Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2010 Jun 8.
Published in final edited form as: Br Med Bull. 2008 Aug 27;87:97–130. doi: 10.1093/bmb/ldn027

TARGETED THERAPIES FOR PANCREATIC CANCER

S A Danovi 1, H H Wong 1, N R Lemoine 1
PMCID: PMC2882225  EMSID: UKMS31047  PMID: 18753179

Abstract

Introduction

Pancreatic cancer is a devastating malignancy and a leading cause of cancer mortality. Furthermore, early diagnosis represents a serious hurdle for clinicians as symptoms are non-specific and usually manifest in advanced, treatment-resistant stages of the disease.

Sources of data

Here, we review the rationale and progress of targeted therapies currently under investigation.

Areas of agreement

At present, chemoradiation regimes are administered palliatively, and produce only marginal survival benefits, underscoring a desperate need for more effective treatment modalities.

Areas of controversy

Questions have been raised as to whether erlotinib, the only targeted therapy to attain a statistically significant increase in median survival, is cost-effective.

Growing points

The last decade of research has provided us with a wealth of information regarding the molecular nature of pancreatic cancer, leading to the identification of signalling pathways and their respective components which are critical for the maintenance of the malignant phenotype.

Areas timely for developing research

These proteins thus represent ideal targets for novel molecular therapies which embody an urgently needed novel treatment strategy.

Keywords: Pancreatic cancer, targeted therapy, novel therapy, clinical trials

Introduction

With mortality rates almost identical to incidence rates, a diagnosis of pancreatic cancer represents a particularly bleak prognosis for patients and their families. The factors that contribute towards this poor outcome include our current inability to diagnose pancreatic cancer at early stages due to the non-specific presenting symptoms, the inaccessibility of the pancreas and the early occurrence of metastasis. At present, surgical resection represents the only curative option for patients with pancreatic cancer, but at present, only 20% of patients present with early-stage operable tumours1. Patients with unresectable disease consequently make up the overwhelming majority of pancreatic cancer diagnoses and receive palliative chemotherapy and radiotherapy, which do not result in significant long-term survival2. The paucity of curative therapies has translated into an overall 5-year survival rate of less than 5%3, underscoring a desperate need for improved therapeutic options and diagnostic tools. Recent work has led to significant advances in the understanding of the genetic changes characteristic to pancreatic cancer in the hope of identifying molecules for the development of targeted therapies, although progress towards translating this knowledge into effective diagnostic tools and therapeutic agents has been slow. Here, we review the rationale for new therapies currently being developed.

Diagnosis and current treatment

At present, the anti-metabolite gemcitabine (2′,2′-difluorodeoxycytidine) is the standard treatment for patients with pancreatic cancer. Gemcitabine is a fluorinated analogue of deoxycytidine which, when incorporated into DNA, inhibits DNA synthesis and repair, culminating in apoptosis. A landmark study of 126 patients in 1997 demonstrated that gemcitabine produces significant benefits over its predecessor 5-fluorouracil (5-FU) in patients with advanced pancreatic cancer with respect to overall survival and disease-related symptoms4. Overall however, gemcitabine produces only marginal survival benefits and modest improvements in quality of life, representing a necessity for the identification of combination partners in order to improve the clinical outcome5, 6. To this end, several clinical trials using gemcitabine in combination with 5-FU, cisplatin, irinotecan, oxaliplatin and pemetrexed have been conducted, but none of these phase III trials have reported statistically significant improvements in survival over gemcitabine monotherapy1. Thus, novel therapeutic strategies are urgently needed. As locally advanced and metastatic pancreatic cancers are currently incurable with standard therapies, experimental therapy is an acceptable first-line strategy7. To this end, researchers are currently focusing their attentions on targeting the molecular defects characteristic to the malignancy itself, which represents a particularly promising avenue for future research (Table 1).

Table 1.

Molecular targets for pancreatic cancer therapy.

Frequency of mutation/expression (%)
Telomerase12 95
VEGF41 93
KRAS54 95
EGFR (ErbB1/HER1)90 69
ErbB2 (HER2/neu)107, 108 33-52
Gastrin precursors and gastrin121 23-91
CCK/gastrin receptor121 95
COX-2204-206 67-90
AKT2159, 160 10-20
Activated AKT/mTOR167 50-55
RelA (p65 subunit of NFκB)174 67
Sonic hedgehog189 70
Notch3207, 208 69-74
IGF-1R191 64
Mesothelin195, 196 90-100
MUC1197, 198 90

Abbreviations: VEGF, vascular endothelial growth factor; EGFR, epidermal growth factor receptor; CCK, cholecystokinin; COX-2, cyclooxygenase-2; mTOR, mammalian target of rapamycin; IGF-1R, insulin-like growth factor-1 receptor.

An Introduction to targeted therapies

The recent success of novel cancer therapeutics based on the specific inhibition of tumour-associated pathways has breathed new life into the area of cancer drug discovery. As these agents have been designed to inhibit specific pathways, they are commonly referred to as targeted agents. Conventional therapies also target cellular processes, but fail to distinguish tumour cells from their normal counterparts. This lack of discrimination subsequently manifests in unavoidable, often unacceptable side-effects which can be as devastating to the patient as the disease itself. In contrast, targeted therapies disable specific cellular pathways that are absolutely required for the maintenance of cancer. As a consequence, normal cells are left relatively unharmed, leading to a different and generally less severe repertoire of drug-related toxicities. A further requirement of targeted therapies is that their effects should be quantifiable, with a correlation between target inhibition and clinical outcome8, although this does not always occur due to the complexity of the living organisms.

Telomerase

Unlimited proliferation is an acquired trait intrinsic to the survival of cancer cells9, and mammalian cells have evolved sophisticated, cell-autonomous mechanisms to limit their replicative potential. In vitro, human cells undergo a certain number of population doublings (PD) before entering an irreversible post-mitotic state known as replicative senescence. Senescence represents the first of two checkpoints that serve as barriers against limitless proliferation and is dependent on the p53 and retinoblastoma tumour suppressor pathways and can thus be abrogated through either genetic mutation or the expression of viral oncoproteins such as the SV40 T-antigen. Such events allow cells to proliferate for a further 20-40 PD, although the population eventually enters replicative crisis, the second checkpoint, characterised by chromosome instability and a decline of the cell population as apoptosis exceeds proliferation. The rampant genomic instability observed during crisis can be attributed to the attrition of telomeres, which are specialised, tandemly repeated sequences (TTAGGG in humans), located at the ends of chromosomes. The inability of DNA polymerase to replicate the extreme 5′ termini of chromosomes results in progressive telomere shortening with each replicative cycle. Once telomeres become critically short, the cell loses the ability to protect the ends of its chromosomes and enters crisis. Thus, telomere erosion poses a finite lifespan on the cell and represents a serious hurdle for any cell with malignant aspirations. Tellingly however, almost all immortal cells express telomerase, a unique ribonucleoprotein which catalyses the addition of de novo telomeric sequences to the ends of chromosomes and are consequently able to bypass crisis. In contrast to healthy somatic cells, most malignant cells show detectable telomerase activity10. Furthermore, the inhibition of telomerase in tumour cells leads to rapid telomere shortening and apoptosis11. Taken together, these data provide a rationale for the development of anti-telomerase therapies for the treatment of cancer.

Several groups have demonstrated the up-regulation of telomerase in pancreatic malignancies but not in normal tissues or in benign disease12, 13. These in vitro studies thus argued in favour of the development of telomerase inhibition for pancreatic cancer treatment.

One strategy currently in development is the use of immune recognition technology to target and destroy telomerase-positive cells. hTERT, the telomerase reverse transcriptase catalytic subunit, is degraded into short peptides by tumour cells of different histological origins and types and is presented on the cell surface on major histocompatibility complex (MHC) molecules, leading to the activation of cytotoxic T-lymphocytes (CTLs), resulting in the elimination of tumour cells bearing the same peptide-MHC complex on their surfaces14. A significant concern regarding telomerase-based therapies is the possibility of adverse effects of telomere inhibition on the small number of tissue types that do express telomerase such as germline cells, haematopoietic cells and those in stem cell compartments of the skin and intestine15. Importantly, no CTL effect was observed towards CD34-positive haematopoietic cells16, 17, suggesting that the high expression of telomerase in malignant tissue compared to normal tissue does indeed provide the required therapeutic window for the immune system to distinguish between healthy and tumour cells.

The telomerase-derived 16-mer peptide GV1001 (Pharmexa A/S) has previously been demonstrated to elicit T-cell responses in non-small cell lung cancer patients without exhibiting cytotoxicity towards telomerase-positive bone marrow stem cells18. GV1001 was recently subjected to a phase I/II study to investigate its safety and tolerability in patients with non-resectable pancreatic adenocarcinoma19. A rapid and significant immune response was observed in 75% of the group receiving an intermediate dose of the peptide. There was no overall increase in immune response in the patient group receiving high peptide doses, possibly due to high local concentration of peptide leading to leakage into surrounding tissue, uptake and presentation of vaccine by non-professional antigen-presenting cells and the subsequent selection of T-cells with low-affinity T-cell receptors or anergy. Importantly, the vaccine was well-tolerated, with no serious adverse side effects or withdrawals due to treatment-related events. Furthermore, although survival was not the primary endpoint of the study, a significant correlation was observed between overall survival and immune response with a median survival of 8.6 months for patients receiving intermediate doses compared to 4.5 months for those receiving high or low doses. Accordingly, after 300 days, all surviving patients were those who had demonstrated an immune response to the peptide. These encouraging results suggest that GV1001 is immunogenic, safe and may offer a new therapeutic option for the treatment of pancreatic cancer either alone, or in conjunction with current conventional therapies. The large (>1000 patients) phase III TeloVac trial is currently ongoing. It aims to examine gemcitabine and capecitabine chemotherapy with concurrent or sequential GV1001 in patients with locally advanced and metastatic pancreatic cancers.

Inhibition of angiogenesis

Angiogenesis, the establishment of new vasculature, is essential during embryogenesis, wound healing and normal reproductive function in the female20. Inappropriate angiogenesis however contributes towards several diseases such as rheumatoid arthritis, age-related macular degeneration and cancer21. Research over the last three decades has provided a body of evidence demonstrating that the requirement for vasculature to provide oxygen, nutrients and to remove waste products at both primary and secondary sites obliges tumour cells to reside within 100-200μm of a capillary blood vessel or face apoptosis and necrosis9.

The transition between vascular quiescence and angiogenesis, known as the “angiogenic switch”, results from dominance of pro-angiogenic over anti-angiogenic signals and represents a significant obstacle for the growing tumour22. The angiogenic switch is activated in tumours by the over-production of angiogenic inducers, and by neutralising inhibitors23. Although the development of vessel walls is an extremely complex process, requiring a tightly-choreographed series of receptor-ligand interactions and the participation of several signalling pathways, the vascular endothelial growth factor (VEGF) is generally accepted to represent a critical rate-limiting step24 and has thus far become a focus in the quest for anti-angiogenic agents.

The VEGF family consists of six dimeric glycoproteins: VEGF-A, -B, -C, -D, -E and placental growth factor (PGF). VEGF-A is the most extensively studied member of the family and comprises five isoforms, generated by alternative splicing of 121, 145, 165, 189 and 206 amino acids.

VEGF activity is a potent mitogen for micro- and macro-vascular endothelial cells derived from arteries, veins and lymphatics20, and an effective activator of angiogenesis in vivo25-27. Additionally, VEGF is a regulator of vascular permeability, vasodilatation, endothelial migration and gene expression20. VEGF-A mediates these biological effects through binding to two VEGF receptors: vascular endothelial growth factor receptor (VEGFR)-1 and VEGFR-228-31.

Although pancreatic tumours are not grossly vascular, VEGF-A, VEGFR-1 and VEGFR-2 are often co-expressed implying an autocrine effect of VEGF on pancreatic cells expressing VEGF receptors, and paracrine effects on microvascular endothelial cells32-34. Studies in animal models have shown that perturbation of the VEGF signalling pathway inhibits pancreatic tumour growth and angiogenesis35-38. Finally, several studies have demonstrated a correlation between tumour VEGF-A levels, blood vessel density, tumour size and local metastasis of pancreatic adenocarcinoma39-41. Taken together, these studies indicate that VEGF-A promotes tumour growth and metastasis in pancreatic cancer by encouraging angiogenesis, and thus provide a rationale for the targeting of VEGF in pancreatic cancer.

The humanised monoclonal antibody bevacizumab (Avastin®, Genentech) targets all known isoforms of VEGF-A42. In preclinical studies, bevacizumab showed encouraging inhibition of tumour cell line growth in nude mice42, 43 and was deemed to be non-toxic when either used alone, or in combination with standard chemotherapy in early-phase clinical trials44, 45. Based on exciting phase III results46, bevacizumab, in combination with standard therapeutic regimes, has been approved by the United States Food and Drug Administration (FDA) as a first-line therapy for colorectal cancer, demonstrating that VEGF inhibition is a clinically effective strategy to treat at least one type of human cancer. Consequently, a non-randomised phase II study involving 52 patients to assess the efficacy of bevacizumab with gemcitibine as a first-line treatment for metastatic pancreatic cancer was initiated47. Gemcitabine was administered at 1000mg/m2 weekly for 3 out of 4 weeks and bevacizumab at 10mg/m2 on days 1 and 15 in a 28-day treatment cycle. Results of this study demonstrated that combination therapy resulted in a superior objective response rate, progression-free survival (5.4 months), median survival (8.8.months) as well as 1-year survival rate (29%) when compared to gemcitabine alone, warranting further clinical assessment. The combination of gemcitabine and bevacizumab was relatively well-tolerated, although 4 out of the 52 patients developed severe gastrointestinal complications including bowel perforations and oesophageal tears. In response to these encouraging data, a phase III trial involving 602 patients receiving either gemcitabine with placebo or bevacizumab was conducted48. Disappointingly however, preliminary results have indicated that the combination therapy has no significant clinical benefit over gemcitabine alone.

As VEGF inhibition can overcome radioresistance, possibly by attenuating the protection afforded to endothelial cells by VEGF following radiation49, 50, a phase I study was initiated to evaluate the safety of bevacizumab in combination with the oral fluoropyrimidine capecitabine-based chemoradiotherapy in patients with locally advanced pancreatic cancer7. As bevacizumab did not significantly increase the toxicity of chemoradiation therapy, further studies are justified and indeed trials are currently being conducted to investigate the benefit of bevacizumab in pancreatic cancer in a number of clinical contexts such as combination therapy with other chemotherapeutic agents and as an adjuvant therapy following surgical resection.

Inhibition of RAS

The 21kDa RAS family proteins were first identified as cellular homologues of oncoproteins encoded by the genomes of the Harvey and Kirsten murine sarcoma retroviruses. Four oncogenic RAS proteins have been described thus far, HRAS, NRAS, KRAS-4A and KRAS-4B, with the two KRAS proteins arising from alternative splicing of a single gene transcript. All family members exert their biological functions by assembling signalling complexes at the cell membrane that activate intracellular signalling cascades, altered transcriptional profiles and changes in diverse cellular processes such as migration, survival, proliferation, endocytosis, differentiation, senescence and more51. As RAS proteins are anchored to the cell membrane and toggle between an inactive guanosine diphosphate (GDP)-bound and an active guanosine triphosphate (GTP)-bound state, they act as molecular switches in the cell, transmitting signals from the cell membrane to the nucleus. In quiescent cells, RAS is usually found in the “OFF” state, until activation by upstream signalling components such as the binding of epidermal growth factor (EGF) to its cognate receptor tyrosine kinase, the epidermal growth factor receptor (EGFR). Such events initiate signalling casades resulting in a conformational change in RAS, the disassociation of GDP and the binding of GTP, thereby switching RAS into its “ON” state. In this state, RAS induces activation of distinct effector signalling cascades such as the RAF serine/threonine kinase, the phosphoinositide 3-kinase (PI3K) or the three RAL exchange proteins: RAL guanine nucleotide disassociation stimulator (RALGDS), RALGDS-like gene (RGL/RSB2) and RGL2/RLF. The signalling cascades elicited by RAS are transient and short-lived by virtue of RAS’ own intrinsic GTPase activity which is stimulated by cytosolic GTPase activating proteins (GAPs).

