, FGF, HGF, and IGF-1. Results from in vitro and in vivo studies suggest that cancer cells recruit PSCs to tumors, where PSCs promote cancer cell proliferation and facilitate their invasion and metastasis, and that FGF and PDGF mediate these interactions between the two cell types (reviewed in [55]).

Phenotypic changes also occur in cancer cells, as represented by epithelial-mesenchymal transition (EMT) at the interface between tumor and stroma, in which epithelial cells undergo morphologic changes characterized by a transition from epithelial to fibroblastic (mesenchymal cell) phenotypes. Most factors involved in pancreatic cancer-stromal interactions have the potential to induce EMT in cancer cells. This process involves loss of cell-to-cell adhesion and E-cadherin expression, actin cytoskeleton reorganization, and increased expression of mesenchymal molecules (e.g., vimentin, fibronectin, α-smooth muscle actin, N-cadherin). In this way, EMT facilitates the invasion and metastasis of cancer cells and renders them resistant to chemotherapy and radiation [56,57]. Accordingly, growth factors such as TGF-β and hepatocyte growth factor (HGF, or its receptor c-Met) that are involved in cancer-stromal interactions and EMT have been well studied in order to develop therapeutic strategies targeting these factors [53,54].

4. Targeting Molecular Pathways Deregulated in Pancreatic Cancer

Most cases of pancreatic cancer are resistant to conventional chemotherapy and radiation therapy [5-7]; therefore, new strategies are needed to enhance the antitumor effects of gemcitabine, which is the standard chemotherapeutic agent used to treat pancreatic cancer [58]. These new classes of biology-based treatment modalities include molecular target-directed therapies.

4.1. Deregulated Molecular Pathways Mediated by Receptor-Type Tyrosine Kinases

Molecular studies have investigated the complex genetic mechanisms of cancer, which involve multidirectional signal transduction pathways [3,59,60]. As shown in Figure 2, the major signal transduction pathways in pancreatic cancer pathogenesis and progression are RAS/mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR, and hedgehog pathways [3]. The receptor tyrosine kinase family and their ligands, which include epidermal growth factor (EGF) receptor (EGFR), vascular endothelial growth factor (VEGF) receptor (VEGFR), and PDGF receptor (PDGFR), are targets of therapy because they are overexpressed in many tumor types, including pancreatic cancer [61].

4.2. Pharmacologic Agents that Target Deregulated Kinases

Currently available agents that target these factors include anti-EGFR antibodies (cetuximab, panitumumab), small molecule EGFR inhibitors (gefitinib, erlotinib), an anti-VEGF antibody (bevacizumab), and a small molecule VEGFR inhibitor (axitinib). A number of phase III clinical trials have tested kinase inhibitors as monotherapy or in combination therapy with gemcitabine for pancreatic cancer, but other than the combination of erlotinib and gemcitabine [62], these approaches have produced few therapeutic benefits [63]. Characterization of new molecular targets is necessary in order to develop strategies that enhance the effect of gemcitabine and improve the survival rate. Recent studies have pursued potential kinases as targets for new anticancer agents [64] and evaluated agents targeting known kinases (e.g., EGFR, check-point kinase 1) to combine with gemcitabine in order to improve its antitumor effects [65].

5. GSK3β, an Emerging Therapeutic Target in Cancer

GSK3β has emerged as a critical factor that plays distinct pathologic roles in glucose intolerance, inflammation, and in various cancer types (e.g., pancreatic cancer). Here, we briefly review recent studies, including our own, that demonstrate the direct anticancer effects of GSK3β inhibition, alone or in combination with chemotherapy and radiation.

5.1. Outline of GSK3β and its Involvement in Chronic Progressive Diseases

GSK3β was first identified as a serine/threonine protein kinase that regulates glucose/glycogen metabolism under the control of insulin signaling. Unlike most protein kinases, GSK3β is active in normal cells, and its activity is controlled by subcellular localization, differential phosphorylation at serine 9 (S9) and tyrosine 216 (Y216) residues, and different binding partners. In addition to regulating its primary target, glycogen synthase, GSK3β is involved in other fundamental cellular pathways depending on its substrates and binding partners [66-68]. GSK3β is a potential therapeutic target for common chronic diseases including type 2 DM and Alzheimer's disease, given the causative associations with glucose intolerance, neurodegenerative disorders, and inflammation [69-71].

5.2. Pathologic Role of GSK3β in Various Cancer Types

Under physiologic conditions, GSK3β phosphorylates several transcription factors (e.g., c-Jun, c-Myc), cell cycle regulators (e.g., cyclin D1), and proto-oncoproteins (e.g., β-catenin), thereby triggering their degradation via the ubiquitin-proteasome system. GSK3β is, therefore, hypothesized to inhibit tumor development by interfering with oncogenic signaling (e.g., Wnt, hedgehog) [72]. However, there is little evidence that links GSK3β inactivation or loss of GSK3β expression with tumor development.

