Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2019 Aug 5;286(18):3510–3539. doi: 10.1111/febs.15011

Targeting the complexity of Src signalling in the tumour microenvironment of pancreatic cancer: from mechanism to therapy

Ashleigh Parkin 1, Jennifer Man 1, Paul Timpson 1,2,, Marina Pajic 1,2,
PMCID: PMC6771888  PMID: 31330086

Abstract

Pancreatic cancer, a disease with extremely poor prognosis, has been notoriously resistant to virtually all forms of treatment. The dynamic crosstalk that occurs between tumour cells and the surrounding stroma, frequently mediated by intricate Src/FAK signalling, is increasingly recognised as a key player in pancreatic tumourigenesis, disease progression and therapeutic resistance. These important cues are fundamental for defining the invasive potential of pancreatic tumours, and several components of the Src and downstream effector signalling have been proposed as potent anticancer therapeutic targets. Consequently, numerous agents that block this complex network are being extensively investigated as potential antiinvasive and antimetastatic therapeutic agents for this disease. In this review, we will discuss the latest evidence of Src signalling in PDAC progression, fibrotic response and resistance to therapy. We will examine future opportunities for the development and implementation of more effective combination regimens, targeting key components of the oncogenic Src signalling axis, and in the context of a precision medicine‐guided approach.

Keywords: focal adhesion kinase, integrin, microenvironment, pancreatic cancer, Src kinase, stroma


Abbreviations

Bcl2

B‐cell lymphoma 2

Cdk

cyclin‐dependent kinase

ECM

extracellular matrix

EGF

epidermal growth factor

EGFR

epidermal growth factor receptor

EMT

epithelial–mesenchymal transition

ERK

extracellular signal‐regulated kinase

ERK

extracellular signal‐regulated kinases

FAK

focal adhesion kinase

GLUT1

glucose transporter 1

GSK3beta

glycogen synthase kinase 3 beta

GTP

guanosine triphosphate

HA

hyaluronic acid

HGF

hepatocyte growth factor

HNSCC

head and neck squamous cell carcinoma

IL10

interleukin 10

IL6

interleukin 6

ITGA

integrin alpha‐3

JNK

Jun kinase

LAMA

laminin

MAPK

mitogen‐activated protein kinase

MAPK

mitogen‐activated protein kinases

MDM

minute 2 homolog

Mdm2

mouse double minute 2 homolog

MDSC

myeloid‐derived suppressor cell

MEK

mitogen‐activated protein kinase kinase

MMPs

metalloproteinases

mTOR

mammalian target of rapamycin

NF2

neurofibromin 2

NFkappaB

nuclear factor kappa‐light‐chain‐enhancer of activated B cells

PARP

poly‐ADP ribose polymerase

PD1

programmed cell death protein

PDAC

pancreatic ductal adenocarcinoma

PDGF

platelet‐derived growth factor

PIP

phosphatidylinositol 4,5‐bisphosphate

PTEN

phosphatase and tensin homolog

QCMG

Queensland Centre of Medical Genomics

Raf

rapidly accelerated fibrosarcoma

Rho

Ras homolog gene family

ROCK

Rho‐associated coiled‐coil containing protein kinase

TAM

tumour‐associated macrophage

TCGA

The Cancer Genome Atlas

TME

tumour microenvironment

TNF

Tumour necrosis factor

VEGF

vascular endothelial growth factor

VEGFR

vascular endothelial growth factor receptor

WGS

whole genome sequencing

Introduction

Our definition of ‘cancer’ is constantly being revised, with the traditional definition of a malignancy derived from epithelial cells now being inapplicable 1. It is now well recognised that carcinomas are not simply collections of individual clonal tumour cells, but rather comprise a complex environment of distinct cell types including molecularly diverse malignant cells and supporting nontransformed components that promote cancer development, spread and therapeutic resistance 2. These include resident cancer‐associated fibroblasts, pericytes, endothelial cells, adipocytes, nerves and infiltrating immune cells, which through dynamic communication with tumour cells, collectively regulate tumour growth and progression 2.

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a dismal 5‐year survival of < 8%, and this statistic has remained largely unchanged for the past 50 years 3, 4. PDAC is the third leading cause of all cancer deaths and is predicted to become the second by 2030 3, representing a significant burden in the Western society 3, 4, 5. Combination of chemotherapy agents, fluorouracil [5‐FU], leucovorin, irinotecan and oxaliplatin (FOLFIRINOX) or gemcitabine and nanoparticle albumin‐bound paclitaxel (Abraxane) represent current first‐line treatments for advanced PDAC 6, 7, 8. As most recent data indicate, their efficacy may also be of significant benefit in both adjuvant 9 and neoadjuvant settings 10. However, due to the toxicity associated with multiagent chemotherapy, there is a discernible need for novel, more tailored treatment combinations, as well as the identification of biomarkers to help rationalise treatment selection 5.

PDAC has a high molecular heterogeneity despite being morphologically indistinguishable 11, 12. Characterisation of this complex molecular landscape has revealed key insights into the biology of tumours 11, 13, 14, enabling us to build upon the traditional anatomical definition of cancer and further includes molecular subtyping or ‘omic’ stratification as a foundation for developing approaches for early detection and improved treatment options 11, 15, 16, as well as identification of mechanisms of therapeutic resistance 11, 12, 17. With new advances in sequencing and analytical methodologies, PDAC has been genomically and transcriptomically characterised to an incredible depth, as reviewed recently 14. Building on early studies which have identified the 12 key pathways and oncogenes genetically altered in most pancreatic cancers 18, this disease has since been stratified into distinct molecular subtypes using gene expression profiling 17, and comprehensive whole genome sequencing (WGS) approaches 11, 12, 19. For example, these analyses have led to the identification of a PDAC subtype characterised by high structural variation (> 200 structural rearrangements per tumour), that may be preferentially sensitive to DNA‐damaging agents, including PARP inhibitors and cisplatin 11. Subsequent integrative analysis of genomic and transcriptomic signatures has further characterised an ‘immunogenic’ subtype in PDAC 12, associated with a significant immune infiltrate, with predominant expression profiles related to infiltrating B and T cells, upregulation of CTLA4 and PD1 immunosuppressive pathways, suggesting that a proportion of PDAC tumours may potentially be targeted with immune‐modulating agents. Further work by Connor et al. 19 has described an interesting correlation between signatures that define double‐stranded DNA break repair and mismatch repair deficiencies and specific immune profiles in pancreatic cancer, highlighting that similar to other solid cancers 20, a subset of pancreatic cancers with a high mutation burden may present a viable target for immune‐modulating combination therapies.

Moreover, comprehensive genomic and transcriptomic studies in more frequently occurring cancers, such as breast cancer, have not only transformed and improved our understanding of the tumour landscape, but have been utilised to refine breast cancer classification, assess prognosis and response to therapy 21, 22. These examples demonstrate how the identification of key mutations can clearly benefit a larger number of selected cancer patients, and illustrate the need to include a molecular taxonomy when establishing effective treatment plans.

In addition to the novel approaches to cancer treatment developed from the genomic characterisation of cancer cells within tumours, the equally complex and dynamic tumour microenvironment (TME) has been shown to play a significant role in promoting cancer development, progression and treatment failure. Of note, PDAC is characterised by a hypoxic, immunosuppressive and highly fibrotic environment, with stromal components outnumbering pancreatic cancer cells 23, 24. Intricate communication between pancreatic cancer cells and their surrounding environment, driven by a dynamic signalling network of cellular and matrix remodelling enzymes, cytokines, chemokines and growth factors, collectively promotes tumour growth and treatment resistance 25, 26, 27, 28.

A key pathway that regulates the tumour microenvironment is the Src signalling network. The c‐Src non‐receptor tyrosine kinase is frequently overexpressed in numerous human malignancies, including PDAC 29, where it has been shown to promote tumour development and progression to distant metastases, leading to poor patient survival. Moreover, Src kinase is a mediator of integrin signalling in pancreatic cancer cells 30, and plays an important role in the regulation of several proteins that are frequently deregulated in cancer including focal adhesion kinase (FAK), epidermal growth factor receptor (EGFR), Akt/PI 3‐kinase, and Rho/ROCK signalling. These pathways directly drive tumour‐cell to stromal‐cell crosstalk, 31, 32, 33, 34, 35 and play a prominent role in regulating pancreatic tumour cell survival, adhesion, migration and invasion 29. In this review, we summarise and discuss the current understanding of the diverse and complex roles of aberrant Src signalling in the complex niche of a rapidly developing and metastasising pancreatic tumour, highlighting challenges with and new avenues for the utilisation of inhibitors that target this dynamic network.

The Src signalling axis promotes pancreatic cancer progression

The proto‐oncogene tyrosine‐protein kinase Src or cellular Src (c‐Src) belongs to a family of nine nonreceptor tyrosine kinases that share similar structure and function 36. Src kinase localises at cell–matrix adhesions, and is readily activated by positive migratory growth factor signalling, including, but not limited to, epidermal growth factor (EGF), hepatocyte growth factor (HGF), platelet‐derived growth factor (PDGF), vascular endothelial growth factor (VEGF) and integrin 37 and Eph receptor (EphA2) activation 38. In turn, Src can phosphorylate substrates from numerous molecular pathways and consequently promotes tumour cell survival, proliferation, cell adhesion, migration, invasion and angiogenesis, key hallmarks of cancer (Fig. 1) 29, 30, 39, 40, 41, 42, 43, 44. The roles of Src in tumourigenesis and metastasis are well established, with constitutive activation of Src being observed in a variety of cancers including breast, lung, colon, prostate and pancreas 29, 42, 45.

Figure 1.

Figure 1

Schematic of the canonical Integrin/Src/FAK signalling network. Src and FAK interact with, and are activated by, numerous receptor tyrosine kinases (RTKs), including epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), fibroblast growth factor receptor (FGFR), and platelet‐derived growth factor receptor (PDGFR), as well as the ‘matrix receptor’ integrins, which all facilitate their downstream signalling. (a) Phosphorylation and activation of RAS, RAF, MEK1/2 and ERK1/2 leads to the transcriptional regulation of genes associated with cell growth and proliferation. (b) Phosphorylation of signal transducer and activator of transcription 3 (STAT3), enables STAT3 dimerisation and translocation into the nucleus where it regulates gene expression of VEGF, IL10 and FoxP3, stimulating angiogenesis and immunosuppression. (c) PI3K assists in the recruitment of Akt to the plasma membrane, where it is phosphorylated and activated by PDK1/2, and then translocates to the cytosol or nucleus. Through its downstream mediators, Akt promotes RNA translation and protein synthesis, and cell survival. (d) Activation of Rho GTPases results in the binding of Rho‐associated protein kinase (ROCK) leading to actin cytoskeleton remodelling and cell motility. Rho GTPases can also activate myosin‐light chain (MLC) which is involved in the maintenance of stromal feedback and extracellular matrix deposition. Activation of Rac GTPases leads to the recruitment and activation of Arp2/3 via WAVE, leading to the formation of new actin polymers, whilst Rac can also activate PAK, leading to the inhibition of depolarisation of actin, key processes affecting actin dynamics and lamellipodia formation.

Src modulates integrin adhesions, cadherin‐mediated cell–cell adhesions and metalloproteinase expression, and it is this disruption of intercellular adhesion that results in the detachment of tumour cells from the tumour mass, allowing them to invade through the extracellular matrix (ECM), penetrate the blood vessels and metastasize to other sites 43. Furthermore, Src kinase activity is required for mesenchymal invasion (involving integrin and protease‐dependent stromal remodelling) as it controls the turnover of integrin‐based adhesions 46. In addition, Src has been suggested as a mechanistic link between inflammation and cancer 47. Specifically, Src activation in tumour‐associated macrophages, leads to their increased motility and infiltration into the tumour, a process which is driven by the secretion of pro‐inflammatory cytokines within the tumour microenvironment 47, 48, 49. Src also plays a role in the metabolic reprogramming of cancers by promoting the Warburg effect. This involves activation of hexokinases and upregulation of glycolysis, which in turn promotes tumourigenesis 45.

The significance of Src in PDAC tumourigenesis is also well established 29, 48, 50. Src kinase expression and activity is upregulated in PDAC, increased further during progression to invasive and metastatic (advanced) PDAC and is associated with poor survival 29, 50, 51. Src also plays a role in the progression of pancreatitis, an inflammatory condition that presents a risk for development of pancreatic cancer 52. Similar to other cancers, Src inhibition has been shown to reduce proliferation, migration and invasion in PDAC cell lines, as well as inhibits tumour progression and metastasis in vivo 43, 53, 54, 55, 56, 57. Src can also promote the progression of PDAC by reducing tumour response to gemcitabine, one of the current standards of care chemotherapies for this cancer 58.

In addition to SRC, the integrin–focal adhesion signalling‐mediated modulation of ECM mechanics and cytoskeleton stability involves several important sensor proteins that are also frequently deregulated in cancer, including integrins, FAK and downstream Akt/PI 3‐kinase, LIM kinase, and Rho/ROCK activation 59, 60, 61, 62 (Fig. 1). Integrins are composed of two noncovalently associated transmembrane glycoprotein subunits, and can be divided into several subtypes 63. These molecules can signal bidirectionally: through the recruitment of adaptor proteins the integrin receptor becomes activated and has a high affinity for ECM ligands, which in turn leads to the recruitment of signalling proteins and the assembly of focal adhesions 63. Integrins bind to, and remodel ECM components such as vitronectin, laminin, fibronectin and collagen, thereby providing the traction required for tumour cell motility and invasion. Increased deposition and cross‐linking of ECM proteins can also further promote tumour progression via mechanical force‐induced clustering of integrin receptors 64.

The crosstalk between integrins, growth factor receptors and SRC oncogene is readily exploited by cancer cells during both tumour initiation and disease progression 59. Furthermore, integrins also play a role in angiogenesis, by providing a docking site for several cell types, including endothelial cells, endothelial stem cells and inflammatory cells, at the site of angiogenesis 65. Upregulation of ανβ6‐integrins occurs in a variety of tumours, including PDAC, where it has been shown to activate TGF‐β, stimulating tumour cell epithelial‐to‐mesenchymal transition (EMT) and stromal myofibroblast differentiation 66, which has in turn been shown to either promote 67 or restrict tumour growth and progression 68. The association between ανβ6‐integrins and increased migration, invasion and cell survival is partly due to the regulation of proteases (MMPs), and urokinase‐type plasminogen activator (uPA) 63, 66, 69, 70, 71. In PDAC specifically, overexpression of integrin ανβ3/ανβ6 has been previously shown to associate with poor survival of patients as well as lymph node metastasis 59, 72, and recent findings indicate that the stromal localisation and levels of active α5β1‐integrin and FAK can identify two readily distinguishable desmoplastic phenotypes in pancreatic cancer. Tumours with high stromal pSMAD2/3 levels were found to be prognostic of poor outcome, whilst increased stromal levels of active α,β‐integrin constituted a patient‐protective PDAC‐associated desmoplastic phenotype 73. In addition, integrins also play a role in regulating cancer stem cell properties leading to metastasis as well as resistance to tyrosine kinase inhibitors in PDAC 74.

Focal adhesion kinase (FAK) is a ubiquitously expressed nonreceptor tyrosine kinase that regulates integrin‐mediated cell‐ECM signalling, and its phosphorylation and activation is dependent on Src. The Src‐FAK multiprotein complex localises at cell–matrix attachment sites and influences several downstream pathways including cell motility, migration, invasion, survival, immunosuppression and apoptosis 25, 29, 75, 76. The mechanisms involved are complex but often include the regulation of downstream effectors, including TGFβ, as well as regulators of ERK, Jun kinase (JNK) and Rho signalling pathways 34, 35, 42, 77, 78, 79. FAK is overexpressed in a variety of cancers including PDAC, and overexpression is associated with poor prognosis 76, 80. It has recently been shown that FAK plays an important role in regulating pro‐inflammatory pathway activation and cytokine production during wound healing 25, 44, 80, 81, 82, 83. In PDAC specifically, FAK activity has been shown to correlate with high levels of fibrosis and poor CD8+ cytotoxic T‐cell infiltration, making it a promising target to overcome the highly fibrotic and immunosuppressive nature of PDAC 25, 84.

Src‐family kinases (SFKs) not only promote cell–matrix adhesion turnover through FAK, but also regulate Rho family of small GTPases, in particular RhoA and Rac1 activation 85, 86. Rho GTPases are often hijacked by cancers because they regulate diverse cellular processes that are important for tumour growth and metastasis including cytoskeletal dynamics, motility, contractility, cell polarity, membrane transport, gene transcription, as well as regulating the interaction between stromal cells and cancer cells 87, 88, 89, 90, 91, 92, 93. SFKs control the regulatory molecules of Rho GTPases (guanine nucleotide exchange factors (GEFs), GTPase‐activating proteins (GAPs) and guanine dissociation inhibitors (GDIs)), and it is the tight regulation and extensive crosstalk between Src/FAK and Src/RhoA/Rac1 that controls integrin‐mediated cell adhesion and migration 94, 95, 96. We have recently reviewed the role of Rho‐associated kinase signalling in cancers including PDAC 87, 88.

PI 3‐kinase (PI3K) signalling is another relevant, tumour‐promoting and potentially druggable effector network activated through FAK/SFK 97, 98, 99. Activated PI3K phosphorylates phosphatidylinositol 4,5‐biphosphate (PIP2) to produce PIP3, and this process is negatively regulated by PTEN 100. Activation of PIP3 can then further activate Akt (Akt activation occurs in ~ 59% PDAC samples 101) and additional downstream targets such as Bcl‐2, Mdm2, GSK3beta, NF‐kappaB and mTOR 97, 102, ultimately promoting cancer cell survival, growth, and motility and inhibiting apoptosis 97, 100, 103, 104. The PI3K‐Akt‐mTOR pathway is also responsible for controlling cellular metabolism. Oncogenic K‐Ras can enhance the activity of the metabolic enzyme ATP citrate lyase in an Akt‐dependent manner leading to histone acetylation and alteration of the acetyl‐CoA pool, subsequently leading to changes in gene expression, DNA damage response and DNA replication 105. The PI3K/Akt pathway can also inhibit glucose metabolism by blocking glycogen synthase kinase 3β and can alter glucose uptake by mediating expression of glucose transporters such as GLUT1 105, 106. Furthermore, Akt signalling is present in preneoplastic lesions during pancreatic carcinogenesis induced by mutated Kras, and is associated with progression towards higher grade tumours and poorer patient survival 99, 107, 108, 109.