In tumours, RAS proteins are frequently found in the constitutively “ON” states. This is often due to activating mutations in RAS genes themselves which abolish GAP-induced GTPase activity and hence render RAS unable to switch back to the “OFF” state. De-regulated RAS signalling can lead to uncontrolled proliferation, enhanced migration and invasion, resistance to apoptosis and angiogenesis52, and it is thus unsurprising that RAS mutations, which almost always occur at codons 12, 13 and 61, are found in about 30% of all human tumours53. KRAS mutations are exceptionally frequent events in pancreatic cancers and predominantly occur at codon 1254. Inhibition of RAS activity is thus an appealing anti-cancer endeavour, and several approaches have been developed to achieve this goal.

Much attention has been given to the inhibition of post-translational events that are absolutely required for RAS activation and functional specificity. Newly synthesised RAS and RHO proteins are cytosolic, and require further processing in order to associate with the lipid membrane. A pivotal step in this pathway is the covalent addition of a hydrophobic 15-carbon farnesyl to the cysteine residue within the CAAX motif (C, cysteine; A, any aliphatic amino acid; X, typically methionine or serine) at the extreme C-terminus of the protein. This event, known as farnesylation, is mediated by farnesyltransferase (FT). The -AAX amino acids are cleaved off by RAS-converting enzyme I, and the farnesylated cysteine is carboxymethylated by isoprenylcysteine carboxy methyl transferase. Finally, HRAS, NRAS and KRAS-4A are palmitoylated by palmitoyltransferase, which facilitates the addition of two long-chain fatty-acid groups to their C-terminal cysteine residue and anchors the proteins to the cell membrane. KRAS-4B is not a substrate for palmitoylation but its interaction with the cell membrane is stabilised by the presence of a polybasic domain. The absolute requirement for these modifications was demonstrated by the fact that mutant RAS proteins which lack the isoprenylated cysteine could not associate with the membrane and could not transform fibroblasts55. These results prompted the search for farnesyltransferase inhibitors (FTIs) for use as anti-cancer agents. The first generation FTIs were largely peptides designed to mimic the CAAX motif and thus compete for binding to farnesyltransferase (CAAX peptidomimetics). Newer compounds were designed to compete with the farnesyl pyrophosphate or share characteristics of both farnesyl pyrophosphate and the CAAX motif (bisubstrate analogues). Given the necessity for RAS signalling in normal growth factor signalling and proliferation, it was somewhat surprising that FTIs displayed negligible toxicity and did not appear to interfere with normal cell proliferation in early in vitro studies56. Preclinical studies using these compounds produced very encouraging data and FTIs demonstrated impressive anti-proliferative effects against several different tumour cell lines including those of pancreatic origin57. Moreover, the FTI R115777 (tipifarnib/Zarnestra®, Johnson and Johnson Pharmaceutical Research and Development) demonstrated anti-proliferative effects in pancreatic cancer cell lines at clinically relevant concentrations, and also showed significant tumour growth inhibition and anti-angiogenic activity in a pancreatic xenograft model58. Critically, the CAAX analogue L-744,832 potently reversed mammary tumour development in transgenic mice expressing oncogenic H-Ras under the control of the MMTV promoter59. FTIs also showed promise in early-phase clinical trials. Prolonged administration of single-agent R115777 was deemed to be non-toxic and well-tolerated60-62. However, these promising results were not recapitulated in later-stage trials. R115777 was found to be ineffective as first-line monotherapy for advanced pancreatic cancer63. The phase II study, conducted by the South West Oncology Group, reported that administration of R115777 resulted in a median time to progression of 1.4 months and a median survival of 2.6 months. As these data were inferior to those obtained with gemcitabine, the study concluded that R115777 was not an active agent in pancreatic cancer.

A recent phase III study involving 688 patients also deemed R115777 ineffective when used in combination with gemcitabine for advanced pancreatic cancer64. Although the combination of R115777 and gemcitabine was well-tolerated, no statistically significant improvement in overall survival was observed when compared to gemcitabine monotherapy (median survival of 193 versus 182 days respectively).

The unimpressive clinical trial data of FTIs may in part be attributed to the fact that KRAS and NRAS proteins are alternatively prenylated by the addition of a 20-carbon geranylgeranyl isoprenoid moiety by geranylgeranyltransferase I (GGT)65. Given that the majority of RAS mutations in human cancers occur in the KRAS gene66, the lack of toxicity and clinical efficacy of FTIs is thus unsurprising. Subsequent preclinical studies of concomitant FT and GGT inhibition have demonstrated efficacy in pancreatic cancer cell lines, but revealed high levels of toxicity67. In spite of this, a phase I study of the dual FT and GGT inhibitor L-778,123 in combination with radiotherapy was conducted in patients with locally advanced pancreatic cancer68. Although the combination of radiation and L-778,123 displayed acceptable toxicity at low doses, further development of this compound has been halted due to adverse cardiac effects observed in other studies69, 70. Despite their debatable mode of action, and their largely disappointing results in clinical trials to date, FTIs have shown some early potential in the treatment of leukaemias71. If shown to be useful, at least in certain clinical settings, FTIs will not only benefit patients, but will also strengthen the premise that targeting signalling molecules represents an exciting strategy for future cancer drug development. Clearly, further work is needed to ascertain whether this will indeed be the case.

A further strategy to inhibit RAS function has been through the use of antisense technology. This method utilises antisense oligonucleotides which are typically 13-25 nucleotides long and have been designed to hybridise with the target mRNA. Subsequent gene silencing occurs when protein translation is blocked by RNAse H-mediated cleavage of the mRNA-oligonucleotide duplex72, 73.

Despite the predominance of KRAS mutations in pancreatic cancer, the selective targeting of HRAS has also been explored as a therapeutic option. ISIS-2503 is a 20-base antisense oligonucleotide that prevents translation initiation of HRAS mRNA. In vitro studies have shown ISIS-2503 to be a potent and selective inhibitor of HRAS74, which, in a phase I study, was deemed to be safe and non-toxic when combined with gemcitabine and used against advanced pancreatic cancer75. This was followed by a phase II study involving 48 patients with locally advanced or metastatic disease76. ISIS-2503 combined with gemcitabine produced a response rate of 10.4%, including one complete response, and median survival of 6.6 months. In agreement with the phase I data, the combination treatment was relatively well-tolerated with an acceptable toxicity profile. Given that these results are superior to those obtained by FTI inhibition, this antisense technology currently represents the RAS-targeting strategy with the most promise.

MEK inhibition

The mitogen-activated extracellular kinase (MEK) is a principal component of the RAS-RAF-MEK-extracellular signal-regulated kinase (ERK) which plays a core role in several cellular processes including proliferation, apoptosis, differentiation and angiogenesis77. The pathway is also known to be de-regulated in a number of cancers including those of the breast, lung, colon and pancreas78, and thus represents an appealing target for the development of novel therapies.

CI-1040 is a low-molecular weight inhibitor of MEK1/2 which demonstrated impressive anti-proliferative activity against pancreatic cancer xenografts79. Unsurprisingly, the efficacy of CI-1040 is dependent on constitutive activation of the MEK-ERK pathway, making pancreatic cancer an ideal clinical setting for application of CI-1040 due to the high frequency of RAS mutations80. In a phase I study of 77 patients with advanced malignancies including those of colorectal, lung, kidney, skin and pancreas, CI-1040 was well-tolerated with a satisfactory safety profile81. The most common treatment-associated toxicities were low-grade diarrhoea, acneiform rash, nausea and vomiting. Even more encouraging was the fact that one pancreatic cancer patient demonstrated a partial response, with improvements in pain, fatigue and anorexia. Testing of CI-1040 progressed to a phase II study of 67 patients with advanced colorectal, lung, breast or pancreatic cancer82. In this setting, CI-1040 failed to demonstrate sufficient anti-tumoral activity and development was thus discontinued.

Inhibition of RAF has also been investigated in the context of pancreatic cancer. The small molecule MEK inhibitor sorafenib (BAY 43-9006, Bayer) also inhibits VEGFR-2, VEGFR-3 and platelet-derived growth factor receptor-β83, and was tested in a phase II trial for the treatment of advanced solid tumours in combination with gemcitabine84. The combination of 400mg sorafenib bi-daily with 1000mg/m2 gemcitabine was found to be relatively well-tolerated and 13 pancreatic cancer patients (56.5%) showed evidence of disease stabilisation. However, a more recent phase II trial of 17 patients failed to show any objective response in patients with advanced pancreatic cancer85. A phase III trial is currently in progress.

EGFR inhibition

The epidermal growth factor receptor (EGFR) is a member of the ErbB family of transmembrane receptor tyrosine kinases which comprises four members: ErbB1/HER1/EGFR, ErbB2/HER2/neu, ErbB3/HER3 and ErbB4/HER4. Structurally, ErbB receptors consist of an extracellular ligand-binding domain, a transmembrane domain and an intracellular protein kinase domain containing a regulatory carboxyl terminal segment. Engagement of receptor with cognate ligands such as EGF, transforming growth factor-alpha (TGF-α), heparin-binding growth factor, amphiregulin, betacellulin or epiregulin results in homo- or hetero-dimerisation among the ErbB family, leading to activation of intrinsic protein kinase activity, followed by tyrosine trans-auto-phosphorylation, accompanied by the recruitment of intra-cellular protein substrates. Phosphorylated tyrosine residues on the receptor also create binding sites for various adaptor and docking proteins.

Receptor tyrosine kinases play pivotal roles in the regulation of a number of cellular processes such as survival, proliferation, migration, apoptosis, invasion and metastasis. The first indication of a role for the EGFR gene in cellular transformation came from the finding that EGFR is the cellular homologue of the avian erythroblastosis virus v-erbB oncogene86. Since this landmark discovery, several studies have shown that inappropriate EGFR signalling arising from gene amplification, receptor or ligand over-expression, activating mutations or inactivation of negative regulatory pathways, is a feature of several human tumour types including pancreatic cancer87-89. Co-expression of EGFR (found in 69% of pancreatic adenocarcinoma90) and its ligands is frequently observed in pancreatic cancer and functions as an autocrine loop, constitutively stimulating pancreatic cancer cell proliferation91. A correlation between co-expression of EGFR and TGF-α with increased tumour size, advanced clinical stage and poor prognosis has been reported, and thus may represent a molecular signature of a more aggressive disease92, 93. Studies have also suggested an association between EGFR expression and metastasis, particularly to the liver94. Moreover, EGFR and TGF-α have also been implicated in tumour angiogenesis by virtue of their ability to induce VEGF secretion95.

Taken together, these data suggest that attenuation of the EGFR pathway may not only block tumour cell proliferation, but may also inhibit both angiogenesis and metastasis. In the first preclinical studies of EGFR inhibition, a panel of monoclonal antibodies directed against the EGF-binding site on EGFR inhibited the growth of cancer cells expressing high levels of EGFR in both cell culture and xenograft models96-98. These data were the first to demonstrate a potent anti-proliferative effect of EGFR inhibition and paved the way for a serious effort to develop EGFR inhibitors for use as anti-cancer agents. To date, two main classes of EGFR inhibitors have been developed, the first being small molecule inhibitors which interfere with adenosine triphosphate (ATP) binding and the second being monoclonal antibodies which prevent association with ligands.

Cetuximab (IMC-C225/Erbitux®, ImClone Systems/Merck KGaA) is a chimeric human-mouse monoclonal antibody directed against EGFR. In May 2004, cetuximab received FDA approval for the treatment of irinotecan-resistant or - intolerant colorectal cancer. Cetuximab has been tested against pancreatic cancer in a number of preclinical settings, both as single-agent monotherapy and in combination with gemcitabine. Cetuximab potently inhibited EGFR auto-phosphorylation in human pancreatic cancer cell lines and orthotopic tumour models99. The observed anti-proliferative effect of cetuximab alone was modest (only 20%), but was enhanced upon addition of gemcitabine. In the same study of murine orthotopic pancreatic tumour models, cetuximab reduced tumour volume and liver metastasis either alone or with gemcitabine. Systemic administration of cetuximab also resulted in decreased VEGF and interleukin-8 (IL-8) expression, accompanied by increased apoptosis in endothelial cells and a concomitant decrease in microvessel density, suggesting that cetuximab was also exerting an anti-angiogenic effect. Other groups have reported similar findings100-102. These results prompted a phase II trial to evaluate the efficacy of cetuximab in combination with gemcitabine in locally advanced and recurrent pancreatic cancers103. Patients received weekly 1000mg/m2 gemictabine initially for 7 out of 8 weeks and 3 out of 4 weeks thereafter. Cetuximab was administered at a loading dose of 400mg/m2 followed by weekly infusions of 250mg/m2.

In keeping with the results of previous studies, cetuximab was well-tolerated with the most common grade 3 and 4 adverse events being neutropaenia, abdominal pain and asthenia. In agreement with previous data, patients also developed an acne-like rash, which was only severe in 12% of cases, and has been attributed to the inhibition of EGFR in the skin. Significantly, the median survival was 7.1 months and 31.7% of patients were alive after one year, suggesting that the combination of gemcitabine and cetuximab may be more effective than gemcitabine monotherapy. These encouraging results led the design of two further trials. The first, conducted by the Southwest Oncology Group, was a phase III trial to study cetuximab alone or in combination with gemcitabine as first-line treatment for advanced pancreatic cancer104. Disappointingly however, this study failed to show any significant benefit of cetuximab addition for response, progression-free survival and overall survival. The second study, conducted by the Eastern Cooperative Oncology Group, is currently ongoing and aims to investigate the combination of docetaxel and irinotecan with or without cetuximab.

Matuzumab (EMD72000, Merck KGaA) is also a humanised monoclonal antibody, which binds EGFR with high specificity and affinity. Preclinical trials using orthotopic tumour models showed that matuzumab administration resulted in significantly decreased tumour progression and angiogenesis105. A subsequent phase I trial was launched to assess the safety and benefit of using matuzumab in combination with gemcitabine as a first-line treatment for advanced pancreatic cancer. Results of this study demonstrated that the combination was relatively well-tolerated at the biologically active dose of 800mg matuzumab per week, with stable disease observed in 65% of patients106. Further clinical trials with this compound are being planned.

Trastuzumab (Herceptin®, Genentech) is a recombinant humanised antibody against ErbB2 (HER2/neu) and received FDA approval for breast cancer treatment in 1998. Encouraged by the success of trastuzumab in the treatment of breast cancer, researchers began to explore the benefit of using this compound for the treatment of other malignancies, including pancreatic cancer where ErbB2 is over-expressed in about 50% of cases107, 108. However, in a phase II clinical trial the combination of gemcitabine and trastuzumab was not significantly superior to gemcitabine alone109. A second trial to study trastuzumab in combination with chemoradiation has recently been completed and results are awaited.

Small molecule inhibitors of EGFR were developed on the basis of the observation that mutation of the ATP-binding site of EGFR potently attenuated its receptor tyrosine kinase activity110. These findings led to the large-scale screening of compound libraries for molecules able to compete for the ATP-binding pocket of the tyrosine kinase domain of EGFR. One such molecule is erlotinib (OSI-774/Tarceva®, Genentech) which received FDA approval in 2004 for the treatment of patients with advanced non-small cell lung cancer, and is being increasingly tested for use in other cancers including colorectal, ovarian and pancreatic cancers.

In a recently published, landmark phase III trial, 569 patients with advanced pancreatic cancer were randomised to receive gemcitabine (1000mg/m2/week for 7 out of 8 weeks) with placebo or 100mg erlotinib daily, although a small cohort of patient received a higher dose of 150mg111. The results of this study demonstrated that erlotinib and gemcitabine combination therapy resulted in a small but statistically significant increase in median survival (6.24 versus 5.91 months respectively) and 1-year overall survival (23 versus 17%). Response rates and quality of life analysis gave similar results for both patient groups. Importantly, the combination of gemcitabine and erlotinib was well-tolerated with diarrhoea and rash being the most commonly observed toxicities associated with the addition of erlotinib. Interestingly, patients with grade 2 or higher skin rash experienced statistically significant survival. This study was the first to show a statistically significant benefit of combination therapy over gemcitabine monotherapy, and consequently, erlotinib became the first targeted therapy approved by the FDA for use in pancreatic cancer in 2005. Recently however, Grubbs and colleagues112 have questioned whether this combination is in fact cost-effective. Although the increase in median survival gained by the addition of erlotinib to gemcitabine was statistically significant, it was modest (an increase of 0.4 months over gemcitabine alone), and is therefore not cost-effective even at the highest year-per life gained parameters. Thus, the overall clinical impact of erlotinib with gemcitabine therapy on the treatment of pancreatic cancer remains to be seen.