In the last five years, we demonstrated that deregulated expression, phosphorylation of S9 and Y216, and GSK3β activity are distinct features of gastrointestinal cancers including pancreatic cancers and glioblastoma, and that GSK3β sustains the survival and proliferation of these tumor cells. A pathologic role for GSK3β is supported by observations that inhibition of its activity reduced the survival and proliferation of different cancer cell types, predisposing them to apoptosis both in vitro and in tumor xenografts [73-76]. We also found that GSK3β inhibition in cancer cells was accompanied by restoration of p53 and Rb tumor suppressor pathways [75,76] and downregulation of human telomerase reverse transcriptase (hTERT), resulting in cell senescence [76]. This led us to propose GSK3β as a potential target for cancer treatment and to apply for domestic and international patents [77].

Simultaneously and following our studies on the antitumor effects of GSK3β inhibition, similar observations from other laboratories were reported for various cancer types with underlying mechanisms that included regulation by GSK3β inhibition of several pathways mediated by p53, Rb, p27Kip1, cyclin-dependent kinase (CDK), cyclin D1, c-Myc, and NF-κB [78]. Although the putative role of GSK3β in cancer is still debated [79,80], the overall results indicate that aberrant expression and activity of GSK3β is likely to be a common and fundamental characteristic of a broad spectrum of cancers (Figure 3) [78].

Based on the potential involvement of GSK3β in NF-κB-mediated cell survival [81,82], a number of studies demonstrated that GSK3β is involved in pancreatic cancer cell survival via the NF-κB pathway [83-85]. Few studies had focused on the role of GSK3β in the cellular response to chemotherapy until we reported that GSK3β inhibition sensitizes glioblastoma cells to chemotherapeutic agents (e.g., temozolomide, ACNU) and ionizing radiation [75]. However, a recent study failed to demonstrate that disrupting NF-κB activity by inhibiting GSK3β sensitizes PANC-1 pancreatic cancer cells to gemcitabine [85]. We previously found that GSK3β inhibition did not affect endogenous NF-κB transcriptional activity in tumor cells established from pancreatic cancers and glioblastoma [75,76]. In a preliminary study, we found that a small-molecule GSK3β inhibitor increased pancreatic cancer cell sensitivity to gemcitabine in cell culture and tumor xenografts when its dose and treatment protocol were optimized, and have identified the molecular mechanisms underlying the increased sensitivity [86]. Our findings indicate that GSK3β inhibition combined with chemotherapy is a novel and promising strategy to sensitize pancreatic cancer cells to gemcitabine.

5.3. Putative Antitumor Effects of GSK3β Inhibition via Modulation of Tumor Biology

Increasing evidence suggests that GSK3β participates in a wide range of physiological processes that determine cell fate, including cell motility, energy metabolism, and transcriptional control (reviewed in [78]). In addition to the pathologic roles of GSK3β in cancer cell survival and proliferation [73-78,83-85] as discussed above, GSK3β may influence critical biological properties of cancer cells, such as their dependence on glycolysis and invasive ability associated with EMT (Figure 3). Modulation of these biological properties by pharmacologic inhibition of GSK3β may sensitize pancreatic cancer cells to standard chemotherapy and radiation. Given the systemic pathology caused by aberrant GSK3β activity in glucose intolerance and chronic inflammation [71], GSK3β inhibition may decrease the risk of developing pancreatic cancer by improving these conditions. Accordingly, investigating the functions and pathologic roles of GSK3β should establish a firm molecular basis for future cancer treatments that target this kinase.

6. Perspectives

Here, we reviewed recent studies on the epidemiologic characteristics of pancreatic cancer and the molecular and biological mechanisms contributing to its development and progression, and highlight advances in alternatives to conventional treatments. Although molecular target-directed therapy is currently attracting considerable attention, especially for cancer refractory to standard chemotherapy and radiation, this therapy produces therapeutic effects that are still far from sufficient for most patients with advanced and recurrent pancreatic cancer. For many cancer patients, including those with pancreatic cancer, resistance to currently available therapeutics presents a major obstacle and is due to the aberrant metabolism of cancer cells, their microenvironment, their ability to invade and metastasize, and the acquisition of gene mutations. Multidisciplinary approaches directed to a more complete understanding of pancreatic cancer pathogenesis hold great promise in improving the outlook of this disease.