Molecular and genomic aberrations of the Src signalling axis in Pancreatic Cancer: Implications for therapeutic targeting

Historically, the documented cases of activating Src mutations are rare, with only one major study in colon cancer documenting 12% of cases with a truncating mutation at codon 531 110, which when functionally validated, was shown to lead to increased Src specificity and transformation of NIH 3T3 cells. Despite this, other studies using larger colon cancer populations document no such mutations 111, 112. In addition, no such mutations have been documented for Src‐implicated cancers, such as haematological malignancies 113. In PDAC specifically, examination of multidimensional publically available cancer genomics datasets (TCGA, PanCan Atlas and QCMG cohorts) revealed that Src mutations occur at a frequency of less than 2% (Fig. 2B) 114, 115, indicating that aberrant intratumoural Src activity occurs through constitutive activation of Src, or by changes in the levels of regulators of Src and amplification of downstream signalling pathways 113, 116, 117, 118.

Figure 2.

Figure 2

Genetic alteration frequency (% of patients) for key Src signalling components, generated from publically available pancreatic cancer genomics datasets. These datasets include The Cancer Genome Atlas (TCGA), PanCan Atlas (TCGA PanCan), University of Texas South Western Medical Centre (UTSW) and Queensland Centre for Medical Genomics (QCMG 2016) cohorts 114, 115. The genetic alterations examined include mutations (green), fusions (purple), amplifications (red), deletions (blue) and multiple alterations (grey) (F). (A) Genetic alteration frequency of integrins, with integrin genes being defined in (E). (B) Genetic alteration frequency of Src. (C) Genetic alteration frequency of FAK (PTK2). (D) Genetic alteration frequency of the PI3K/AKT pathway. (E) The list of genes used to define the PI3K/AKT pathway. Figure reproduced from Refs 114, 115

Integrins are key regulators of Src signalling, and are also deregulated in cancers, but are rarely mutated. Several cancers, including glioblastoma, show modifications of the integrin pattern to be associated with tumour progression and poor patient survival, including α6β4, α6β1, αvβ6 and αvβ3 119. An early sequencing study demonstrated a positive association between mutations in subunit α7 (encoded by ITGA7 gene), identified in 57% of prostate cancers, and increased cancer recurrence 120. The mutation also occurred in 21% of hepatocellular carcinomas and 83% of glioblastomas, as well as leiomyosarcomas 120. Decreased integrin expression has also been correlated with cancer progression. In mesothelioma, reduced expression of ITGA7 was associated with promoter methylation and was identified as an important mechanism for the aggressive migratory transformation of mesothelioma 121, 122. Similar results have also been seen with α2β1 in breast cancer, and α6β4/ α6β1 in oesophageal carcinoma 59. In PDAC, early sequencing studies identified genetic alterations in the integrin signalling pathway (ITGA4, ITGA9, ITGA11, LAMA1, LAMA4, LAMA5, FN1 and ILK) in 67% of tumours 18. However, these alterations appear less frequent (67% versus 13%) when compared to the findings of the TCGA, UTSW, ICGC and QCMG 114, 115, 123 (Fig. 2A). This inconsistency may be explained through the study design of Jones et al. 124, where only small cohorts derived from cell lines (commercial and patient‐derived; = 24); and xenograft models (= 90) were used to analyse the mutational cancer landscape. Recent findings suggest that molecular landscapes of patient‐derived models may diverge from their parental tumours during long‐term propagation. More recently, the integrin β4 subunit was found to be commonly overexpressed in PDAC and is an adverse prognostic marker; however, it is not commonly mutated 125. An alternate mechanism involving a mutation in TP53 is thought to promote integrin α6β4‐mediated tumour cell survival 125.

In addition, recent large‐scale, pan‐cancer proteogenomic studies have identified molecular alterations in several Src effector networks including PI3K/Akt/mTOR and FAK 80, 126, 127, 128. Of > 7000 tumours examined, 63% harboured nonsilent somatic mutations or copy number alterations within the PI3K/AKT/mTOR pathway 127. In PDAC specifically, ~ 17% of tumours carried alterations, the majority of which involved gene amplification, and this finding is consistent across multiple cohorts 114, 115 (Fig. 2D). The PI3KCA gene mutations present in 3–5% of pancreatic cancer patients can act as activating mutations initiating pancreatic tumour formation 129. Further, inactivating aberrations in PTEN (negative regulator of PI3K/PI3K pathway) occur in up to 70% of human PDAC, and have been shown to activate the tumour‐promoting stromal and immune cell components that shape the PDAC TME 130. FAK is also frequently overexpressed and deregulated in PDAC, with genomics alterations occurring at a frequency of ~ 6%, the majority of which are gene amplifications (Fig. 2C) 114, 115. FAK inhibitor monotherapy has shown mixed clinical efficacy in mesothelioma tumours that harbour loss of specific tumour suppressive signals, such as Merlin (encoded by NF2 gene; 131, 132, 133). Although mutations at the NF2 locus are rare (~ 10%) in human PDAC 12, 19, Merlin expression is lost in > 40% of PDAC and is negatively correlated with tumour stage, regional lymph node metastasis and differentiation 134. Assessment into the efficacy of FAK inhibition in the context of Merlin loss, and combined with additional biomarkers, in PDAC may be of interest.

A personalised treatment strategy using pharmacological inhibition of Src, Src‐associated regulators or downstream targets, in tumour subtypes carrying these aberrations, could be beneficial and remains to be examined. Currently there are no FDA‐approved prognostic or predictive biomarkers for PDAC 7. Importantly, moving forward, the integration of DNA copy‐number alterations, methylome, mRNA and protein, metabolomics and clinical information may help to further delineate the extent of Src signalling deregulation in pancreatic and other cancers, and could potentially lay the foundation for more accurate and rapid implementation of therapeutic inhibitors of Src as personalised cancer therapeutics.

Targeting Src kinase in pancreatic cancer

Recognising the established role of Src in cancer initiation and progression led to the rapid development of several small molecule inhibitors (Table 1) 135. Inhibitors including bosutinib, saracatinib and dasatinib have shown measurable antitumour activity in several in vitro and in vivo models of cancer 47, 53, 56, 136, 137, 138. Dasatinib is a potent adenosine triphosphate‐competitive inhibitor of Src and Abl kinases, as well as c‐KIT, PDGFR and ephrin‐A2, which works by competitive inhibition of the ATP binding site. Its activity results in inhibition of cell proliferation (causing G0/G1 arrest), as well as inhibition of cell adhesion, migration, invasion and tumour metastasis 44, 53, 139, 140, 141, 142, 143. These results were particularly promising in models of advanced PDAC, presenting dasatinib as an encouraging antimetastatic agent for this disease 29, 56, 144. Despite the encouraging clinical results for the use of dasatinib as a standalone therapy in CML, clinical findings with dasatinib or alternative Src/ABL‐kinase inhibitors (saracatinib, bosutinib) 145, 146 in PDAC were predominately negative, partially due to poor drug tolerance, but also due to the highly aggressive and adaptable nature of this disease to single‐agent targeted therapies and rapid onset of resistance 53, 138, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156. Moreover, the presumption that these biologic agents would significantly improve survival in nonstratified cohorts, particularly in PDAC, is inconsistent with prior preclinical data, which suggests that therapeutic response may correlate with biological markers. For example, Saracatinib effectively inhibited the growth of three patient‐derived pancreatic xenografts characterised by decreased FAK, paxillin and STAT3 signalling 136. In addition Bosutinib sensitivity was shown to correlate with caveolin 1 expression 138, and clinical trial data indicate that selected individuals experienced durable and sustained responses to dasatinib treatment 102, 150, 151. Collectively, these data highlight the need for further investigation into the biological ‘omics’ of patients prior to treatment in order to identify the mechanistic rationale that can predict which patients may most optimally respond to Src‐based therapies.

Table 1.

Clinical trials in pancreatic cancer associated with targeting Src kinase.

Signalling pathway Agent Molecular target Cancer type Phase Combination therapy Findings/status Protocol ID Reference
Src Dasatinib Src, Abl, PDGFR Metastatic pancreatic cancer II (single arm) Monotherapy Completed: no significant clinical activity measured (= 34); 1 durable sustained response on therapy (> 20 months), plus 6 long‐term survivors noted (> 20 months) NCT00474812 150
Metastatic pancreatic cancer II (single arm) Monotherapy Terminated: Due to toxicity (= 7) NCT00544908
Molecular analysis for therapy choice (MATCH), multiple solid cancers incl metastatic or recurrent pancreatic cancer II (personalised) Monotherapy‐targeted against DDR2 mutations Recruiting NCT02465060
Metastatic pancreatic cancer I Gemcitabine Terminated: Due to low accrual NCT00598091
Locally advanced pancreatic cancer II (randomised) Gemcitabine Completed: no significant improvement in PFS, OS in unselected patient cohort (= 202). High dose regimen utilised leading to significant adverse events NCT01395017 155
Resected pancreatic cancer (adjuvant) II (randomised) Gemcitabine Completed: awaiting results NCT01234935
Advanced pancreatic cancer I Erlotinib + gemcitabine Active, not recruiting. Well tolerated. Early clinical activity with reported OS 8 months and disease control rate 69% vs historical control OS 5.9 months and 58% respectively. Small patient cohort (= 19) NCT01660971 185
Metastatic pancreatic cancer II (single arm) mFOLFOX6 Active, not recruiting (= 38) NCT01652976 137
Bosutinib Src, Abl Advanced solid cancers (incl pancreatic) I Monotherapy Completed: MTD determined; no significant efficacy observed NCT00195260 154
Resected pancreatic cancer I Gemcitabine Terminated: Due to slow accrual NCT01025570
Locally advanced/metastatic solid cancers (incl pancreatic) I/II Capecitabine Terminated: Tolerated, limited efficacy overall (= 5 pancreatic cancer patients) NCT00959946 156
Saracatinib (AZD0530) Src Recurrent metastatic pancreatic cancer II (single arm) Monotherapy Completed: no objective response observed in unselected cohort (= 19) NCT00735917 138
Advanced pancreatic cancer I/II (Single Arm) Gemcitabine Completed: well tolerated but no improvement in efficacy over Gemcitabine alone NCT00265876 153
Advanced solid cancers (incl pancreatic) I Cediranib (VEGFR1 inhibitor) Completed: tolerated. Demonstrated stable disease as best response in 22/35 evaluable patients NCT00475956 256
TNO155 SHP‐2 Advanced solid cancers I Monotherapy Recruiting NCT03114319
RMC‐4630 Advanced refractory solid cancers I Monotherapy Recruiting NCT03634982

Given that in pancreatic (and other) cancers, multiple mechanisms often work in synchrony to lead to chemoresistance, considering more tailored treatment combinations that involve inhibition of Src, other molecular targets, plus tumour‐debulking cytotoxic agents may present a more effective approach. The rationale behind this includes the finding that Src is associated with increased chemoresistance in PDAC, and that inhibition of Src can overcome resistance to gemcitabine 58, 137, 143. Furthermore, Src inhibition is associated with decreased thymidylate synthase, which in turn is associated with the reversal of 5‐fluorouracil resistance 137. Src inhibition can also increase oxaliplatin activity, and inhibit oxaliplatin‐induced Src activation 137. When dasatinib was combined with gemcitabine in locally advanced pancreatic cancer, there was no improvement in progression‐free or overall survival (NCT01395017) (Table 1) 157. However, newer combination chemotherapy regimens, such as FOLFIRINOX 6, lead to significantly higher response rates and disease control in patients with metastatic disease. Hence, a potentially more appropriate future study design may involve sequential administration of dasatinib as ‘maintenance’ therapy, after optimal disease control is achieved with this highly active chemotherapy regimen (similar to successful previous studies utilising sunitinib 152), or alternatively a ‘priming regimen’ could be applied 92, thus limiting toxicity associated with chronic dosing.

The Src signalling network is also known to play an important role in the movement and infiltration of immune cells into the tumour. In addition Src activation is mediated by inflammatory cytokines within the tumour microenvironment, whilst also being involved in intercellular communication 47. Although there is minimal evidence in pancreatic cancer, research into other solid cancers including melanoma, sarcoma, colon and breast cancer demonstrates that Src‐inhibitors such as dasatinib have potent immunomodulatory functions 158, and consequently may present a promising adjunct to immunotherapy. Dasatinib may enhance cellular immunity through a number of mechanisms including T‐cell immunomodulation, whereby treatment has been shown to reduce the number of intratumoural regulatory T cells, in various solid tumour mouse models and haematological malignancies, promoting natural killer (NK) cell expansion and differentiation 158, 159, 160. In chronic myeloid leukaemia (CML) cancer models, dasatinib may increase the number of Granzyme B (GrB) expressing memory CD4+ T cells (GrB+CD4+ T‐cells) and promote their differentiation into Th1‐type T‐cells, which in turn produce interferon‐gamma, a powerful tumour‐suppressive cytokine 161. Moreover, in CML and head and neck cancers, dasatinib has been shown to reduce the number of myeloid‐derived suppressor cells (MDSCs), and induce anti‐inflammatory macrophages (defined by increased production of IL‐10, decreased production of IL6, IL‐12p40 and TNF‐alpha, and high expression of LIGHT, SPHK1 and arginase 1), via the inhibition of salt‐inducible kinases 160, 162, 163. Surprisingly, the potential in combining the immunomodulatory effects of Src‐inhibitors with other immunomodulatory therapies has not been extensively studied. Preclinical data in head and neck squamous cell carcinoma (HNSCC) showed inhibition of tumour growth, suggesting that combining dasatinib with anti‐CTLA4 immunotherapy may be a viable treatment approach 164. However in a clinical study of gastrointestinal stromal tumours (GIST), dasatinib and anti‐CTLA4 antibody ipilimumab were well tolerated yet the combination was not synergistic, potentially due to the lack of a biomarker‐driven approach 165. At present there is only one phase II trial underway examining the combination of dasatinib and anti‐PD‐1 therapy nivolumab in nonsmall cell lung cancer (NCT02750514). However due to the strong immunomodulatory effects of Src inhibition seen in vivo, assessment of synergistic combinatorial therapies including dasatinib and other immunomodulatory drugs is warranted. This could be particularly relevant in pancreatic cancer where immunotherapy provides no therapeutic benefit as a result of the immunosuppressive microenvironment that defines these tumours 166.

Combining Src inhibition with additional targeted therapies is another potentially beneficial approach aimed at enhancing antitumour efficacy, while minimising inherent and acquired resistance. This strategy has already shown promise in several cancers 167. Almost 30 years ago, Src tyrosine kinase and EGFR were found to synergistically stimulate EGF‐induced mitogenic cellular responses in fibroblast cultures 168. Since then, Src has been shown to directly phosphorylate EGFR and may also mediate transactivation of EGFR by other receptor signalling pathways 37, 169, 170. The EGF‐mediated RAS/RAF/MEK/ERK pathway (Fig. 1) is one of the major players in the regulation of tumour growth, survival, proliferation, inhibition of apoptosis and autophagy 171, 172, with deregulated activation associated with poor prognosis in solid tumours 173, including PDAC 174.

Targeting this key pro‐tumourigenic molecular pathway has been explored in PDAC with the combination of standard therapy gemcitabine and small molecule EGFR inhibitor erlotinib revealing a modest but significant improvement in patient survival in advanced disease 175, 176, 177. However, significance was lost when this combination was trialled in all‐comers in the adjuvant setting 178. Further analyses revealed that therapeutic benefit of combined gemcitabine/EGFR inhibition associated with KRAS wild‐type tumour status 179, 180 or development of skin rash in patients, which represents another measure of EGFR inhibitor activity 181. Dasatinib has been combined with the EGFR inhibitor, erlotinib in NSCLC, resulting in two partial responses, and a disease control rate of 63% 182. Collectively, these studies highlight the potential utility of this treatment combination when applied in small, but potentially well‐defined subgroups of patients with pancreatic cancer. Moreover, the combination of dasatinib, erlotinib and gemcitabine showed significant synergy in preclinical studies, with potent inhibition of cancer cell proliferation, viability and xenograft tumour growth 183. The triple combination was also shown to overcome constitutive activation of STAT3‐mediated signalling, a key player in PDAC chemoresistance 27, 55, 183, 184, and was shown to be well tolerated, with promising preliminary clinical activity in advanced pancreatic cancer 185. The potential of this therapeutic combination also provides support for the development of a novel multikinase inhibitor (SKLB261) that potently inhibits EGFR, Src and VEGFR2 kinases. In the context of PDAC, this inhibitor effectively inhibited cancer cell proliferation, migration, invasion and induced apoptosis in vitro, and demonstrated potent antiangiogenic effects in pancreatic cancer xenografts, with stronger antitumour activity when compared to dasatinib, erlotinib and gemcitabine monotherapies 186.

Dual Src/MEK blockade using saracatinib/selumetinib presents another interesting therapeutic strategy shown to induce apoptosis of dormant cancer cells and limit tumour recurrence in breast cancer models 187 that may potentially be applied to other solid cancers, including PDAC. Dual targeting of Src and the protein tyrosine phosphatase SHP‐2, required for full activation of the RAS/ERK1/2 pathway, has also shown promise in in vitro and in vivo models of pancreatic cancer. Combined Src/SHP‐2 inhibition resulted in a supra‐additive loss of phosphorylation of Akt and ERK‐1/2, and led to an increase in apoptotic marker expression in L3.6pl and PANC‐1 pancreatic cancer cells. The combination also led to a reduction in cell viability, adhesion, migration and invasion in vitro and reduction in pancreatic tumour formation in vivo, using the L3.6pl orthotopic model 188. The central role for SHP‐2 in oncogenic KRAS‐driven tumours has been therapeutically exploited in other contexts, with most recent data demonstrating potent synergistic antitumour effects of combined SHP‐2 and MEK inhibition in multiple cancer types 189, including genetically engineered models of KRAS‐mutant lung and pancreatic cancer 190. Further exploration of these targeted therapeutic combinations, particularly in molecularly enriched patient subsets, is warranted, with early dose‐finding clinical studies underway (NCT03114319, NCT03634982; Table 1).