In addition, erlotinib has been investigated in combination with capecitabine in gemcitabine-refractory advanced pancreatic cancer113. Patients were administered 1000mg/m2 capecitabine twice daily for 14 out of 21 days, and 150mg erlotinib daily. The regimen was well-tolerated with rash and diarrhoea being the most frequently observed toxicities. Given that the study reported an overall response rate of 10% and a median survival of 6.5 months, it was concluded that this combination may represent an appropriate treatment for patients for whom gemcitabine is either ineffective or inappropriate, and further analysis is required to establish whether this is indeed the case.

Preclinical studies have revealed a role for erlotinib as a potent radiosensitising agent114. Thus, the feasibility of using the novel combination of erlotinib, gemcitabine, paclitaxel and concomitant radiotherapy followed by maintenance erlotinib for locally advanced pancreatic cancer was the subject of a phase I trial conducted by Iannitti and colleagues115. Here, the maximum tolerated dose of erlotinib during chemoradiation therapy was 50mg daily due to dose-limiting diarrhoea. Given that this trial reported an encouraging partial response rate of 46% and median survival of 14 months, this treatment regimen merits further investigation.

Gefitinib (ZD1839/Iressa,® AstraZeneca) is a low molecular weight EGFR inhibitor. It was the first tyrosine kinase inhibitor to receive FDA approval for the treatment of advanced lung cancer in 2003, and is currently under investigation as treatment for several other tumour types including pancreatic cancer. In early-phase clinical trials, gefitinib used in combination with capecitabine and radiation therapy resulted in significant toxicity even at the lowest administered doses116. A phase II trial of gefitinib and gemcitabine in patients with inoperable or metastatic pancreatic cancer has shown results similar to those of erlotinib with gemcitabine117. However, results from phase II trials of gefitinib and docetaxel as second-line therapy were unremarkable118, 119.

Gastrin inhibition

The peptide hormone gastrin is produced in large amounts by G cells of the gastric antrum, and in smaller amounts by the upper small intestine, colon and pancreas, and mediates the secretion of pancreatic juice120.

Expression of cholecystokinin (CCK)/gastrin receptor and the gastrin precursors progastrin, glycine-extended gastrin (Ggly) and amidated gastrin (Gamide) has been observed by immunohistochemistry in 95%, 91%, 55% and 23% of pancreatic tumours respectively121. Gastrin stimulates the proliferation of pancreatic carcinoma122 which, conversely can be blocked by the administration of antisense oligonucleotides to gastrin123. Further experimentation revealed that the over-expression of CCK-2 in transgenic mice increased pancreatic weight by 40% and resulted in a 15% rate of malignant transformation when these mice were crossed with mice expressing Gamide in pancreatic β-cells124. Together, these data provided the rationale for the development of anti-gastrin therapy to treat pancreatic cancer.

The gastrin receptor antagonist gastrazole (JB95008, James Black Foundation) was recently the subject of two trials involving pancreatic cancer. The first study recruited 18 patients and compared gastrazole with placebo125. Overall, gastrazole resulted in a significant improvement in median survival (7.9 versus 4.5 months) and 1-year survival (33 versus 11%), while displaying minimal toxicity. The second study compared gastrazole with 5-FU but failed to demonstrate meaningful difference between the two patient groups, although gastrazole was associated with fewer toxicities125.

An alternative strategy is the use of immunogens to stimulate the production of neutralising antibodies against the serum- and tumour-associated forms of gastrin-17. One such immunogen is G17DT (gastrimmune/Insegia®, Aphton Corporation) which is administered by intramuscular injection and was the subject of a phase II trial with 30 patients with advanced pancreatic cancer126. While the majority (82.4%) of patients mounted an antibody response to the immungen, G17DT resulted in severe adverse reaction in 3 patients, although interestingly these patients survived for longer than the median survival of the group (244 days). Furthermore, the median survival of responders (217 days) was significantly longer than that of non-responders (121 days) indicating that G17DT does in fact possess anti-tumoral properties. Nevertheless, a subsequent phase III trial of G17DT in combination with gemcitabine failed to improve overall survival or secondary endpoints (e.g. response rate, time to progression)127.

Proteinase inhibition

The aggressive nature of pancreatic cancer is partly attributed to its early and frequent invasion of local structures, neural invasion and metastasis to the liver and lymph nodes, all of which preclude surgical resection and contribute towards the lethality of the disease128-130. Metastasis, which is a multi-step process during which cells within a primary tumour disengage and migrate to secondary organs, can only occur once cells are endowed with the ability to escape the primary tumour, invade surrounding tissue, enter and exit blood vessels and extravasate and establish metastatic foci at secondary sites. During the metastatic process, cells require the ability to degrade their surrounding matrix9, 131, 132.

The matrix metalloproteinases (MMPs) are a family of zinc-dependent proteolytic enzymes that are involved in the degradation of the extracellular matrix (ECM). The family can broadly be sub-classified on the basis of substrate specificity: collagenases (MMP-1, -8 and -13), gelatinases (MMP-2 and -9), stromelysins (MMP-3, -7, -10) membrane types (MMP-14/MT-MMP1, MMP-15/MT-MMP2, MMP-16/MT-MMP3, MMP-17/MT-MMP4, MMP-24/MT-MMP5) and others (MMP-18, MMP-19, MMP-20/enamelysin). Based on this ability to destroy the ECM, MMPs were immediately implicated in tumour progression and a strong association between MMP expression patterns with tumour progression is now widely accepted133. Studies in pancreatic cancer have shown that MMP-2, -7, -8, -9 and -11 are over-expressed in malignant tissue, with high MMP-7 levels correlating with shortened survival134, 135. Of note, MMP-7 expression is also increased during metaplastic transition in a murine model of pancreatic acinar-to-ductal metaplasia136. Taken together, these observations supported the development of MMP inhibitors (MPIs) for use as therapeutic agents in pancreatic cancer.

Preclinical results for MPIs were encouraging. The MPI SC44463 proved to be effective in blocking metastasis in an early mouse model137. Moreover, in orthotopic murine models of pancreatic cancer, administration of the MPI batimastat (BB-94, British Biotech) reduced metastases and also significantly prolonged overall survival with or without gemcitabine therapy138, 139.

Unfortunately clinical trials with MPIs have not lived up to the initial high expectations. Of particular concern was the fact that one trial of the MPI tanomastat (BAY 12-9566, Bayer) in patients with advanced pancreatic cancer was terminated early as patients receiving tanomastat showed significantly poorer survival than those receiving placebo140. However, these negative effects on survival were not recapitulated in subsequent studies of another MPI, marimastat (BB2516, British Biotech), indicating that the adverse effects of tanomastat were probably mediated by an as yet unclear mechanism. Marimastat has in fact been the subject of several clinical trials in pancreatic cancer patients. Early-phase trials demonstrated that marimastat administration was associated with a decrease or stabilisation in pain, mobility and analgesia scores in 51% of patients and was relatively well-tolerated, with the major toxicity being musculoskeletal141. Based on these findings, two randomised phase III trials were conducted with the aim of comparing marimastat with gemcitabine as first-line therapy for patients with unresectable pancreatic cancer. A total of 414 patients received marimastat (5mg, 10mg or 25mg twice daily) or 1000mg/m2 gemcitabine142. No significant difference in 1-year survival was observed between patients receiving marimastat and those on the standard gemcitabine therapy. Furthermore, a subsequent study failed to show any significant clinical benefit of using marimastat in combination with gemcitabine over gemcitabine monotherapy143.

It has however been argued that the failure of marimastat and other MPIs to demonstrate clinical efficacy in advanced pancreatic cancer may be attributed to an inherent flaw in design of the clinical trials. Critics argue that trials of marimastat have included large numbers of patients with metastatic disease, yet the cytostatic nature of these agents suggests that MPIs decrease the rate of tumour progression, and this would be of maximum therapeutic benefit in early stages of the disease131. In agreement with this notion, it has been noted in pancreatic cancer patients that marimastat increased overall survival time in patients without overt metastatic disease compared to those with metastases at trial entry142, 143.

A further problem is the failure of phase II trials to define adequately the biologically active dose of MPIs, due to the lack of a suitable endpoint measurement. As MPIs are cytostatic and not cytotoxic agents, reduction in tumour size is unlikely and therefore an inappropriate parameter. Several studies have used changes in serum concentrations of the tumour antigen CA 19-9 as a measure of MPI efficacy, but has attracted criticisms, as alterations in biomarker levels do not always represent tumour regression144. Supporters of MPI therefore maintain that the true potential of these compounds can only be fairly assessed once these limitations have been addressed. Until then, the role of MMP inhibition in the clinic remains uncertain.

COX-2 inhibition

An association between inflammation and cancer has been demonstrated by several epidemiological studies although the molecular nature of this relationship is yet to be completely understood. It is known however that mediators of the inflammatory process such as growth factors, cytokines and chemokines can contribute towards the development and progression of tumours145.

The last decade of research has highlighted the importance of cyclooxygenases (COXs) in cancer. Two isoforms have been identified thus far; COX-1, which is required for the supply of prostaglandins to organs and COX-2, which is dramatically up-regulated following inflammation and exposure to cytokines, tumour promoters and growth factors. COX-2 plays a principal role in the metabolism of arachidonic acid to prostaglandins and thromboxanes, and thus mediates resistance to apoptosis, adhesion, motility, immunosuppression and angiogenesis146, 147. Several lines of evidence have implicated COX-2, but not COX-1 in pancreatic cancer including the observation that its over-expression at both mRNA and protein levels has been observed in both pancreatic cancer cell lines and primary tumours, with one study finding COX-2 protein expression in 90% of pancreatic adenocarcinoma cases148-150. Significantly, inhibition of COX-2 activity by non-steroidal anti-inflammatory drugs and COX-2-specific inhibitors such as celecoxib and rofecoxib all exert anti-proliferative effects on pancreatic cancer cell lines in vitro and in vivo149, 151, 152, and a recently published study showed that administration of the COX-2 inhibitor nimesulide delayed pancreatic intraepithelial neoplasia progression in a murine model of pancreatic cancer153. Taken together, these data suggested that COX-2 inhibition represents a rational strategy for the treatment of pancreatic cancer. In a phase I study of patients with locally advanced disease, celecoxib (Celebrex®, Pfizer) increased the efficacy of a gemcitabine-radiation regimen, but significantly augmented its toxicity154. Another study reported minimal toxicities, but failed to demonstrate any clinical benefit when combining celecoxib with 5-FU in gemcitabine-refractory disease155. Similarly, celecoxib failed to increase the sensitivity of tumours to the chemotherapy combination of cisplatin and gemcitabine156. However, other studies yielded more promising data. Preliminary results from a phase II trial using celecoxib and gemcitabine reported a median survival of 6.2 months, 3-month survival rate of 72% and an acceptable toxicity profile103. A second phase II trial involving 42 patients with locally advanced or metastatic disease found that the combination of gemcitabine and celecoxib (400mg bi-daily) produced an encouraging median survival rate of 9.1 months, with an overall clinical benefit response of 54.7%157. Overall, the treatment was well-tolerated, with renal toxicity, heartburn and anaemia (all grade 1) being the most common adverse events associated with celecoxib. Importantly however, no dose-limiting toxicities were observed. Celecoxib has also been tested in combination with gemcitabine and irinotecan, resulting in a median survival of 13 months, an 8-month median time to progression and a 1-year overall survival of 64%158. Additionally, 76% of patients reported an improvement on overall quality of life.

Taken together, these data suggest that this class of inhibitors does display biological activity against pancreatic cancer and thus warrants further clinical investigation.

Novel targets

Novel drugs against newly identified targets are slowly entering the clinical trial pipeline following exciting preclinical evaluation. Taking centre stage are inhibitors of the PI3K/AKT and NFκB pathways which are emerging as critical to chemoresistance in pancreatic cancer.

The serine/threonine kinase AKT is up-regulated through gene amplification in several tumour types including pancreatic cancer159, 160. De-regulation of the AKT pathway can also be achieved through inactivation of the lipid phosphatase PTEN (phosphatase and tensin homologue deleted on chromosome 10), which is also frequently observed in pancreatic cancer cell lines and tumours161. The requirement of the AKT pathway in pancreatic cancer is demonstrated by the observation that its inhibition abrogates pancreatic cancer cell proliferation, induces apoptosis and restores gemcitabine-mediated chemosensitivity in several, but not all pancreatic cancer cell lines162-165. Thus, AKT inhibition may represent an exciting therapeutic strategy, although no specific compounds have commenced clinical development thus far.

The mammalian target of rapamycin (mTOR) lies downstream of AKT and regulates cell growth by exerting effects on protein synthesis, metabolism, autophagy and ribosome biogenesis166. An investigation of surgically resected tumours showed detectable levels of activated mTOR and its downstream kinase S6K1 in 55% and 65% of samples respectively167, providing the rationale for mTOR inhibition as a therapeutic avenue for pancreatic cancer. In preclinical studies, the mTOR inhibitor CCI-779 (temsirolimus/Torisel®, Wyeth) suppressed proliferation and induced apoptosis in pancreatic cancer cell lines, and also enhanced gemcitabine-dependent apoptosis in xenograft models167, 168.

Studies have also shown that rapamycin (sirolimus/Rapamune®, Wyeth) exhibits potent anti-proliferative activity by blocking not only mTOR, but also the hypoxia-inducible factor 1α and VEGF169, 170, and was reported to exert significant anti-angiogenic and anti-proliferative effects on orthotopic tumours when used alone, and resulted in profound growth inhibition of tumour growth and local metastasis when combined with gemcitabine171. Similarly, inhibition of mTOR in pancreatic cancer cell lines with RAD001 (everolimus/Certican®, Novartis) suppressed proliferation, induced apoptosis and sensitised cells to gemcitabine172, 173. Based on these promising results, early-stage clinical trials of mTOR inhibitors for pancreatic cancer treatment are currently in progress and their results are eagerly anticipated.

Under normal conditions, the principal function of the NFκB transcription factor is to regulate immune and inflammatory responses, although it has become a key target for therapeutic intervention due to its role in promoting proliferation, survival and chemoresistance. De-regulated NFκB expression has been reported in pancreatic tumours and cancer cell lines, but has not been observed in immortalised, non-tumorigenic pancreatic epithelial cells174. Indeed, it has been proposed that the differential sensitivities of pancreatic cancer cell lines to gemcitabine may in fact be attributed to the level of NFκB activation, as NFκB inhibition restored chemosensitivity175, 176. At present however, no specific NFκB inhibitors have been developed, although the proteosome antagonist bortezomib (PS-341/Velcade®, Millennium Pharmaceuticals) blocks NFκB activity by preventing degradation of its regulator IκBα and has been granted FDA approval for its use in refractory multiple myeloma. In vitro and in vivo studies have demonstrated that bortezomib significantly inhibited tumour growth of pancreatic cancer xenografts in athymic nude mice and displayed impressive anti-proliferative effects on some, but not all, pancreatic cancer cell lines. It also acts as an efficient chemosensitiser177-179, although sensitivity does not necessarily correlate with the level of NFκB activation180. Nevertheless, bortezomib performed well in phase I trials in patients with advanced solid tumours when combined with gemcitabine181, displaying an acceptable toxicity profile and subsequently quickly progressed to a phase II study. Unexpectedly, this study failed to demonstrate superiority of bortezomib alone or in combination with gemcitabine compared to gemcitabine monotherapy in patients with advanced pancreatic cancer, and resulted in an unacceptable toxicity profile182. It has thus been concluded that this compound is not suitable for use in pancreatic cancer treatment.

