文档库 最新最全的文档下载
当前位置:文档库 › Current concepts and noveltargets in advanced pancreatic cancer

Current concepts and noveltargets in advanced pancreatic cancer

Current concepts and novel targets in advanced pancreatic cancer Patrick Michl,Thomas M Gress

Department of Gastroenterology,Endocrinology and Metabolism,Philipps University Marburg,Marburg, Germany Correspondence to Professor Dr Thomas Gress, Division of Gastroenterology, Endocrinology and Metabolism, Philipps University Marburg, Baldinger Strasse,

Marburg35043,Germany; gress@med.uni-marburg.de ABSTRACT

Pancreatic cancer remains one of the most aggressive

tumours with a5-year survival rate of less than5%.The

dismal prognosis of this tumour entity that is associated

with a high degree of drug resistance has not changed

over the past decades.Since1997,gemcitabine-based

regimens have been the therapy of choice for advanced

pancreatic cancer.Recently,however,new combination

chemotherapy regimens achieved a signi?cant survival

bene?t compared to gemcitabine-based therapies.In

addition,novel approaches to improve drug delivery are

currently being developed,and new drugs targeting

signalling pathways both within the tumour cells and the

tumour microenvironment are undergoing preclinical and

clinical validation.Furthermore,efforts are being made to

identify predictive markers for individualised treatment

approaches based on molecular tumour characteristics.

This review provides an overview on current and

emerging concepts as well as novel targets for systemic

treatment of advanced pancreatic https://www.wendangku.net/doc/7f11408523.html,bination

therapies incorporating drugs directed against these new

targets may open new avenues for improving the ef?cacy

of current treatment approaches and overcoming the

devastating prognosis of pancreatic cancer patients.

INTRODUCTION

Pancreatic ductal adenocarcinoma(PDAC)is asso-

ciated with a5-year survival rate of less than5%

and a median survival of6months after diagnosis,

thereby exhibiting the poorest prognosis of all

solid tumours.12It is characterised by a high pro-

pensity for local invasion and distant metastasis as

well as a largely drug-resistant phenotype.At pres-

entation,only approximately20%of PDAC

patients qualify for surgical resection in curative

intent.Even for those,the5-year survival rate

rarely exceeds20%due to early relapse or meta-

static spread of the disease.The vast majority of

patients,however,already present with locally

advanced or metastatic disease,which is associated

with an extremely poor prognosis.3

The current management of PDAC is guided by

tumour stage,comorbidities and performance

status of the patients.Surgical resection followed

by a6-month-course of adjuvant gemcitabine-

based chemotherapy is the standard of care for

early-stage disease(?gure1).3In contrast,patients

with metastatic disease are candidates for systemic

palliative chemotherapy.For patients with locally

advanced disease without evidence of metastasis,

optimal treatment remains to be de?ned,with

chemotherapy alone as well as chemoradiation to

be considered(?gure1).

This review focuses on current and emerging

therapeutic concepts for patients presenting with

metastatic or locally advanced disease,for which

surgery in curative intent is not an option.

CURRENT MANAGEMENT

Metastatic disease

Since1997,the nucleoside analogue gemcitabine

has been established as standard of care in meta-

static PDAC.Burris et al4published the results of a

phase III trial comparing5-?uorouracil(5-FU),

which had been used frequently by that time,

with gemcitabine as a single agent administered as

a weekly intravenous injection.T reatment with

gemcitabine resulted in a superior clinical response

compared to5-FU and was associated with a sig-

ni?cant,but modest survival bene?t(median sur-

vival5.65vs4.41months).In addition,the clinical

bene?t response,a composite score for pain(anal-

gesic consumption and pain intensity),Karnofsky

performance score and weight,was in favour of

gemcitabine leading to US Food and Drug

Administration approval and the establishment of

gemcitabine as standard of care in?rst-line pallia-

tive therapy for advanced PDAC.3

Over the past decade,numerous trials have been

conducted to improve the outcome in patients

with metastatic disease by combination therapies

using gemcitabine as backbone.Most trials used a

second cytotoxic agent such as5-FU,5capecita-

bine,6oxaliplatin,78cisplatin,910irinotecan,11

exatecan12or pemetrexed13administered in com-

bination with gemcitabine.However,despite a

modest improvement in progression-free survival

in some trials,a signi?cant bene?t in overall sur-

vival could not be demonstrated for any of these

combination therapies.3

Despite these discouraging results of single

trials,a recent meta-analysis by Heinemann et al14

suggests a bene?t of combination therapies in

patients with good performance status.By analys-

ing15randomised trials of combination therapies,

the authors showed a signi?cant survival bene?t

when gemcitabine was combined with either a

platinum derivative or?uoropyrimidines.This

was,however,restricted to patients with good per-

formance status.Another meta-analysis by

Sultana et al15con?rmed the bene?t of combin-

ation therapy with gemcitabine and a platinum

compound.In that meta-analysis,however,com-

bination with?uoropyrimidines only resulted in

improved survival when the oral analogue capeci-

tabine was used as the partner drug.T aken

together,these data indicate that there might be a

Recent advances in clinical practice

signi?cant,albeit small,survival bene?t for gemcitabine-based combination therapies in patients with good performance status.

Conroy et al16recently reported the?rst signi?cant improve-ment in overall survival using a gemcitabine-free combination therapy regimen.In the landmark PRODIGE trial published in 2011,the authors used the FOLFIRINOX protocol(folinic acid,?uorouracil,irinotecan and oxaliplatin),which resulted in a remarkably improved overall survival of11.1months compared to6.8months in the control arm using gemcitabine alone.16As expected,the side effects of this new combination regimen were signi?cant,including a grade3and4neutropenia rate of 45.7%.Interestingly,pancreatic head tumours requiring biliary stents accounted for only14.3%of all patients,which might explain the low observed incidence of cholangitis.T aken together,this new regimen represents a new treatment stand-ard for patients with metastatic PDAC that should be preferred to gemcitabine-based combination regimens in carefully selected patients with good performance status and without contraindications.