Figure 1. Molecular and biological pathways involved in the development and progression of pancreatic cancer and putative mechanisms underlying the anticancer effects of metformin, 2-deoxyglucose (2-DG), and dichloroacetate (DCA). The gray triangle in a box indicates a molecular target. Abbreviations: EMT, epithelial-to-mesenchymal transition; PSCs, pancreatic stellate cells; Warburg, Warburg effect.
Figure 1. Molecular and biological pathways involved in the development and progression of pancreatic cancer and putative mechanisms underlying the anticancer effects of metformin, 2-deoxyglucose (2-DG), and dichloroacetate (DCA). The gray triangle in a box indicates a molecular target. Abbreviations: EMT, epithelial-to-mesenchymal transition; PSCs, pancreatic stellate cells; Warburg, Warburg effect.
Cancers 03 00446f1 1024
Figure 2. Critical molecular pathways leading to the development and progression of pancreatic cancer. Abbreviations: ARRB2, arrestin β2 ; COS2, kinesin-related protein Costal 2; DUSP6, dual specificity phosphatase 6; EGF, epidermal growth factor; EGFR, EGF receptor; GRK2, G protein-coupled receptor kinase-2; IRS, insulin receptor substrate 1; M, cell membrane; MAPK, mitogen-activated protein kinase; MAP2K, MAP kinase kinases; mTOR, mammalian target of rapamycin; PI3K, phosphatidylinositol 3-kinase; PTCH, patched; PTEN, phosphatase and tensin homolog deleted in chromosome 10; Shh, sonic hedgehog; SMO, smoothened; SUFU, suppressor of fused; TSC, tuberous sclerosis complex; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor. The gray triangle in a box indicates a target for drug development.
Figure 2. Critical molecular pathways leading to the development and progression of pancreatic cancer. Abbreviations: ARRB2, arrestin β2 ; COS2, kinesin-related protein Costal 2; DUSP6, dual specificity phosphatase 6; EGF, epidermal growth factor; EGFR, EGF receptor; GRK2, G protein-coupled receptor kinase-2; IRS, insulin receptor substrate 1; M, cell membrane; MAPK, mitogen-activated protein kinase; MAP2K, MAP kinase kinases; mTOR, mammalian target of rapamycin; PI3K, phosphatidylinositol 3-kinase; PTCH, patched; PTEN, phosphatase and tensin homolog deleted in chromosome 10; Shh, sonic hedgehog; SMO, smoothened; SUFU, suppressor of fused; TSC, tuberous sclerosis complex; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor. The gray triangle in a box indicates a target for drug development.
Cancers 03 00446f2 1024
Figure 3. Systemic and local effects of aberrant GSK3β on risk factors (glucose intolerance and chronic inflammation) and progression of pancreatic cancer. Molecular mechanisms leading to the pathways indicated by the dotted arrows are not well characterized.
Figure 3. Systemic and local effects of aberrant GSK3β on risk factors (glucose intolerance and chronic inflammation) and progression of pancreatic cancer. Molecular mechanisms leading to the pathways indicated by the dotted arrows are not well characterized.
Cancers 03 00446f3 1024

Acknowledgments

This work was supported in part by Grants-in-Aids for Scientific Research from the Japanese Ministry of Education, Science, Sports, Technology and Culture (to Y.M., T.M.), Grant for Collaborative Research from Kanazawa Medical University (C2008-3), and by a Project Research Grant from the High-Tech Research Center of Kanazawa Medical University (H2010 -11).