Curcumin, a phytochemical derivative of the Curcuma longa plant is an inhibitor of several molecules involved in proliferation, but also blocks NFκB-dependent transcription and subsequent activation of target genes such as COX-2 and IL-8183. Administration of curcumin results in growth inhibition and apoptosis in pancreatic cancer cells in vitro, and can also potentiate gemcitabine-dependent apoptosis in cancer cell lines and in xenograft models184-186. These data provided the rationale for a phase II trial in pancreatic cancer patients. Curcumin was relatively well-tolerated and in spite of low bioavailability, a down-regulation of NFκB, COX-2, and phosphorylated STAT3 was observed in patients’ peripheral blood187. It was thus concluded that curcumin is safe and biologically active, and further trials are currently ongoing.

Other novel targets that are the subjects of intense research include the Hedgehog and Notch signalling pathways, insulin-like growth factor-1 receptor (IGF-1R), TGF-β, mesothelin and MUC1 (mucin 1, cell surface associated). Physiologically, Hedgehog and Notch signalling is important in organ development, but abnormal expression and activation could lead to pancreatic tumourigenesis188, 189. Although still in the early developmental stages, treatments such as the Hedgehog pathway inhibitor cyclopamine has shown promising results in vivo190. IGF-1R has anti-apoptotic and growth-promoting effects, and is found in 64% of pancreatic cancer191. Its inhibition by the AMG-479 antibody was found to enhance the anti-tumour effects of gemcitabine and erlotinib in xenograft models192. TGF-β signalling is tumour suppressive in epithelial cells, but it can promote invasion and metastasis during the later stages of cancer progression193. Early data from an ongoing phase I/II study using AP 12009, an antisense oligonucleotide specific for TGF-β2 in patients with pancreatic carcinoma, malignant melanoma and colorectal carcinoma, has been encouraging194. The cell membrane proteins mesothelin and MUC1 are over-expressed in the majority of pancreatic cancer195-198. They have been studied as targets for immunotherapy197, 199, 200 but further research is still required.

Outlook and future perspectives

The failure of conventional chemotherapeutic regimes to produce any meaningful impact on survival in patients with pancreatic cancer highlights a desperate need for novel treatment strategies. New-generation, targeted drugs offered an obvious new avenue for hope, owing to their successes in the treatment of chronic myeloid leukaemia and cancers of the breast and colon. However, despite numerous clinical trials involving thousands of patients, these drugs have yet to revolutionise the treatment of advanced pancreatic cancer.

The advent of molecular medicine has transformed the ways in which cancer is approached both clinically and intellectually. Technologies such as high-throughput gene expression profiling using microarray analysis have allowed investigators to identify genes whose expression patterns correlate with drug sensitivity, disease staging and prognosis, as well as molecular targets against which novel drugs may be directed, offering clinicians a greater arsenal of treatment options. This, in the case of several malignancies, has translated successfully into the clinic. The routine genetic profiling of tumour samples for key indicator genes such as HER2/neu in breast cancer provides not only important prognostic information, but also guidance as to whether a patient will respond to a particular treatment regime. Thus, an individualised treatment plan can be tailored to suit the specific profile of the patient. Trastuzumab has transformed the bleak landscape of breast cancer for thousands of women and provides an excellent example of how the successful integration of targeted therapies and tumour profiling can be when applied to the appropriate clinical context. The development of both trastuzumab and bevacizumab also illustrates the importance of appropriate patient selection into clinical trials, as indiscriminate recruitment may potentially mask the efficacy of a particular drug. Tumours are heterogenous and complex, making it extremely unlikely that all tumours within a particular subtype will harbour the same activating mutation, or indeed that the tumour will be entirely dependent on the de-regulation of one particular pathway. Thus, while the pursuit of novel targeted agents is mandatory, this must be coupled with the development of new strategies to allow us to identify patients who will most benefit from these drugs.

Herein perhaps lies the reason behind the failure of targeted therapies to provide any meaningful impact on the management of pancreatic cancer. While our knowledge of the molecular characteristics of this disease has risen exponentially over the past decade, we have been less successful in applying this information in a clinical context. This may in part be due to the unique nature of pancreatic cancer where the majority of patients present with inoperable disease, and tissue samples obtained for diagnosis do not provide enough material for profiling techniques outlined above. This profound lack of tumour material has precluded the retrospective identification of biomarkers associated with clinical response to a particular drug, and the subsequent selection of appropriate patients into a clinical trial, as well as the limited identification of a gene-expression pattern present in individuals predisposed to the disease. The development of improved methods to readily obtain tumour material sufficient for genetic analyses must therefore be a high priority.

Future trials may also benefit from a shift away from conventional chemotherapy combinations and instead focus on combining targeted therapies. Some studies have been performed on the basis of this hypothesis, both combining EGFR and VEGF inhibitors. A phase II trial, conducted by the University of Chicago, aimed to evaluate gemcitabine and bevacizumab in combination with either erlotinib or cetuximab in 139 patients with advanced pancreatic cancer201. Results, however, were far from impressive, although there was a trend for improved overall survival in patients who developed grade 2 or higher skin rash after treatment with gemcitabine, bevacizumab and erlotinib. The AVITA/BO17706 trial, sponsored by Roche, is a randomised, double-blind, placebo-controlled phase III study examining the benefit of adding bevacizumab to gemcitabine and erlotinib in 607 patients with metastatic pancreatic cancer202. Preliminary results indicated that the addition of bevacizumab did not significantly prolong overall survival, but there was a trend towards improved survival. Significant improvement in progression-free survival was noted. A recent phase II study of bevacizumab and erlotinib in 26 patients with gemcitabine-refractory metastatic pancreatic cancer has demonstrated modest benefit203.

Despite these failures, our collective effort has not only created a substantial platform of knowledge from which future studies can spring, but has also moulded a robust clinical trial framework which will allow us to better address these issues in the future. It is hoped that the next decade will bring improved diagnostic tools and newer, more effective therapies to patients who will benefit the most. Together, this may finally allow us to improve the currently gloomy outlook for pancreatic cancer patients.

Table 2.

Phase III pancreatic cancer clinical trials with targeted and standard therapies.

Study Patient
number
Disease
stage
Treatment Mechanism MS
(months)
PFS
(months)
1-year
survival
(%)
CR +
PR
(%)
Stable
disease
(%)
CALGB 80303
(2007)48
602 Advanced Bevacizumab +
gemcitabine
Anti-VEGF antibody 5.7 4.8 13.1 40.7
Gemcitabine 6.0 4.3 11.3 35.7

Van Custem et
al. (2004)64
688 Advanced Tipifarnib
(R115777) +
gemcitabine
RAS
farnesyltransferase
inhibitor
6.3 3.7 27.0 6.0 53.0
Gemcitabine 6.0 3.6 24.0 8.0 52.0

SWOG S0205
(2007)209
766 Advanced Cetuximab +
gemcitabine
Anti-EGFR antibody 6.5 3.5 12.0
Gemcitabine 6.0 3.0 14.0

NCIC CTG
(2007)111
569 Advanced Erlotinib +
gemcitabine
EGFR tyrosine kinase
inhibitor
6.24* 3.75* 23.0* 8.6 48.9
Gemcitabine 5.91 3.55 17.0 8.0 41.2

Chau et al.
(2006)125
A – 18 Advanced Gastrazole
Placebo
Gastrin receptor
antagonist
7.9*
4.5
33.0*
11.0
B – 98 Gastrazole
5-FU
3.6
4.2
13.2
26.2
0.0
5.0
15.0
12.0

Shapiro et al.
(2005)127
383 Advanced G17DT +
gemcitabine
Immunogen – induces
antibody against
gastrin-17
5.9 3.9 11.0
Gemcitabine 6.7 3.9 13.0

NCIC CTG
(2003)140
277 Advanced Tanomastat MMP inhibitor 3.74 1.68 10.0 0.9 28.7
Gemcitabine 6.59* 3.5* 25.0 5.2 53.9

Bramhall et al.
(2001)142
414 Unresectable Marimastat
(25mg twice
daily)
MMP inhibitor 4.1 1.9 20.0 2.8
Gemcitabine 5.5* 3.8* 19.0 25.8

Bramhall et al.
(2002)143
239 Unresectable Marimastat +
gemcitabine
MMP inhibitor 5.4 3.0 18.0 11.0 50.0
Gemcitabine 5.4 3.1 17.0 16.0 56.0

AVITA/BO17706
(2008)202
607 Metastatic Bevacizumab +
erlotinib +
gemcitabine
Anti-VEGF antibody
and EGFR tyrosine
kinase inhibitor
7.1 4.6* 13.5
Erlotinib +
gemcitabine
6.0 3.6 8.6
*

Statistically significant

Abbreviations: MS, median survival; PFS, progression-free survival; PR + CR, partial and complete response; CALGB, Cancer and Leukaemia Group B; SWOG, Southwest Oncology Group; NCIC CTG, National Cancer Institute of Canada Clinical Trials Group; VEGF, vascular endothelial growth factor; EGFR, epithelial growth factor receptor; MMP, matrix metalloproteinase.