Second-line therapy in metastatic disease

Only a few trials have evaluated the use of second-line regi-mens after the failure of?rst-line therapy.The only randomised trial in this context tested the combination of oxaliplatin and 5-FU versus best supportive care after failure of gemcitabine-based?rst-line therapy.Although the trial was prematurely ter-minated due to insuf?cient accrual,the oxaliplatin and5-FU protocol resulted in a signi?cantly prolonged median second-line survival of4.82months compared to2.30months with best supportive care.317This suggests that a sequential admin-istration of a5-FU and platinum-based combination regimen after gemcitabine-based monotherapy is able to provide a sur-vival bene?t.3

Targeted therapies in metastatic disease

During the past decade,numerous targeted agents have been evaluated alone or in combination with chemotherapy in meta-static PDAC.Unfortunately,most agents have so far failed to improve patient survival signi?cantly.The long list of agents tested in trials as futile include antiangiogenic drugs such as the vascular endothelial growth factor(VEGF)antibody bevazi-cumab and multikinase inhibitors with antiangiogenic activity such as axitinib,sunitinib and sorafenib.18–20The same applies for various other targeted agents directed against secreted matrix proteases21or against intracellular targets such as farne-syl transferase inhibitors.22

T o date,inhibition of the epidermal growth factor receptor (EGFR)pathway by the small molecule inhibitor erlotinib represents the only targeted therapy approved for metastatic PDAC.A trial conducted by the National Cancer Institute of Canada Clinical T rials Group demonstrated a small,but signi?-cant bene?t in median overall survival of approximately 2weeks in patients receiving erlotinib in combination with gemcitabine compared to patients treated with gemcitabine plus placebo.23This marginal bene?t clearly raises questions about the clinical signi?cance of erlotinib.However,in a small subgroup of patients who developed grade II or more skin rash as a side effect of erlotinib treatment,a prolonged median survival of10.5months was observed—almost the median sur-vival that has been described with FOLFIRINOX treatment.23 The underlying molecular mechanisms behind this striking observation remain to be fully elucidated.At this point,it is not entirely clear whether the erlotinib-induced rash is predict-ive of treatment response to erlotinib or serves merely as a prognostic factor re?ecting a more favourable tumour biology.24 In view of these data,it is presently recommended to stop erlo-tinib and to continue gemcitabine as monotherapy in the absence of skin toxicity.

Locally advanced pancreatic cancer

Locally advanced pancreatic cancer(LAPC)is de?ned as surgi-cally unresectable disease without evidence of distant metasta-ses.25The optimal treatment for these patients remains to be de?ned.In addition to systemic chemotherapy,chemoradiation has to be considered in order to achieve locoregional control.26 In contrast,radiotherapy alone has been largely abandoned since early studies demonstrated a lack of bene?t from radiation

Figure1Overview on current stage-dependent treatment strategies and new approaches for improvement of pancreatic cancer

therapy.

Recent advances in clinical practice

alone compared to chemoradiation.2728The impact of chemor-adiation versus chemotherapy alone on the survival of patients presenting with locally advanced disease still has to be clari?ed. Several small early studies using older radiation techniques and chemotherapy regimens resulting in high toxicities may no longer be representative of state-of-the-art clinical care.29–31 However,recent studies continue to produce con?icting data.

A randomised trial conducted by French groups used a fairly aggressive regimen including60Gy of radiation combined with simultaneous continuous infusions of5-FU and cisplatin. Compared with gemcitabine alone,survival was signi?cantly inferior in the chemoradiation group(8.6vs13months).32In contrast,a recent trial conducted by the Eastern Cooperative Oncology Group compared a less toxic regimen consisting of radiotherapy(50.4Gy)and gemcitabine(600mg/m2per week) with gemcitabine alone.In that study,radiochemotherapy resulted in a slight,but signi?cant,survival bene?t (11.1months(95%CI7.6to15.5months)vs9.2months(95% CI7.9to11.4months),one-sided p=0.017).33

In light of these con?icting data,it has to be taken into account that approximately30%of the patients treated with chemoradiation for LAPC develop metastatic disease soon after diagnosis and may thus have little bene?t from this type of therapy.34Therefore,a sequential approach has been proposed consisting of an initial chemotherapy regimen followed by restaging and subsequent chemoradiation only for those patients with no evidence of early metastatic progression,thus avoiding unnecessary toxic treatment.34

The ongoing GERCOR LAP-07study,a multi-institutional phase III trial,was designed to test the impact of this sequen-tial approach on survival(https://www.wendangku.net/doc/7f11408523.html,,identi-?er code NCT00634725).Patients are randomly assigned to receive either gemcitabine alone or gemcitabine/erlotinib.After 4months of chemotherapy,patients with stable disease are ran-domly assigned a second time either to continue their chemo-therapy or to receive chemoradiation to54Gy with concurrent capecitabine.The results of this largest randomised trial in LAPC are eagerly awaited.35

Current trial concepts are not only aiming to identify the optimal sequence of radiation and chemotherapy but also to de?ne the most ef?cient chemotherapy regimens in LAPC. Given the profound impact of the FOLFIRINOX regimen in metastatic PDAC,it may be speculated that an intensi?ed chemotherapy preceding chemoradiation might also be of bene?t to patients with locally advanced tumours.

In addition,it remains unclear to date which drug is most effective as radiosensitiser given concurrently with radiation. Earlier studies have established chemoradiation with5-FU as standard.Based on in-vitro data indicating that gemcitabine is a more potent radiosensitiser compared to5-FU,several smaller trials using gemcitabine together with radiotherapy revealed a trend towards better survival compared to5-FU;36however,at the cost of a higher systemic toxicity.Therefore,efforts have been made to decrease toxicity by reducing the dose of gemci-tabine37or by improving the radiotherapy techniques.2638 Currently,5-FU or gemcitabine at a reduced dose are equally applied as radiosensitiser.

Neoadjuvant therapy in LAPC

Radiochemotherapy as well as intensi?ed chemotherapy with or without subsequent chemoradiation are concepts that may offer the possibility of downstaging patients to achieve second-ary resectability in locally advanced or borderline resectable cases.Several retrospective and small prospective trials indicate that a subgroup of patients who are initially unresectable can successfully undergo resection after neoadjuvant therapy. According to recent meta-analyses by Gillen et al39and Assi?et al40approximately one-third of pancreatic cancer patients initially staged as unresectable or borderline resectable had resectable tumours following neoadjuvant chemoradiation, with survival rates comparable to initially resectable patients. Chemotherapeutic regimens included gemcitabine,5-FU(and oral analogues),mitomycin C and platinum compounds.In the meta-analysis by Gillen et al,39however,neither increased resec-tion frequencies nor improved survival by neoadjuvant therapy were observed in patients with initially resectable tumours. These meta-analysis data demonstrating a survival bene?t of neoadjuvant chemoradiation in patients with LAPC could be corroborated by a recent large retrospective series reported on 215patients with LAPC at a single institution.41In that series, radiotherapy was delivered with a median dose of52.2Gy in single fractions of1.8Gy,with gemcitabine being applied con-comitantly at a dose of300mg/m2weekly.Following radioche-motherapy,26%of the patients could be resected and had a median overall survival of22months,which was similar to patients with primary resection.41

As discussed above,a sequential approach with upfront chemotherapy followed by radiochemotherapy after restaging might prevent unnecessary radiotherapy in patients with early metastatic disease.However,evidence for this approach is limited and further trials for patients with locally advanced tumours are urgently required(NCT01359007).