Modulation of the upstream and downstream Src signalling components in pancreatic cancer

Modulation of the downstream mediators and interacting partners of Src represents another potentially viable therapeutic approach that is increasingly being investigated (Table 2). Inhibition of FAK decreased PDAC cell growth and migration in vitro 191, 192, and limited pancreatic tumour progression in vivo, doubling the survival in the p48‐Cre;LSL‐KrasG12D;Trp53flox/+ (KPC) mouse model of PDAC 25, 193, 194. FAK inhibitor VS‐4718 treatment further reduced tumour fibrosis and numbers of infiltrating immunosuppressive populations of myeloid‐derived suppressor cells (MDSCs), tumour‐associated macrophages (TAMs) and regulatory T‐cells, sensitising the KPC mouse model to checkpoint immunotherapy 25. As a result, several trials are now focused on combining FAK inhibition with immunotherapies such as trametinib, and pembrolizumab in PDAC (NCT02428270 195, NCT02758587) (Table 2). In addition, FAK inhibitors such as PF‐00562271 are well tolerated and hence show significant promise for the treatment of PDAC 131, 196. Promising preclinical data in malignant pleural mesothelioma, ovarian and other solid tumours suggest that therapeutic responsiveness to FAK inhibition may be guided by Merlin loss 197, 198 or E‐cadherin levels 199. This is supported by positive data from two phase I studies (NCT01138033, NCT01938443) in advanced solid tumours, where improved response to the FAK inhibitor GSK2256098 was observed in Merlin‐negative mesothelioma 131, 133. However, findings of a recent prospective phase II trial in malignant pleural mesothelioma (MPM; COMMAND study), has since failed to confirm Merlin expression as a predictive biomarker of efficacy to a different FAK inhibitor, defactinib 132. The observed discordance in the findings of these studies could potentially be due to a substantial difference in the cut‐offs utilised to define Merlin‐negative or Merlin‐low tumour status, with the Soria et al. 131 and Mak et al. 133 trials more stringently defining Merlin‐negative cancers. These studies also differ in terms of their patient selection and cohort size, with the larger COMMAND trial 132 being a prospective study examining defactinib efficacy as a maintenance therapy in chemo‐responsive advanced MPM, whereas the smaller phase I and Ib studies of the GSK2256098 compound examined efficacy in advanced chemo‐resistant solid tumours, including mesothelioma. Moreover, as defactinib targets both FAK and Pyk2 200 while GSK2256098 is selective for FAK alone, this difference in target selectivity between the two compounds may potentially lead to divergent antitumour activity, and mechanism of action on tumour cells, as well as the distinct components of the tumour microenvironment. Further assessment into the efficacy of FAK inhibition in the context of Merlin loss may still be of interest, particularly in pancreatic cancer where it has yet to be examined. Future trials would however need to consider standardisation of the biomarker analysis and interpretation of Merlin loss, sampling of multiple tumour areas where possible to account for potential intratumoural heterogeneity of molecular marker(s) of interest and incorporation of additional promising biomarkers to aid identification of clinical responders to FAK inhibitor‐based treatment regimens.

Table 2.

Clinical trials in pancreatic cancer associated with targeting downstream mediators and interacting partners of Src kinase.

Signalling pathway Agent Molecular target Cancer type Phase Combination therapy Findings/status Protocol ID Reference
EGFR Erlotinib EGFR Advanced pancreatic cancer III Gemcitabine Completed: modest significant improvement in OS (0.33 months) (= 569). Association between rash and a better outcome was observed NCT00026338 175
Locally advanced pancreatic cancer III Gemcitabine Completed: no significant improvement in OS in combination arm (1.7 months; P = 0.09; = 449) NCT00634725 257
Advanced pancreatic cancer II (Single Arm) Gemcitabine Completed: well tolerated, no significant improvement in PFS as primary measure in unselected cohort (= 30) NCT00810719 258
Advanced pancreatic cancer III Cross‐over design (Gemcitabine vs Capecitabine) Completed: well tolerated, comparable efficacy between the two Erlotinib‐based regimens (= 274). KRAS wild‐type status was associated with an improved overall survival (HR 1.68, P = 0.005) NCT00440167 176, 177
Resected pancreatic cancer (adjuvant) III (open label) Gemcitabine Completed: no improvement in patient survival observed (= 436) and occurrence of rash was not associated with response CONKO‐005 178
Metastatic pancreatic cancer II (single arm) Gemcitabine Completed: improved survival in rash‐positive patients, comparable 1% survival rate to FOLFIRINOX NCT0172948 181
Cetuximab Chimeric monoclonal IgG1 antibody against extracellular III domain of EGFR Advanced pancreatic cancer III Gemcitabine Completed: no significant improvement in survival (= 745) and no association with EGFR IHC NCT00075686 259
Nimotuzumab Humanised IgG2 mAb against extracellular III domain of EGFR Advanced pancreatic cancer IIb (randomised) Gemcitabine Completed: safe and well tolerated. One‐year OS and PFS were significantly improved (= 192). Particularly of benefit in KRAS wild‐type patients NCT00561990 180
FAK PF‐00562271 FAK Advanced solid cancers (incl pancreatic) I Monotherapy Completed: tolerated, MTD established. (= 99; 14% pancreatic) NCT00666926 196
VS‐4718 Advanced pancreatic cancer I Gemcitabine/ Nab‐paclitaxel Terminated: Company de‐prioritised drug development NCT02651727
Defactinib Molecular analysis for therapy choice (MATCH), multiple solid cancers (incl metastatic/ recurrent pancreatic cancer) II (personalised) Monotherapy‐targeted against NF2 inactivation Recruiting NCT02465060
Advanced solid cancers (incl pancreatic) I/II Pembrolizumab (anti‐PD1) Recruiting NCT02758587
Advanced solid cancers (incl pancreatic) I Pembrolizumab and Gemcitabine Phase I Completed (= 17). Well tolerated. Recruiting: Expansion cohort NCT02546531 260
GSK2256098 Recurrent pancreatic cancer II (Single Arm) Trametinib (MEK1/2 inhibitor) Completed: no objective response measured in unselected cohort (= 16). 1 patient with KRAS amplification showed stable disease for 5 months after rapid progression on First‐line FOLFIRINOX; Correlative biomarker studies ongoing from collected material NCT02428270 195
Integrin Cilengitide Cyclic peptide inhibitor of ανβ3/ανβ5 integrins Advanced pancreatic cancer II (randomised, open label) Gemcitabine Completed: well tolerated, no improvements in OS, PFS and response rate in unselected cohort (= 89) EMD 121974 233
Volociximab (M200) Chimeric mAb against human α5β1 integrin Metastatic pancreatic cancer II (single arm, open label) Gemcitabine Completed: well tolerated, awaiting further results NCT00401570 236
IMGN388 Human IgG1 anti‐integrin Ab conjugated to maytansinoid (DM4) Advanced solid cancers I Monotherapy Completed: well tolerated, safety data reported on 26 patients; awaiting final results NCT00721669 261
Hyaluronan PEGPH20 Hyaluronan Metastatic pancreatic cancer Ib/II (randomised) Gemcitabine Completed: tolerated combination therapy, with promising early clinical activity, particularly in patients with HA‐high tumours (IHC). Phase II terminated due to change in standard‐of‐care chemotherapy treatment NCT01453153 262
Metastatic pancreatic cancer II (randomised, open label) Gemcitabine/ Nab‐paclitaxel Completed: improved PFS as primary endpoint in the overall cohort (= 279), with the greatest improvement in PFS observed in patients with HA‐high tumours (prevalence of 34%) NCT01839487 248
Advanced pancreatic cancer NA (non‐randomised, open label) Gemcitabine/ Nab‐paclitaxel Recruiting: Interim results indicate adding Rivaroxaban is safe and effectively controls thromboembolic events, with PEGPH20‐combination therapy showing encouraging early responses (= 28) NCT02921022 252
Borderline resectable pancreatic cancer (neoadjuvant) II (single arm, open label) Gemcitabine/ Nab‐paclitaxel Recruiting NCT02487277 263
Metastatic pancreatic cancer III (randomised) Gemcitabine/ Nab‐paclitaxel Recruiting NCT02715804
Locally advanced pancreatic cancer II (single arm, open label) Gemcitabine and radiation No longer recruiting, no results posted NCT02910882
Metastatic pancreatic cancer I/II modified (m) FOLFIRINOX Phase II closed as PEGPH20 with mFFOX caused significantly increased toxicity and decreased treatment duration compared to mFFOX alone NCT01959139 253
Resectable pancreatic cancer (neoadjuvant) NA Cetuximab Study closed due to slow accrual NCT02241187 264
Advanced (chemotherapy‐resistant) pancreatic cancer I Avelumab Recruiting NCT03481920
Advanced (chemotherapy‐resistant) pancreatic cancer: HA high II (single arm, open label) Pembrolizumab Not yet recruiting NCT03634332
Metastatic pancreatic cancer Ib/II (randomised, open label) Atezolizumab Recruiting NCT03193190
Rho/ROCK AT13148 AGC Kinase Advanced solid cancers I Monotherapy Completed: tolerable, dose escalation ongoing (= 30), awaiting final results NCT01585701 201
PI3K/Akt Pathway MK2206 Akt (pan) Advanced pancreatic cancer I/Ib (randomised, open label) Dinaciclib (CDK inhibitor) Completed: results pending NCT01783171
Recurrent metastatic pancreatic cancer II (randomised, open label) Selumetinib (MEK1/2 inhibitor) Completed:No improvement in OS, and increased rate of adverse events in experimental arm, compared to mFOLFOX standard therapy (= 137) NCT01658943 223
Afuresertib (GSK2110183) Akt (pan) Advanced solid cancers (incl pancreatic) I/II (open label) Trametinib (MEK1/2 inhibitor) Completed: Poor tolerability with daily dosing. Potential for intermittent administration discussed within study NCT01476137 224
Uprosertib (GSK2141795) Akt (pan) Advanced solid cancers (incl pancreatic) I Trametinib (MEK1/2 inhibitor) Completed: results pending NCT01138085
Oleandrin (PBI‐05204) Akt (pan) Metastatic pancreatic cancer II (single arm, open label) Monotherapy Active, not recruiting NCT02329717
AZD5363 Akt (pan) Molecular analysis for therapy choice (MATCH), multiple solid cancers (incl metastatic/ recurrent pancreatic cancer) II (personalised) Monotherapy‐targeted against Akt mutations Recruiting NCT02465060
Perifosine Akt (pan) Advanced pancreatic cancer II (single arm, open label) Monotherapy Completed: no results posted NCT00053924
Advanced pancreatic cancer II (single arm, open label) Monotherapy Terminated: Significant treatment‐related toxicity (= 10). Disease progression noted NCT00059982 265
Alpelisib (BYL719) PI3Kα Advanced solid cancers (incl pancreatic neuroendocrine neoplasms) Ib Everolimus (mTOR) + Exemestane (Aromatase) Active, not recruiting NCT02077933
Advanced pancreatic cancer I/Ib (single arm, open label) Gemcitabine/ Nab‐paclitaxel Active, not recruiting NCT02155088
Buparlisib (BKM120) PI3K (pan) Metastatic pancreatic cancer I (single arm, open label) mFOLFOX6 Completed: results pending NCT01571024
Advanced solid cancers (incl pancreatic) Ib (single arm, open label) Trametinib (MEK1/2 inhibitor) Completed: long‐term tolerability of the combination was challenging, with promising efficacy in select tumour types (ovarian) (= 113; 47 patients in the expansion cohort) NCT01155453 222
Advanced solid cancers (incl pancreatic) Ib (single arm, open label) MEK162 (MEK1/2 inhibitor) Completed: results pending NCT01363232
Sirolimus (Rapamycin) mTORC1 Advanced (gemcitabine‐resistant) pancreatic cancer II (single arm, open label) Monotherapy Completed: well tolerated, marginal efficacy, examined biomarker (p70S6K IHC) did not correlate with activity (= 31) NCT00499486 204
Advanced pancreatic cancer II (single arm, open label) Monotherapy Recruiting NCT03662412
Advanced solid cancers (incl pancreatic ductal and acinar adenocarcinoma) I Vismodegib (Hedgehog inhibitor) Suspended: results pending NCT01537107
Advanced solid cancers I Sunitinib (RTK inhibitor) Completed: results pending NCT00583063
Advanced solid cancers I Sorafenib (Raf, VEGFR inhibitor) Completed: results pending NCT00449280
Metastatic pancreatic cancer I/II (randomised, open label) Metformin Active, not recruiting NCT02048384
SM‐88 Combination: metyrosine‐derivative + low‐dose sirolimus, phenytoin + methoxsalen Metastatic (chemotherapy‐resistant) pancreatic cancer II (randomised) Monotherapy Recruiting: Preliminary results are promising, with therapy well tolerated (= 28), with a median of 4.3 months of follow‐up after treatment initiation, 67.8% still alive (trial ongoing), promising compared with historical data NCT03512756 213
Temsirolimus mTORC1 Metastatic pancreatic cancer II (single arm, open label) Gemcitabine Terminated NCT00593008
Advanced solid cancers (incl pancreatic) I/II (single arm, open label) Nivolumab Terminated: Investigator no longer at site to enrol patients or write up data NCT02423954
Advanced pancreatic cancer II (single arm, open label) Monotherapy Terminated: Study closed due to significant treatment‐related toxicity (= 5). Disease progression noted in 2 patients NCT00075647 266
Everolimus (RAD001) mTORC1 Advanced or metastatic pancreatic cancer II (single arm, open label) Erlotinib Terminated: Study closed due to significant treatment‐related toxicity (= 15). Lack of objective responses noted. Study suggests activation of negative feedback loops following mTOR inhibition may explain lack of efficacy, and which may require simultaneous inhibition of multiple PI3K pathway components to elicit response NCT00640978 266
Metastatic (gemcitabine‐resistant) pancreatic cancer II (single arm, open label) Monotherapy Completed: well tolerated, minimal clinical activity as monotherapy in unselected cohort (= 33) NCT00409292 267
Advanced or metastatic pancreatic cancer I/II (randomised, open label) Irinotecan and Cetuximab Terminated: emergence of FOLFIRINOX and slow recruitment. Triple combination showed similar PFS but increased OS compared to Capecitabine + Oxaliplatin (7.7 vs 4.5 months = 0.04) (= 26) NCT01042028 268
Metastatic pancreatic cancer II (non‐randomised, open label) Capecitabine and Cetuximab Completed: MTD determined; partial response documented in 2 patients (6.5%), and 5 (16.1%) had stable disease. Considerable epidermal and mucosal toxicities NCT01077986 269
Metastatic (gemcitabine refractory) pancreatic cancer I/II (single arm, open label) Sorafenib Completed: awaiting results NCT00981162
Advanced and/or metastatic pancreatic cancer I/II (single arm, open label) Gemcitabine Completed: MTD determined. Clinical benefit (CR, PR or stable disease) observed in 78% patients (= 21) NCT00560963 270
Pancreatic neuroendocrine tumours I/II (open label) X‐82 (VEGFR/PDGFR inhibitor) Active, not recruiting. Prolonged stable disease (3‐23 months) (= 10) NCT01784861 271
Advanced GI neuroendocrine tumours (incl pancreatic) II (single arm, open label) Monotherapy Active, recruitment complete (= 25). Early data indicate therapy is well tolerated with signs of efficacy (high rate of PR) NCT01648465 272
Vistusertib mTORC1/2 Advanced solid cancers (incl pancreatic) II (personalised, single arm) Monotherapy‐targeted against RICTOR amplifications Not yet recruiting NCT03166904
Advanced solid cancers (incl pancreatic) II (personalised, single arm) Monotherapy‐targeted against TSC1/2 mutations Not yet recruiting NCT03166176
Dactolisib PI3K/mTOR Advanced solid cancers (incl pancreatic) Ib (open label) MEK162 (MEK1/2 inhibitor) Completed: results pending NCT01337765
Gedatolisib PI3K/mTOR Advanced solid cancers (incl pancreatic) I (single arm, open label) Palbociclib Recruiting NCT03065062

Several inhibitors that target Rho GTPase or its downstream effectors including Rho‐associated kinases (ROCK) have shown antitumour activity in preclinical models, which we have reviewed previously 87, 88. Most recently, fasudil, an inexpensive, off‐patent ROCK inhibitor, may present a promising new treatment approach for PDAC. It has recently been shown that using a short‐term ‘priming’ treatment approach to inhibit ROCK signalling can reduce tissue stiffness, improve vascular patency, increase tumour perfusion, decrease in vivo primary tumour growth, metastasis and improve response to standard of care therapy 23, 92, similar to chronic fasudil treatment 89. Newer ROCK inhibitors (such as ripasudil, CCT129254 or AT13148), are currently being trialled, and utilise a similar ‘priming’ 92, 93 or intermittent regime 201. The rationale behind this novel treatment scheduling involves modulating or ‘loosening’ the ECM, via ROCK inhibition, prior to chemotherapy administration in order to improve chemotherapy drug perfusion and reduce toxicity 92. Potentially, this regime could be applied for the use of other stromal‐based therapies in PDAC as well as other stromal‐driven cancers.