References

  • 1.Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet. 2004;363:1049–57. doi: 10.1016/S0140-6736(04)15841-8. [DOI] [PubMed] [Google Scholar]
  • 2.Bardeesy N, DePinho RA. Pancreatic cancer biology and genetics. Nat Rev Cancer. 2002;2:897–909. doi: 10.1038/nrc949. [DOI] [PubMed] [Google Scholar]
  • 3.Warshaw AL, Fernandez-del Castillo C. Pancreatic carcinoma. N Engl J Med. 1992;326:455–65. doi: 10.1056/NEJM199202133260706. [DOI] [PubMed] [Google Scholar]
  • 4.Burris HA, 3rd, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, Nelson R, Dorr FA, Stephens CD, Von Hoff DD. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–13. doi: 10.1200/JCO.1997.15.6.2403. [DOI] [PubMed] [Google Scholar]
  • 5.Kleespies A, Jauch KW, Bruns CJ. Tyrosine kinase inhibitors and gemcitabine: new treatment options in pancreatic cancer? Drug Resist Updat. 2006;9:1–18. doi: 10.1016/j.drup.2006.02.002. [DOI] [PubMed] [Google Scholar]
  • 6.Saif MW, Karapanagiotou L, Syrigos K. Genetic alterations in pancreatic cancer. World J Gastroenterol. 2007;13:4423–30. doi: 10.3748/wjg.v13.i33.4423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Crane CH, Ellis LM, Abbruzzese JL, Amos C, Xiong HQ, Ho L, Evans DB, Tamm EP, Ng C, Pisters PW, Charnsangavej C, Delclos ME, O’Reilly M, Lee JE, Wolff RA. Phase I trial evaluating the safety of bevacizumab with concurrent radiotherapy and capecitabine in locally advanced pancreatic cancer. J Clin Oncol. 2006;24:1145–51. doi: 10.1200/JCO.2005.03.6780. [DOI] [PubMed] [Google Scholar]
  • 8.Sledge GW., Jr. What is targeted therapy? J Clin Oncol. 2005;23:1614–5. doi: 10.1200/JCO.2005.01.016. [DOI] [PubMed] [Google Scholar]
  • 9.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  • 10.Shay JW, Bacchetti S. A survey of telomerase activity in human cancer. Eur J Cancer. 1997;33:787–91. doi: 10.1016/S0959-8049(97)00062-2. [DOI] [PubMed] [Google Scholar]
  • 11.Zhang X, Mar V, Zhou W, Harrington L, Robinson MO. Telomere shortening and apoptosis in telomerase-inhibited human tumor cells. Genes Dev. 1999;13:2388–99. doi: 10.1101/gad.13.18.2388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hiyama E, Kodama T, Shinbara K, Iwao T, Itoh M, Hiyama K, Shay JW, Matsuura Y, Yokoyama T. Telomerase activity is detected in pancreatic cancer but not in benign tumors. Cancer Res. 1997;57:326–31. [PubMed] [Google Scholar]
  • 13.Suehara N, Mizumoto K, Muta T, Tominaga Y, Shimura H, Kitajima S, Hamasaki N, Tsuneyoshi M, Tanaka M. Telomerase elevation in pancreatic ductal carcinoma compared to nonmalignant pathological states. Clin Cancer Res. 1997;3:993–8. [PubMed] [Google Scholar]
  • 14.Nguyen B, Elmore LW, Holt SE. Telomerase as a target for cancer immunotherapy. Cancer Biol Ther. 2003;2:131–6. doi: 10.4161/cbt.2.2.255. [DOI] [PubMed] [Google Scholar]
  • 15.Forsyth NR, Wright WE, Shay JW. Telomerase and differentiation in multicellular organisms: turn it off, turn it on, and turn it off again. Differentiation. 2002;69:188–97. doi: 10.1046/j.1432-0436.2002.690412.x. [DOI] [PubMed] [Google Scholar]
  • 16.Vonderheide RH, Hahn WC, Schultze JL, Nadler LM. The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity. 1999;10:673–9. doi: 10.1016/s1074-7613(00)80066-7. [DOI] [PubMed] [Google Scholar]
  • 17.Minev B, Hipp J, Firat H, Schmidt JD, Langlade-Demoyen P, Zanetti M. Cytotoxic T cell immunity against telomerase reverse transcriptase in humans. Proc Natl Acad Sci U S A. 2000;97:4796–801. doi: 10.1073/pnas.070560797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Brunsvig PF, Aamdal S, Gjertsen MK, Kvalheim G, Markowski-Grimsrud CJ, Sve I, Dyrhaug M, Trachsel S, Moller M, Eriksen JA, Gaudernack G. Telomerase peptide vaccination: a phase I/II study in patients with non-small cell lung cancer. Cancer Immunol Immunother. 2006;55:1553–64. doi: 10.1007/s00262-006-0145-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bernhardt SL, Gjertsen MK, Trachsel S, Moller M, Eriksen JA, Meo M, Buanes T, Gaudernack G. Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: A dose escalating phase I/II study. Br J Cancer. 2006;95:1474–82. doi: 10.1038/sj.bjc.6603437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ferrara N, Davis-Smyth T. The biology of vascular endothelial growth factor. Endocr Rev. 1997;18:4–25. doi: 10.1210/edrv.18.1.0287. [DOI] [PubMed] [Google Scholar]
  • 21.Roy H, Bhardwaj S, Yla-Herttuala S. Biology of vascular endothelial growth factors. FEBS Lett. 2006;580:2879–87. doi: 10.1016/j.febslet.2006.03.087. [DOI] [PubMed] [Google Scholar]
  • 22.Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 2003;3:401–10. doi: 10.1038/nrc1093. [DOI] [PubMed] [Google Scholar]
  • 23.Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86:353–64. doi: 10.1016/s0092-8674(00)80108-7. [DOI] [PubMed] [Google Scholar]
  • 24.Ferrara N. VEGF and the quest for tumour angiogenesis factors. Nat Rev Cancer. 2002;2:795–803. doi: 10.1038/nrc909. [DOI] [PubMed] [Google Scholar]
  • 25.Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science. 1989;246:1306–9. doi: 10.1126/science.2479986. [DOI] [PubMed] [Google Scholar]
  • 26.Plouet J, Schilling J, Gospodarowicz D. Isolation and characterization of a newly identified endothelial cell mitogen produced by AtT-20 cells. Embo J. 1989;8:3801–6. doi: 10.1002/j.1460-2075.1989.tb08557.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tolentino MJ, Miller JW, Gragoudas ES, Chatzistefanou K, Ferrara N, Adamis AP. Vascular endothelial growth factor is sufficient to produce iris neovascularization and neovascular glaucoma in a nonhuman primate. Arch Ophthalmol. 1996;114:964–70. doi: 10.1001/archopht.1996.01100140172010. [DOI] [PubMed] [Google Scholar]
  • 28.de Vries C, Escobedo JA, Ueno H, Houck K, Ferrara N, Williams LT. The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. Science. 1992;255:989–91. doi: 10.1126/science.1312256. [DOI] [PubMed] [Google Scholar]
  • 29.Terman BI, Dougher-Vermazen M, Carrion ME, Dimitrov D, Armellino DC, Gospodarowicz D, Bohlen P. Identification of the KDR tyrosine kinase as a receptor for vascular endothelial cell growth factor. Biochem Biophys Res Commun. 1992;187:1579–86. doi: 10.1016/0006-291x(92)90483-2. [DOI] [PubMed] [Google Scholar]
  • 30.Millauer B, Wizigmann-Voos S, Schnurch H, Martinez R, Moller NP, Risau W, Ullrich A. High affinity VEGF binding and developmental expression suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis. Cell. 1993;72:835–46. doi: 10.1016/0092-8674(93)90573-9. [DOI] [PubMed] [Google Scholar]
  • 31.Quinn TP, Peters KG, De Vries C, Ferrara N, Williams LT. Fetal liver kinase 1 is a receptor for vascular endothelial growth factor and is selectively expressed in vascular endothelium. Proc Natl Acad Sci U S A. 1993;90:7533–7. doi: 10.1073/pnas.90.16.7533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Itakura J, Ishiwata T, Shen B, Kornmann M, Korc M. Concomitant over-expression of vascular endothelial growth factor and its receptors in pancreatic cancer. Int J Cancer. 2000;85:27–34. doi: 10.1002/(sici)1097-0215(20000101)85:1<27::aid-ijc5>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
  • 33.von Marschall Z, Cramer T, Hocker M, Burde R, Plath T, Schirner M, Heidenreich R, Breier G, Riecken EO, Wiedenmann B, Rosewicz S. De novo expression of vascular endothelial growth factor in human pancreatic cancer: evidence for an autocrine mitogenic loop. Gastroenterology. 2000;119:1358–72. doi: 10.1053/gast.2000.19578. [DOI] [PubMed] [Google Scholar]
  • 34.Luo J, Guo P, Matsuda K, Truong N, Lee A, Chun C, Cheng SY, Korc M. Pancreatic cancer cell-derived vascular endothelial growth factor is biologically active in vitro and enhances tumorigenicity in vivo. Int J Cancer. 2001;92:361–9. doi: 10.1002/ijc.1202. [DOI] [PubMed] [Google Scholar]
  • 35.Tsuzuki Y, Carreira C Mouta, Bockhorn M, Xu L, Jain RK, Fukumura D. Pancreas microenvironment promotes VEGF expression and tumor growth: novel window models for pancreatic tumor angiogenesis and microcirculation. Lab Invest. 2001;81:1439–51. doi: 10.1038/labinvest.3780357. [DOI] [PubMed] [Google Scholar]
  • 36.Solorzano CC, Baker CH, Bruns CJ, Killion JJ, Ellis LM, Wood J, Fidler IJ. Inhibition of growth and metastasis of human pancreatic cancer growing in nude mice by PTK 787/ZK222584, an inhibitor of the vascular endothelial growth factor receptor tyrosine kinases. Cancer Biother Radiopharm. 2001;16:359–70. doi: 10.1089/108497801753354267. [DOI] [PubMed] [Google Scholar]
  • 37.Baker CH, Solorzano CC, Fidler IJ. Blockade of vascular endothelial growth factor receptor and epidermal growth factor receptor signaling for therapy of metastatic human pancreatic cancer. Cancer Res. 2002;62:1996–2003. [PubMed] [Google Scholar]
  • 38.Bockhorn M, Tsuzuki Y, Xu L, Frilling A, Broelsch CE, Fukumura D. Differential vascular and transcriptional responses to anti-vascular endothelial growth factor antibody in orthotopic human pancreatic cancer xenografts. Clin Cancer Res. 2003;9:4221–6. [PubMed] [Google Scholar]
  • 39.Itakura J, Ishiwata T, Friess H, Fujii H, Matsumoto Y, Buchler MW, Korc M. Enhanced expression of vascular endothelial growth factor in human pancreatic cancer correlates with local disease progression. Clin Cancer Res. 1997;3:1309–16. [PubMed] [Google Scholar]
  • 40.Ikeda N, Adachi M, Taki T, Huang C, Hashida H, Takabayashi A, Sho M, Nakajima Y, Kanehiro H, Hisanaga M, Nakano H, Miyake M. Prognostic significance of angiogenesis in human pancreatic cancer. Br J Cancer. 1999;79:1553–63. doi: 10.1038/sj.bjc.6690248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Seo Y, Baba H, Fukuda T, Takashima M, Sugimachi K. High expression of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer. 2000;88:2239–45. doi: 10.1002/(sici)1097-0142(20000515)88:10<2239::aid-cncr6>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
  • 42.Presta LG, Chen H, O’Connor SJ, Chisholm V, Meng YG, Krummen L, Winkler M, Ferrara N. Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res. 1997;57:4593–9. [PubMed] [Google Scholar]
  • 43.Kim KJ, Li B, Houck K, Winer J, Ferrara N. The vascular endothelial growth factor proteins: identification of biologically relevant regions by neutralizing monoclonal antibodies. Growth Factors. 1992;7:53–64. doi: 10.3109/08977199209023937. [DOI] [PubMed] [Google Scholar]
  • 44.Gordon MS, Margolin K, Talpaz M, Sledge GW, Jr., Holmgren E, Benjamin R, Stalter S, Shak S, Adelman D. Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J Clin Oncol. 2001;19:843–50. doi: 10.1200/JCO.2001.19.3.843. [DOI] [PubMed] [Google Scholar]
  • 45.Margolin K, Gordon MS, Holmgren E, Gaudreault J, Novotny W, Fyfe G, Adelman D, Stalter S, Breed J. Phase Ib trial of intravenous recombinant humanized monoclonal antibody to vascular endothelial growth factor in combination with chemotherapy in patients with advanced cancer: pharmacologic and long-term safety data. J Clin Oncol. 2001;19:851–6. doi: 10.1200/JCO.2001.19.3.851. [DOI] [PubMed] [Google Scholar]
  • 46.Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–42. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
  • 47.Kindler HL, Friberg G, Singh DA, Locker G, Nattam S, Kozloff M, Taber DA, Karrison T, Dachman A, Stadler WM, Vokes EE. Phase II trial of bevacizumab plus gemcitabine in patients with advanced pancreatic cancer. J Clin Oncol. 2005;23:8033–40. doi: 10.1200/JCO.2005.01.9661. [DOI] [PubMed] [Google Scholar]
  • 48.Kindler HL, Niedzwiecki D, Hollis D, Oraefo E, Schrag D, Hurwitz H, McLeod HL, Mulcahy MF, Schilsky RL, Goldberg RM, Cancer and Leukemia Group B A double-blind, placebo-controlled, randomized phase III trial of gemcitabine (G) plus bevacizumab (B) versus gemcitabine plus placebo (P) in patients (pts) with advanced pancreatic cancer (PC): A preliminary analysis of Cancer and Leukemia Group B (CALGB) J Clin Oncol (Meeting Abstracts) 2007;25:4508. [Google Scholar]
  • 49.Gorski DH, Beckett MA, Jaskowiak NT, Calvin DP, Mauceri HJ, Salloum RM, Seetharam S, Koons A, Hari DM, Kufe DW, Weichselbaum RR. Blockage of the vascular endothelial growth factor stress response increases the antitumor effects of ionizing radiation. Cancer Res. 1999;59:3374–8. [PubMed] [Google Scholar]
  • 50.Geng L, Donnelly E, McMahon G, Lin PC, Sierra-Rivera E, Oshinka H, Hallahan DE. Inhibition of vascular endothelial growth factor receptor signaling leads to reversal of tumor resistance to radiotherapy. Cancer Res. 2001;61:2413–9. [PubMed] [Google Scholar]
  • 51.Rajalingam K, Schreck R, Rapp UR, Albert S. Ras oncogenes and their downstream targets. Biochim Biophys Acta. 2007;1773:1177–95. doi: 10.1016/j.bbamcr.2007.01.012. [DOI] [PubMed] [Google Scholar]
  • 52.Shields JM, Pruitt K, McFall A, Shaub A, Der CJ. Understanding Ras: ‘it ain’t over ‘til it’s over’. Trends Cell Biol. 2000;10:147–54. doi: 10.1016/s0962-8924(00)01740-2. [DOI] [PubMed] [Google Scholar]
  • 53.Bos JL. ras oncogenes in human cancer: a review. Cancer Res. 1989;49:4682–9. [PubMed] [Google Scholar]
  • 54.Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell. 1988;53:549–54. doi: 10.1016/0092-8674(88)90571-5. [DOI] [PubMed] [Google Scholar]
  • 55.Willumsen BM, Norris K, Papageorge AG, Hubbert NL, Lowy DR. Harvey murine sarcoma virus p21 ras protein: biological and biochemical significance of the cysteine nearest the carboxy terminus. Embo J. 1984;3:2581–5. doi: 10.1002/j.1460-2075.1984.tb02177.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.James GL, Brown MS, Cobb MH, Goldstein JL. Benzodiazepine peptidomimetic BZA-5B interrupts the MAP kinase activation pathway in H-Ras-transformed Rat-1 cells, but not in untransformed cells. J Biol Chem. 1994;269:27705–14. [PubMed] [Google Scholar]
  • 57.Sepp-Lorenzino L, Ma Z, Rands E, Kohl NE, Gibbs JB, Oliff A, Rosen N. A peptidomimetic inhibitor of farnesyl:protein transferase blocks the anchorage-dependent and -independent growth of human tumor cell lines. Cancer Res. 1995;55:5302–9. [PubMed] [Google Scholar]
  • 58.End DW, Smets G, Todd AV, Applegate TL, Fuery CJ, Angibaud P, Venet M, Sanz G, Poignet H, Skrzat S, Devine A, Wouters W, Bowden C. Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer Res. 2001;61:131–7. [PubMed] [Google Scholar]
  • 59.Kohl NE, Omer CA, Conner MW, Anthony NJ, Davide JP, deSolms SJ, Giuliani EA, Gomez RP, Graham SL, Hamilton K, et al. Inhibition of farnesyltransferase induces regression of mammary and salivary carcinomas in ras transgenic mice. Nat Med. 1995;1:792–7. doi: 10.1038/nm0895-792. [DOI] [PubMed] [Google Scholar]