EMERGING CONCEPTS

Improving drug delivery

The impact of current chemotherapeutic regimens on the overall survival of pancreatic cancer patients is clearly unsatis-fying.Several new approaches aim to improve the delivery of known chemotherapeutic agents into the tumour.Based on recent preclinical data in mouse models that will be discussed below in detail,it has been hypothesised that currently avail-able cytotoxic drugs can not access tumour cells at an effective concentration due to the extensive hypovascular stroma reac-tion that acts as a fence around tumour cells protecting them from therapeutic agents administered to the systemic circula-tion.42Therefore,numerous efforts have been made to improve drug formulations in the aim of enhancing accessibility to the tumour cells.

Nanoparticle albumin-bound(nab)-paclitaxel is an albumin-stabilised paclitaxel formulation that was initially developed to avoid toxicities associated with oil-based solvents required to solubilise paclitaxel.Nab-paclitaxel has been approved in patients with metastatic breast cancer.In addition,it has shown promising activity in a recent phase I/II trial with pan-creatic cancer patients,43and several trials using nab-paclitaxel in combination chemotherapies are currently ongoing (NCT01161186,NCT01470417,NCT01010945).The molecular mechanism of nab-paclitaxel has not been fully elucidated.In addition to the hypothesis that the tumour cells themselves are albumin-avid leading to enhanced tumoral concentration,it has been hypothesised that secreted protein,acidic and rich in cyst-eine,also known as osteonectin,which is highly expressed and secreted by pancreatic peritumoral?broblasts,may serve as an albumin-binding protein that sequesters nab-paclitaxel and con-centrates the drug intratumorally.44Interestingly,a recent pre-clinical study that used a combination of nab-paclitaxel and gemcitabine in a genetic mouse model of pancreatic cancer45 showed that coadministration of nab-paclitaxel and gemcitabine

Recent advances in clinical practice

uniquely led to tumour regression that was associated with increased intratumoral gemcitabine levels and a marked decrease in cytidine deaminase,the primary gemcitabine metabolising enzyme.The authors could demonstrate that paclitaxel reduced the levels of cytidine deaminase protein in cultured cells through reactive oxygen species-mediated degradation,45which suggests a synergistic effect of(nab)paclitaxel and gemcitabine in com-bination regimens.

In addition to(nab)paclitaxel,several other formulations have been developed in order to facilitate drug delivery to the tumour, among them coating drugs with liposomal phospholipid vesi-cles:EndoTAG-1is a novel cationic liposomal formulation of paclitaxel,which targets tumour endothelial cells that express negatively charged cell-surface molecules.Therapy with cationic liposomal paclitaxel led to reduced tumour vessel density and endothelial cell mitosis resulting in diminished tumour perfu-sion and growth.46A randomised phase II trial showed signi?-cantly improved survival in patients treated with liposomal paclitaxel in combination with gemcitabine compared to gemci-tabine alone.47Similarly,a liposomal formulation of irinotecan (PEP02)has shown activity in gemcitabine-refractory pancreatic cancer.48Further trials are warranted to investigate the activity of liposomal formulations and other modi?cations aiming to improve drug delivery in pancreatic cancer.

Novel targeted approaches

In addition to improving drug delivery,novel targeted therapies are urgently needed.So far,targeted agents have largely failed to provide a substantial survival bene?t in advanced pancreatic cancer.Therefore,it is of utmost importance to identify novel candidates for drug development that are able to impact signi?-cantly on the outcome of this disease.Most novel compounds can be classi?ed functionally into four main groups:(1)drugs interfering with downstream signalling cascades within the tumours cells;(2)drugs targeting the stromal response;(3)drugs modulating tumour vasculature thereby enhancing drug delivery; and(4)drugs targeting the immune response(?gure2).Targeting survival and angiogenesis pathways

Therapeutic inhibition of receptor tyrosine kinases such as EGFR and VEGFR regulating tumour cell survival,proliferation and angiogenesis both by monoclonal antibodies and small molecule inhibitors has been evaluated extensively and approved for clinical use in several solid tumours including colon cancer.49In pancreatic cancer,however,EGFR inhibition with the small molecule inhibitor erlotinib is the only targeted approach that has been approved for clinical use,but only pro-vides a marginal survival bene?t,as discussed above.23 Although robust data on the role of activating K-Ras mutations as predictors of response to EGFR inhibitors in pancreatic cancer are missing,it may be speculated that due to the high percentage of activating K-Ras mutations occurring in up to 90%of pancreatic cancer patients,pharmacological inhibition of EGFR upstream of K-Ras remains only marginally effective in this cancer type.

T argeting VEGF by the monoclonal antibody bevacizumab has also been demonstrated as ineffective in pancreatic cancer.18In addition,antiangiogenic multikinase inhibitors such as axitinib targeting the VEGF family,50sorafenib target-ing VEGF receptor,platelet-derived growth factor receptor and Raf51as well as the VEGF inhibitor a?ibercept52have all shown negative results in recent randomised trials.It can be speculated that the futility of all antiangiogenetic approaches tested so far is due to the largely hypovascular nature of the stroma sur-rounding cancer cells in this disease.In line with the disap-pointing clinical data,recent reports in genetically engineered mouse models suggest that decreasing the stromal density by the inhibition of stromal signalling pathways leads to enhanced intratumoral perfusion.This improves rather than impairs the ef?cacy of chemotherapy by facilitating drug accessibility to the tumour cells.53

Despite these disappointing results with anti-EGFR and anti-VEGR approaches,several novel targets within receptor tyrosine kinase signalling cascades are under evaluation in clinical trials.Among those are components of the insulin-like

Figure2Schematic overview of different therapeutic approaches for advanced pancreatic cancer.CTLA-4, cytotoxic T lymphocyte-associated antigen4;ECM,extracellular matrix; IGFR,insulin-like growth factor receptor;IL2,interleukin2;nab, nanoparticle albumin-bound;mTOR, mammalian target of rapamycin; PEGPH20,PEGylated human recombinant PH20hyaluronidase; TGFβ,transforming growth factorβ

.