Furthermore, there has been significant research dedicated to targeting the PI3K/AKT signalling pathway in PDAC due to its role in cell metabolism, cell cycle, protein synthesis and apoptosis 202. Rapamycin, an mTORC1 inhibitor, showed promising preclinical results in PDAC, significantly halting disease progression in PI3K/AKT‐activated tumours 203. However clinical data failed to demonstrate a benefit, particularly when administered as monotherapy (Table 2) 204. This may further be explained by mTORC1 being involved in complex negative feedback loops that restrain upstream signalling. For example, inhibition of mTORC1 drives activation of PI3K‐, AKT‐ or ERK pathways 205, which in turn limits the efficacy of mTORC‐inhibitors as targeted therapies 206. More recently developed dual ATP‐competitive agents that target mTORC1/mTORC2 have shown favourable results 207, 208 with AZD2014 effectively inhibiting PDAC cell division (G1 arrest), proliferation, and invasion in vitro 158, 160 and prolonging survival in the KPC mouse model of PDAC 109, 208, 209. However there is still some debate as to whether blocking mTORC1/2 leads to the adaptive activation of the PI3K‐AKT pathway 209, and consequently whether multiple targeting of this network is required to effectively interfere with both branches of adaptive signalling and to elicit a durable therapeutic response.

The combination of Cyclin‐dependent Kinase (CDK) inhibitors with PI3K pathway inhibition has been shown to inhibit tumour growth and metastasis in a variety of cancers including PDAC 210, 211, with a need for molecular stratification into responsive subtypes 212. Furthermore, multitarget, unique formulations, including SM‐88, a combination of a tyrosine derivative (D,L‐alpha‐metyrosine), mTOR inhibitor (sirolimus), CYP3a4 inducer (phenytoin) and oxidative stress catalyst (methoxsalen), are showing encouraging efficacy in early stage trials, particularly in patients with advanced pancreatic cancer (Table 2) 213, who have frequently exhausted all options. There is also ample evidence supporting the combination of PI3K/AKT/mTOR inhibitors with tyrosine kinase inhibitors (TKIs). Cancers with active/overexpressed TKIs often display resistance to TKIs through PI3K signalling 214. In addition, targeting RAS/RAF/MEK/ERK pathway in combination with PI3K/AKT/mTOR inhibitors is another promising strategy because there is significant stimulatory crosstalk 214. Synergy has previously been shown between a MEK‐inhibitor and PI3K/mTOR inhibitor in a lung cancer model, where inhibition of MEK/ERK was shown to stabilise BIM, and PI3K/AKT inhibition upregulated PUMA via FOXO, all of which are key mediators of apoptosis 215, 216. Inhibition of the MAPK pathway has also been shown to associate with increased PI3K pathway activity 217, 218. This therapeutic combination could also be beneficial in PDAC, as an alternative approach for inhibiting oncogenic Kras, which is located upstream of MEK/ERK and PI3K. Thus far, attempts at targeting the most frequently mutated protein in PDAC, KRAS, have been unsuccessful 14, 219. Whilst the combination of MEK inhibitors with alternative pathway inhibitors such as PI3K or Src has shown early promise 218, 220, 221, the combinations, including addition of chemotherapies, may require an alternative, intermittent dosing regimen design due to issues with chronic administration 222, 223, 224, and are yet to be systematically examined in PDAC. Preclinical data suggest that therapeutic efficacy may be dependent on PDAC subtype, as well as MEK activity and expression 225, with further investigation, including determination of biologically effective dose(s) of targeted therapies, testing and implementation of alternative dosing regimens, warranted.

Given the importance of the integrin/Src/FAK signalling in diverse cancer types, significant research has also gone into targeting molecules upstream of Src, including integrins, which critically modulates ECM mechanics and cytoskeleton stability, stellate cell activation 226, cancer cell survival and angiogenesis 59 and most recently, production of tumour‐promoting cytokines and chemokines 227. With each integrin comprising an α and β transmembrane subunit, most studies have focused on testing avβ1, avβ3, avβ5 integrin antagonists, the most promising of which is cilengitide. Cilengitide is an RGD (arginine‐glycine‐aspartic acid) peptide which is selective against avβ3, avβ5 integrins 228. Cilengitide was shown to have antitumour activity in recurrent and newly diagnosed glioblastoma 229, 230, 231, 232; however, further phase III studies showed no significant differences in median overall survival 231, with similar negative findings in PDAC when examined in all‐comers 233. In contrast, results from a phase I study suggest promising early signals of activity with cilengitide and chemoradiotherapy combination in advanced nonsmall cell lung cancer 234. Clinical trials of further integrin antagonists, including intetumumab, volociximab, ATN‐161 (Ac‐PHSCN‐NH2 peptide), abituzumab and etaracizumab, all of which are antibodies or peptide mimetics, have largely yielded no improvements in patient progression‐free or overall survival (Table 2) 235, 236; however, specific studies in colon cancer suggest that their antitumour activity may be linked to the presence of a biomarker 237, and, alternatively, may specifically inhibit the progression of bone‐associated metastases in prostate cancer 238. Adding to the complexity, anti‐integrin compounds may increase intratumoural hypoxia, leading to increased tumour growth, metastasis and chemoresistance in certain settings 239, 240, process that is dose‐ and/or tumour type‐dependent 65, 241. Reynolds et al. 241 showed that in fact, low (nanomolar) concentrations of avβ3, avβ5 inhibitors can paradoxically promote VEGF‐mediated angiogenesis by altering avβ3 integrin and VEGFR‐2 trafficking, stimulating cancer growth.

Hence, more recent research efforts have focussed on utilising these agents as part of ‘vascular normalisation’, whereby improved tumour blood flow increases drug delivery 242. However as this approach is highly time‐ and dose‐dependent, its clinical implementation may be challenging 243. Specifically, in pancreatic cancer, cilengitide has been effectively applied in combination with chemotherapy using a strategy called ‘vascular promotion’, aimed at improving delivery of chemotherapy to the tumour 244. Although the combination has yet to be trialled in the clinic, preclinical evidence is positive. Co‐administration of low‐dose therapy regimen of cilengitide and verapamil increased tumour blood flow and perfusion, promoted gemcitabine delivery inside growing pancreatic tumours, ultimately leading to reduced primary tumour growth, metastasis and significantly improved survival in multiple models of PDAC with minimal side effects 244. This dual therapy also increased levels of proteins involved in active transport of gemcitabine into cells, and production of active metabolites, further enhancing gemcitabine potency. Vascular promotion is also associated with reduced hypoxia and desmoplasia, salient features of PDAC 244. In addition, volociximab, an integrin α5β1 blocking antibody, has completed phase II trials in combination with gemcitabine in metastatic pancreatic cancer, with results pending (NCT00401570). Of note, mutant P53 has been shown to regulate α5β1 signalling and EGFR, which suggests there may also be potential for molecular stratification 245.

Another major advance in ECM‐targeting is the development of agents that break down hyaluronic acid (HA). HA is a large, linear, glycosaminoglycan that plays an important structural role in the ECM, and accumulates in conditions involving rapid and invasive cell division, including cancer. HA regulates interstitial gel fluid pressure within tumours, often impacting on drug delivery. Pegylated recombinant human hyaluronidase (PEGPH20) and 4‐methylumbelliferone are two key examples of compounds that inhibit and/or break down HA. Of note, PEGPH20 has already shown significant promise in PDAC. HA degradation following PEGPH20 treatment has been shown to normalise interstitial fluid pressures and re‐expand the microvasculature by increasing the diameter but not the total number of blood vessels within PDAC tumours 246. This in turn significantly improved chemotherapeutic response in the KPC murine model of PDAC, resulting in a near doubling of overall survival 246, 247. Clinical studies of PEGPH20 are also promising with phase II data already demonstrating significant efficacy of this agent when combined with chemotherapy, effect particularly prominent in patients with HA‐high tumours 248, highlighting the potential utility of intratumoural HA as a predictive biomarker of response 248, 249, 250. Favourable results are particularly observed when PEGPH20 is combined with Gemcitabine and Abraxane 248, 251, 252, whereas FOLFIRINOX in contrast may be better utilised in other settings 253. Development of a liquid biopsy‐based companion diagnostic for selecting potential PEGPH20 responders is also underway 254. Consequently several phase II/III clinical trials are now investigating further the clinical efficacy of PEGPH20, in combination with standard of care chemotherapies (Table 2) (NCT02487277, NCT02715804), or immune checkpoint inhibition (NCT03481920; NCT03634332, NCT03193190) in HA‐high molecular subgroups of PDAC 248, 255. These encouraging early clinical findings highlight the potential of stromal components as viable therapeutic targets, supporting further clinical development of PEGPH20 as well as detailed exploration of new biomarker‐driven therapeutic combinations utilising this agent.

Future perspectives for inhibition of Src signalling in pancreatic cancer

The extraordinary and constantly expanding understanding of the role of Src signalling in pancreatic cancer biology and treatment supports the foundation for the specific inhibition of this complex network in PDAC. However, the presumption that a single‐targeted therapy will improve survival in such an aggressive disease is unrealistic. Unfortunately, most targeted therapies are at best only transiently effective, with cancer cells rapidly acquiring resistance, often leading to more rapid disease progression. This is supported by the numerous unsuccessful nonbiomarker‐driven clinical trials that have been summarised in this review.

Further understanding of the intricacies in integrin/Src/FAK and downstream signalling in the various tumour compartments will determine whether the inhibitors of this complex network may serve as effective treatments for newly diagnosed or recurrent tumours and will establish optimal combinations with radiation, cytotoxic chemotherapy and other targeted molecular compounds. Given the need for co‐targeting of multiple cancer capabilities to overcome the high therapeutic resistance of pancreatic tumours, future clinical applications of multiagent therapies will likely require a more innovative approach to dosing, including use of biologically effective doses of targeted agents (integrin/Src/FAK), and alternative dosing schedules such as ‘priming’ or ‘maintenance therapy’ to ensure maximal benefit to the patient 152. Finally, the emerging efficacy of Src pathway inhibitors in combination with other targeted and/or cytotoxic therapies, when examined in a molecular subtype‐specific context 248, 249, and with longitudinal tracking of long‐term therapeutic responsiveness, reveals significant potential as a personalised medicine strategy for pancreatic cancer, and provides real hope for patients in the future.

Conflict of interest

The authors declare no conflict of interest.

Acknowledgements

Marina Pajic acknowledges fellowship support from the NHMRC 1162556, Cancer Institute NSW and Philip Hemstritch philanthropic fellowship, with project grant support from the NHMRC 1162860 and Cancer Australia 1143699. Paul Timpson acknowledges support from a Len Ainsworth fellowship in pancreatic cancer, and an NHMRC Senior Research fellowship, with project grant support from the Avner Pancreatic Cancer Foundation. This work was also supported by Suttons Motors and Sydney Catalyst.

Paul Timpson and Marina Pajic contributed equally to this article.

Contributor Information

Paul Timpson, Email: p.timpson@garvan.org.au.

Marina Pajic, Email: m.pajic@garvan.org.au.