  • 60.Zujewski J, Horak ID, Bol CJ, Woestenborghs R, Bowden C, End DW, Piotrovsky VK, Chiao J, Belly RT, Todd A, Kopp WC, Kohler DR, Chow C, Noone M, Hakim FT, Larkin G, Gress RE, Nussenblatt RB, Kremer AB, Cowan KH. Phase I and pharmacokinetic study of farnesyl protein transferase inhibitor R115777 in advanced cancer. J Clin Oncol. 2000;18:927–41. doi: 10.1200/JCO.2000.18.4.927. [DOI] [PubMed] [Google Scholar]
  • 61.Crul M, de Klerk GJ, Swart M, van’t Veer LJ, de Jong D, Boerrigter L, Palmer PA, Bol CJ, Tan H, de Gast GC, Beijnen JH, Schellens JH. Phase I clinical and pharmacologic study of chronic oral administration of the farnesyl protein transferase inhibitor R115777 in advanced cancer. J Clin Oncol. 2002;20:2726–35. doi: 10.1200/JCO.2002.09.116. [DOI] [PubMed] [Google Scholar]
  • 62.Johnston SR, Hickish T, Ellis P, Houston S, Kelland L, Dowsett M, Salter J, Michiels B, Perez-Ruixo JJ, Palmer P, Howes A. Phase II study of the efficacy and tolerability of two dosing regimens of the farnesyl transferase inhibitor, R115777, in advanced breast cancer. J Clin Oncol. 2003;21:2492–9. doi: 10.1200/JCO.2003.10.064. [DOI] [PubMed] [Google Scholar]
  • 63.Macdonald JS, McCoy S, Whitehead RP, Iqbal S, Wade JL, 3rd, Giguere JK, Abbruzzese JL. A phase II study of farnesyl transferase inhibitor R115777 in pancreatic cancer: a Southwest oncology group (SWOG 9924) study. Invest New Drugs. 2005;23:485–7. doi: 10.1007/s10637-005-2908-y. [DOI] [PubMed] [Google Scholar]
  • 64.Van Cutsem E, van de Velde H, Karasek P, Oettle H, Vervenne WL, Szawlowski A, Schoffski P, Post S, Verslype C, Neumann H, Safran H, Humblet Y, Ruixo J Perez, Ma Y, Von Hoff D. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol. 2004;22:1430–8. doi: 10.1200/JCO.2004.10.112. [DOI] [PubMed] [Google Scholar]
  • 65.Walker K, Olson MF. Targeting Ras and Rho GTPases as opportunities for cancer therapeutics. Curr Opin Genet Dev. 2005;15:62–8. doi: 10.1016/j.gde.2004.11.001. [DOI] [PubMed] [Google Scholar]
  • 66.Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3:11–22. doi: 10.1038/nrc969. [DOI] [PubMed] [Google Scholar]
  • 67.Lobell RB, Omer CA, Abrams MT, Bhimnathwala HG, Brucker MJ, Buser CA, Davide JP, deSolms SJ, Dinsmore CJ, Ellis-Hutchings MS, Kral AM, Liu D, Lumma WC, Machotka SV, Rands E, Williams TM, Graham SL, Hartman GD, Oliff AI, Heimbrook DC, Kohl NE. Evaluation of farnesyl:protein transferase and geranylgeranyl:protein transferase inhibitor combinations in preclinical models. Cancer Res. 2001;61:8758–68. [PubMed] [Google Scholar]
  • 68.Martin NE, Brunner TB, Kiel KD, DeLaney TF, Regine WF, Mohiuddin M, Rosato EF, Haller DG, Stevenson JP, Smith D, Pramanik B, Tepper J, Tanaka WK, Morrison B, Deutsch P, Gupta AK, Muschel RJ, McKenna WG, Bernhard EJ, Hahn SM. A phase I trial of the dual farnesyltransferase and geranylgeranyltransferase inhibitor L-778,123 and radiotherapy for locally advanced pancreatic cancer. Clin Cancer Res. 2004;10:5447–54. doi: 10.1158/1078-0432.CCR-04-0248. [DOI] [PubMed] [Google Scholar]
  • 69.Britten CD, Rowinsky EK, Soignet S, Patnaik A, Yao SL, Deutsch P, Lee Y, Lobell RB, Mazina KE, McCreery H, Pezzuli S, Spriggs D. A phase I and pharmacological study of the farnesyl protein transferase inhibitor L-778,123 in patients with solid malignancies. Clin Cancer Res. 2001;7:3894–903. [PubMed] [Google Scholar]
  • 70.Lobell RB, Liu D, Buser CA, Davide JP, DePuy E, Hamilton K, Koblan KS, Lee Y, Mosser S, Motzel SL, Abbruzzese JL, Fuchs CS, Rowinsky EK, Rubin EH, Sharma S, Deutsch PJ, Mazina KE, Morrison BW, Wildonger L, Yao SL, Kohl NE. Preclinical and clinical pharmacodynamic assessment of L-778,123, a dual inhibitor of farnesyl:protein transferase and geranylgeranyl:protein transferase type-I. Mol Cancer Ther. 2002;1:747–58. [PubMed] [Google Scholar]
  • 71.Cortes J, Albitar M, Thomas D, Giles F, Kurzrock R, Thibault A, Rackoff W, Koller C, O’Brien S, Garcia-Manero G, Talpaz M, Kantarjian H. Efficacy of the farnesyl transferase inhibitor R115777 in chronic myeloid leukemia and other hematologic malignancies. Blood. 2003;101:1692–7. doi: 10.1182/blood-2002-07-1973. [DOI] [PubMed] [Google Scholar]
  • 72.Crooke ST. Molecular mechanisms of action of antisense drugs. Biochim Biophys Acta. 1999;1489:31–44. doi: 10.1016/s0167-4781(99)00148-7. [DOI] [PubMed] [Google Scholar]
  • 73.Tamm I, Dorken B, Hartmann G. Antisense therapy in oncology: new hope for an old idea? Lancet. 2001;358:489–97. doi: 10.1016/S0140-6736(01)05629-X. [DOI] [PubMed] [Google Scholar]
  • 74.Monia BP, Johnston JF, Ecker DJ, Zounes MA, Lima WF, Freier SM. Selective inhibition of mutant Ha-ras mRNA expression by antisense oligonucleotides. J Biol Chem. 1992;267:19954–62. [PubMed] [Google Scholar]
  • 75.Adjei AA, Dy GK, Erlichman C, Reid JM, Sloan JA, Pitot HC, Alberts SR, Goldberg RM, Hanson LJ, Atherton PJ, Watanabe T, Geary RS, Holmlund J, Dorr FA. A phase I trial of ISIS 2503, an antisense inhibitor of H-ras, in combination with gemcitabine in patients with advanced cancer. Clin Cancer Res. 2003;9:115–23. [PubMed] [Google Scholar]
  • 76.Alberts SR, Schroeder M, Erlichman C, Steen PD, Foster NR, Moore DF, Jr., Rowland KM, Jr., Nair S, Tschetter LK, Fitch TR. Gemcitabine and ISIS-2503 for patients with locally advanced or metastatic pancreatic adenocarcinoma: a North Central Cancer Treatment Group phase II trial. J Clin Oncol. 2004;22:4944–50. doi: 10.1200/JCO.2004.05.034. [DOI] [PubMed] [Google Scholar]
  • 77.McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, Chang F, Lehmann B, Terrian DM, Milella M, Tafuri A, Stivala F, Libra M, Basecke J, Evangelisti C, Martelli AM, Franklin RA. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta. 2007;1773:1263–84. doi: 10.1016/j.bbamcr.2006.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hoshino R, Chatani Y, Yamori T, Tsuruo T, Oka H, Yoshida O, Shimada Y, Ari-i S, Wada H, Fujimoto J, Kohno M. Constitutive activation of the 41-/43-kDa mitogen-activated protein kinase signaling pathway in human tumors. Oncogene. 1999;18:813–22. doi: 10.1038/sj.onc.1202367. [DOI] [PubMed] [Google Scholar]
  • 79.Allen LF, Sebolt-Leopold J, Meyer MB. CI-1040 (PD184352), a targeted signal transduction inhibitor of MEK (MAPKK) Semin Oncol. 2003;30:105–16. doi: 10.1053/j.seminoncol.2003.08.012. [DOI] [PubMed] [Google Scholar]
  • 80.Sebolt-Leopold JS, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer. 2004;4:937–47. doi: 10.1038/nrc1503. [DOI] [PubMed] [Google Scholar]
  • 81.Lorusso PM, Adjei AA, Varterasian M, Gadgeel S, Reid J, Mitchell DY, Hanson L, DeLuca P, Bruzek L, Piens J, Asbury P, Van Becelaere K, Herrera R, Sebolt-Leopold J, Meyer MB. Phase I and pharmacodynamic study of the oral MEK inhibitor CI-1040 in patients with advanced malignancies. J Clin Oncol. 2005;23:5281–93. doi: 10.1200/JCO.2005.14.415. [DOI] [PubMed] [Google Scholar]
  • 82.Rinehart J, Adjei AA, Lorusso PM, Waterhouse D, Hecht JR, Natale RB, Hamid O, Varterasian M, Asbury P, Kaldjian EP, Gulyas S, Mitchell DY, Herrera R, Sebolt-Leopold JS, Meyer MB. Multicenter phase II study of the oral MEK inhibitor, CI-1040, in patients with advanced non-small-cell lung, breast, colon, and pancreatic cancer. J Clin Oncol. 2004;22:4456–62. doi: 10.1200/JCO.2004.01.185. [DOI] [PubMed] [Google Scholar]
  • 83.Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, Chen C, Zhang X, Vincent P, McHugh M, Cao Y, Shujath J, Gawlak S, Eveleigh D, Rowley B, Liu L, Adnane L, Lynch M, Auclair D, Taylor I, Gedrich R, Voznesensky A, Riedl B, Post LE, Bollag G, Trail PA. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004;64:7099–109. doi: 10.1158/0008-5472.CAN-04-1443. [DOI] [PubMed] [Google Scholar]
  • 84.Siu LL, Awada A, Takimoto CH, Piccart M, Schwartz B, Giannaris T, Lathia C, Petrenciuc O, Moore MJ. Phase I trial of sorafenib and gemcitabine in advanced solid tumors with an expanded cohort in advanced pancreatic cancer. Clin Cancer Res. 2006;12:144–51. doi: 10.1158/1078-0432.CCR-05-1571. [DOI] [PubMed] [Google Scholar]
  • 85.Wallace JA, Locker G, Nattam S, Kasza K, Wade-Oliver K, Stadler WM, Vokes EE, Kindler HL. Sorafenib (S) plus gemcitabine (G) for advanced pancreatic cancer (PC): A phase II trial of the University of Chicago Phase II Consortium. J Clin Oncol (Meeting Abstracts) 2007;25:4608. [Google Scholar]
  • 86.Downward J, Yarden Y, Mayes E, Scrace G, Totty N, Stockwell P, Ullrich A, Schlessinger J, Waterfield MD. Close similarity of epidermal growth factor receptor and v-erb-B oncogene protein sequences. Nature. 1984;307:521–7. doi: 10.1038/307521a0. [DOI] [PubMed] [Google Scholar]
  • 87.Barton CM, Hall PA, Hughes CM, Gullick WJ, Lemoine NR. Transforming growth factor alpha and epidermal growth factor in human pancreatic cancer. J Pathol. 1991;163:111–6. doi: 10.1002/path.1711630206. [DOI] [PubMed] [Google Scholar]
  • 88.Lemoine NR, Hughes CM, Barton CM, Poulsom R, Jeffery RE, Kloppel G, Hall PA, Gullick WJ. The epidermal growth factor receptor in human pancreatic cancer. J Pathol. 1992;166:7–12. doi: 10.1002/path.1711660103. [DOI] [PubMed] [Google Scholar]
  • 89.Moscatello DK, Holgado-Madruga M, Godwin AK, Ramirez G, Gunn G, Zoltick PW, Biegel JA, Hayes RL, Wong AJ. Frequent expression of a mutant epidermal growth factor receptor in multiple human tumors. Cancer Res. 1995;55:5536–9. [PubMed] [Google Scholar]
  • 90.Bloomston M, Bhardwaj A, Ellison EC, Frankel WL. Epidermal growth factor receptor expression in pancreatic carcinoma using tissue microarray technique. Dig Surg. 2006;23:74–9. doi: 10.1159/000093497. [DOI] [PubMed] [Google Scholar]
  • 91.Funatomi H, Itakura J, Ishiwata T, Pastan I, Thompson SA, Johnson GR, Korc M. Amphiregulin antisense oligonucleotide inhibits the growth of T3M4 human pancreatic cancer cells and sensitizes the cells to EGF receptor-targeted therapy. Int J Cancer. 1997;72:512–7. doi: 10.1002/(sici)1097-0215(19970729)72:3<512::aid-ijc21>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
  • 92.Yamanaka Y, Friess H, Kobrin MS, Buchler M, Beger HG, Korc M. Coexpression of epidermal growth factor receptor and ligands in human pancreatic cancer is associated with enhanced tumor aggressiveness. Anticancer Res. 1993;13:565–9. [PubMed] [Google Scholar]
  • 93.Ueda S, Ogata S, Tsuda H, Kawarabayashi N, Kimura M, Sugiura Y, Tamai S, Matsubara O, Hatsuse K, Mochizuki H. The correlation between cytoplasmic overexpression of epidermal growth factor receptor and tumor aggressiveness: poor prognosis in patients with pancreatic ductal adenocarcinoma. Pancreas. 2004;29:e1–8. doi: 10.1097/00006676-200407000-00061. [DOI] [PubMed] [Google Scholar]
  • 94.Tobita K, Kijima H, Dowaki S, Kashiwagi H, Ohtani Y, Oida Y, Yamazaki H, Nakamura M, Ueyama Y, Tanaka M, Inokuchi S, Makuuchi H. Epidermal growth factor receptor expression in human pancreatic cancer: Significance for liver metastasis. Int J Mol Med. 2003;11:305–9. [PubMed] [Google Scholar]
  • 95.Goldman CK, Kim J, Wong WL, King V, Brock T, Gillespie GY. Epidermal growth factor stimulates vascular endothelial growth factor production by human malignant glioma cells: a model of glioblastoma multiforme pathophysiology. Mol Biol Cell. 1993;4:121–33. doi: 10.1091/mbc.4.1.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Sato JD, Kawamoto T, Le AD, Mendelsohn J, Polikoff J, Sato GH. Biological effects in vitro of monoclonal antibodies to human epidermal growth factor receptors. Mol Biol Med. 1983;1:511–29. [PubMed] [Google Scholar]
  • 97.Masui H, Kawamoto T, Sato JD, Wolf B, Sato G, Mendelsohn J. Growth inhibition of human tumor cells in athymic mice by anti-epidermal growth factor receptor monoclonal antibodies. Cancer Res. 1984;44:1002–7. [PubMed] [Google Scholar]
  • 98.Goldstein NI, Prewett M, Zuklys K, Rockwell P, Mendelsohn J. Biological efficacy of a chimeric antibody to the epidermal growth factor receptor in a human tumor xenograft model. Clin Cancer Res. 1995;1:1311–8. [PubMed] [Google Scholar]
  • 99.Bruns CJ, Harbison MT, Davis DW, Portera CA, Tsan R, McConkey DJ, Evans DB, Abbruzzese JL, Hicklin DJ, Radinsky R. Epidermal growth factor receptor blockade with C225 plus gemcitabine results in regression of human pancreatic carcinoma growing orthotopically in nude mice by antiangiogenic mechanisms. Clin Cancer Res. 2000;6:1936–48. [PubMed] [Google Scholar]
  • 100.Overholser JP, Prewett MC, Hooper AT, Waksal HW, Hicklin DJ. Epidermal growth factor receptor blockade by antibody IMC-C225 inhibits growth of a human pancreatic carcinoma xenograft in nude mice. Cancer. 2000;89:74–82. [PubMed] [Google Scholar]
  • 101.Buchsbaum DJ, Bonner JA, Grizzle WE, Stackhouse MA, Carpenter M, Hicklin DJ, Bohlen P, Raisch KP. Treatment of pancreatic cancer xenografts with Erbitux (IMC-C225) anti-EGFR antibody, gemcitabine, and radiation. Int J Radiat Oncol Biol Phys. 2002;54:1180–93. doi: 10.1016/s0360-3016(02)03788-4. [DOI] [PubMed] [Google Scholar]
  • 102.Arnoletti JP, Buchsbaum DJ, Huang ZQ, Hawkins AE, Khazaeli MB, Kraus MH, Vickers SM. Mechanisms of resistance to Erbitux (anti-epidermal growth factor receptor) combination therapy in pancreatic adenocarcinoma cells. J Gastrointest Surg. 2004;8:960–9. doi: 10.1016/j.gassur.2004.09.021. discussion 969-70. [DOI] [PubMed] [Google Scholar]
  • 103.Xiong HQ, Rosenberg A, LoBuglio A, Schmidt W, Wolff RA, Deutsch J, Needle M, Abbruzzese JL. Cetuximab, a monoclonal antibody targeting the epidermal growth factor receptor, in combination with gemcitabine for advanced pancreatic cancer: a multicenter phase II Trial. J Clin Oncol. 2004;22:2610–6. doi: 10.1200/JCO.2004.12.040. [DOI] [PubMed] [Google Scholar]
  • 104.Philip PA, Benedetti J, Fenoglio-Preiser C, Zalupski M, Lenz H, O’Reilly E, Wong R, Atkins J, Abruzzese J, Blanke C. Phase III study of gemcitabine [G] plus cetuximab [C] versus gemcitabine in patients [pts] with locally advanced or metastatic pancreatic adenocarcinoma [PC]: SWOG S0205 study. ASCO Meeting Abstracts. 2007;25:4509. [Google Scholar]
  • 105.Bangard C, Gossmann A, Papyan A, Tawadros S, Hellmich M, Bruns CJ. Magnetic resonance imaging in an orthotopic rat model: blockade of epidermal growth factor receptor with EMD72000 inhibits human pancreatic carcinoma growth. Int J Cancer. 2005;114:131–8. doi: 10.1002/ijc.20696. [DOI] [PubMed] [Google Scholar]