References

  1. Li, D.; Xie, K.; Wolff, R.; Abbruzzese, J.L. Pancreatic cancer. Lancet 2004, 363, 1049–1057. [Google Scholar]
  2. Schneider, G.; Siveke, J.T.; Eckel, F.; Schmid, R.M. Pancreatic cancer: Basic and clinical aspects. Gastroenterology 2005, 128, 1606–1625. [Google Scholar]
  3. Bardeesy, N.; DePinho, R.A. Pancreatic cancer biology and genetics. Nat. Rev. Cancer 2002, 2, 897–909. [Google Scholar]
  4. Lockhart, A.C.; Rothenberg, M.L.; Berlin, J.D. Treatment for pancreatic cancer: Current therapy and continued progress. Gastroenterology 2005, 128, 1642–1654. [Google Scholar]
  5. Wray, C.J.; Ahmad, S.A.; Matthews, J.B.; Lowy, A.M. Surgery for pancreatic cancer: Recent controversies and current practice. Gastroenterology 2005, 128, 1626–1641. [Google Scholar]
  6. Ben-Josef, E.; Lawrence, T.S. Chemotherapy for unresectable pancreatic cancer. Int. J. Clin. Oncol. 2008, 13, 121–126. [Google Scholar]
  7. Okusaka, T.; Ito, Y.; Furuse, J.; Yamada, S.; Ishii, H.; Shibuya, K.; Ioka, T.; Shinchi, H. Current status of chemoradiotherapy for locally advanced pancreatic cancer in Japan. Int. J. Clin. Oncol. 2008, 13, 127–131. [Google Scholar]
  8. Jamel, A.; Siegel, R.; Ward, E.; Hao, Y.; Xu, J.; Thun, M.J. Cancer statistics, 2009. CA Cancer J. Clin. 2009, 59, 225–249. [Google Scholar]
  9. Raimondi, S.; Maisonneuve, P.; Lowenfels, A.B. Epidemiology of pancreatic cancer: An overview. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 699–708. [Google Scholar]
  10. Li, D.; Abbruzzese, J.L. New strategies in pancreatic cancer: Emerging epidemiologic and therapeutic concepts. Clin. Cancer Res. 2010, 16, 4313–4318. [Google Scholar]
  11. Smallbone, K.; Gatenby, R.A.; Gillies, R.J.; Maini, P.K.; Gavaghan, D.J. Metabolic changes during carcinogenesis: Potential impact on invasiveness. J. Theor. Biol. 2007, 244, 703–713. [Google Scholar]
  12. DeBerardinis, R.J.; Sayed, N.; Ditsworth, D.; Thompson, C.B. Brick by brick: Metabolism and tumor cell growth. Curr. Opin. Genet. Dev. 2008, 18, 54–61. [Google Scholar]
  13. Gatenby, R.A.; Gillies, R.J. Glycolysis in cancer: A potential target for therapy. Int. J. Biochem. Cell Biol. 2007, 39, 1358–1366. [Google Scholar]
  14. Engelman, J.A.; Cantley, L.C. Chemoprevention meets glucose control. Cancer Prev. Res. 2010, 3, 1049–1052. [Google Scholar]
  15. Larsson, S.C.; Orsini, N.; Wolk, A. Body mass index and pancreatic cancer risk: A meta-analysis of prospective studies. Int. J. Cancer 2007, 120, 1993–1998. [Google Scholar]
  16. Renehan, A.G.; Tyson, M.; Egger, M.; Heller, R.F.; Zwahlen, M. Body-mass index and incidence of cancer: A systemic review and meta-analysis of prospective observation studies. Lancet 2008, 371, 569–578. [Google Scholar]
  17. Huxley, R.; Ansary-Moghaddam, A.; Berrington de González, A.; Barzi, F.; Woodward, M. Type-II diabetes and pancreatic cancer: A meta-analysis of 36 studies. Br. J. Cancer 2005, 92, 2076–2083. [Google Scholar]
  18. Luo, J.; Iwasaki, M.; Inoue, M.; Sasazuki, S.; Otani, T.; Ye, W.; Tsugane, S.; JPHC Study Group. Body mass index, physical activity and the risk of pancreatic cancer in relation to smoking status and history of diabetes: A large-scale population-based cohort study in Japan—the JPHC study. Cancer Causes Contr. 2007, 18, 603–612. [Google Scholar]
  19. Li, D.; Tang, H.; Hassan, M.M.; Holly, E.A.; Bracci, P.M.; Silverman, D.T. Diabetes and risk of pancreatic cancer: A pooled analysis of three large case-control studies. Cancer Causes Contr. 2010. [Google Scholar] [CrossRef]
  20. Li, D.; Morris, J.S.; Liu, J.; Hassan, M.M.; Day, R.S.; Bondy, M.L.; Abbruzzese, J.L. Body mass index and risk, age of onset, and survival in patients with pancreatic cancer. JAMA 2009, 301, 2553–2562. [Google Scholar]
  21. Li, D.; Yeung, S.C.; Hassan, M.M.; Konopleva, M.; Abbruzzese, J.L. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology 2009, 137, 482–488. [Google Scholar]
  22. Bolen, S.; Feldman, L.; Vassy, J.; Wilson, L.; Yeh, H.C.; Marinopoulos, S.; Wiley, C.; Selvin, E.; Wilson, R.; Bass, E.B.; Brancati, F.L. Systematic review: Comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann. Intern. Med. 2007, 147, 386–399. [Google Scholar]