References

  • 1. Albini A & Sporn MB (2007) The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer 7, 139–147. [DOI] [PubMed] [Google Scholar]
  • 2. Maman S & Witz IP (2018) A history of exploring cancer in context. Nat Rev Cancer 18, 359–376. [DOI] [PubMed] [Google Scholar]
  • 3. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM & Matrisian LM (2014) Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Can Res 74, 2913–2921. [DOI] [PubMed] [Google Scholar]
  • 4. Siegel RL, Miller KD & Jemal A (2017) Cancer statistics, 2017. CA Cancer J Clin 67, 7–30. [DOI] [PubMed] [Google Scholar]
  • 5. Chin V, Nagrial A, Sjoquist K, O'Connor CA, Chantrill L, Biankin AV, Scholten RJ & Yip D (2018) Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev 3, CD011044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou‐Bourgade S, de la Fouchardière C et al (2011) FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 364, 1817–1825. [DOI] [PubMed] [Google Scholar]
  • 7. Yu IS & Cheung WY (2018) A contemporary review of the treatment landscape and the role of predictive and prognostic biomarkers in pancreatic adenocarcinoma. Can J Gastroenterol Hepatol 2018, 1863535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN et al (2013) Increased survival in pancreatic cancer with nab‐paclitaxel plus gemcitabine. N Engl J Med 369, 1691–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Conroy T (2018) Unicancer GI PRODIGE 24/CCTG PA.6 trial: A multicenter international randomized phase III trial of adjuvant mFOLFIRINOX versus gemcitabine (gem) in patients with resected pancreatic ductal adenocarcinomas. in ASCO Annual Meeting.
  • 10. Vogel JA, Rombouts SJ, de Rooij T, van Delden OM, Dijkgraaf MG, van Gulik TM, van Hooft JE, van Laarhoven HW, Martin RC, Schoorlemmer A et al (2017) Induction chemotherapy followed by resection or irreversible electroporation in locally advanced pancreatic cancer (IMPALA): a prospective cohort study. Ann Surg Oncol 24, 2734–2743. [DOI] [PubMed] [Google Scholar]
  • 11. Waddel N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K et al (2015) Whole Genome Sequencing redefines the mutational landscape of Pancreatic Cancer. Nature 518, 495–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, Miller DK, Christ AN, Bruxner TJ, Quinn MC et al (2016) Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 531, 47–52. [DOI] [PubMed] [Google Scholar]
  • 13. Alexandrov LB, Nik‐Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, Bignell GR, Bolli N, Borg A, Borresen‐Dale AL et al (2013) Signatures of mutational processes in human cancer. Nature 500, 415–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Parkin A, Man J, Chou A, Nagrial AM, Samra J, Gill AJ, Timpson P & Pajic M (2018) The evolving understanding of the molecular and therapeutic landscape of pancreatic ductal adenocarcinoma. Diseases 6, 103 10.3390/diseases6040103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Balachandran VP, Luksza M, Zhao JN, Makarov V, Moral JA, Remark R, Herbst B, Askan G, Bhanot U, Senbabaoglu Y et al (2017) Identification of unique neoantigen qualities in long‐term survivors of pancreatic cancer. Nature 551, 512–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Humphrey ES, Su SP, Nagrial AM, Hochgrafe F, Pajic M, Lehrbach GM, Parton RG, Yap AS, Horvath LG, Chang DK et al (2016) Resolution of novel pancreatic ductal adenocarcinoma subtypes by global phosphotyrosine profiling. Mol Cell Proteomics 15, 2671–2685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Collisson EA, Sadanandam A, Olson P, Gibb WJ, Truitt M, Gu S, Cooc J, Weinkle J, Kim GE, Jakkula L et al (2011) Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med 17, 500–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A et al (2008) Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321, 1801–1806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Connor AA, Denroche RE, Jang GH, Timms L, Kalimuthu SN, Selander I, McPherson T, Wilson GW, Chan‐Seng‐Yue MA, Borozan I et al (2017) Association of distinct mutational signatures with correlates of increased immune activity in pancreatic ductal adenocarcinoma. JAMA Oncol 3, 774–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS et al (2017) Mismatch repair deficiency predicts response of solid tumors to PD‐1 blockade. Science 357, 409–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Nik‐Zainal S, Davies H, Staaf J, Ramakrishna M, Glodzik D, Zou X, Martincorena I, Alexandrov LB, Martin S, Wedge DC et al (2016) Landscape of somatic mutations in 560 breast cancer whole‐genome sequences. Nature 534, 47–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Yates LR, Knappskog S, Wedge D, Farmery JHR, Gonzalez S, Martincorena I, Alexandrov LB, Van Loo P, Haugland HK, Lilleng PK et al (2017) Genomic evolution of breast cancer metastasis and relapse. Cancer Cell 32, 169–184 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Vennin C, Murphy KJ, Morton JP, Cox TR, Pajic M & Timpson P (2018) Reshaping the tumor stroma for treatment of pancreatic cancer. Gastroenterology 154, 820–838. [DOI] [PubMed] [Google Scholar]
  • 24. Neesse A, Michl P, Frese KK, Feig C, Cook N, Jacobetz MA, Lolkema MP, Buchholz M, Olive KP, Gress TM et al (2011) Stromal biology and therapy in pancreatic cancer. Gut 60, 861–868. [DOI] [PubMed] [Google Scholar]
  • 25. Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, Nywening TM, Hawkins WG, Shapiro IM, Weaver DT et al (2016) Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med 22, 851–860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Sinha S & Leach SD (2016) New insights in the development of pancreatic cancer. Curr Opin Gastroenterol 32, 394–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Wormann SM, Song L, Ai J, Diakopoulos KN, Kurkowski MU, Gorgulu K, Ruess D, Campbell A, Doglioni C, Jodrell D et al (2016) Loss of P53 function activates JAK2‐STAT3 signaling to promote pancreatic tumor growth, stroma modification, and gemcitabine resistance in mice and is associated with patient survival. Gastroenterology 151, 180–193 e12. [DOI] [PubMed] [Google Scholar]
  • 28. Apte MV, Wilson JS, Lugea A & Pandol SJ (2013) A starring role for stellate cells in the pancreatic cancer microenvironment. Gastroenterology 144, 1210–1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Morton JP, Karim SA, Graham K, Timpson P, Jamieson N, Athineos D, Doyle B, McKay C, Heung MY, Oien KA et al (2010) Dasatinib inhibits the development of metastases in a mouse model of pancreatic ductal adenocarcinoma. Gastroenterology 139, 292–303. [DOI] [PubMed] [Google Scholar]
  • 30. Frame MC (2002) Src in cancer: deregulation and consequences for cell behaviour. Biochem Biophys Acta 1602, 114–130. [DOI] [PubMed] [Google Scholar]
  • 31. Canel M, Serrels A, Miller D, Timpson P, Serrels B, Frame MC & Brunton VG (2010) Quantitative in vivo imaging of the effects of inhibiting integrin signaling via Src and FAK on cancer cell movement: effects on E‐cadherin dynamics. Can Res 70, 9413–9422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Erami Z, Herrmann D, Warren SC, Nobis M, McGhee EJ, Lucas MC, Leung W, Reischmann N, Mrowinska A, Schwarz JP et al (2016) Intravital FRAP imaging using an E‐cadherin‐GFP mouse reveals disease‐ and drug‐dependent dynamic regulation of cell‐cell junctions in live tissue. Cell Rep 14, 152–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Nobis M, McGhee EJ, Morton JP, Schwarz JP, Karim SA, Quinn J, Edward M, Campbell AD, McGarry LC, Evans TR et al (2013) Intravital FLIM‐FRET imaging reveals dasatinib‐induced spatial control of src in pancreatic cancer. Can Res 73, 4674–4686. [DOI] [PubMed] [Google Scholar]
  • 34. Nobis M, Herrmann D, Warren SC, Kadir S, Leung W, Killen M, Magenau A, Stevenson D, Lucas MC, Reischmann N et al (2017) A RhoA‐FRET biosensor mouse for intravital imaging in normal tissue homeostasis and disease contexts. Cell Rep 21, 274–288. [DOI] [PubMed] [Google Scholar]
  • 35. Timpson P, McGhee EJ, Morton JP, von Kriegsheim A, Schwarz JP, Karim SA, Doyle B, Quinn JA, Carragher NO, Edward M et al (2011) Spatial regulation of RhoA activity during pancreatic cancer cell invasion driven by mutant p53. Can Res 71, 747–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Stehelin D, Varmus HE, Bishop JM & Vogt PK (1976) DNA related to the transforming gene(s) of avian sarcoma viruses is present in normal avian DNA. Nature 260, 170–173. [DOI] [PubMed] [Google Scholar]
  • 37. Chen Z, Oh D, Dubey AK, Yao M, Yang B, Groves JT & Sheetz M (2018) EGFR family and Src family kinase interactions: mechanics matters? Curr Opin Cell Biol 51, 97–102. [DOI] [PubMed] [Google Scholar]
  • 38. Chen Z, Oh D, Biswas KH, Yu CH, Zaidel‐Bar R & Groves JT (2018) Spatially modulated ephrinA1:EphA2 signaling increases local contractility and global focal adhesion dynamics to promote cell motility. Proc Natl Acad Sci USA 115, E5696–E5705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Je DW, O YM, Ji YG, Cho Y & Lee DH (2014) The inhibition of SRC family kinase suppresses pancreatic cancer cell proliferation, migration, and invasion. Pancreas 43, 768–776. [DOI] [PubMed] [Google Scholar]
  • 40. Ischenko I, Guba M, Yezhelyev M, Papyan A, Schmid G, Green T, Fennell M, Jauch KW & Bruns CJ (2007) Effect of Src kinase inhibition on metastasis and tumor angiogenesis in human pancreatic cancer. Angiogenesis 10, 167–182. [DOI] [PubMed] [Google Scholar]
  • 41. Thomas SM & Brugge JS (1997) Cellular functions regulated by Src family kinases. Annu Rev Cell Dev Biol 13, 513–609. [DOI] [PubMed] [Google Scholar]
  • 42. Ishizawar R & Parsons SJ (2004) c‐Src and cooperating partners in human cancer. Cancer Cell 6, 209–214. [DOI] [PubMed] [Google Scholar]
  • 43. Yeatman TJ (2004) A renaissance for SRC. Nat Rev Cancer 4, 470–480. [DOI] [PubMed] [Google Scholar]
  • 44. Serrels A, Macpherson IR, Evans TR, Lee FY, Clark EA, Sansom OJ, Ashton GH, Frame MC & Brunton VG (2006) Identification of potential biomarkers for measuring inhibition of Src kinase activity in colon cancer cells following treatment with dasatinib. Mol Cancer Ther 5, 3014–3022. [DOI] [PubMed] [Google Scholar]
  • 45. Zhang J, Wang S, Jiang B, Huang L, Ji Z, Li X, Zhou H, Han A, Chen A, Wu Y et al (2017) c‐Src phosphorylation and activation of hexokinase promotes tumorigenesis and metastasis. Nat Commun 8, 13732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Carragher NO, Walker SM, Scott Carragher LA, Harris F, Sawyer TK, Brunton VG, Ozanne BW & Frame MC (2006) Calpain 2 and Src dependence distinguishes mesenchymal and amoeboid modes of tumour cell invasion: a link to integrin function. Oncogene 25, 5726–5740. [DOI] [PubMed] [Google Scholar]
  • 47. Liu ST, Pham H, Pandol SJ & Ptasznik A (2013) Src as the link between inflammation and cancer. Front Physiol 4, 416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Yokoi K, Hawke D, Oborn CJ, Jang JY, Nishioka Y, Fan D, Kim SW, Kim SJ & Fidler IJ (2011) Identification and validation of SRC and phospho‐SRC family proteins in circulating mononuclear cells as novel biomarkers for pancreatic cancer. Transl Oncol 4, 83–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Ramnath RD, Sun J & Bhatia M (2009) Involvement of SRC family kinases in substance P‐induced chemokine production in mouse pancreatic acinar cells and its significance in acute pancreatitis. J Pharmacol Exp Ther 329, 418–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Lutz MP, Esser IB, Flossmann‐Kast BB, Vogelmann R, Luhrs H, Friess H, Buchler MW & Adler G (1998) Overexpression and activation of the tyrosine kinase Src in human pancreatic carcinoma. Biochem Biophys Res Comm 243, 503–508. [DOI] [PubMed] [Google Scholar]
  • 51. Hakam A, Fang Q, Karl R & Coppola D (2003) Coexpression of IGF‐1R and c‐Src proteins in human pancreatic ductal adenocarcinoma. Dig Dis Sci 48, 1972–1978. [DOI] [PubMed] [Google Scholar]
  • 52. Nuche‐Berenguer B, Ramos‐Alvarez I & Jensen RT (2016) Src kinases play a novel dual role in acute pancreatitis affecting severity but no role in stimulated enzyme secretion. Am J Physiol Gastrointest Liver Physiol 310, G1015–G1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Aleshin A & Finn RS (2010) SRC: a century of science brought to the clinic. Neoplasia 12, 599–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Ito H, Gardner‐Thorpe J, Zinner MJ, Ashley SW & Whang EE (2003) Inhibition of tyrosine kinase Src suppresses pancreatic cancer invasiveness. Surgery 134, 221–226. [DOI] [PubMed] [Google Scholar]
  • 55. Nagaraj NS, Smith JJ, Revetta F, Washington MK & Merchant NB (2010) Targeted inhibition of SRC kinase signaling attenuates pancreatic tumorigenesis. Mol Cancer Ther 9, 2322–2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Trevino JG, Summy JM, Lesslie DP, Parikh NU, Hong DS, Lee FY, Donato NJ, Abbruzzese JL, Baker CH & Gallick GE (2006) Inhibition of SRC expression and activity inhibits tumor progression and metastasis of human pancreatic adenocarcinoma cells in an orthotopic nude mouse model. Am J Pathol 168, 962–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Yezhelyev MV, Koehl G, Guba M, Brabletz T, Jauch KW, Ryan A, Barge A, Green T, Fennell M & Bruns CJ (2004) Inhibition of SRC tyrosine kinase as treatment for human pancreatic cancer growing orthotopically in nude mice. Clin Cancer Res 10, 8028–8036. [DOI] [PubMed] [Google Scholar]
  • 58. Duxbury MS, Ito H, Zinner MJ, Ashley SW & Whang EE (2004) Inhibition of SRC tyrosine kinase impairs inherent and acquired gemcitabine resistance in human pancreatic adenocarcinoma cells. Clin Cancer Res 10, 2307–2318. [DOI] [PubMed] [Google Scholar]
  • 59. Desgrosellier JS & Cheresh DA (2010) Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer 10, 9–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Samuel MS, Lopez JI, McGhee EJ, Croft DR, Strachan D, Timpson P, Munro J, Schroder E, Zhou J, Brunton VG et al (2011) Actomyosin‐mediated cellular tension drives increased tissue stiffness and beta‐catenin activation to induce epidermal hyperplasia and tumor growth. Cancer Cell 19, 776–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ibbetson SJ, Pyne NT, Pollard AN, Olson MF & Samuel MS (2013) Mechanotransduction pathways promoting tumor progression are activated in invasive human squamous cell carcinoma. Am J Pathol 183, 930–937. [DOI] [PubMed] [Google Scholar]
  • 62. Timpson P, Jones GE, Frame MC & Brunton VG (2001) Coordination of cell polarization and migration by the Rho family GTPases requires Src tyrosine kinase activity. Curr Biol 11, 1836–1846. [DOI] [PubMed] [Google Scholar]
  • 63. Hamidi H, Pietila M & Ivaska J (2016) The complexity of integrins in cancer and new scopes for therapeutic targeting. Br J Cancer 115, 1017–1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A, Reinhart‐King CA, Margulies SS, Dembo M, Boettiger D et al (2005) Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254. [DOI] [PubMed] [Google Scholar]
  • 65. Carter A (2010) Integrins as target: first phase III trial launches, but questions remain. J Natl Cancer Inst 102, 675–677. [DOI] [PubMed] [Google Scholar]
  • 66. Tod J, Hanley CJ, Morgan MR, Rucka M, Mellows T, Lopez MA, Kiely P, Moutasim KA, Frampton SJ, Sabnis D et al (2017) Pro‐migratory and TGF‐beta‐activating functions of alphavbeta6 integrin in pancreatic cancer are differentially regulated via an Eps8‐dependent GTPase switch. J Pathol 243, 37–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Krebs AM, Mitschke J, Lasierra Losada M, Schmalhofer O, Boerries M, Busch H, Boettcher M, Mougiakakos D, Reichardt W, Bronsert P et al (2017) The EMT‐activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat Cell Biol 19, 518–529. [DOI] [PubMed] [Google Scholar]
  • 68. David CJ, Huang YH, Chen M, Su J, Zou Y, Bardeesy N, Iacobuzio‐Donahue CA & Massague J (2016) TGF‐beta tumor suppression through a lethal EMT. Cell 164, 1015–1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Moore KM, Thomas GJ, Duffy SW, Warwick J, Gabe R, Chou P, Ellis IO, Green AR, Haider S, Brouilette K et al (2014) Therapeutic targeting of integrin αvβ6 in breast cancer. J Natl Cancer Inst 106, dju169 10.1093/jnci/dju169 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Croucher DR, Saunders DN, Lobov S & Ranson M (2008) Revisiting the biological roles of PAI2 (SERPINB2) in cancer. Nat Rev Cancer 8, 535–545. [DOI] [PubMed] [Google Scholar]
  • 71. Harris NLE, Vennin C, Conway JRW, Vine KL, Pinese M, Cowley MJ, Shearer RF, Lucas MC, Herrmann D, Allam AH et al (2017) SerpinB2 regulates stromal remodelling and local invasion in pancreatic cancer. Oncogene 36, 4288–4298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Zhu GH, Huang C, Qiu ZJ, Liu J, Zhang ZH, Zhao N, Feng ZZ & Lv XH (2011) Expression and prognostic significance of CD151, c‐Met, and integrin alpha3/alpha6 in pancreatic ductal adenocarcinoma. Dig Dis Sci 56, 1090–1098. [DOI] [PubMed] [Google Scholar]
  • 73. Franco‐Barraza J, Francescone R, Luong T, Shah N, Madhani R, Cukierman G, Dulaimi E, Devarajan K, Egleston BL, Nicolas E et al (2017) Matrix-regulated integrin αvβ5 maintains α5β1-dependent desmoplastic traits prognostic of neoplastic recurrence. eLife 6, 10.7554/eLife.20600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Seguin L, Desgrosellier JS, Weis SM & Cheresh DA (2015) Integrins and cancer: regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol 25, 234–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Hsia DA, Mitra SK, Hauck CR, Streblow DN, Nelson JA, Ilic D, Huang S, Li E, Nemerow GR, Leng J et al (2003) Differential regulation of cell motility and invasion by FAK. J Cell Biol 160, 753–767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Sulzmaier FJ, Jean C & Schlaepfer DD (2014) FAK in cancer: mechanistic findings and clinical applications. Nat Rev Cancer 14, 598–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Kong DB, Chen F & Sima N (2017) Focal adhesion kinases crucially regulate TGFbeta‐induced migration and invasion of bladder cancer cells via Src kinase and E‐cadherin. Onco Targets Ther 10, 1783–1792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Schlaepfer DD & Mitra SK (2004) Multiple connections link FAK to cell motility and invasion. Curr Opin Genet Dev 14, 92–101. [DOI] [PubMed] [Google Scholar]