Recent advances in clinical practice

growth factor 1receptor (IGF1R)pathway.IGF1R mediates a strong survival effect through both K-Ras-dependent and inde-pendent downstream signalling cascades and is highly expressed in pancreatic cancer tissues (?gure 3).IGF1R there-fore represents a promising survival target,which might be functionally relevant even in K-Ras mutated tumours.54Currently,several monoclonal antibodies targeting IGF1R,among them AMG-479,are under investigation in clinical trials (table 1).Unfortunately,the IGF1R inhibitor cixutumumab (IMC-A12)has already failed to show an effect on progression-free or overall survival in a recent randomised phase II trial when administered in combination with erlotinib and gemcita-bine.55In addition,another trial exploring the IGF1R inhibitor AMG-479has very recently been stopped after an interim ana-lysis revealed that the trial was unlikely to meet its ef ?cacy endpoint.

Activated K-Ras belongs to the predominant oncogenes in pancreatic adenocarcinomas that are mutated in a high percent-age of cases.This mutation can already be detected in early pre-invasive lesions during pancreatic carcinogenesis.56Earlier efforts to target K-Ras by farnesyltransferase inhibitors have failed.22In addition to preclinical research evaluating K-Ras as target using newer molecules,numerous trials are currently investigating kinases acting downstream of mutant K-Ras as therapeutic targets to overcome K-ras-induced drug resistance.The mitogen-activated protein kinase MEK belongs to the most important downstream effectors of K-Ras signalling,and has been shown to mediate K-Ras induced effects on proliferation and survival (?gure 3).MEK inhibition therefore represents an innovative therapeutic avenue particularly in tumours carrying an activating K-Ras mutation.Several small molecule inhibitors have been developed and have been validated in vitro for their therapeutic ef ?cacy in pancreatic cancer cells,including the compounds GSK1120212,BAY86-9766,MSC1936369B and AZD6244.Based on promising preclinical data,phase II trials with GSK1120212and MSC1936369B in combination with gemcitabine in advanced pancreatic cancer are currently in pro-gress (table 1).In addition,AZD6244is being tested in

combination with the EGFR inhibitor erlotinib in patients with pancreatic cancer refractory to gemcitabine (table 1).

The PI3K/Akt signalling pathway represents another promis-ing candidate whose inhibition might offer a signi ?cant thera-peutic potential.57PI3K/Akt is implicated in the progression of numerous human malignancies including pancreatic cancer by enhancing tumour cell proliferation,survival and metabolism.Activation of PI3K/Akt by upstream signals through receptor tyrosine kinases such as IGF1R has been reported in a high pro-portion of pancreatic cancers and serves as an independent negative prognostic factor (?gure 3).58In addition,its activity may be dependent on the phosphatase and tensin homologue,which normally inactivates Akt but whose expression is fre-quently lost in pancreatic cancers.59

V arious small molecules have been identi ?ed as potential inhibitors of PI3K/Akt signalling,some of which are being eval-uated in clinical trials as monotherapy or in combination with chemotherapeutic agents.Among them,the Akt antisense oligonucleotide RX-0201is being tested in a phase I/II trial in patients with metastatic pancreatic cancer 60(table 1).PI3K inhibitors such as BKM120or the combined PI3K/mammalian target of rapamycin (mTOR)inhibitor BEZ235are currently being evaluated in phase I trials in combination with two dif-ferent MEK inhibitors GSK1120212and MEK162in advanced solid tumours including pancreatic cancer carrying K-Ras,N-Ras and/or B-Raf mutations (table 1).This approach aims to overcome drug resistance mediated by upstream mutations of Ras and/or Raf through simultaneous blockade of the two major downstream cascades,MEK/ERK and PI3K/Akt signal-ling.The results of this combined approach are eagerly awaited.mTOR represents another promising target for therapeutic intervention.A serine/threonine kinase like Akt,it is

activated

Figure 3Schematic overview of major survival and proliferation

pathways and targeted agents currently evaluated in pancreatic cancer.mTOR,mammalian target of rapamycin;PTEN,phosphatase and tensin homologue;RTK,receptor tyrosine kinase.

Table 1Overview of selected targeted therapies currently under investigation in clinical trials

Compound

Clinical trial Survival signalling IGF1R AMG-479NCT01231347MEK

GSK1120212NCT01231581MSC1936369B NCT01016483AZD6244NCT01222689MEK162NCT01363232PI3K BKM120NCT01571024BEZ235NCT01337765AKT/PKB RX-0201NCT01028495mTOR RAD001

NCT00560963NCT01077986Immune response CTLA-4

Ipilimumab NCT01473940Tremelimumab NCT00556023CD40

CP870,893NCT01456585Stromal response Fibronectin ED-B L19-IL2NCT01198522TGF βRI LY2157299NCT01373164Hyaluronan

PEGPH20NCT01453153Oncofetal pathways

γ-Secretase (Notch pathway)RO4929097NCT01232829MK0752NCT01098344Epigenetics HDAC

Vorinostat NCT00948688DNA methyltransferase

5-Azacitidine

NCT01167816

CTLA-4,cytotoxic T lymphocyte-associated antigen 4;HDAC,histone deacetylases;IGF1R,insulin-like growth factor 1receptor;mTOR,mammalian target of rapamycin;TGF βRI;transforming growth factor βtype I receptor.

Recent advances in clinical practice

by PI3K/Akt signalling and is known as a regulator of gene tran-scription and cell cycle progression(?gure3).Although a recent phase II trial using the mTOR inhibitor RAD001as monother-apy had minimal clinical activity in patients with gemcitabine-refractory metastatic pancreatic cancer,61trials testing the effect of mTOR inhibition in combination with cytotoxic drugs or EGFR inhibitors are currently ongoing(table1).Interestingly, metformin,a commonly used antidiabetic drug,is known to inhibit the mTOR pathway through activation of the AMP-activated protein kinase(AMPK)that negatively regulates mTOR activity via phosphorylation and stabilisation of the tumour suppressor gene TSC2.62Based on these preclinical?nd-ings and supported by epidemiological studies suggesting that use of the antidiabetic drug metformin might be associated with a reduced life-time risk of developing cancers,6364metfor-min is currently being tested in a randomised phase II trial together with gemcitabine and erlotinib in patients with advanced pancreatic cancer(NCT01210911).In this context, tumour cell metabolism has gained increasing interest as a potential therapeutic avenue with several preclinical studies investigating the feasibility of interfering with glucose and glu-tamine utilisation,65and trials aiming to investigate interference with tumour cell metabolism and to use metabolic pro?les to predict therapeutic outcome(NCT01196247).