  • 106.Graeven U, Kremer B, Sudhoff T, Killing B, Rojo F, Weber D, Tillner J, Unal C, Schmiegel W. Phase I study of the humanised anti-EGFR monoclonal antibody matuzumab (EMD 72000) combined with gemcitabine in advanced pancreatic cancer. Br J Cancer. 2006;94:1293–9. doi: 10.1038/sj.bjc.6603083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lei S, Appert HE, Nakata B, Domenico DR, Kim K, Howard JM. Overexpression of HER2/neu oncogene in pancreatic cancer correlates with shortened survival. Int J Pancreatol. 1995;17:15–21. doi: 10.1007/BF02788354. [DOI] [PubMed] [Google Scholar]
  • 108.Yamanaka Y, Friess H, Kobrin MS, Buchler M, Kunz J, Beger HG, Korc M. Overexpression of HER2/neu oncogene in human pancreatic carcinoma. Hum Pathol. 1993;24:1127–34. doi: 10.1016/0046-8177(93)90194-l. [DOI] [PubMed] [Google Scholar]
  • 109.Safran H, Iannitti D, Ramanathan R, Schwartz JD, Steinhoff M, Nauman C, Hesketh P, Rathore R, Wolff R, Tantravahi U, Hughes TM, Maia C, Pasquariello T, Goldstein L, King T, Tsai JY, Kennedy T. Herceptin and gemcitabine for metastatic pancreatic cancers that overexpress HER-2/neu. Cancer Invest. 2004;22:706–12. doi: 10.1081/cnv-200032974. [DOI] [PubMed] [Google Scholar]
  • 110.Redemann N, Holzmann B, von Ruden T, Wagner EF, Schlessinger J, Ullrich A. Anti-oncogenic activity of signalling-defective epidermal growth factor receptor mutants. Mol Cell Biol. 1992;12:491–8. doi: 10.1128/mcb.12.2.491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, 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;25:1960–6. doi: 10.1200/JCO.2006.07.9525. [DOI] [PubMed] [Google Scholar]
  • 112.Grubbs SS, Grusenmeyer PA, Petrelli NJ, Gralla RJ. Is it cost-effective to add erlotinib to gemcitabine in advanced pancreatic cancer? ASCO Meeting Abstracts. 2006;24:6048. [Google Scholar]
  • 113.Kulke MH, Blaszkowsky LS, Ryan DP, Clark JW, Meyerhardt JA, Zhu AX, Enzinger PC, Kwak EL, Muzikansky A, Lawrence C, Fuchs CS. Capecitabine plus erlotinib in gemcitabine-refractory advanced pancreatic cancer. J Clin Oncol. 2007;25:4787–92. doi: 10.1200/JCO.2007.11.8521. [DOI] [PubMed] [Google Scholar]
  • 114.Chinnaiyan P, Huang S, Vallabhaneni G, Armstrong E, Varambally S, Tomlins SA, Chinnaiyan AM, Harari PM. Mechanisms of enhanced radiation response following epidermal growth factor receptor signaling inhibition by erlotinib (Tarceva) Cancer Res. 2005;65:3328–35. doi: 10.1158/0008-5472.CAN-04-3547. [DOI] [PubMed] [Google Scholar]
  • 115.Iannitti D, Dipetrillo T, Akerman P, Barnett JM, Maia-Acuna C, Cruff D, Miner T, Martel D, Cioffi W, Remis M, Kennedy T, Safran H. Erlotinib and chemoradiation followed by maintenance erlotinib for locally advanced pancreatic cancer: a phase I study. Am J Clin Oncol. 2005;28:570–5. doi: 10.1097/01.coc.0000184682.51193.00. [DOI] [PubMed] [Google Scholar]
  • 116.Czito BG, Willett CG, Bendell JC, Morse MA, Tyler DS, Fernando NH, Mantyh CR, Blobe GC, Honeycutt W, Yu D, Clary BM, Pappas TN, Ludwig KA, Hurwitz HI. Increased toxicity with gefitinib, capecitabine, and radiation therapy in pancreatic and rectal cancer: phase I trial results. J Clin Oncol. 2006;24:656–62. doi: 10.1200/JCO.2005.04.1749. [DOI] [PubMed] [Google Scholar]
  • 117.Fountzilas G, Murray S, Xiros N, Karayannopoulou G, Dafni U, Linardou H, Kalogera-Fountzila A, Bobos M, Koumarianou A, Kosmidis P. Gemcitabine (G) combined with gefitinib in patients with inoperable or metastatic pancreatic cancer. A phase II trial. J Clin Oncol (Meeting Abstracts) 2007;25:15016. [Google Scholar]
  • 118.Blaszkowsky LS, Ryan DP, Earle C, Kwak E, Fuchs C, Meyerhardt JA, Stuart K, Zhu AX, Enzinger P, Kulke MH. A phase II study of docetaxel in combination with ZD1839 (gefitinib) in previously treated patients with metastatic pancreatic cancer. J Clin Oncol (Meeting Abstracts) 2007;25:15080. [Google Scholar]
  • 119.Ignatiadis M, Polyzos A, Stathopoulos GP, Tselepatiotis E, Christophylakis C, Kalbakis K, Vamvakas L, Kotsakis A, Potamianou A, Georgoulias V. A multicenter phase II study of docetaxel in combination with gefitinib in gemcitabine-pretreated patients with advanced/metastatic pancreatic cancer. Oncology. 2006;71:159–63. doi: 10.1159/000106064. [DOI] [PubMed] [Google Scholar]
  • 120.Aly A, Shulkes A, Baldwin GS. Gastrins, cholecystokinins and gastrointestinal cancer. Biochim Biophys Acta. 2004;1704:1–10. doi: 10.1016/j.bbcan.2004.01.004. [DOI] [PubMed] [Google Scholar]
  • 121.Caplin M, Savage K, Khan K, Brett B, Rode J, Varro A, Dhillon A. Expression and processing of gastrin in pancreatic adenocarcinoma. Br J Surg. 2000;87:1035–40. doi: 10.1046/j.1365-2168.2000.01488.x. [DOI] [PubMed] [Google Scholar]
  • 122.Smith JP, Fantaskey AP, Liu G, Zagon IS. Identification of gastrin as a growth peptide in human pancreatic cancer. Am J Physiol. 1995;268:R135–41. doi: 10.1152/ajpregu.1995.268.1.R135. [DOI] [PubMed] [Google Scholar]
  • 123.Smith JP, Verderame MF, Zagon IS. Antisense oligonucleotides to gastrin inhibit growth of human pancreatic cancer. Cancer Lett. 1999;135:107–12. doi: 10.1016/s0304-3835(98)00279-1. [DOI] [PubMed] [Google Scholar]
  • 124.Clerc P, Leung-Theung-Long S, Wang TC, Dockray GJ, Bouisson M, Delisle MB, Vaysse N, Pradayrol L, Fourmy D, Dufresne M. Expression of CCK2 receptors in the murine pancreas: proliferation, transdifferentiation of acinar cells, and neoplasia. Gastroenterology. 2002;122:428–37. doi: 10.1053/gast.2002.30984. [DOI] [PubMed] [Google Scholar]
  • 125.Chau I, Cunningham D, Russell C, Norman AR, Kurzawinski T, Harper P, Harrison P, Middleton G, Daniels F, Hickish T, Prendeville J, Ross PJ, Theis B, Hull R, Walker M, Shankley N, Kalindjian B, Murray G, Gillbanks A, Black J. Gastrazole (JB95008), a novel CCK2/gastrin receptor antagonist, in the treatment of advanced pancreatic cancer: results from two randomised controlled trials. Br J Cancer. 2006;94:1107–15. doi: 10.1038/sj.bjc.6603058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Brett BT, Smith SC, Bouvier CV, Michaeli D, Hochhauser D, Davidson BR, Kurzawinski TR, Watkinson AF, Van Someren N, Pounder RE, Caplin ME. Phase II study of anti-gastrin-17 antibodies, raised to G17DT, in advanced pancreatic cancer. J Clin Oncol. 2002;20:4225–31. doi: 10.1200/JCO.2002.11.151. [DOI] [PubMed] [Google Scholar]
  • 127.Shapiro J, Marshall J, Karasek P, Figer A, Oettle H, Couture F, Jeziorski K, Broome P, Hawkins R. G17DT+gemcitabine [Gem] versus placebo+Gem in untreated subjects with locally advanced, recurrent, or metastatic adenocarcinoma of the pancreas: Results of a randomized, double-blind, multinational, multicenter study. J Clin Oncol (Meeting Abstracts) 2005;23:4012. [Google Scholar]
  • 128.Mollenhauer J, Roether I, Kern HF. Distribution of extracellular matrix proteins in pancreatic ductal adenocarcinoma and its influence on tumor cell proliferation in vitro. Pancreas. 1987;2:14–24. doi: 10.1097/00006676-198701000-00003. [DOI] [PubMed] [Google Scholar]
  • 129.Nagakawa T, Konishi I, Higashino Y, Ueno K, Ohta T, Kayahara M, Ueda N, Maeda K, Miyazaki I. The spread and prognosis of carcinoma in the region of the pancreatic head. Jpn J Surg. 1989;19:510–8. doi: 10.1007/BF02471656. [DOI] [PubMed] [Google Scholar]
  • 130.Nagakawa T, Kayahara M, Ueno K, Ohta T, Konishi I, Ueda N, Miyazaki I. A clinicopathologic study on neural invasion in cancer of the pancreatic head. Cancer. 1992;69:930–5. doi: 10.1002/1097-0142(19920215)69:4<930::aid-cncr2820690416>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
  • 131.Coussens LM, Fingleton B, Matrisian LM. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science. 2002;295:2387–92. doi: 10.1126/science.1067100. [DOI] [PubMed] [Google Scholar]
  • 132.Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006;25:9–34. doi: 10.1007/s10555-006-7886-9. [DOI] [PubMed] [Google Scholar]
  • 133.Chambers AF, Matrisian LM. Changing views of the role of matrix metalloproteinases in metastasis. J Natl Cancer Inst. 1997;89:1260–70. doi: 10.1093/jnci/89.17.1260. [DOI] [PubMed] [Google Scholar]
  • 134.Bramhall SR, Neoptolemos JP, Stamp GW, Lemoine NR. Imbalance of expression of matrix metalloproteinases (MMPs) and tissue inhibitors of the matrix metalloproteinases (TIMPs) in human pancreatic carcinoma. J Pathol. 1997;182:347–55. doi: 10.1002/(SICI)1096-9896(199707)182:3<347::AID-PATH848>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
  • 135.Jones LE, Humphreys MJ, Campbell F, Neoptolemos JP, Boyd MT. Comprehensive analysis of matrix metalloproteinase and tissue inhibitor expression in pancreatic cancer: increased expression of matrix metalloproteinase-7 predicts poor survival. Clin Cancer Res. 2004;10:2832–45. doi: 10.1158/1078-0432.ccr-1157-03. [DOI] [PubMed] [Google Scholar]
  • 136.Crawford HC, Scoggins CR, Washington MK, Matrisian LM, Leach SD. Matrix metalloproteinase-7 is expressed by pancreatic cancer precursors and regulates acinar-to-ductal metaplasia in exocrine pancreas. J Clin Invest. 2002;109:1437–44. doi: 10.1172/JCI15051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Reich R, Thompson EW, Iwamoto Y, Martin GR, Deason JR, Fuller GC, Miskin R. Effects of inhibitors of plasminogen activator, serine proteinases, and collagenase IV on the invasion of basement membranes by metastatic cells. Cancer Res. 1988;48:3307–12. [PubMed] [Google Scholar]
  • 138.Zervox EE, Franz MG, Salhab KF, Shafii AE, Menendez J, Gower WR, Rosemurgy AS. Matrix metalloproteinase inhibition improves survival in an orthotopic model of human pancreatic cancer. J Gastrointest Surg. 2000;4:614–9. doi: 10.1016/s1091-255x(00)80111-0. [DOI] [PubMed] [Google Scholar]
  • 139.Haq M, Shafii A, Zervos EE, Rosemurgy AS. Addition of matrix metalloproteinase inhibition to conventional cytotoxic therapy reduces tumor implantation and prolongs survival in a murine model of human pancreatic cancer. Cancer Res. 2000;60:3207–11. [PubMed] [Google Scholar]
  • 140.Moore MJ, Hamm J, Dancey J, Eisenberg PD, Dagenais M, Fields A, Hagan K, Greenberg B, Colwell B, Zee B, Tu D, Ottaway J, Humphrey R, Seymour L. Comparison of gemcitabine versus the matrix metalloproteinase inhibitor BAY 12-9566 in patients with advanced or metastatic adenocarcinoma of the pancreas: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2003;21:3296–302. doi: 10.1200/JCO.2003.02.098. [DOI] [PubMed] [Google Scholar]
  • 141.Evans JD, Stark A, Johnson CD, Daniel F, Carmichael J, Buckels J, Imrie CW, Brown P, Neoptolemos JP. A phase II trial of marimastat in advanced pancreatic cancer. Br J Cancer. 2001;85:1865–70. doi: 10.1054/bjoc.2001.2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Bramhall SR, Rosemurgy A, Brown PD, Bowry C, Buckels JA. Marimastat as first-line therapy for patients with unresectable pancreatic cancer: a randomized trial. J Clin Oncol. 2001;19:3447–55. doi: 10.1200/JCO.2001.19.15.3447. [DOI] [PubMed] [Google Scholar]
  • 143.Bramhall SR, Schulz J, Nemunaitis J, Brown PD, Baillet M, Buckels JA. A double-blind placebo-controlled, randomised study comparing gemcitabine and marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer. Br J Cancer. 2002;87:161–7. doi: 10.1038/sj.bjc.6600446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Gore M, A’Hern R, Stankiewicz M, Slevin M. Tumour marker levels during marimastat therapy. Lancet. 1996;348:263–4. doi: 10.1016/s0140-6736(96)24030-9. [DOI] [PubMed] [Google Scholar]
  • 145.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–7. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Fosslien E. Review: molecular pathology of cyclooxygenase-2 in cancer-induced angiogenesis. Ann Clin Lab Sci. 2001;31:325–48. [PubMed] [Google Scholar]
  • 147.Trifan OC, Hla T. Cyclooxygenase-2 modulates cellular growth and promotes tumorigenesis. J Cell Mol Med. 2003;7:207–22. doi: 10.1111/j.1582-4934.2003.tb00222.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Okami J, Yamamoto H, Fujiwara Y, Tsujie M, Kondo M, Noura S, Oshima S, Nagano H, Dono K, Umeshita K, Ishikawa O, Sakon M, Matsuura N, Nakamori S, Monden M. Overexpression of cyclooxygenase-2 in carcinoma of the pancreas. Clin Cancer Res. 1999;5:2018–24. [PubMed] [Google Scholar]
  • 149.Molina MA, Sitja-Arnau M, Lemoine MG, Frazier ML, Sinicrope FA. Increased cyclooxygenase-2 expression in human pancreatic carcinomas and cell lines: growth inhibition by nonsteroidal anti-inflammatory drugs. Cancer Res. 1999;59:4356–62. [PubMed] [Google Scholar]
  • 150.Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, Soslow RA, Masferrer JL, Woerner BM, Koki AT, Fahey TJ., 3rd Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer. Cancer Res. 1999;59:987–90. [PubMed] [Google Scholar]
  • 151.Ding XZ, Tong WG, Adrian TE. Blockade of cyclooxygenase-2 inhibits proliferation and induces apoptosis in human pancreatic cancer cells. Anticancer Res. 2000;20:2625–31. [PubMed] [Google Scholar]
  • 152.Tseng WW, Deganutti A, Chen MN, Saxton RE, Liu CD. Selective cyclooxygenase-2 inhibitor rofecoxib (Vioxx) induces expression of cell cycle arrest genes and slows tumor growth in human pancreatic cancer. J Gastrointest Surg. 2002;6:838–43. doi: 10.1016/s1091-255x(02)00061-6. discussion 844. [DOI] [PubMed] [Google Scholar]
  • 153.Funahashi H, Satake M, Dawson D, Huynh NA, Reber HA, Hines OJ, Eibl G. Delayed progression of pancreatic intraepithelial neoplasia in a conditional Kras(G12D) mouse model by a selective cyclooxygenase-2 inhibitor. Cancer Res. 2007;67:7068–71. doi: 10.1158/0008-5472.CAN-07-0970. [DOI] [PubMed] [Google Scholar]
  • 154.Crane CH, Mason K, Janjan NA, Milas L. Initial experience combining cyclooxygenase-2 inhibition with chemoradiation for locally advanced pancreatic cancer. Am J Clin Oncol. 2003;26:S81–4. doi: 10.1097/00000421-200308002-00009. [DOI] [PubMed] [Google Scholar]
  • 155.Milella M, Gelibter A, Di Cosimo S, Bria E, Ruggeri EM, Carlini P, Malaguti P, Pellicciotta M, Terzoli E, Cognetti F. Pilot study of celecoxib and infusional 5-fluorouracil as second-line treatment for advanced pancreatic carcinoma. Cancer. 2004;101:133–8. doi: 10.1002/cncr.20338. [DOI] [PubMed] [Google Scholar]
  • 156.El-Rayes BF, Zalupski MM, Shields AF, Ferris AM, Vaishampayan U, Heilbrun LK, Venkatramanamoorthy R, Adsay V, Philip PA. A phase II study of celecoxib, gemcitabine, and cisplatin in advanced pancreatic cancer. Invest New Drugs. 2005;23:583–90. doi: 10.1007/s10637-005-1028-z. [DOI] [PubMed] [Google Scholar]
  • 157.Ferrari V, Valcamonico F, Amoroso V, Simoncini E, Vassalli L, Marpicati P, Rangoni G, Grisanti S, Tiberio GA, Nodari F, Strina C, Marini G. Gemcitabine plus celecoxib (GECO) in advanced pancreatic cancer: a phase II trial. Cancer Chemother Pharmacol. 2006;57:185–90. doi: 10.1007/s00280-005-0028-1. [DOI] [PubMed] [Google Scholar]