  23. DeCensi, A.; Puntoni, M.; Goodwin, P.; Cazzaniga, M.; Gennari, A.; Bonanni, B.; Gandini, S. Metformin and cancer risk in diabetic patients: A systemic review and meta-analysis. Cancer Prev. Res. 2010, 3, 1452–1461. [Google Scholar]
  24. Ben Sahra, I.; Le Marchand-Brustel, Y.; Tanti, J.-F.; Bost, F. Metformin in cancer therapy: A new perspective for an old antidiabetic drug? Mol. Cancer Ther. 2010, 9, 1092–1099. [Google Scholar]
  25. Pollak, M. Metformin and other biguanides in oncology: Advancing the research agenda. Cancer Prev. Res. 2010, 3, 1060–1065. [Google Scholar]
  26. Pollak, M. Insulin and insulin-like growth factor signalling in neoplasia. Nat. Rev. Cancer 2008, 8, 915–928. [Google Scholar]
  27. Shackelford, D.B.; Shaw, R.J. The LKB1-MAPK pathway: Metabolism and growth control in tumor suppression. Nat. Rev. Cancer 2009, 9, 563–575. [Google Scholar]
  28. Rozengurt, E.; Sinnett-Smith, J.; Kisfalvi, K. Crosstalk between insulin/insulin-like growth factor-1 receptors and G protein-coupled receptor signaling systems: A novel target for the antidiabetic drug metformin in pancreatic cancer. Clin. Cancer Res. 2010, 16, 2505–2511. [Google Scholar]
  29. Kim, J.; Dang, C.V. Cancer's molecular sweet tooth and the Warburg effect. Cancer Res. 2006, 66, 8927–8930. [Google Scholar]
  30. Samudio, I.; Fiegl, M.; Andreeff, M. Mitochondrial uncoupling and the Warburg effect: Molecular basis for the reprogrammimg of cancer cell metabolism. Cancer Res. 2009, 69, 2163–2166. [Google Scholar]
  31. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2010, 324, 1029–1033. [Google Scholar]
  32. Kroemer, G.; Pouyssegur, J. Tumor cell metabolism: Cancer's Achilles' heel. Cancer Cell 2008, 13, 472–482. [Google Scholar]
  33. Mankoff, D.A.; Eary, J.F.; Link, J.M.; Muzi, M.; Rajendran, J.G.; Spence, A.M.; Krohn, K.A. Tumor-specific positron emission tomography imaging in patients: [18F] Fruolodeoxyglucose and beyond. Clin. Cancer Res. 2007, 13, 3460–3469. [Google Scholar]
  34. Dwarakanath, B.S.; Jain, V. Targeting glucose metabolism with 2-deoxy-D-glucose for improving cancer therapy. Future Oncol. 2009, 5, 581–585. [Google Scholar]
  35. Stacpoole, P.W.; Nagaraja, N.V.; Hutson, A.D. Efficacy of dichloroacetate as a lactate-lowering drug. J. Clin. Pharmacol. 2003, 43, 683–691. [Google Scholar]
  36. Stacpoole, P.W.; Kerr, D.S.; Barnes, C.; Bunch, S.T.; Carney, P.R.; Fennell, E.M.; Felitsyn, N.M.; Gilmore, R.L.; Greer, M.; Henderson, G.N.; Hutson, A.D.; Neiberger, R.E.; O'Brien, R.G.; Perkins, L.A.; Quisling, R.G.; Shroads, A.L.; Shuster, J.J.; Silverstein, J.H.; Theriaque, D.W.; Valenstein, E. Controlled clinical trial of dichloroacetate for treatment of congenital lactic acidosis in children. Pediatrics 2006, 117, 1519–1531. [Google Scholar]
  37. Bonnet, S.; Archer, S.L.; Allalunis-Turner, J.; Haromy, A.; Beaulieu, C.; Thompson, R.; Lee, C.T.; Lopaschuk, G.D.; Puttagunta, L.; Bonnet, S.; Harry, G.; Hashimoto, K.; Porter, C.J.; Andrade, M.A.; Thebaud, B.; Michelakis, E.D. A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell 2007, 11, 37–51. [Google Scholar]
  38. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar]
  39. McKay, C.J.; Glen, P.; McMillan, D.C. Chronic inflammation and pancreatic cancer. Best Pract. Res. Clin. Gastroenterol. 2008, 22, 65–73. [Google Scholar]
  40. Hart, A.R.; Kennedy, H.; Harvey, I. Pancreatic cancer: A review of the evidence on causation. Clin. Gastroenterol. Hepatol. 2008, 6, 275–282. [Google Scholar]
  41. Greer, J.B.; Whitcomb, D.C. Inflammation and pancreatic cancer: An evidence-based review. Curr. Opin. Pharmacol. 2009, 9, 411–418. [Google Scholar]
  42. Raimondi, S.; Lowenfels, A.B.; Morselli-Labate, A.M.; Maisonneuve, P.; Pezzilli, R. Pancreatic cancer in chronic pancreatitis: Aetiology, incidence, and early detection. Best Practice Res. Clin. Gastroenterol. 2010, 24, 349–358. [Google Scholar]
  43. Howes, N.; Lerch, M.M.; Greenhalf, W.; Stocken, D.D.; Ellis, I.; Simon, P.; Truninger, K.; Ammann, R.; Cavallini, G.; Charnley, R.M.; Uomo, G.; Delhaye, M.; Spicak, J.; Drumm, B.; Jansen, J.; Mountford, R.; Whitcomb, D.C.; Neoptolemos, J.P.; European Registry of Hereditary Pancreatitis and Pancreatic Cancer (EUROPAC). Clinical and genetic characteristics of hereditary pancreatitis in Europe. Clin. Gastroenterol. Hepatol. 2004, 2, 252–261. [Google Scholar]