Poly(ADP-ribose)polymerase(PARP)is a family of proteins involved in a number of cellular processes mainly involving DNA repair and apoptosis.Sensitivity to therapeutic PARP inhibition is known to be associated with defects in the breast cancer DNA repair pathway.A subset of pancreatic cancer patients including a proportion of patients with familial pan-creatic cancer,have defects in the breast cancer DNA repair pathway or other defects in homologous repair.These cancers might respond to PARP inhibitors when given in combination with the DNA damaging agents.One ongoing trial is currently evaluating the PARP inhibitor olaparib in combination with cis-platin and irinotecan(NCT01296763).Moreover,several groups are investigating another PARP inhibitor,veliparib,with differ-ent drug combinations for advanced pancreatic cancer (NCT01489865,NCT01585805).

Targeting the immune response

In recent years,evidence has accumulated indicating that local and systemic immune response represents a major determinant of tumour resistance and progression.66In contrast to targeted approaches that aim to inhibit molecular pathways crucial for tumour growth and maintenance,immunotherapy endeavours to stimulate a host immune response that results in long-term tumour destruction.67The stroma of pancreatic cancer is par-ticularly rich in in?ammatory cells that are proposed to mediate drug resistance and tumour progression.68Therefore, immune cells such as T cells and macrophages in?ltrating the peritumoral stroma represent a promising target for immu-notherapeutic approaches.

T-cell-mediated immunity includes multiple sequential steps that are regulated by counterbalancing stimulatory and inhibi-tory signals that?ne-tune the response.66Inhibitory pathways are referred to as immune checkpoints that are crucial for main-taining self-tolerance and modulating the duration and ampli-tude of physiological immune responses.It has been recognised that many tumours co-opt certain immune checkpoint path-ways as a major mechanism of immune resistance,particularly against T cells that are speci?c for tumour antigens.66 Cytotoxic T-lymphocyte-associated antigen4(CTLA-4)is one of these immune checkpoints that plays a critical role in regulating and limiting immune responses and can be blocked by speci?c antibodies such as ipilimumab,a fully human anti-body.Binding to CTLA-4blocks its activity,thereby sustaining an active immune response in its attack on cancer cells.After successful clinical trials and approval of ipilimumab for advanced melanoma,early phase trials are currently underway in pancreatic cancer for ipilimumab and a second monoclonal antibody targeting CTLA-4,tremelimumab(table1).It remains to be seen whether the therapeutic ef?cacy of this immu-notherapeutic approach demonstrated in melanoma can be extended to pancreatic cancer.

Another promising target in this context was recently pub-lished in a study by Beatty et al.69In this paper,the authors tested the combination of an agonist CD40antibody with gem-citabine in a phase I trial with patients with advanced pancre-atic cancer.CD40is a tumour necrosis factor receptor superfamily member that has been shown to be a key regulatory step in the development of T-cell-dependent antitumor immun-ity,which relies on CD40-mediated‘licensing’of antigen-presenting cells for tumour-speci?c T-cell priming and activa-tion.69T reatment with the CD40agonist CP870,893resulted in tumour regression in some patients,in particular in those with macrophage-dominated in?ammatory tumour in?ltrates. The authors reproduced this treatment effect in a genetically engineered mouse model of pancreatic cancer and found unex-pectedly that CD40-induced tumour regression required macro-phages rather than T cells or gemcitabine.CD40-activated tumoricidal macrophages rapidly in?ltrated tumours and facili-tated the depletion of tumour stroma.69Following these results, the effect of the CD40agonist CP870,893is currently being tested in a neoadjuvant and adjuvant setting in patients with resectable pancreatic cancer(table1).

In addition to enhancing the systemic immune response, attracting selected antitumour cytokines to the site of the tumour is a promising concept that is currently under intense preclinical and clinical investigation.One of the most tumour-selective antigens that can be used to guide cytokines to the site of the tumour is the extradomain B(ED-B)of?bronectin. Expression of ED-B itself is associated with neoangiogenesis and tumour growth.A human single-chain Fv antibody frag-ment L19has recentlyb been generated,which displays high binding af?nity for ED-B.70This antibody fragment can be linked to interleukin(IL)-2,one of the most potent antitumor cytokines,which is,however,too toxic for systemic administra-tion and is only suitable for local therapy.71The fusion product consisting of the ED-B antibody fragment L19and IL-2 (L19-IL2)is currently being tested in a phase I/II trial in pancre-atic cancer(table1).The targeted accumulation of IL-2to the tumour microenvironment by conjugating it to the tumour-selective L19antibody appears to be an attractive concept to enhance the therapeutic index of IL-2.70

Targeting the stromal reaction

The extensive desmoplastic reaction that comprises up to90% of the tumour volume is the predominant histological feature of pancreatic cancer.The stroma consists of a variety of cellular components such as in?ammatory cells,stellate cells and acti-vated?broblasts that are embedded within a dense extracellular matrix.7273Over decades,the majority of research efforts had focussed on the tumour cells thereby largely neglecting the impact of the stromal response.Only recently,the role of the stroma as a barrier fencing off the tumour cells against system-ically applied drugs has been recognised.73Furthermore,it was hypothesised that inef?cient drug delivery due to the intense

Recent advances in clinical practice

stromal reaction may be an important contributor to chemore-sistance in pancreatic cancer.5373

During recent years,several landmark papers have investi-gated the impact of stroma-related signalling pathways on des-moplastic reaction,tumour progression and drug resistance in genetically engineered mouse models.Inhibition of these path-ways is considered a promising tool to decrease stromal density and to facilitate the access of cytotoxic drugs to the tumour cells.The sonic hedgehog pathway is one of the predominant signalling cascades known to stimulate stromal reaction.In a genetic mouse model,systemic administration of the hedgehog inhibitor IPI926resulted in a signi?cant depletion of tumour-associated stroma.This was associated with an increase in the intratumoral vascular density,enhanced concentration of the coadministered cytotoxic agent gemcitabine and increased sur-vival of the animals.53Based on these preclinical data,trials investigating hedgehog inhibitors have been initiated in patients with advanced pancreatic cancer,among them trials evaluating the hedgehog inhibitor IPI926.Recently,however, this trial was stopped.According to a preliminary press release, an interim analysis found that overall survival in patients on the gemcitabine plus IPI926arm was inferior compared to gem-citabine plus placebo(https://www.wendangku.net/doc/7f11408523.html,).Despite these sobering results,further clinical trials evaluating other hedgehog inhibitors such as GDC-0449in combination with gemcitabine or LDE225in combination with FOLFIRINOX or gemcitabine are still ongoing(table1).