  • 79. Serrels A & Frame MC (2016) FAK goes nuclear to control antitumor immunity‐a new target in cancer immuno‐therapy. Oncoimmunology 5, e1119356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Kanteti R, Mirzapoiazova T, Riehm JJ, Dhanasingh I, Mambetsariev B, Wang J, Kulkarni P, Kaushik G, Seshacharyulu P, Ponnusamy MP et al (2018) Focal adhesion kinase a potential therapeutic target for pancreatic cancer and malignant pleural mesothelioma. Cancer Biol Ther 19, 316–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Stokes JB, Adair SJ, Slack‐Davis JK, Walters DM, Tilghman RW, Hershey ED, Lowrey B, Thomas KS, Bouton AH, Hwang RF et al (2011) Inhibition of focal adhesion kinase by PF‐562,271 inhibits the growth and metastasis of pancreatic cancer concomitant with altering the tumor microenvironment. Mol Cancer Ther 10, 2135–2145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Tavora B, Reynolds LE, Batista S, Demircioglu F, Fernandez I, Lechertier T, Lees DM, Wong PP, Alexopoulou A, Elia G et al (2014) Endothelial‐cell FAK targeting sensitizes tumours to DNA‐damaging therapy. Nature 514, 112–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Zhao XK, Cheng Y, Liang Cheng M, Yu L, Mu M, Li H, Liu Y, Zhang B, Yao Y, Guo H et al (2016) Focal adhesion kinase regulates fibroblast migration via integrin beta‐1 and plays a central role in fibrosis. Sci Rep 6, 19276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Serrels A, Lund T, Serrels B, Byron A, McPherson RC, von Kriegsheim A, Gomez‐Cuadrado L, Canel M, Muir M, Ring JE et al (2015) Nuclear FAK controls chemokine transcription, Tregs, and evasion of anti‐tumor immunity. Cell 163, 160–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Burridge K (2017) Focal adhesions: a personal perspective on a half century of progress. FEBS J 284, 3355–3361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Romanova LY & Mushinski JF (2011) Central role of paxillin phosphorylation in regulation of LFA‐1 integrins activity and lymphocyte migration. Cell Adh Migr 5, 457–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Chin VT, Nagrial AM, Chou A, Biankin AV, Gill AJ, Timpson P & Pajic M (2015) Rho‐associated kinase signalling and the cancer microenvironment: novel biological implications and therapeutic opportunities. Expert Rev Mol Med 17, e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Pajic M, Herrmann D, Vennin C, Conway JR, Chin VT, Johnsson AK, Welch HC & Timpson P (2015) The dynamics of Rho GTPase signaling and implications for targeting cancer and the tumor microenvironment. Small GTPases 6, 123–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Rath N, Morton JP, Julian L, Helbig L, Kadir S, McGhee EJ, Anderson KI, Kalna G, Mullin M, Pinho AV et al (2017) ROCK signaling promotes collagen remodeling to facilitate invasive pancreatic ductal adenocarcinoma tumor cell growth. EMBO Mol Med 9, 198–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Kagawa Y, Matsumoto S, Kamioka Y, Mimori K, Naito Y, Ishii T, Okuzaki D, Nishida N, Maeda S, Naito A et al (2013) Cell cycle‐dependent Rho GTPase activity dynamically regulates cancer cell motility and invasion in vivo. PLoS ONE 8, e83629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Rodriguez‐Hernandez I, Cantelli G, Bruce F & Sanz‐Moreno V (2016) Rho, ROCK and actomyosin contractility in metastasis as drug targets F1000Research, 5, F1000 Faculty Rev-783. 10.12688/f1000research.7909.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Vennin C, Chin VT, Warren SC, Lucas MC, Herrmann D, Magenau A, Melenec P, Walters SN, Del Monte‐Nieto G, Conway JR et al (2017) Transient tissue priming via ROCK inhibition uncouples pancreatic cancer progression, sensitivity to chemotherapy, and metastasis. Sci Transl Med 9, eaai8504 10.1126/scitranslmed.aai8504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Rath N, Munro J, Cutiongco MF, Jagiello A, Gadegaard N, McGarry L, Unbekandt M, Michalopoulou E, Kamphorst JJ, Sumpton D et al (2018) Rho kinase inhibition by AT13148 blocks pancreatic ductal adenocarcinoma invasion and tumor growth. Can Res 78, 3321–3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Huveneers S & Danen EH (2009) Adhesion signaling ‐ crosstalk between integrins, Src and Rho. J Cell Sci 122, 1059–1069. [DOI] [PubMed] [Google Scholar]
  • 95. Joshi B, Strugnell SS, Goetz JG, Kojic LD, Cox ME, Griffith OL, Chan SK, Jones SJ, Leung SP, Masoudi H et al (2008) Phosphorylated caveolin‐1 regulates Rho/ROCK‐dependent focal adhesion dynamics and tumor cell migration and invasion. Can Res 68, 8210–8220. [DOI] [PubMed] [Google Scholar]
  • 96. Sadok A, McCarthy A, Caldwell J, Collins I, Garrett MD, Yeo M, Hooper S, Sahai E, Kuemper S, Mardakheh FK et al (2015) Rho kinase inhibitors block melanoma cell migration and inhibit metastasis. Can Res 75, 2272–2284. [DOI] [PubMed] [Google Scholar]
  • 97. Massihnia D, Galvano A, Fanale D, Perez A, Castiglia M, Incorvaia L, Listi A, Rizzo S, Cicero G, Bazan V et al (2016) Triple negative breast cancer: shedding light onto the role of pi3k/akt/mtor pathway. Oncotarget 7, 60712–60722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Liu P, Cheng H, Roberts TM & Zhao JJ (2009) Targeting the phosphoinositide 3‐kinase pathway in cancer. Nat Rev Drug Discovery 8, 627–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Conway JR, Herrmann D, Evans TJ, Morton JP & Timpson P (2018) Combating pancreatic cancer with PI3K pathway inhibitors in the era of personalised medicine. Gut 68, 742–758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Mayer IA & Arteaga CL (2016) The PI3K/AKT pathway as a target for cancer treatment. Annu Rev Med 67, 11–28. [DOI] [PubMed] [Google Scholar]
  • 101. Schlieman MG, Fahy BN, Ramsamooj R, Beckett L & Bold RJ (2003) Incidence, mechanism and prognostic value of activated AKT in pancreas cancer. Br J Cancer 89, 2110–2115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Garrido‐Laguna I, Tometich D, Hu N, Ying J, Geiersbach K, Whisenant J, Wang K, Ross JS & Sharma S (2015) N of 1 case reports of exceptional responders accrued from pancreatic cancer patients enrolled in first‐in‐man studies from 2002 through 2012. Oncoscience 2, 285–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Dancey J (2010) mTOR signaling and drug development in cancer. Nat Rev Clin Oncol 7, 209–219. [DOI] [PubMed] [Google Scholar]
  • 104. Manning BD & Cantley LC (2007) AKT/PKB signaling: navigating downstream. Cell 129, 1261–1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Murthy D, Attri KS & Singh PK (2018) Phosphoinositide 3‐kinase signaling pathway in pancreatic ductal adenocarcinoma progression, pathogenesis, and therapeutics. Front Physiol 9, 335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Thillai K, Lam H, Sarker D & Wells CM (2017) Deciphering the link between PI3K and PAK: an opportunity to target key pathways in pancreatic cancer? Oncotarget 8, 14173–14191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Baer R, Cintas C, Therville N & Guillermet‐Guibert J (2015) Implication of PI3K/Akt pathway in pancreatic cancer: when PI3K isoforms matter? Adv Biol Regul 59, 19–35. [DOI] [PubMed] [Google Scholar]
  • 108. Kennedy AL, Morton JP, Manoharan I, Nelson DM, Jamieson NB, Pawlikowski JS, McBryan T, Doyle B, McKay C, Oien KA et al (2011) Activation of the PIK3CA/AKT pathway suppresses senescence induced by an activated RAS oncogene to promote tumorigenesis. Mol Cell 42, 36–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Conway JRW, Warren SC, Herrmann D, Murphy KJ, Cazet AS, Vennin C, Shearer RF, Killen MJ, Magenau A, Melenec P et al (2018) Intravital imaging to monitor therapeutic response in moving hypoxic regions resistant to PI3K pathway targeting in pancreatic cancer. Cell Rep 23, 3312–3326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Irby RB, Mao W, Coppola D, Kang J, Loubeau JM, Trudeau W, Karl R, Fujita DJ, Jove R & Yeatman TJ (1999) Activating SRC mutation in a subset of advanced human colon cancers. Nat Genet 21, 187–190. [DOI] [PubMed] [Google Scholar]
  • 111. Daigo Y, Furukawa Y, Kawasoe T, Ishiguro H, Fujita M, Sugai S, Nakamori S, Liefers GJ, Tollenaar RA, van de Velde CJ et al (1999) Absence of genetic alteration at codon 531 of the human c‐src gene in 479 advanced colorectal cancers from Japanese and Caucasian patients. Can Res 59, 4222–4224. [PubMed] [Google Scholar]
  • 112. Laghi L, Bianchi P, Orbetegli O, Gennari L, Roncalli M & Malesci A (2001) Lack of mutation at codon 531 of SRC in advanced colorectal cancers from Italian patients. Br J Cancer 84, 196–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Siveen KS, Prabhu KS, Achkar IW, Kuttikrishnan S, Shyam S, Khan AQ, Merhi M, Dermime S & Uddin S (2018) Role of non receptor tyrosine kinases in hematological malignances and its targeting by natural products. Mol Cancer 17, 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E et al (2012) The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2, 401–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E et al (2013) Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6, pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Ku M, Wall M, MacKinnon RN, Walkley CR, Purton LE, Tam C, Izon D, Campbell L, Cheng HC & Nandurkar H (2015) Src family kinases and their role in hematological malignancies. Leuk Lymphoma 56, 577–586. [DOI] [PubMed] [Google Scholar]
  • 117. Donahue TR, Tran LM, Hill R, Li Y, Kovochich A, Calvopina JH, Patel SG, Wu N, Hindoyan A, Farrell JJ et al (2012) Integrative survival‐based molecular profiling of human pancreatic cancer. Clin Cancer Res 18, 1352–1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Kelber JA, Reno T, Kaushal S, Metildi C, Wright T, Stoletov K, Weems JM, Park FD, Mose E, Wang Y et al (2012) KRas induces a Src/PEAK1/ErbB2 kinase amplification loop that drives metastatic growth and therapy resistance in pancreatic cancer. Can Res 72, 2554–2564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Malric L, Monferran S, Gilhodes J, Boyrie S, Dahan P, Skuli N, Sesen J, Filleron T, Kowalski‐Chauvel A, Cohen‐Jonathan Moyal E et al (2017) Interest of integrins targeting in glioblastoma according to tumor heterogeneity and cancer stem cell paradigm: an update. Oncotarget 8, 86947–86968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Ren B, Yu YP, Tseng GC, Wu C, Chen K, Rao UN, Nelson J, Michalopoulos GK & Luo JH (2007) Analysis of integrin alpha7 mutations in prostate cancer, liver cancer, glioblastoma multiforme, and leiomyosarcoma. J Natl Cancer Inst 99, 868–880. [DOI] [PubMed] [Google Scholar]
  • 121. Burkin DJ & Fontelonga TM (2015) Mesothelioma cells breaking bad: loss of integrin alpha7 promotes cell motility and poor clinical outcomes in patients. J Pathol 237, 282–284. [DOI] [PubMed] [Google Scholar]
  • 122. Laszlo V, Hoda MA, Garay T, Pirker C, Ghanim B, Klikovits T, Dong YW, Rozsas A, Kenessey I, Szirtes I et al (2015) Epigenetic down‐regulation of integrin alpha7 increases migratory potential and confers poor prognosis in malignant pleural mesothelioma. J Pathol 237, 203–214. [DOI] [PubMed] [Google Scholar]
  • 123. International Cancer Genome Consortium , Hudson TJ, Anderson W, Artez A, Barker AD, Bell C, Bernabe RR, Bhan MK, Calvo F, Eerola I et al (2010) International network of cancer genome projects. Nature 464, 993–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Ben‐David U, Siranosian B, Ha G, Tang H, Oren Y, Hinohara K, Strathdee CA, Dempster J, Lyons NJ, Burns R et al (2018) Genetic and transcriptional evolution alters cancer cell line drug response. Nature 560, 325–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Stewart RL & O'Connor KL (2015) Clinical significance of the integrin alpha6beta4 in human malignancies. Lab Invest 95, 976–986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Lee BY, Timpson P, Horvath LG & Daly RJ (2015) FAK signaling in human cancer as a target for therapeutics. Pharmacol Ther 146, 132–149. [DOI] [PubMed] [Google Scholar]
  • 127. Zhang Y, Kwok‐Shing Ng P, Kucherlapati M, Chen F, Liu Y, Tsang YH, de Velasco G, Jeong KJ, Akbani R, Hadjipanayis A et al (2017) A pan‐cancer proteogenomic Atlas of PI3K/AKT/mTOR pathway alterations. Cancer Cell 31, 820–832 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Zhou B, Wang G-Z, Wen Z-S, Zhou Y-C, Huang Y-C, Chen Y & Zhou G-B (2018) Somatic mutations and splicing variants of focal adhesion kinase in non‐small cell lung cancer. J Natl Cancer Inst 110, 195–204. [DOI] [PubMed] [Google Scholar]
  • 129. Payne SN, Maher ME, Tran NH, Van De Hey DR, Foley TM, Yueh AE, Leystra AA, Pasch CA, Jeffrey JJ, Clipson L et al (2015) PIK3CA mutations can initiate pancreatic tumorigenesis and are targetable with PI3K inhibitors. Oncogenesis 4, e169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Ying H, Elpek KG, Vinjamoori A, Zimmerman SM, Chu GC, Yan H, Fletcher‐Sananikone E, Zhang H, Liu Y, Wang W et al (2011) PTEN is a major tumor suppressor in pancreatic ductal adenocarcinoma and regulates an NF‐kappaB‐cytokine network. Cancer Discov 1, 158–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Soria JC, Gan HK, Blagden SP, Plummer R, Arkenau HT, Ranson M, Evans TR, Zalcman G, Bahleda R, Hollebecque A et al (2016) A phase I, pharmacokinetic and pharmacodynamic study of GSK2256098, a focal adhesion kinase inhibitor, in patients with advanced solid tumors. Ann Oncol 27, 2268–2274. [DOI] [PubMed] [Google Scholar]
  • 132. Fennell DA, Baas P, Taylor P, Nowak AK, Gilligan D, Nakano T, Pachter JA, Weaver DT, Scherpereel A, Pavlakis N et al (2019) Maintenance defactinib versus placebo after first‐line chemotherapy in patients with merlin‐stratified pleural mesothelioma: COMMAND‐A double‐blind, randomized, phase II study. J Clin Oncol 37, 790–798. [DOI] [PubMed] [Google Scholar]
  • 133. Mak G, Soria JC, Blagden SP, Plummer R, Fleming RA, Nebot N, Zhang J, Mazumdar J, Rogan D, Gazzah A et al (2019) A phase Ib dose‐finding, pharmacokinetic study of the focal adhesion kinase inhibitor GSK2256098 and trametinib in patients with advanced solid tumours. Br J Cancer 120, 975–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Quan M, Cui J, Xia T, Jia Z, Xie D, Wei D, Huang S, Huang Q, Zheng S & Xie K (2015) Merlin/NF2 suppresses pancreatic tumor growth and metastasis by attenuating the FOXM1‐mediated Wnt/beta‐catenin signaling. Can Res 75, 4778–4789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Patel A, Sabbineni H, Clarke A & Somanath PR (2016) Novel roles of Src in cancer cell epithelial‐to‐mesenchymal transition, vascular permeability, microinvasion and metastasis. Life Sci 157, 52–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Rajeshkumar NV, Tan AC, De Oliveira E, Womack C, Wombwell H, Morgan S, Warren MV, Walker J, Green TP, Jimeno A et al (2009) Antitumor effects and biomarkers of activity of AZD0530, a Src inhibitor, in pancreatic cancer. Clin Cancer Res 15, 4138–4146. [DOI] [PubMed] [Google Scholar]
  • 137. George TJ Jr, Trevino JG & Liu C (2014) Src inhibition is still a relevant target in pancreatic cancer. Oncologist 19, 211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Messersmith WA, Rajeshkumar NV, Tan AC, Wang XF, Diesl V, Choe SE, Follettie M, Coughlin C, Boschelli F, Garcia‐Garcia E et al (2009) Efficacy and pharmacodynamic effects of bosutinib (SKI‐606), a Src/Abl inhibitor, in freshly generated human pancreas cancer xenografts. Mol Cancer Ther 8, 1484–1493. [DOI] [PubMed] [Google Scholar]
  • 139. Demetri GD, Lo Russo P, MacPherson IR, Wang D, Morgan JA, Brunton VG, Paliwal P, Agrawal S, Voi M & Evans TR (2009) Phase I dose‐escalation and pharmacokinetic study of dasatinib in patients with advanced solid tumors. Clin Cancer Res 15, 6232–6240. [DOI] [PubMed] [Google Scholar]
  • 140. Khoury HJ, Guilhot F, Hughes TP, Kim DW & Cortes JE (2009) Dasatinib treatment for Philadelphia chromosome‐positive leukemias: practical considerations. Cancer 115, 1381–1394. [DOI] [PubMed] [Google Scholar]
  • 141. Lombardo LJ, Lee FY, Chen P, Norris D, Barrish JC, Behnia K, Castaneda S, Cornelius LA, Das J, Doweyko AM et al (2004) Discovery of N‐(2‐chloro‐6‐methyl‐ phenyl)‐2‐(6‐(4‐(2‐hydroxyethyl)‐ piperazin‐1‐yl)‐2‐methylpyrimidin‐4‐ ylamino)thiazole‐5‐carboxamide (BMS‐354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays. J Med Chem 47, 6658–6661. [DOI] [PubMed] [Google Scholar]
  • 142. Nam S, Kim D, Cheng JQ, Zhang S, Lee JH, Buettner R, Mirosevich J, Lee FY & Jove R (2005) Action of the Src family kinase inhibitor, dasatinib (BMS‐354825), on human prostate cancer cells. Can Res 65, 9185–9189. [DOI] [PubMed] [Google Scholar]
  • 143. Montero JC, Seoane S, Ocana A & Pandiella A (2011) Inhibition of SRC family kinases and receptor tyrosine kinases by dasatinib: possible combinations in solid tumors. Clin Cancer Res 17, 5546–5552. [DOI] [PubMed] [Google Scholar]
  • 144. Park SI, Zhang J, Phillips KA, Araujo JC, Najjar AM, Volgin AY, Gelovani JG, Kim SJ, Wang Z & Gallick GE (2008) Targeting SRC family kinases inhibits growth and lymph node metastases of prostate cancer in an orthotopic nude mouse model. Can Res 68, 3323–3333. [DOI] [PubMed] [Google Scholar]
  • 145. Abbas R, Hug BA, Leister C, Gaaloul ME, Chalon S & Sonnichsen D (2012) A phase I ascending single‐dose study of the safety, tolerability, and pharmacokinetics of bosutinib (SKI‐606) in healthy adult subjects. Cancer Chemother Pharmacol 69, 221–227. [DOI] [PubMed] [Google Scholar]
  • 146. Campone M, Bondarenko I, Brincat S, Hotko Y, Munster PN, Chmielowska E, Fumoleau P, Ward R, Bardy‐Bouxin N, Leip E et al (2012) Phase II study of single‐agent bosutinib, a Src/Abl tyrosine kinase inhibitor, in patients with locally advanced or metastatic breast cancer pretreated with chemotherapy. Ann Oncol 23, 610–617. [DOI] [PubMed] [Google Scholar]