  44. Rebours, V.; Boutron-Ruault, M.C.; Schnee, M.; Férec, C.; Maire, F.; Hammel, P.; Ruszniewski, P.; Lévy, P. Risk of pancreatic adenocarcinoma in patients with hereditary pancreatitis: A national exhaustive series. Am. J. Gastroenterol. 2008, 103, 111–119. [Google Scholar]
  45. Greer, J.B.; Lynch, H.T.; Brand, R.E. Herediatry pancreatic cancer: A clinical perspective. Best Pract. Res. Clin. Gastroenterol. 2009, 23, 159–170. [Google Scholar]
  46. Whitcomb, D.C.; Gorry, M.C.; Preston, R.A.; Furey, W.; Sossenheimer, M.J.; Ulrich, C.D.; Martin, S.P.; Gates, L.K., Jr.; Amann, S.T.; Toskes, P.P.; Liddle, R.; McGrath, K.; Uomo, G.; Post, J.C.; Ehrlich, G.D. Herediatry pancreatitis is caused by a mutation in the cationic trypsinogen gene. Nat. Genet. 1996, 14, 141–145. [Google Scholar]
  47. Farrow, B.; Evers, B.M. Inflammation and the development of pancreatic cancer. Surg. Oncol. 2002, 10, 153–169. [Google Scholar]
  48. Garcea, G.; Dennison, A.R.; Steward, W.P.; Berry, D.P. Role of inflammation in pancreatic carcinogenesis and the implications for future therapy. Pancreatology 2005, 5, 514–529. [Google Scholar]
  49. Uomo, I.; Miraglia, S.; Pastorello, M. Inflammation and pancreatic ductal adenocarcinoma: A potential scenario for novel drug targets. J. Pancreatol. (JOP) 2010, 11, 199–202. [Google Scholar]
  50. Algül, H.; Treiber, M.; Lesina, M.; Schmid, R.M. Mechanisms of disease: Chronic inflammation and cancer in the pancreas—a potential role for pancreatic stellate cells? Nat. Clin. Pract. Gastroenterol. Hepatol. 2007, 4, 454–462. [Google Scholar]
  51. Apte, M.; Pirola, R.; Wilson, J. New insights into alcoholic pancreatitis and pancreatic cancer. J. Gastroenterol. Hepatol. 2009, 24, S51–S56. [Google Scholar]
  52. Masamune, A.; Shimosegawa, T. Signal transduction in pancreatic stellate cells. J. Gastroenterol. 2009, 44, 249–260. [Google Scholar]
  53. Mahadevan, D.; Von Hoff, D.D. Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol. Cancer Ther. 2007, 6, 1186–1197. [Google Scholar]
  54. Joyce, A.C. Therapeutic targeting of the tumor microenvironment. Cancer Cell 2005, 7, 513–520. [Google Scholar]
  55. Vonlaufen, A.; Phillips, P.A.; Xu, Z.; Goldstein, D.; Pirola, R.C.; Wilson, J.S.; Apte, M.V. Pancreatic stellate cells and pancreatic cancer cells: An unholy alliance. Cancer Res. 2008, 68, 7707–7710. [Google Scholar]
  56. Yilmaz, M.; Christofori, G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009, 28, 15–33. [Google Scholar]
  57. Polyak, K.; Weiberg, R.A. Transition between epithelial and mesenchymal states: Acquisition of malignant and stem cell traits. Nat. Rev. Cancer 2009, 9, 265–273. [Google Scholar]
  58. Giovannetti, E.; Mey, V.; Nannizzi, S.; Pasqualetti, G.; Tacca, M.D.; Danesi, R. Pharmacogenetics of anticancer drug sensitivity in pancreatic cancer. Mol. Cancer Ther. 2006, 5, 1387–1395. [Google Scholar]
  59. Hruban, R.H.; Goggins, M.; Parsons, J.; Kern, S.E. Progression model for pancreatic cancer. Clin. Cancer Res. 2000, 6, 2969–2972. [Google Scholar]
  60. Furukawa, T.; Sunamura, M.; Horii, A. Moecular mechanisms of pancreatic carcinogenesis. Cancer Sci. 2006, 97, 1–7. [Google Scholar]
  61. Giroux, V.; Dagorn, J.C.; Iovanna, J.L. A review of kinases implicated in pancreatic cancer. Pancreatology 2009, 9, 738–754. [Google Scholar]
  62. Moore, M.J.; Goldstein, D.; Hamm, J.; Figer, A.; Hecht, J.R.; Gallinger, S.; Au, H.J.; Murawa, P.; Walde, D.; Wolff, R.A.; Campos, D.; Lim, R.; Ding, K.; Clark, G.; Voskoglou-Nomikos, T.; Ptasynski, M.; Parulekar, W. 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. J. Clin. Oncol. 2007, 25, 1960–1966. [Google Scholar]
  63. Furukawa, T. Molecular targeting therapy for pancreatic cancer: Current knowledge and perspectives from bench to bedside. J. Gastroenterol. 2008, 43, 905–911. [Google Scholar]
  64. Giroux, V.; Iovanna, J.; Dagorn, J.-C. Probing the human kinome for kinases involved in pancreatic cancer cell survival and gemcitabine resistance. FASEB J. 2006, 20, 1982–1991. [Google Scholar]
  65. Morgan, M.A.; Parsels, L.A.; Maybaum, J.; Lawrence, T.S. Improving gemcitabine-mediated radiosensitization using molecularly targeted therapy: A review. Clin. Cancer Res. 2008, 14, 6744–6750. [Google Scholar]