The transforming growth factorβ(TGFβ)-dependent signal-ling cascade is known as another key pathway implicated in stromal reaction.74TGFβplays a crucial role in promoting stroma production and invasion,metastasis,angiogenesis and escape from immunosurveillance in pancreatic cancer.75Several drugs have been developed to target TGFβsignalling,among them the antisense oligodeoxynucleotide trabedersen(AP 12009),which speci?cally inhibits TGFβ2expression.This compound demonstrated an excellent safety pro?le and encour-aging survival results when administered as monotherapy in refractory solid tumours including pancreatic cancer76(table1). TGFβsignalling is induced after TGFβbinds as dimers to the TGFβtype II receptor,which recruits and phosphorylates the TGFβtype I receptor.During recent years,efforts have been made to develop compounds that bind to and inhibit TGFβreceptors.A phase I/II trial is currently ongoing that tests the impact of LY2157299,a small molecule that has been designed to inhibit TGFβtype I receptor selectively,in combination with gemcitabine for patients with advanced or metastatic pancreatic cancer(table1).

In addition to targeting stromal-related signalling pathways, acellular matrix components are currently being evaluated as targets for therapeutic intervention:T wo groups recently iden-ti?ed hyaluronan,a non-sulphated glycosaminoglycan,as highly abundant in the extracellular matrix of both human and murine pancreatic cancer tissues.7778In a genetically engi-neered mouse model the authors could show that a PEGylated human recombinant PH20hyaluronidase(PEGPH20)enzymati-cally depletes hyalouronan,thereby inducing re-expansion of tumour blood vessels and increasing the concentration of gem-citabine within the tumour,which resulted in signi?cantly diminished tumour growth and prolonged survival in mice. Based on these promising preclinical data,a phase1b/2trial comparing PEGPH20combined with gemcitabine versus gemci-tabine alone is currently recruiting to evaluate the impact of depleting hyaluronan in patients with metastatic pancreatic cancer(table1).Targeting oncofetal signalling

Signalling pathways that are essential for embryonic develop-ment and tissue homeostasis are frequently reactivated in cancers.These developmental or oncofetal signals have been demonstrated to accelerate tumour progression and mediate resistance to chemotherapy in pancreatic cancer.79A recent study by Jones et al80using a comprehensive genome sequen-cing approach revealed12major signalling pathways that are genetically affected in pancreatic cancer.Interestingly,four of them have been implicated in embryonic development:Notch, Hedgehog,TGFβand Wntβ–catenin pathways are altered in the majority of cases.80Accumulating evidence indicates that these genetic alterations translate into functional reactivation of these pathways in both human tumours and murine models.8182

The Hedgehog and TGFβsignalling pathway are also major determinants of the stromal reactions and have been discussed above.The Notch pathway is thought to maintain pancreatic progenitor cells in an undifferentiated state by enhancing their survival and persistence,in analogy to its function during embryogenesis.81Both the Notch ligand and receptor are highly expressed in pancreatic cancer.83In a genetic mouse model,Notch activation has been shown to synergise with K-Ras in inducing pancreatic intraepithelial neoplasias and invasive carcinoma,84making the Notch pathway a promising therapeutic target.V arious inhibitors have been developed at different levels of the pathway,including inhibitors of the enzymeγ-secretase that plays a key role in the activation of Notch signalling by proteolytic cleavage and release of the intracellular domain of Notch.81Currently,trials evaluating the γ-secretase inhibitors RO4929097and MK0752in patients with metastatic pancreatic cancer are ongoing(table1).

Targeting epigenetic changes

In contrast to genetic alterations leading to altered expression or activity of proteins,epigenetic alterations are heritable,yet not accompanied by changes in DNA sequence.In contrast to genetic mutations,epigenetic changes are potentially reversible and therefore amenable to therapeutic interventions.During recent years,various epigenetic mechanisms affecting gene expression at the chromatin level have been implicated in car-cinogenesis and tumour progression of many human malignan-cies including pancreatic cancer,among them DNA methylation and histone acetylation as most prominent and therapeutically exploitable features.85

Acetylation of histones removes the positive charge on the histones,leading to a more relaxed chromatin structure (euchromatin)that is associated with transcriptionally active DNA.This relaxation can be reversed by histone deacetylases (HDAC).HDAC activity leads to reduced transcription of mul-tiple genes,among them several crucial tumour suppressor genes.This results in enhanced proliferation,resistance to apop-tosis and tumour progression.In pancreatic cancer,several members of the HDAC family such as HDAC2and HDAC6are highly expressed and are known to mediate resistance to apop-tosis.8687Several drugs inhibiting HDAC such as vorinostat have been developed and demonstrate signi?cant antitumor activity in a variety of malignancies.88In pancreatic cancer,vor-inostat is currently being evaluated in combination with radi-ation therapy and chemotherapy in two trials enrolling patients with locally advanced disease(table1).

In contrast to histone acetylation that activates gene tran-scription,DNA methylation is associated with gene silencing.

Recent advances in clinical practice

Globally,the DNA of cancer cells is considered to be hypo-methylated.This is associated with genomic instability and transcription of silenced transposable sequences.89However, the CpG islands of multiple promoter regions of tumour sup-pressor genes frequently undergo DNA hypermethylation, leading to gene silencing and promotion of cancer develop-ment.89DNA hypermethylation of tumour suppressor genes is therefore an attractive target for therapeutic intervention. 5-Azacitidine is a chemical cytosine analogue known to inhibit DNA methyltransferase at low doses.Based on promising in-vitro data,90a phase I trial of5-azacitidine plus gemcitabine in patients with advanced pancreatic cancer is currently recruit-ing(table1).It remains to be determined whether the net effect of a drug like5-azacitidine on hypermethylated tumour suppressors outweighs potentially harmful interference with methylation signatures of other genes in humans.

THE CHALLENGE:DEFINING PERSONALISED THERAPIES The ultimate goal to overcome the appalling resistance of pan-creatic cancer to numerous systemic therapeutic approaches may be achieved not only by successfully developing new tar-geted agents but also by de?ning which patient subgroups might obtain the greatest bene?t from them.Accumulating evi-dence indicates that PDAC is characterised by a marked genetic heterogeneity.By combining comprehensive transcriptional and genomic analyses,Jones et al80de?ned a core set of12cellular signalling pathways and processes that were each genetically altered in the majority of investigated PDAC cases.However, for each individual case,the genes and pathways affected varied considerably,suggesting that an individualised approach to therapy is likely to be required.3

Facing the long list of futile clinical trials using targeted ther-apies,it may be speculated that some of the agents that failed to bene?t unselected populations of patients with PDAC might have had an impact in a more individualised strategy using selected patients with distinct molecular subtypes responsive to a given targeted agent.