  • 147. Mayer EL, Baurain JF, Sparano J, Strauss L, Campone M, Fumoleau P, Rugo H, Awada A, Sy O & Llombart‐Cussac A (2011) A phase 2 trial of dasatinib in patients with advanced HER2‐positive and/or hormone receptor‐positive breast cancer. Clin Cancer Res 17, 6897–6904. [DOI] [PubMed] [Google Scholar]
  • 148. Pusztai L, Moulder S, Altan M, Kwiatkowski D, Valero V, Ueno NT, Esteva FJ, Avritscher R, Qi Y, Strauss L et al (2014) Gene signature‐guided dasatinib therapy in metastatic breast cancer. Clin Cancer Res 20, 5265–5271. [DOI] [PubMed] [Google Scholar]
  • 149. Reddy SM, Kopetz S, Morris J, Parikh N, Qiao W, Overman MJ, Fogelman D, Shureiqi I, Jacobs C, Malik Z et al (2015) Phase II study of saracatinib (AZD0530) in patients with previously treated metastatic colorectal cancer. Invest New Drugs 33, 977–984. [DOI] [PubMed] [Google Scholar]
  • 150. Chee CE, Krishnamurthi S, Nock CJ, Meropol NJ, Gibbons J, Fu P, Bokar J, Teston L, O'Brien T, Gudena V et al (2013) Phase II study of dasatinib (BMS‐354825) in patients with metastatic adenocarcinoma of the pancreas. Oncologist 18, 1091–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Hong DS, Choe JH, Naing A, Wheler JJ, Falchook GS, Piha‐Paul S, Moulder SL, George GC, Choe JM, Strauss LC et al (2013) A phase 1 study of gemcitabine combined with dasatinib in patients with advanced solid tumors. Invest New Drugs 31, 918–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Reni M, Cereda S, Milella M, Novarino A, Passardi A, Mambrini A, Di Lucca G, Aprile G, Belli C, Danova M et al (2013) Maintenance sunitinib or observation in metastatic pancreatic adenocarcinoma: a phase II randomised trial. Eur J Cancer 49, 3609–3615. [DOI] [PubMed] [Google Scholar]
  • 153. Renouf DJ, Moore MJ, Hedley D, Gill S, Jonker D, Chen E, Walde D, Goel R, Southwood B, Gauthier I et al (2012) A phase I/II study of the Src inhibitor saracatinib (AZD0530) in combination with gemcitabine in advanced pancreatic cancer. Invest New Drugs 30, 779–786. [DOI] [PubMed] [Google Scholar]
  • 154. Hsyu PH, Mould DR, Abbas R & Amantea M (2014) Population pharmacokinetic and pharmacodynamic analysis of bosutinib. Drug Metab Pharmacokinet 29, 441–448. [DOI] [PubMed] [Google Scholar]
  • 155. Evans TRJ, Van Cutsem E, Moore MJ, Bazin IS, Rosemurgy A, Bodoky G, Deplanque G, Harrison M, Melichar B, Pezet D et al (2017) Phase 2 placebo‐controlled, double‐blind trial of dasatinib added to gemcitabine for patients with locally‐advanced pancreatic cancer. Ann Oncol 28, 354–361. [DOI] [PubMed] [Google Scholar]
  • 156. Isakoff SJ, Wang D, Campone M, Calles A, Leip E, Turnbull K, Bardy‐Bouxin N, Duvillie L & Calvo E (2014) Bosutinib plus capecitabine for selected advanced solid tumours: results of a phase 1 dose‐escalation study. Br J Cancer 111, 2058–2066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Evans TRJ, Cutsem EV, Moore MJ, Purvis JD, Strauss LC, Rock EP, Lee J, Lin C, Rosemurgy A, Arena FP et al (2012) Dasatinib combined with gemcitabine (Gem) in patients (pts) with locally advanced pancreatic adenocarcinoma (PaCa): design of CA180‐375, a placebo‐controlled, randomized, double‐blind phase II trial. J Clin Oncol 30, TPS4134. [Google Scholar]
  • 158. Hekim C, Ilander M, Yan J, Michaud E, Smykla R, Vaha‐Koskela M, Savola P, Tahtinen S, Saikko L, Hemminki A et al (2017) Dasatinib changes immune cell profiles concomitant with reduced tumor growth in several murine solid tumor models. Cancer Immunol Res 5, 157–169. [DOI] [PubMed] [Google Scholar]
  • 159. Najima Y, Yoshida C, Iriyama N, Fujisawa S, Wakita H, Chiba S, Okamoto S, Kawakami K, Takezako N, Kumagai T et al (2018) Regulatory T cell inhibition by dasatinib is associated with natural killer cell differentiation and a favorable molecular response‐The final results of the D‐first study. Leuk Res 66, 66–72. [DOI] [PubMed] [Google Scholar]
  • 160. Christiansson L, Soderlund S, Mangsbo S, Hjorth‐Hansen H, Hoglund M, Markevarn B, Richter J, Stenke L, Mustjoki S, Loskog A et al (2015) The tyrosine kinase inhibitors imatinib and dasatinib reduce myeloid suppressor cells and release effector lymphocyte responses. Mol Cancer Ther 14, 1181–1191. [DOI] [PubMed] [Google Scholar]
  • 161. Kreutzman A, Ilander M, Porkka K, Vakkila J & Mustjoki S (2014) Dasatinib promotes Th1‐type responses in granzyme B expressing T‐cells. Oncoimmunology 3, e28925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Mao L, Deng WW, Yu GT, Bu LL, Liu JF, Ma SR, Wu L, Kulkarni AB, Zhang WF & Sun ZJ (2017) Inhibition of SRC family kinases reduces myeloid‐derived suppressor cells in head and neck cancer. Int J Cancer 140, 1173–1185. [DOI] [PubMed] [Google Scholar]
  • 163. Ozanne J, Prescott AR & Clark K (2015) The clinically approved drugs dasatinib and bosutinib induce anti‐inflammatory macrophages by inhibiting the salt‐inducible kinases. Biochem J 465, 271–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Yu GT, Mao L, Wu L, Deng WW, Bu LL, Liu JF, Chen L, Yang LL, Wu H, Zhang WF et al (2018) Inhibition of SRC family kinases facilitates anti‐CTLA4 immunotherapy in head and neck squamous cell carcinoma. Cell Mol Life Sci 75, 4223–4234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. D'Angelo SP, Shoushtari AN, Keohan ML, Dickson MA, Gounder MM, Chi P, Loo JK, Gaffney L, Schneider L, Patel Z et al (2017) Combined KIT and CTLA‐4 blockade in patients with refractory GIST and other advanced sarcomas: a phase Ib study of dasatinib plus ipilimumab. Clin Cancer Res 23, 2972–2980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Wu AA, Jaffee E & Lee V (2019) Current status of immunotherapies for treating pancreatic cancer. Curr Oncol Rep 21, 60. [DOI] [PubMed] [Google Scholar]
  • 167. Gotwals P, Cameron S, Cipolletta D, Cremasco V, Crystal A, Hewes B, Mueller B, Quaratino S, Sabatos‐Peyton C, Petruzzelli L et al (2017) Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat Rev Cancer 17, 286–301. [DOI] [PubMed] [Google Scholar]
  • 168. Luttrell DK, Luttrell LM & Parsons SJ (1988) Augmented mitogenic responsiveness to epidermal growth factor in murine fibroblasts that overexpress pp60c‐src. Mol Cell Biol 8, 497–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Amos S, Martin PM, Polar GA, Parsons SJ & Hussaini IM (2005) Phorbol 12‐myristate 13‐acetate induces epidermal growth factor receptor transactivation via protein kinase Cdelta/c‐Src pathways in glioblastoma cells. J Biol Chem 280, 7729–7738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Fischgrabe J, Gotte M, Michels K, Kiesel L & Wulfing P (2010) Targeting endothelin A receptor enhances anti‐proliferative and anti‐invasive effects of the HER2 antibody trastuzumab in HER2‐overexpressing breast cancer cells. Int J Cancer 127, 696–706. [DOI] [PubMed] [Google Scholar]
  • 171. Sooro MA, Zhang N & Zhang P (2018) Targeting EGFR‐mediated autophagy as a potential strategy for cancer therapy. Int J Cancer 143, 2116–2125. [DOI] [PubMed] [Google Scholar]
  • 172. Lemmon MA & Schlessinger J (2010) Cell signaling by receptor tyrosine kinases. Cell 141, 1117–1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Zhang H, Berezov A, Wang Q, Zhang G, Drebin J, Murali R & Greene MI (2007) ErbB receptors: from oncogenes to targeted cancer therapies. J Clin Investig 117, 2051–2058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Sheahan AV, Biankin AV, Parish CR & Khachigian LM (2018) Targeted therapies in the management of locally advanced and metastatic pancreatic cancer: a systematic review. Oncotarget 9, 21613–21627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA et al (2007) 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 25, 1960–1966. [DOI] [PubMed] [Google Scholar]
  • 176. Boeck S, Wittwer C, Heinemann V, Haas M, Kern C, Stieber P, Nagel D & Holdenrieder S (2013) Cytokeratin 19‐fragments (CYFRA 21‐1) as a novel serum biomarker for response and survival in patients with advanced pancreatic cancer. Br J Cancer 108, 1684–1694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Heinemann V, Vehling‐Kaiser U, Waldschmidt D, Kettner E, Marten A, Winkelmann C, Klein S, Kojouharoff G, Gauler TC, von Weikersthal LF et al (2013) Gemcitabine plus erlotinib followed by capecitabine versus capecitabine plus erlotinib followed by gemcitabine in advanced pancreatic cancer: final results of a randomised phase 3 trial of the ‘Arbeitsgemeinschaft Internistische Onkologie’ (AIO‐PK0104). Gut 62, 751–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Sinn M, Bahra M, Liersch T, Gellert K, Messmann H, Bechstein W, Waldschmidt D, Jacobasch L, Wilhelm M, Rau BM et al (2017) CONKO‐005: adjuvant chemotherapy with gemcitabine plus erlotinib versus gemcitabine alone in patients after R0 resection of pancreatic cancer: a multicenter randomized phase III trial. J Clin Oncol 35, 3330–3337. [DOI] [PubMed] [Google Scholar]
  • 179. Boeck S, Jung A, Laubender RP, Neumann J, Egg R, Goritschan C, Vehling‐Kaiser U, Winkelmann C, Fischer von Weikersthal L, Clemens MR et al (2013) EGFR pathway biomarkers in erlotinib‐treated patients with advanced pancreatic cancer: translational results from the randomised, crossover phase 3 trial AIO‐PK0104. Br J Cancer 108, 469–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Schultheis B, Reuter D, Ebert MP, Siveke J, Kerkhoff A, Berdel WE, Hofheinz R, Behringer DM, Schmidt WE, Goker E et al (2017) Gemcitabine combined with the monoclonal antibody nimotuzumab is an active first‐line regimen in KRAS wildtype patients with locally advanced or metastatic pancreatic cancer: a multicenter, randomized phase IIb study. Ann Oncol 28, 2429–2435. [DOI] [PubMed] [Google Scholar]
  • 181. Haas M, Siveke JT, Schenk M, Lerch MM, Caca K, Freiberg‐Richter J, Fischer von Weikersthal L, Kullmann F, Reinacher‐Schick A, Fuchs M et al (2018) Efficacy of gemcitabine plus erlotinib in rash‐positive patients with metastatic pancreatic cancer selected according to eligibility for FOLFIRINOX: a prospective phase II study of the ‘Arbeitsgemeinschaft Internistische Onkologie’. Eur J Cancer 94, 95–103. [DOI] [PubMed] [Google Scholar]
  • 182. Fornier MN, Morris PG, Abbruzzi A, D'Andrea G, Gilewski T, Bromberg J, Dang C, Dickler M, Modi S, Seidman AD et al (2011) A phase I study of dasatinib and weekly paclitaxel for metastatic breast cancer. Ann Oncol 22, 2575–2581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Nagaraj NS, Washington MK & Merchant NB (2011) Combined blockade of Src kinase and epidermal growth factor receptor with gemcitabine overcomes STAT3‐mediated resistance of inhibition of pancreatic tumor growth. Clin Cancer Res 17, 483–493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, Preall J & Tuveson DA (2018) IL‐1‐induced JAK/STAT signaling is antagonized by TGF‐beta to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov 9, 282–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Cardin DB, Goff LW, Chan E, Whisenant JG, Dan Ayers G, Takebe N, Arlinghaus LR, Yankeelov TE, Berlin J & Merchant N (2018) Dual Src and EGFR inhibition in combination with gemcitabine in advanced pancreatic cancer: phase I results : a phase I clinical trial. Invest New Drugs 36, 442–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Pan Y, Zheng M, Zhong L, Yang J, Zhou S, Qin Y, Xiang R, Chen Y & Yang SY (2015) A preclinical evaluation of SKLB261, a multikinase inhibitor of EGFR/Src/VEGFR2, as a therapeutic agent against pancreatic cancer. Mol Cancer Ther 14, 407–418. [DOI] [PubMed] [Google Scholar]
  • 187. El Touny LH, Vieira A, Mendoza A, Khanna C, Hoenerhoff MJ & Green JE (2014) Combined SFK/MEK inhibition prevents metastatic outgrowth of dormant tumor cells. J Clin Investig 124, 156–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Gomes EG, Connelly SF & Summy JM (2013) Targeting the yin and the yang: combined inhibition of the tyrosine kinase c‐Src and the tyrosine phosphatase SHP‐2 disrupts pancreatic cancer signaling and biology in vitro and tumor formation in vivo. Pancreas 42, 795–806. [DOI] [PubMed] [Google Scholar]
  • 189. Fedele C, Ran H, Diskin B, Wei W, Jen J, Geer MJ, Araki K, Ozerdem U, Simeone DM, Miller G et al (2018) SHP2 inhibition prevents adaptive resistance to MEK inhibitors in multiple cancer models. Cancer Discov 8, 1237–1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Ruess DA, Heynen GJ, Ciecielski KJ, Ai J, Berninger A, Kabacaoglu D, Gorgulu K, Dantes Z, Wormann SM, Diakopoulos KN et al (2018) Mutant KRAS‐driven cancers depend on PTPN11/SHP2 phosphatase. Nat Med 24, 954–960. [DOI] [PubMed] [Google Scholar]
  • 191. Zhang J, He DH, Zajac‐Kaye M & Hochwald SN (2014) A small molecule FAK kinase inhibitor, GSK2256098, inhibits growth and survival of pancreatic ductal adenocarcinoma cells. Cell Cycle 13, 3143–3149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Begum A, Ewachiw T, Jung C, Huang A, Norberg KJ, Marchionni L, McMillan R, Penchev V, Rajeshkumar NV, Maitra A et al (2017) The extracellular matrix and focal adhesion kinase signaling regulate cancer stem cell function in pancreatic ductal adenocarcinoma. PLoS ONE 12, e0180181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, Ross S, Conrads TP, Veenstra TD, Hitt BA et al (2003) Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4, 437–450. [DOI] [PubMed] [Google Scholar]
  • 194. Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, Rustgi AK, Chang S & Tuveson DA (2005) Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483. [DOI] [PubMed] [Google Scholar]
  • 195. Aung KL, McWhirter E, Welch S, Wang L, Lovell S, Stayner L‐A, Ali S, Malpage A, Makepeace B, Ramachandran M et al (2018) A phase II trial of GSK2256098 and trametinib in patients with advanced pancreatic ductal adenocarcinoma (PDAC) (MOBILITY‐002 Trial, NCT02428270). J Clin Oncol 36, 409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Infante JR, Camidge DR, Mileshkin LR, Chen EX, Hicks RJ, Rischin D, Fingert H, Pierce KJ, Xu H, Roberts WG et al (2012) Safety, pharmacokinetic, and pharmacodynamic phase I dose‐escalation trial of PF‐00562271, an inhibitor of focal adhesion kinase, in advanced solid tumors. J Clin Oncol 30, 1527–1533. [DOI] [PubMed] [Google Scholar]
  • 197. Shapiro IM, Kolev VN, Vidal CM, Kadariya Y, Ring JE, Wright Q, Weaver DT, Menges C, Padval M, McClatchey AI et al (2014) Merlin deficiency predicts FAK inhibitor sensitivity: a synthetic lethal relationship. Sci Transl Med 6, 237ra68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Shah NR, Tancioni I, Ward KK, Lawson C, Chen XL, Jean C, Sulzmaier FJ, Uryu S, Miller NL, Connolly DC et al (2014) Analyses of merlin/NF2 connection to FAK inhibitor responsiveness in serous ovarian cancer. Gynecol Oncol 134, 104–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Kato T, Sato T, Yokoi K & Sekido Y (2017) E‐cadherin expression is correlated with focal adhesion kinase inhibitor resistance in Merlin‐negative malignant mesothelioma cells. Oncogene 36, 5522–5531. [DOI] [PubMed] [Google Scholar]
  • 200. Jones SF, Siu LL, Bendell JC, Cleary JM, Razak AR, Infante JR, Pandya SS, Bedard PL, Pierce KJ, Houk B et al (2015) A phase I study of VS‐6063, a second‐generation focal adhesion kinase inhibitor, in patients with advanced solid tumors. Invest New Drugs 33, 1100–1107. [DOI] [PubMed] [Google Scholar]
  • 201. Papadatos‐Pastos D, Kumar R, Yap TA, Ruddle R, Decordova S, Jones P, Halbert G, Garrett MD, McLeod R, Backholer Z et al (2015) A first‐in‐human study of the dual ROCK I/II inhibitor, AT13148, in patients with advanced cancers. J Clin Oncol 33, 2566. [Google Scholar]
  • 202. Zhang Y, Yang C, Cheng H, Fan Z, Huang Q, Lu Y, Fan K, Luo G, Jin K, Wang Z et al (2018) Novel agents for pancreatic ductal adenocarcinoma: emerging therapeutics and future directions. J Hematol Oncol 11, 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Morran DC, Wu J, Jamieson NB, Mrowinska A, Kalna G, Karim SA, Au AY, Scarlett CJ, Chang DK, Pajak MZ et al (2014) Targeting mTOR dependency in pancreatic cancer. Gut 63, 1481–1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Garrido‐Laguna I, Tan AC, Uson M, Angenendt M, Ma WW, Villaroel MC, Zhao M, Rajeshkumar NV, Jimeno A, Donehower R et al (2010) Integrated preclinical and clinical development of mTOR inhibitors in pancreatic cancer. Br J Cancer 103, 649–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Rozengurt E, Soares HP & Sinnet‐Smith J (2014) Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: an unintended consequence leading to drug resistance. Mol Cancer Ther 13, 2477–2488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Chandarlapaty S (2012) Negative feedback and adaptive resistance to the targeted therapy of cancer. Cancer Discov 2, 311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Basu B, Dean E, Puglisi M, Greystoke A, Ong M, Burke W, Cavallin M, Bigley G, Womack C, Harrington EA et al (2015) First‐in‐human pharmacokinetic and pharmacodynamic study of the dual m‐TORC 1/2 inhibitor AZD2014. Clin Cancer Res 21, 3412–3419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Driscoll DR, Karim SA, Sano M, Gay DM, Jacob W, Yu J, Mizukami Y, Gopinathan A, Jodrell DI, Evans TR et al (2016) mTORC2 signaling drives the development and progression of pancreatic cancer. Can Res 76, 6911–6923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Hassan Z, Schneeweis C, Wirth M, Veltkamp C, Dantes Z, Feuerecker B, Ceyhan GO, Knauer SK, Weichert W, Schmid RM et al (2018) MTOR inhibitor‐based combination therapies for pancreatic cancer. Br J Cancer 118, 366–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Chiron D, Di Liberto M, Martin P, Huang X, Sharman J, Blecua P, Mathew S, Vijay P, Eng K, Ali S et al (2014) Cell‐cycle reprogramming for PI3K inhibition overrides a relapse‐specific C481S BTK mutation revealed by longitudinal functional genomics in mantle cell lymphoma. Cancer Discov 4, 1022–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A et al (2015) Whole‐exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun 6, 6744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Chou A, Froio D, Nagrial AM, Parkin A, Murphy KJ, Chin VT, Wohl D, Steinmann A, Stark R, Drury A et al (2017) Tailored first‐line and second‐line CDK4‐targeting treatment combinations in mouse models of pancreatic cancer. Gut 67, 2142–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Noel MS, Wang‐Gillam A, Ocean AJ, Chawla SP, Priore GD & Picozzi VJ (2019) Phase II trial of SM‐88 in patients with metastatic pancreatic cancer: preliminary results of the first stage. J Clin Oncol 37, 200. [Google Scholar]