  66. Harwood, A.J. Regulation of GSK-3: A cellular multiprocessor. Cell 2001, 105, 821–824. [Google Scholar]
  67. Doble, B.W.; Woodgett, J.R. GSK-3: Tricks of the trade for a multi-tasking kinase. J. Cell. Sci. 2003, 116, 1175–1186. [Google Scholar]
  68. Jope, R.S.; Johnson, G.V. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem. Sci. 2004, 29, 95–102. [Google Scholar]
  69. Cohen, P.; Goedert, M. GSK3 inhibitors: Development and therapeutic potential. Nat. Rev. Drug Discov. 2004, 3, 479–487. [Google Scholar]
  70. Meijer, L.; Flajolet, M.; Greengard, P. Pharmacological inhibitors of glycogen synthase kinase 3. Trends Pharmacol. Sci. 2004, 25, 471–480. [Google Scholar]
  71. Jope, R.S.; Yuskaitis, C.J.; Beurel, E. Glycogen synthase kinase-3 (GSK3): Inflammation, diseases, and therapeutics. Neurochem. Res. 2007, 32, 577–595. [Google Scholar]
  72. Manoukian, A.S.; Woodgett, J.R. Role of GSK-3 in cancer: Regulation by Wnts and other signaling pathways. Adv. Cancer Res. 2002, 84, 203–229. [Google Scholar]
  73. Shakoori, A.; Ougolkov, A.; Yu, Z.W.; Zhang, B.; Modarressi, M.H.; Billadeau, D.D.; Mai, M.; Takahashi, Y.; Minamoto, T. Deregulated GSK3β activity in colorectal cancer: Its association with tumor cell survival and proliferation. Biochem. Biophys. Res. Commun. 2005, 334, 1365–1373. [Google Scholar]
  74. Shakoori, A.; Mai, W.; Miyashita, K.; Yasumoto, K.; Takahashi, Y.; Ooi, A.; Kawakami, K.; Minamoto, T. Inhibition of GSK-3β activity attenuates proliferation of human colon cancer cells in rodents. Cancer Sci. 2007, 98, 1388–1393. [Google Scholar]
  75. Miyashita, K.; Kawakami, K.; Nakada, M.; Mai, W.; Shakoori, A.; Fujisawa, H.; Hayashi, Y.; Hamada, J.; Minamoto, T. Potential therapeutic effect of glycogen synthase kinase 3β inhibition against human glioblastoma. Clin. Cancer Res. 2009, 15, 887–897. [Google Scholar]
  76. Mai, W.; Kawakami, K.; Shakoori, A.; Kyo, S.; Miyashita, K.; Yokoi, K.; Jin, M.; Shimasaki, T.; Motoo, Y.; Minamoto, T. Deregulated GSK3β sustains gastrointestinal cancer cells survival by modulating human telomerase reverse transcriptase and telomerase. Clin. Cancer Res. 2009, 15, 6810–6819. [Google Scholar]
  77. Minamoto, T. Suppression of cancer and method for evaluating anticancer agent based on the effect of inhibiting GSK3β.
  78. Miyashita, K.; Nakada, M.; Shakoori, A.; Ishigaki, Y.; Shimasaki, T.; Motoo, Y.; Kawakami, K.; Minamoto, T. An emerging strategy for cancer treatment targeting aberrant glycogen synthase kinase 3β. Anti-Cancer Agents Med. Chem. 2009, 9, 1114–1122. [Google Scholar]
  79. Patel, S.; Woodgett, J. Glycogen synthase kinase-3 and cancer: Good cop, bad cop? Cancer Cell 2008, 14, 351–353. [Google Scholar]
  80. Luo, J. Glycogen synthase kinase 3β (GSK3β) in tumorigenesis and cancer chemotherapy. Cancer Lett. 2009, 273, 194–200. [Google Scholar]
  81. Hoeflich, K.P.; Luo, J.; Rubie, E.A.; Tsao, M.S.; Jin, O.; Woodgett, J.R. Requirement for glycogen synthase kinase-3β in cell survival and NF-κB activation. Nature 2000, 406, 86–90. [Google Scholar]
  82. Schwabe, R.F.; Brenner, D.A. Role of glycogen synthase kinase-3 in TNF-α-induced NF-κB activation and apoptosis in hepatocytes. Am. J. Physiol. Gastrointest. Liver Physiol. 2002, 283, G204–G211. [Google Scholar]
  83. Ougolkov, A.V.; Fernandez-Zapico, M.E.; Savoy, D.N.; Urrutia, R.A.; Billadeau, D.D. Glycogen synthase kinase-3β participates in nuclear factor κB-mediated gene transcription and cell survival in pancreatic cancer cells. Cancer Res. 2005, 65, 2076–2081. [Google Scholar]
  84. Wilson, W., 3rd.; Baldwin, A.S. Maintenance of constitutive IκB kinase activity by glycogen synthase kinase-3α/β in pancreatic cancer. Cancer Res. 2008, 68, 8156–8163. [Google Scholar]
  85. Mamaghani, S.; Patel, S.; Hedley, D.W. Glycogen synthase kinase-3 inhibition disrupts nuclear factor-κB activity in pancreatic cancer, but fails to sensitize to gemcitabine chemotherapy. BMC Cancer 2009, 9, 132–132. [Google Scholar]
  86. Shimasaki, T.; Ishigaki, Y.; Nakamura, Y.; Takata, T.; Nakaya, N.; Nakajima, H.; Sato, I.; Zhao, X.; Kitano, A.; Kawakami, K.; Tanaka, T.; Takegami, T.; Tomosugi, N.; Minamoto, T.; Motoo, Y. Glycogen synthase kinase 3β inhibition sensitizes pancreatic cancer cells to gemcitabine. J. Gastroenterol. 2011. submitted. [Google Scholar]