Based on transcriptional pro?les of PDAC tissues,Collisson and coworkers91recently de?ned three PDAC subtypes(classic, quasimesenchymal and exocrine-like),which differed signi?-cantly in clinical outcome and therapeutic response to gemcita-bine and erlotinib.Although currently not ready for routine clinical use,the gene signatures for these subtypes may have utility in stratifying patients for subtype-speci?c therapies. CONCLUSIONS

Numerous novel therapeutic avenues targeting tumour cells, tumour vasculature or stromal response are currently under evaluation in clinical trials.T o date,it is unclear which of these avenues—if any—can be translated into clinical routine care for patients with advanced pancreatic cancer.Generally,clinical trials for pancreatic cancer face several obstacles such as short overall survival time and heterogeneity of the disease.Apart from the marginal bene?t demonstrated by erlotinib,all trials testing novel targeted approaches have failed so far.The advent of the FOLFIRINOX regimen has signi?cantly widened the armamentarium of chemotherapeutic options that had been largely restricted to gemcitabine-based regimens before.In clin-ical practice,treatment choice between aggressive combination therapy and gemcitabine has to be made individually depending on the patient’s performance status and comorbidities.For locally advanced disease,chemoradiation is an option to improve local control although data in this regard are con?icting.In addition,the right timing of chemoradiation remains to be de?ned(?gure1).

Despite some improvements in chemotherapeutic strategies, the prognosis of advanced pancreatic cancer still remains appal-ling.In order to develop new therapeutic strategies successfully for this disease,preclinical and clinical research has to focus on three main areas essential to overcome drug resistance:(1)opti-misation of drug delivery;(2)identi?cation of novel targets; and(3)identi?cation of molecular markers to individualise therapy decisions.

Numerous novel compounds with promising preclinical ef?-cacy data are currently being evaluated in clinical trials.Given the long list of futile trials with novel targeted agents in the past,it remains speculative at present which—if any—of these agents will be able to make a difference in improving the prog-nosis of patients with advanced pancreatic cancer.Most of the drugs that are currently in clinical evaluation have been identi-?ed and preclinically validated in xenograft models using human pancreatic cancer cell lines injected orthotopically or subcutaneously into immune-de?cient mice.However,given the high stroma content and the strong immune in?ltration fre-quently detected in endogenous pancreatic tumours,the trans-ferability of most in-vitro data and xenograft studies to the human situation is very limited.Therefore,future preclinical testing of therapeutic strategies should be performed in appro-priate genetically engineered mouse models that recapitulate the genetic and morphological characteristics of the human https://www.wendangku.net/doc/7f11408523.html,ing these models,the impact of novel agents not only on tumour cells but also on the tumour microenviron-ment can be evaluated in detail in a preclinical setting. Finally,molecular markers are urgently needed that are able to predict therapeutic responsiveness or resistance to a given drug in an approach towards a personalised medicine.In con-trast to other solid tumours,predictive biomarkers suitable for use in clinical routine are largely missing in pancreatic cancer. All future trials evaluating novel agents should be accompanied by comprehensive translational programmes aiming to identify predictive biomarkers,either in the tissue or in body?uids that are easier to access and to monitor during therapy.

It will be the combination of innovative therapeutic strat-egies and optimised patient selection that hopefully will be able to make a difference and?nally translate into a clinically signi?cant bene?t for patients with advanced pancreatic cancer. Contributors PM drafted the manuscript,TG critically revised the manuscript.

Competing interest None.

Patient consent Obtained.

Provenance and peer review Commissioned;externally peer reviewed. REFERENCES

1.Jemal A,Bray F,Center MM,et al.Global cancer statistics.CA Cancer J Clin

2011;61:69–90.

2.Krejs GJ.Pancreatic cancer:epidemiology and risk factors.Dig Dis2010;28:355–8.

3.Stathis A,Moore MJ.Advanced pancreatic carcinoma:current treatment and

future challenges.Nature reviews.Clin Oncol2010;7:163–72.

4.Burris HA III,Moore MJ,Andersen J,et al.Improvements in survival and clinical

bene?t with gemcitabine as?rst-line therapy for patients with advanced pancreas

cancer:a randomized trial.J Clin Oncol1997;15:2403–13.

5.Berlin JD,Catalano P,Thomas JP,et al.Phase III study of gemcitabine in

combination with?uorouracil versus gemcitabine alone in patients with advanced

pancreatic carcinoma:Eastern Cooperative Oncology Group Trial E2297.J Clin Oncol 2002;20:3270–5.

6.Herrmann R,Bodoky G,Ruhstaller T,et al.Gemcitabine plus capecitabine

compared with gemcitabine alone in advanced pancreatic cancer:a randomized,

multicenter,phase III trial of the Swiss Group for Clinical Cancer Research and the Central European Cooperative Oncology Group.J Clin Oncol2007;25:2212–17.

Recent advances in clinical practice

7.Louvet C,Labianca R,Hammel P,et al.Gemcitabine in combination with oxaliplatin

compared with gemcitabine alone in locally advanced or metastatic pancreatic

cancer:results of a GERCOR and GISCAD phase III trial.J Clin Oncol

2005;23:3509–16.

8.Poplin E,Feng Y,Berlin J,et al.Phase III,randomized study of gemcitabine and

oxaliplatin versus gemcitabine(?xed-dose rate infusion)compared with gemcitabine (30-minute infusion)in patients with pancreatic carcinoma E6201:a trial of the

Eastern Cooperative Oncology Group.J Clin Oncol2009;27:3778–85.

9.Heinemann V,Quietzsch D,Gieseler F,et al.Randomized phase III trial of

gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer.J Clin Oncol2006;24:3946–52.

10.Colucci G,Giuliani F,Gebbia V,et al.Gemcitabine alone or with cisplatin for the

treatment of patients with locally advanced and/or metastatic pancreatic carcinoma:

a prospective,randomized phase III study of the Gruppo Oncologia dell’Italia

Meridionale.Cancer2002;94:902–10.

11.Rocha Lima CM,Green MR,Rotche R,et al.Irinotecan plus gemcitabine results in

no survival advantage compared with gemcitabine monotherapy in patients with

locally advanced or metastatic pancreatic cancer despite increased tumor response rate.J Clin Oncol2004;22:3776–83.