  • 158.Kerr S, Campbell C, Legore K, Witters L, Harvey H, Lipton A. Phase II trial of gemcitabine and irinotecan plus celecoxib in advanced adenocarcinoma of the pancreas. J Clin Oncol (Meeting Abstracts) 2005;23:4155. [Google Scholar]
  • 159.Cheng JQ, Ruggeri B, Klein WM, Sonoda G, Altomare DA, Watson DK, Testa JR. Amplification of AKT2 in human pancreatic cells and inhibition of AKT2 expression and tumorigenicity by antisense RNA. Proc Natl Acad Sci U S A. 1996;93:3636–41. doi: 10.1073/pnas.93.8.3636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Ruggeri BA, Huang L, Wood M, Cheng JQ, Testa JR. Amplification and overexpression of the AKT2 oncogene in a subset of human pancreatic ductal adenocarcinomas. Mol Carcinog. 1998;21:81–6. [PubMed] [Google Scholar]
  • 161.Asano T, Yao Y, Zhu J, Li D, Abbruzzese JL, Reddy SA. The PI 3-kinase/Akt signaling pathway is activated due to aberrant Pten expression and targets transcription factors NF-kappaB and c-Myc in pancreatic cancer cells. Oncogene. 2004;23:8571–80. doi: 10.1038/sj.onc.1207902. [DOI] [PubMed] [Google Scholar]
  • 162.Ng SSW, Tsao MS, Chow S, Hedley DW. Inhibition of phosphatidylinositide 3-kinase enhances gemcitabine-induced apoptosis in human pancreatic cancer cells. Cancer Res. 2000;60:5451–5. [PubMed] [Google Scholar]
  • 163.Perugini RA, McDade TP, Vittimberga FJ, Jr., Callery MP. Pancreatic cancer cell proliferation is phosphatidylinositol 3-kinase dependent. J Surg Res. 2000;90:39–44. doi: 10.1006/jsre.2000.5833. [DOI] [PubMed] [Google Scholar]
  • 164.Ng SS, Tsao MS, Nicklee T, Hedley DW. Wortmannin inhibits pkb/akt phosphorylation and promotes gemcitabine antitumor activity in orthotopic human pancreatic cancer xenografts in immunodeficient mice. Clin Cancer Res. 2001;7:3269–75. [PubMed] [Google Scholar]
  • 165.Bondar VM, Sweeney-Gotsch B, Andreeff M, Mills GB, McConkey DJ. Inhibition of the phosphatidylinositol 3′-kinase-AKT pathway induces apoptosis in pancreatic carcinoma cells in vitro and in vivo. Mol Cancer Ther. 2002;1:989–97. [PubMed] [Google Scholar]
  • 166.Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell. 2007;12:9–22. doi: 10.1016/j.ccr.2007.05.008. [DOI] [PubMed] [Google Scholar]
  • 167.Ito D, Fujimoto K, Mori T, Kami K, Koizumi M, Toyoda E, Kawaguchi Y, Doi R. In vivo antitumor effect of the mTOR inhibitor CCI-779 and gemcitabine in xenograft models of human pancreatic cancer. Int J Cancer. 2006;118:2337–43. doi: 10.1002/ijc.21532. [DOI] [PubMed] [Google Scholar]
  • 168.Asano T, Yao Y, Zhu J, Li D, Abbruzzese JL, Reddy SA. The rapamycin analog CCI-779 is a potent inhibitor of pancreatic cancer cell proliferation. Biochem Biophys Res Commun. 2005;331:295–302. doi: 10.1016/j.bbrc.2005.03.166. [DOI] [PubMed] [Google Scholar]
  • 169.Dormond O, Madsen JC, Briscoe DM. The effects of mTOR-Akt interactions on anti-apoptotic signaling in vascular endothelial cells. J Biol Chem. 2007;282:23679–86. doi: 10.1074/jbc.M700563200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Land SC, Tee AR. Hypoxia-inducible factor 1alpha is regulated by the mammalian target of rapamycin (mTOR) via an mTOR signaling motif. J Biol Chem. 2007;282:20534–43. doi: 10.1074/jbc.M611782200. [DOI] [PubMed] [Google Scholar]
  • 171.Bruns CJ, Koehl GE, Guba M, Yezhelyev M, Steinbauer M, Seeliger H, Schwend A, Hoehn A, Jauch KW, Geissler EK. Rapamycin-induced endothelial cell death and tumor vessel thrombosis potentiate cytotoxic therapy against pancreatic cancer. Clin Cancer Res. 2004;10:2109–19. doi: 10.1158/1078-0432.ccr-03-0502. [DOI] [PubMed] [Google Scholar]
  • 172.Tuncyurek P, Mayer JM, Klug F, Dillmann S, Henne-Bruns D, Keller F, Stracke S. Everolimus and mycophenolate mofetil sensitize human pancreatic cancer cells to gemcitabine in vitro: a novel adjunct to standard chemotherapy? Eur Surg Res. 2007;39:380–7. doi: 10.1159/000107356. [DOI] [PubMed] [Google Scholar]
  • 173.Zitzmann K, De Toni EN, Brand S, Goke B, Meinecke J, Spottl G, Meyer HH, Auernhammer CJ. The novel mTOR inhibitor RAD001 (everolimus) induces antiproliferative effects in human pancreatic neuroendocrine tumor cells. Neuroendocrinology. 2007;85:54–60. doi: 10.1159/000100057. [DOI] [PubMed] [Google Scholar]
  • 174.Wang W, Abbruzzese JL, Evans DB, Larry L, Cleary KR, Chiao PJ. The nuclear factor-kappa B RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells. Clin Cancer Res. 1999;5:119–27. [PubMed] [Google Scholar]
  • 175.Arlt A, Gehrz A, Muerkoster S, Vorndamm J, Kruse ML, Folsch UR, Schafer H. Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene. 2003;22:3243–51. doi: 10.1038/sj.onc.1206390. [DOI] [PubMed] [Google Scholar]
  • 176.Muerkoster S, Arlt A, Witt M, Gehrz A, Haye S, March C, Grohmann F, Wegehenkel K, Kalthoff H, Folsch UR, Schafer H. Usage of the NF-kappaB inhibitor sulfasalazine as sensitizing agent in combined chemotherapy of pancreatic cancer. Int J Cancer. 2003;104:469–76. doi: 10.1002/ijc.10963. [DOI] [PubMed] [Google Scholar]
  • 177.Bold RJ, Virudachalam S, McConkey DJ. Chemosensitization of pancreatic cancer by inhibition of the 26S proteasome. J Surg Res. 2001;100:11–7. doi: 10.1006/jsre.2001.6194. [DOI] [PubMed] [Google Scholar]
  • 178.Nawrocki ST, Sweeney-Gotsch B, Takamori R, McConkey DJ. The proteasome inhibitor bortezomib enhances the activity of docetaxel in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther. 2004;3:59–70. [PubMed] [Google Scholar]
  • 179.Shah SA, Potter MW, McDade TP, Ricciardi R, Perugini RA, Elliott PJ, Adams J, Callery MP. 26S proteasome inhibition induces apoptosis and limits growth of human pancreatic cancer. J Cell Biochem. 2001;82:110–22. doi: 10.1002/jcb.1150. [DOI] [PubMed] [Google Scholar]
  • 180.Nawrocki ST, Bruns CJ, Harbison MT, Bold RJ, Gotsch BS, Abbruzzese JL, Elliott P, Adams J, McConkey DJ. Effects of the proteasome inhibitor PS-341 on apoptosis and angiogenesis in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther. 2002;1:1243–53. [PubMed] [Google Scholar]
  • 181.Ryan DP, Appleman LJ, Lynch T, Supko JG, Fidias P, Clark JW, Fishman M, Zhu AX, Enzinger PC, Kashala O, Cusack J, Jr., Eder JP. Phase I clinical trial of bortezomib in combination with gemcitabine in patients with advanced solid tumors. Cancer. 2006;107:2482–9. doi: 10.1002/cncr.22264. [DOI] [PubMed] [Google Scholar]
  • 182.Alberts SR, Foster NR, Morton RF, Kugler J, Schaefer P, Wiesenfeld M, Fitch TR, Steen P, Kim GP, Gill S. PS-341 and gemcitabine in patients with metastatic pancreatic adenocarcinoma: a North Central Cancer Treatment Group (NCCTG) randomized phase II study. Ann Oncol. 2005;16:1654–61. doi: 10.1093/annonc/mdi324. [DOI] [PubMed] [Google Scholar]
  • 183.Aggarwal BB, Kumar A, Bharti AC. Anticancer potential of curcumin: preclinical and clinical studies. Anticancer Res. 2003;23:363–98. [PubMed] [Google Scholar]
  • 184.Hidaka H, Ishiko T, Furuhashi T, Kamohara H, Suzuki S, Miyazaki M, Ikeda O, Mita S, Setoguchi T, Ogawa M. Curcumin inhibits interleukin 8 production and enhances interleukin 8 receptor expression on the cell surface:impact on human pancreatic carcinoma cell growth by autocrine regulation. Cancer. 2002;95:1206–14. doi: 10.1002/cncr.10812. [DOI] [PubMed] [Google Scholar]
  • 185.Kunnumakkara AB, Guha S, Krishnan S, Diagaradjane P, Gelovani J, Aggarwal BB. Curcumin potentiates antitumor activity of gemcitabine in an orthotopic model of pancreatic cancer through suppression of proliferation, angiogenesis, and inhibition of nuclear factor-kappaB-regulated gene products. Cancer Res. 2007;67:3853–61. doi: 10.1158/0008-5472.CAN-06-4257. [DOI] [PubMed] [Google Scholar]
  • 186.Lev-Ari S, Vexler A, Starr A, Ashkenazy-Voghera M, Greif J, Aderka D, Ben-Yosef R. Curcumin augments gemcitabine cytotoxic effect on pancreatic adenocarcinoma cell lines. Cancer Invest. 2007;25:411–8. doi: 10.1080/07357900701359577. [DOI] [PubMed] [Google Scholar]
  • 187.Dhillon N, Aggarwal BB, Newman RA, Wolff RA, Kunnumakkara AB, Abbruzzese JL, Hong DS, Camacho LH, Ng C, Kurzrock R. Curcumin and pancreatic cancer: Phase II clinical trial experience. ASCO Meeting Abstracts. 2007;25:4599. [Google Scholar]
  • 188.Miyamoto Y, Maitra A, Ghosh B, Zechner U, Argani P, Iacobuzio-Donahue CA, Sriuranpong V, Iso T, Meszoely IM, Wolfe MS, Hruban RH, Ball DW, Schmid RM, Leach SD. Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003;3:565–76. doi: 10.1016/s1535-6108(03)00140-5. [DOI] [PubMed] [Google Scholar]
  • 189.Thayer SP, di Magliano MP, Heiser PW, Nielsen CM, Roberts DJ, Lauwers GY, Qi YP, Gysin S, Fernandez-del Castillo C, Yajnik V, Antoniu B, McMahon M, Warshaw AL, Hebrok M. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature. 2003;425:851–6. doi: 10.1038/nature02009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Feldmann G, Dhara S, Fendrich V, Bedja D, Beaty R, Mullendore M, Karikari C, Alvarez H, Iacobuzio-Donahue C, Jimeno A, Gabrielson KL, Matsui W, Maitra A. Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res. 2007;67:2187–96. doi: 10.1158/0008-5472.CAN-06-3281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Hakam A, Fang Q, Karl R, Coppola D. Coexpression of IGF-1R and c-Src proteins in human pancreatic ductal adenocarcinoma. Dig Dis Sci. 2003;48:1972–8. doi: 10.1023/a:1026122421369. [DOI] [PubMed] [Google Scholar]
  • 192.Beltran PJ, Mitchell P, Moody G, Chung Y, Cajulis E, Belmontes B, Radinsky R, Calzone F. Effect of AMG 479 on anti-tumor effects of gemcitabine and erlotinib against pancreatic carcinoma xenograft models. J Clin Oncol (Meeting Abstracts) 2008;26:4617. [Google Scholar]
  • 193.Bierie B, Moses HL. Tumour microenvironment: TGFbeta: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer. 2006;6:506–20. doi: 10.1038/nrc1926. [DOI] [PubMed] [Google Scholar]
  • 194.Hilbig A, Seufferlein T, Schmid RM, Luger T, Oettle H, Schneider G, Schmaus S, Wuerth G, Heinrichs H, Schlingensiepen KH. Preliminary results of a phase I/II study in patients with pancreatic carcinoma, malignant melanoma, or colorectal carcinoma using systemic i.v. administration of AP 12009. J Clin Oncol (Meeting Abstracts) 2008;26:4621. [Google Scholar]
  • 195.Argani P, Iacobuzio-Donahue C, Ryu B, Rosty C, Goggins M, Wilentz RE, Murugesan SR, Leach SD, Jaffee E, Yeo CJ, Cameron JL, Kern SE, Hruban RH. 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;7:3862–8. [PubMed] [Google Scholar]
  • 196.Hassan R, Laszik ZG, Lerner M, Raffeld M, Postier R, Brackett D. Mesothelin is overexpressed in pancreaticobiliary adenocarcinomas but not in normal pancreas and chronic pancreatitis. Am J Clin Pathol. 2005;124:838–45. [PubMed] [Google Scholar]
  • 197.Qu CF, Li Y, Song YJ, Rizvi SM, Raja C, Zhang D, Samra J, Smith R, Perkins AC, Apostolidis C, Allen BJ. MUC1 expression in primary and metastatic pancreatic cancer cells for in vitro treatment by (213)Bi-C595 radioimmunoconjugate. Br J Cancer. 2004;91:2086–93. doi: 10.1038/sj.bjc.6602232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Yamasaki H, Ikeda S, Okajima M, Miura Y, Asahara T, Kohno N, Shimamoto F. Expression and localization of MUC1, MUC2, MUC5AC and small intestinal mucin antigen in pancreatic tumors. Int J Oncol. 2004;24:107–13. [PubMed] [Google Scholar]
  • 199.Hassan R, Bullock S, Premkumar A, Kreitman RJ, Kindler H, Willingham MC, Pastan I. 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;13:5144–9. doi: 10.1158/1078-0432.CCR-07-0869. [DOI] [PubMed] [Google Scholar]
  • 200.Armstrong DK, Laheru D, Ma WW, Cohen SJ, Phillips M, Brahmer J, Weil SC, Hassan R. A phase 1 study of MORAb-009, a monoclonal antibody against mesothelin in pancreatic cancer, mesothelioma and ovarian adenocarcinoma. J Clin Oncol (Meeting Abstracts) 2007;25:14041. [Google Scholar]
  • 201.Kindler HL, Gangadhar T, Karrison T, Hochster HS, Moore MJ, Micetich K, Sun W, Catenacci DV, Stadler WM, Vokes EE. Final analysis of a randomized phase II study of bevacizumab (B) and gemcitabine (G) plus cetuximab (C) or erlotinib (E) in patients (pts) with advanced pancreatic cancer (PC) J Clin Oncol (Meeting Abstracts) 2008;26:4502. [Google Scholar]
  • 202.Vervenne W, Bennouna J, Humblet Y, Gill S, Moore MJ, Van Laethem J, Shang A, Cosaert J, Verslype C, Van Cutsem E. A randomized, double-blind, placebo (P) controlled, multicenter phase III trial to evaluate the efficacy and safety of adding bevacizumab (B) to erlotinib (E) and gemcitabine (G) in patients (pts) with metastatic pancreatic cancer. J Clin Oncol (Meeting Abstracts) 2008;26:4507. [Google Scholar]
  • 203.Ko AH, Dito E, Schillinger B, Venook AP, Bergsland EK, Korn WM, Tempero MA. A phase II study of bevacizumab (BEV) plus erlotinib (ERL) in patients with gemcitabine (GEM)-refractory metastatic pancreatic cancer (MPC) J Clin Oncol (Meeting Abstracts) 2008;26:4516. [Google Scholar]
  • 204.Molina MA, Sitja-Arnau M, Lemoine MG, Frazier ML, Sinicrope FA. Increased Cyclooxygenase-2 Expression in Human Pancreatic Carcinomas and Cell Lines: Growth Inhibition by Nonsteroidal Anti-Inflammatory Drugs. Cancer Res. 1999;59:4356–4362. [PubMed] [Google Scholar]
  • 205.Okami J, Yamamoto H, Fujiwara Y, Tsujie M, Kondo M, Noura S, Oshima S, Nagano H, Dono K, Umeshita K, Ishikawa O, Sakon M, Matsuura N, Nakamori S, Monden M. Overexpression of Cyclooxygenase-2 in Carcinoma of the Pancreas. Clin Cancer Res. 1999;5:2018–2024. [PubMed] [Google Scholar]
  • 206.Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, Soslow RA, Masferrer JL, Woerner BM, Koki AT, Fahey TJ., III Cyclooxygenase-2 Expression Is Up-Regulated in Human Pancreatic Cancer. Cancer Res. 1999;59:987–990. [PubMed] [Google Scholar]
  • 207.Doucas H, Mann CD, Sutton CD, Garcea G, Neal CP, Berry DP, Manson MM. Expression of nuclear Notch3 in pancreatic adenocarcinomas is associated with adverse clinical features, and correlates with the expression of STAT3 and phosphorylated Akt. J Surg Oncol. 2008;97:63–8. doi: 10.1002/jso.20894. [DOI] [PubMed] [Google Scholar]
  • 208.Dang T, Vo k, Washington K, Berlin J. The role of Notch3 signaling pathway in pancreatic cancer. J Clin Oncol (Meeting Abstracts) 2007;25:21049. [Google Scholar]
  • 209.Philip PA, Benedetti J, Fenoglio-Preiser C, Zalupski M, Lenz H, O’Reilly E, Wong R, Atkins J, Abruzzese J, Blanke C. Phase III study of gemcitabine [G] plus cetuximab [C] versus gemcitabine in patients [pts] with locally advanced or metastatic pancreatic adenocarcinoma [PC]: SWOG S0205 study. J Clin Oncol (Meeting Abstracts) 2007;25:4509. [Google Scholar]

RESOURCES