Abbreviations

AMPK

5′-AMP-activated protein kinase

ATP

adenosine triphosphate

CDK

cyclin-dependent kinase

COX-2

cyclooxygenase-2

CT

computed tomography

DCA

dichloroacetate

2-DG

2-deoxy-D-glucose

DM

diabetes mellitus

DPC

deleted in pancreatic carcinoma

EGF

epidermal growth factor

EGFR

EGF receptor

EMT

epithelial-mesenchymal transition

FDG

[18F] fluoro-2-D-deoxyglucose

FGF

fibroblast growth factor

GSK3β

glycogen synthase kinase-3β

HGF

hepatocyte growth factor

hTERT

human telomerase reverse transcriptase

IGF-1

insulin-like growth factor-1

LKB1

liver kinase B1

MAPK

mitogen-activated protein kinase

mTOR

mammalian target of rapamycin

NADPH

nicotinamide adenine dinucleotide phosphate

NF-κB

nuclear factor-κB

NO

nitric oxide

iNOS

inducible NO synthase

PDGF

platelet-derived growth factor

PDGFR

PDGF receptor

PDH

pyruvate dehydrogenase

PDK1

PDH kinase 1

PET

positron emission tomography

PI3K

phosphatidylinositol 3′-kinase

PPARγ

peroxisome proliferator-activated receptor-γ

PSC

pancreatic stellate cell

ROS

reactive oxygen species

TCA

tricarboxylic acid

TGF

transforming growth factor

VEGF

vascular endothelial growth factor

VEGFR

VEGF receptor

Share and Cite

MDPI and ACS Style

Motoo, Y.; Shimasaki, T.; Ishigaki, Y.; Nakajima, H.; Kawakami, K.; Minamoto, T. Metabolic Disorder, Inflammation, and Deregulated Molecular Pathways Converging in Pancreatic Cancer Development: Implications for New Therapeutic Strategies. Cancers 2011, 3, 446-460. https://doi.org/10.3390/cancers3010446

AMA Style

Motoo Y, Shimasaki T, Ishigaki Y, Nakajima H, Kawakami K, Minamoto T. Metabolic Disorder, Inflammation, and Deregulated Molecular Pathways Converging in Pancreatic Cancer Development: Implications for New Therapeutic Strategies. Cancers. 2011; 3(1):446-460. https://doi.org/10.3390/cancers3010446

Chicago/Turabian Style

Motoo, Yoshiharu, Takeo Shimasaki, Yasuhito Ishigaki, Hideo Nakajima, Kazuyuki Kawakami, and Toshinari Minamoto. 2011. "Metabolic Disorder, Inflammation, and Deregulated Molecular Pathways Converging in Pancreatic Cancer Development: Implications for New Therapeutic Strategies" Cancers 3, no. 1: 446-460. https://doi.org/10.3390/cancers3010446

Article Metrics

Back to TopTop