  • 214. Fruman DA & Rommel C (2014) PI3K and cancer: lessons, challenges and opportunities. Nat Rev Drug Discovery 13, 140–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Bean GR, Ganesan YT, Dong Y, Takeda S, Liu H, Chan PM, Huang Y, Chodosh LA, Zambetti GP, Hsieh JJ et al (2013) PUMA and BIM are required for oncogene inactivation‐induced apoptosis. Sci Signal 6, ra20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, Maira M, McNamara K, Perera SA, Song Y et al (2008) Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med 14, 1351–1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Abel EV, Basile KJ, Kugel CH 3rd, Witkiewicz AK, Le K, Amaravadi RK, Karakousis GC, Xu X, Xu W, Schuchter LM et al (2013) Melanoma adapts to RAF/MEK inhibitors through FOXD3‐mediated upregulation of ERBB3. J Clin Investig 123, 2155–2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218. Pettazzoni P, Viale A, Shah P, Carugo A, Ying H, Wang H, Genovese G, Seth S, Minelli R, Green T et al (2015) Genetic events that limit the efficacy of MEK and RTK inhibitor therapies in a mouse model of KRAS‐driven pancreatic cancer. Can Res 75, 1091–1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Cox AD, Fesik SW, Kimmelman AC, Luo J & Der CJ (2014) Drugging the undruggable RAS: mission possible? Nat Rev Drug Discovery 13, 828–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Shimizu T, Tolcher AW, Papadopoulos KP, Beeram M, Rasco DW, Smith LS, Gunn S, Smetzer L, Mays TA, Kaiser B et al (2012) The clinical effect of the dual‐targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced cancer. Clin Cancer Res 18, 2316–2325. [DOI] [PubMed] [Google Scholar]
  • 221. Simpkins F, Jang K, Yoon H, Hew KE, Kim M, Azzam DJ, Sun J, Zhao D, Ince TA, Liu W et al (2018) Dual Src and MEK inhibition decreases ovarian cancer growth and targets tumor initiating stem‐like cells. Clin Cancer Res 24, 4874–4886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Bedard PL, Tabernero J, Janku F, Wainberg ZA, Paz‐Ares L, Vansteenkiste J, Van Cutsem E, Perez‐Garcia J, Stathis A, Britten CD et al (2015) A phase Ib dose‐escalation study of the oral pan‐PI3K inhibitor buparlisib (BKM120) in combination with the oral MEK1/2 inhibitor trametinib (GSK1120212) in patients with selected advanced solid tumors. Clin Cancer Res 21, 730–738. [DOI] [PubMed] [Google Scholar]
  • 223. Chung V, McDonough S, Philip PA, Cardin D, Wang‐Gillam A, Hui L, Tejani MA, Seery TE, Dy IA, Al Baghdadi T et al (2017) Effect of selumetinib and MK‐2206 vs oxaliplatin and fluorouracil in patients with metastatic pancreatic cancer after prior therapy: SWOG S1115 study randomized clinical trial. JAMA Oncol 3, 516–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Tolcher AW, Patnaik A, Papadopoulos KP, Rasco DW, Becerra CR, Allred AJ, Orford K, Aktan G, Ferron‐Brady G, Ibrahim N et al (2015) Phase I study of the MEK inhibitor trametinib in combination with the AKT inhibitor afuresertib in patients with solid tumors and multiple myeloma. Cancer Chemother Pharmacol 75, 183–189. [DOI] [PubMed] [Google Scholar]
  • 225. Er JL, Goh PN, Lee CY, Tan YJ, Hii LW, Mai CW, Chung FF & Leong CO (2018) Identification of inhibitors synergizing gemcitabine sensitivity in the squamous subtype of pancreatic ductal adenocarcinoma (PDAC). Apoptosis 23, 343–355. [DOI] [PubMed] [Google Scholar]
  • 226. Yan C, Yang Q & Gong Z (2018) Activation of hepatic stellate cells during liver carcinogenesis requires fibrinogen/integrin alphavbeta5 in zebrafish. Neoplasia 20, 533–542. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 227. Zhao W, Ajani JA, Sushovan G, Ochi N, Hwang R, Hafley M, Johnson RL, Bresalier RS, Logsdon CD, Zhang Z et al (2018) Galectin‐3 mediates tumor cell‐stroma interactions by activating pancreatic stellate cells to produce cytokines via integrin signaling. Gastroenterology 154, 1524–1537 e6. [DOI] [PubMed] [Google Scholar]
  • 228. Smith JW (2003) Cilengitide merck. Curr Opin Investig Drugs 4, 741–745. [PubMed] [Google Scholar]
  • 229. Gilbert MR, Kuhn J, Lamborn KR, Lieberman F, Wen PY, Mehta M, Cloughesy T, Lassman AB, Deangelis LM, Chang S et al (2012) Cilengitide in patients with recurrent glioblastoma: the results of NABTC 03‐02, a phase II trial with measures of treatment delivery. J Neurooncol 106, 147–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Nabors LB, Mikkelsen T, Hegi ME, Ye X, Batchelor T, Lesser G, Peereboom D, Rosenfeld MR, Olsen J, Brem S et al (2012) A safety run‐in and randomized phase 2 study of cilengitide combined with chemoradiation for newly diagnosed glioblastoma (NABTT 0306). Cancer 118, 5601–5607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Stupp R, Hegi ME, Gorlia T, Erridge SC, Perry J, Hong YK, Aldape KD, Lhermitte B, Pietsch T, Grujicic D et al (2014) Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071‐22072 study): a multicentre, randomised, open‐label, phase 3 trial. Lancet Oncol 15, 1100–1108. [DOI] [PubMed] [Google Scholar]
  • 232. Reardon DA, Fink KL, Mikkelsen T, Cloughesy TF, O'Neill A, Plotkin S, Glantz M, Ravin P, Raizer JJ, Rich KM et al (2008) Randomized phase II study of cilengitide, an integrin‐targeting arginine‐glycine‐aspartic acid peptide, in recurrent glioblastoma multiforme. J Clin Oncol 26, 5610–5617. [DOI] [PubMed] [Google Scholar]
  • 233. Friess H, Langrehr JM, Oettle H, Raedle J, Niedergethmann M, Dittrich C, Hossfeld DK, Stoger H, Neyns B, Herzog P et al (2006) A randomized multi‐center phase II trial of the angiogenesis inhibitor Cilengitide (EMD 121974) and gemcitabine compared with gemcitabine alone in advanced unresectable pancreatic cancer. BMC Cancer 6, 285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Massabeau C, Khalifa J, Filleron T, Modesto A, Bigay‐Game L, Plat G, Dierickx L, Aziza R, Rouquette I, Gomez‐Roca C et al (2018) Continuous infusion of cilengitide plus chemoradiotherapy for patients with stage III non‐small‐cell lung cancer: a phase I study. Clin Lung Cancer 19, e277–e285. [DOI] [PubMed] [Google Scholar]
  • 235. Pandolfi F, Franza L, Altamura S, Mandolini C, Cianci R, Ansari A & Kurnick JT (2017) Integrins: integrating the biology and therapy of cell‐cell interactions. Clin Ther 39, 2420–2436. [DOI] [PubMed] [Google Scholar]
  • 236. Ricart AD, Tolcher AW, Liu G, Holen K, Schwartz G, Albertini M, Weiss G, Yazji S, Ng C & Wilding G (2008) Volociximab, a chimeric monoclonal antibody that specifically binds alpha5beta1 integrin: a phase I, pharmacokinetic, and biological correlative study. Clin Cancer Res 14, 7924–7929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Elez E, Kocakova I, Hohler T, Martens UM, Bokemeyer C, Van Cutsem E, Melichar B, Smakal M, Csoszi T, Topuzov E et al (2015) Abituzumab combined with cetuximab plus irinotecan versus cetuximab plus irinotecan alone for patients with KRAS wild‐type metastatic colorectal cancer: the randomised phase I/II POSEIDON trial. Ann Oncol 26, 132–140. [DOI] [PubMed] [Google Scholar]
  • 238. Hussain M, Le MS, Gimmi C, Bruns R, Straub J, Miller K & PERSEUS Study Group (2016) Differential effect on bone lesions of targeting integrins: randomized phase II trial of abituzumab in patients with metastatic castration‐resistant prostate cancer. Clin Cancer Res 22, 3192–3200. [DOI] [PubMed] [Google Scholar]
  • 239. Paez‐Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F, Inoue M, Bergers G, Hanahan D & Casanovas O (2009) Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 15, 220–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Ebos JM, Lee CR, Cruz‐Munoz W, Bjarnason GA, Christensen JG & Kerbel RS (2009) Accelerated metastasis after short‐term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15, 232–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Reynolds AR, Hart IR, Watson AR, Welti JC, Silva RG, Robinson SD, Da Violante G, Gourlaouen M, Salih M, Jones MC et al (2009) Stimulation of tumor growth and angiogenesis by low concentrations of RGD‐mimetic integrin inhibitors. Nat Med 15, 392–400. [DOI] [PubMed] [Google Scholar]
  • 242. Carmeliet P & Jain RK (2011) Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat Rev Drug Discovery 10, 417–427. [DOI] [PubMed] [Google Scholar]
  • 243. Webb T (2005) Vascular normalization: study examines how antiangiogenesis therapies work. J Natl Cancer Inst 97, 336–337. [DOI] [PubMed] [Google Scholar]
  • 244. Wong PP, Demircioglu F, Ghazaly E, Alrawashdeh W, Stratford MR, Scudamore CL, Cereser B, Crnogorac‐Jurcevic T, McDonald S, Elia G et al (2015) Dual‐action combination therapy enhances angiogenesis while reducing tumor growth and spread. Cancer Cell 27, 123–137. [DOI] [PubMed] [Google Scholar]
  • 245. Muller PA, Caswell PT, Doyle B, Iwanicki MP, Tan EH, Karim S, Lukashchuk N, Gillespie DA, Ludwig RL, Gosselin P et al (2009) Mutant p53 drives invasion by promoting integrin recycling. Cell 139, 1327–1341. [DOI] [PubMed] [Google Scholar]
  • 246. Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, Feig C, Nakagawa T, Caldwell ME, Zecchini HI et al (2013) Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 62, 112–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD & Hingorani SR (2012) Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21, 418–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Hingorani SR, Zheng L, Bullock AJ, Seery TE, Harris WP, Sigal DS, Braiteh F, Ritch PS, Zalupski MM, Bahary N et al (2018) HALO 202: randomized phase II study of PEGPH20 plus nab‐paclitaxel/gemcitabine versus nab‐paclitaxel/gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. J Clin Oncol 36, 359–366. [DOI] [PubMed] [Google Scholar]
  • 249. Hingorani SR, Harris WP, Beck JT, Berdov BA, Wagner SA, Pshevlotsky EM, Tjulandin SA, Gladkov OA, Holcombe RF, Korn R et al (2016) Phase Ib study of PEGylated recombinant human hyaluronidase and gemcitabine in patients with advanced pancreatic cancer. Clin Cancer Res 22, 2848–2854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Clift R, Thompson BJ, Taverna D, Blouw B, Lee J, Thompson CB & Maneval DC (2018) Abstract 1743: rationale for evaluating PEGylated recombinant human hyaluronidase PH20 (pegvorhyaluronidase alfa; PEGPH20) in patients with hyaluronan (HA)‐accumulating colorectal cancer. Can Res 78, 1743. [Google Scholar]
  • 251. Ko AH, Cinar P, Tempero MA, Nakakura EK, Yeh BM & Chondros D (2016) Perioperative stromal depletion by PEGPH20 in pancreatic ductal adenocarcinoma. J Clin Oncol 34, TPS476‐TPS476. [Google Scholar]
  • 252. Yu KH, Mantha S, Tjan C, Kaufmann ES, Brenner R, Lowery MA, Ku GY, Raj NP, Shcherba M, Goldberg Z et al (2018) Pilot study of gemcitabine, nab‐paclitaxel, PEGPH20, and rivaroxaban for advanced pancreatic adenocarcinoma: an interim analysis. J Clin Oncol 36, 405. [Google Scholar]
  • 253. Ramanathan RK, McDonough S, Philip PA, Hingorani SR, Lacy J, Kortmansky JS, Thumar JR, Chiorean EG, Shields AF, Behl D et al (2018) A phase IB/II randomized study of mFOLFIRINOX (mFFOX) + pegylated recombinant human hyaluronidase (PEGPH20) versus mFFOX alone in patients with good performance status metastatic pancreatic adenocarcinoma (mPC): SWOG S1313 (NCT #01959139). J Clin Oncol 36, 208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Wang S, Willumsen N, Bager C, Karsdal MA, Chondros D & Taverna D (2018) Extracellular matrix (ECM) circulating peptide biomarkers as potential predictors of survival in patients (pts) with untreated metastatic pancreatic ductal adenocarcinoma (mPDA) receiving pegvorhyaluronidase alfa (PEGPH20), nab‐paclitaxel (A), and gemcitabine (G). J Clin Oncol 36, 12030. [Google Scholar]
  • 255. Wong KM, Horton KJ, Coveler AL, Hingorani SR & Harris WP (2017) Targeting the tumor stroma: the biology and clinical development of pegylated recombinant human hyaluronidase (PEGPH20). Curr Oncol Rep 19, 47. [DOI] [PubMed] [Google Scholar]
  • 256. Trarbach T, Schultheis B, Gauler TC, Schneider V, Strumberg D, Eberhardt WE, Le Scouiller S, Marotti M, Brown KH & Drevs J (2012) Phase I open‐label study of cediranib, an oral inhibitor of VEGF signalling, in combination with the oral Src inhibitor saracatinib in patients with advanced solid tumours. Invest New Drugs 30, 1962–1971. [DOI] [PubMed] [Google Scholar]
  • 257. Hammel P, Huguet F, van Laethem JL, Goldstein D, Glimelius B, Artru P, Borbath I, Bouche O, Shannon J, Andre T et al (2016) Effect of chemoradiotherapy vs chemotherapy on survival in patients with locally advanced pancreatic cancer controlled after 4 months of gemcitabine with or without erlotinib: the LAP07 randomized clinical trial. JAMA 315, 1844–1853. [DOI] [PubMed] [Google Scholar]
  • 258. Semrad T, Barzi A, Lenz HJ, Hutchins IM, Kim EJ, Gong IY, Tanaka M, Beckett L, Holland W, Burich RA et al (2015) Pharmacodynamic separation of gemcitabine and erlotinib in locally advanced or metastatic pancreatic cancer: therapeutic and biomarker results. Int J Clin Oncol 20, 518–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Philip PA, Benedetti J, Corless CL, Wong R, O'Reilly EM, Flynn PJ, Rowland KM, Atkins JN, Mirtsching BC, Rivkin SE et al (2010) Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group‐directed intergroup trial S0205. J Clin Oncol 28, 3605–3610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260. Wang‐Gillam A, Lockhart AC, Tan BR, Suresh R, Lim K‐H, Ratner L, Morton A, Huffman J, Marquez S, Boice N et al (2018) Phase I study of defactinib combined with pembrolizumab and gemcitabine in patients with advanced cancer. J Clin Oncol 36, 380. [Google Scholar]
  • 261. Thompson DS, Patnaik A, Bendell JC, Papadopoulos K, Infante JR, Mastico RA, Johnson D, Qin A, O'Leary JJ & Tolcher AW (2010) A phase I dose‐escalation study of IMGN388 in patients with solid tumors. J Clin Oncol 28, 3058. [Google Scholar]
  • 262. Hingorani SR, Bullock AJ, Seery TA, Zheng L, Sigal D, Ritch PS, Braiteh FS, Zalupski M, Bahary N, Harris WP et al (2015) High response rate and PFS with PEGPH20 added to nab‐paclitaxel/gemcitabine in stage IV previously untreated pancreatic cancer patients with high‐HA tumors: interim results of a randomized phase 2 study. ASCO Annual Meeting.
  • 263. Ko AH, Bekaii‐Saab T, Van Ziffle J, Mirzoeva OM, Joseph NM, Talasaz A, Kuhn P, Tempero MA, Collisson EA, Kelley RK et al (2016) A multicenter, open‐label phase II clinical trial of combined MEK plus EGFR inhibition for chemotherapy‐refractory advanced pancreatic adenocarcinoma. Clin Cancer Res 22, 61–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264. Ettrich TJ, Ebert M, Lorenzen S, Moehler M, Vogel A, Witkowski L, Seufferlein T & Reinacher‐Schick A (2018) ASCO‐ and ESMO‐update 2017 ‐ highlights of the 53. meeting of the American Society of Clinical Oncology/ASCO 2017 and European Society for Medical Oncology/ESMO congress 2017. Z Gastroenterol 56, 384–397. [DOI] [PubMed] [Google Scholar]
  • 265. Marsh Rde W, Rocha Lima CM, Levy DE, Mitchell EP, Rowland KM Jr & Benson AB 3rd (2007) A phase II trial of perifosine in locally advanced, unresectable, or metastatic pancreatic adenocarcinoma. Am J Clin Oncol 30, 26–31. [DOI] [PubMed] [Google Scholar]
  • 266. Javle MM, Shroff RT, Xiong H, Varadhachary GA, Fogelman D, Reddy SA, Davis D, Zhang Y, Wolff RA & Abbruzzese JL (2010) Inhibition of the mammalian target of rapamycin (mTOR) in advanced pancreatic cancer: results of two phase II studies. BMC Cancer 10, 368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. Wolpin BM, Hezel AF, Abrams T, Blaszkowsky LS, Meyerhardt JA, Chan JA, Enzinger PC, Allen B, Clark JW, Ryan DP et al (2009) Oral mTOR inhibitor everolimus in patients with gemcitabine‐refractory metastatic pancreatic cancer. J Clin Oncol 27, 193–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Bjerregaard JK, Ladekarl M, Farr KP, Vestermark LW, Jensen HA & Pfeiffer P (2014) A randomized phase I/II study of everolimus, irinotecan, and cetuximab versus capecitabine and oxaliplatin in gemcitabine‐resistant patients with pancreatic cancer. J Clin Oncol 32, 337. [Google Scholar]
  • 269. Kordes S, Richel DJ, Klumpen HJ, Weterman MJ, Stevens AJ & Wilmink JW (2013) A phase I/II, non‐randomized, feasibility/safety and efficacy study of the combination of everolimus, cetuximab and capecitabine in patients with advanced pancreatic cancer. Invest New Drugs 31, 85–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Joka M, Boeck S, Zech CJ, Seufferlein T, Wichert G, Licht T, Krause A, Jauch KW, Heinemann V & Bruns CJ (2014) Combination of antiangiogenic therapy using the mTOR‐inhibitor everolimus and low‐dose chemotherapy for locally advanced and/or metastatic pancreatic cancer: a dose‐finding study. Anticancer Drugs 25, 1095–1101. [DOI] [PubMed] [Google Scholar]
  • 271. Tan BR, Picus J, Chan E, Lockhart AC, Roth BJ, Morton A, Liang C & Wang‐Gillam A (2016) Phase I study of X‐82, an oral dual anti‐VEGFR/PDGFR tyrosine kinase inhibitor, with everolimus in solid tumors. J Clin Oncol 34, 2588. [DOI] [PubMed] [Google Scholar]
  • 272. Koumarianou A, Pectasides DG, Manousou K, Dionysopoulos D, Kaltsas GA, Kolomodi D, Poulios C, Skondra M, Samantas E, Pentheroudakis G et al (2018) 1326PEvaluation of the efficacy and safety of everolimus as a first‐line treatment in newly diagnosed patients with advanced gastroenteropancreatic neuroendocrine tumors. Ann Oncol 29, mdy293.019 10.1093/annonc/mdy293.019 [DOI] [Google Scholar]

Articles from The Febs Journal are provided here courtesy of Wiley

RESOURCES