12.Abou-Alfa GK,Letourneau R,Harker G,et al.Randomized phase III study of

exatecan and gemcitabine compared with gemcitabine alone in untreated advanced pancreatic cancer.J Clin Oncol2006;24:4441–7.

13.Oettle H,Richards D,Ramanathan RK,et al.A phase III trial of pemetrexed plus

gemcitabine versus gemcitabine in patients with unresectable or metastatic

pancreatic cancer.Ann Oncol2005;16:1639–45.

14.Heinemann V,Labianca R,Hinke A,et al.Increased survival using platinum analog

combined with gemcitabine as compared to single-agent gemcitabine in advanced pancreatic cancer:pooled analysis of two randomized trials,the GERCOR/GISCAD

intergroup study and a German multicenter study.Ann Oncol2007;18:1652–9. 15.Sultana A,Smith CT,Cunningham D,et al.Meta-analyses of chemotherapy for

locally advanced and metastatic pancreatic cancer.J Clin Oncol2007;25:2607–15.

16.Conroy T,Desseigne F,Ychou M,et al.FOLFIRINOX versus gemcitabine for

metastatic pancreatic cancer.N Engl J Med2011;364:1817–25.

17.Pelzer U,Schwaner I,Stieler J,et al.Best supportive care(BSC)versus oxaliplatin,

folinic acid and5-?uorouracil(OFF)plus BSC in patients for second-line advanced

pancreatic cancer:a phase III-study from the German CONKO study group.Eur J

Cancer2011;47:1676–81.

18.Kindler HL,Niedzwiecki D,Hollis D,et al.Gemcitabine plus bevacizumab compared

with gemcitabine plus placebo in patients with advanced pancreatic cancer:phase III trial of the Cancer and Leukemia Group B(CALGB80303).J Clin Oncol

2010;28:3617–22.

19.Kindler HL,Wroblewski K,Wallace JA,et al.Gemcitabine plus sorafenib in patients

with advanced pancreatic cancer:a phase II trial of the University of Chicago Phase II Consortium.Invest New Drugs2012;30:382–6.

20.O’Reilly EM,Niedzwiecki D,Hall M,et al.A Cancer and Leukemia Group B phase II

study of sunitinib malate in patients with previously treated metastatic pancreatic

adenocarcinoma(CALGB80603).Oncologist2010;15:1310–19.

21.Bramhall SR,Schulz J,Nemunaitis J,et al.A double-blind placebo-controlled,

randomised study comparing gemcitabine and marimastat with gemcitabine and

placebo as?rst line therapy in patients with advanced pancreatic cancer.Br J

Cancer2002;87:161–7.

22.Van Cutsem E,van de Velde H,Karasek P,et al.Phase III trial of gemcitabine plus

tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer.J Clin Oncol2004;22:1430–8.

23.Moore MJ,Goldstein D,Hamm J,et al.Erlotinib plus gemcitabine compared with

gemcitabine alone in patients with advanced pancreatic cancer:a phase III trial of the National Cancer Institute of Canada Clinical Trials Group.J Clin Oncol

2007;25:1960–6.

24.Heinemann V,Haas M,Boeck S.Systemic treatment of advanced pancreatic

cancer.Cancer Treat Rev2012;38:843–53.

25.Callery MP,Chang KJ,Fishman EK,et al.Pretreatment assessment of resectable

and borderline resectable pancreatic cancer:expert consensus statement.Ann Surg Oncol2009;16:1727–33.

26.Johung K,Saif MW,Chang BW.Treatment of locally advanced pancreatic cancer:

the role of radiation therapy.Int J Radiat Oncol,Biol,Phys2012;82:508–18.

27.Moertel CG,Frytak S,Hahn RG,et al.Therapy of locally unresectable pancreatic

carcinoma:a randomized comparison of high dose(6000rads)radiation alone,

moderate dose radiation(4000rads+5-?uorouracil),and high dose radiation+

5-?uorouracil:the Gastrointestinal Tumor Study Group.Cancer1981;48:1705–10. 28.Sultana A,Tudur Smith C,Cunningham D,et al.Systematic review,including

meta-analyses,on the management of locally advanced pancreatic cancer using

radiation/combined modality therapy.Br J Cancer2007;96:1183–90.

29.The Gastrointestinal Tumor Study Group.Treatment of locally unresectable

carcinoma of the pancreas:comparison of combined-modality therapy

(chemotherapy plus radiotherapy)to chemotherapy alone.Gastrointestinal Tumor

Study Group.J Natl Cancer Inst1988;80:751–5.

30.Klaassen DJ,MacIntyre JM,Catton GE,et al.Treatment of locally unresectable

cancer of the stomach and pancreas:a randomized comparison of5-?uorouracil

alone with radiation plus concurrent and maintenance5-?uorouracil—an Eastern

Cooperative Oncology Group study.J Clin Oncol1985;3:373–8.

31.Hazel JJ,Thirlwell MP,Huggins M,et al.Multi-drug chemotherapy with and

without radiation for carcinoma of the stomach and pancreas:a prospective

randomized trial.J Can Assoc Radiol1981;32:164–5.

32.Chauffert B,Mornex F,Bonnetain F,et al.Phase III trial comparing intensive

induction chemoradiotherapy(60Gy,infusional5-FU and intermittent cisplatin)

followed by maintenance gemcitabine with gemcitabine alone for locally advanced unresectable pancreatic cancer.De?nitive results of the2000–01FFCD/SFRO study.

Ann Oncol2008;19:1592–9.

33.Loehrer PJ Sr,Feng Y,Cardenes H,et al.Gemcitabine alone versus gemcitabine

plus radiotherapy in patients with locally advanced pancreatic cancer:an Eastern

Cooperative Oncology Group trial.J Clin Oncol2011;29:4105–12.

34.Goodman KA,Hajj C.Role of radiation therapy in the management of pancreatic

cancer.J Surg Oncol Published Online First:24Apr2012.doi:10.1002/jso.23137 35.Li J,Ng J,Allendorf J,et al.Locally advanced pancreatic adenocarcinoma:are we

making progress?.Highlights from the“2011ASCO Annual Meeting”.Chicago,IL, USA;June3–7,2011.JOP2011;12:347–50.

36.Li CP,Chao Y,Chi KH,et al.Concurrent chemoradiotherapy treatment of locally

advanced pancreatic cancer:gemcitabine versus5-?uorouracil,a randomized

controlled study.Int J Radiat Oncol,Biol,Phys2003;57:98–104.

37.Crane CH,Abbruzzese JL,Evans DB,et al.Is the therapeutic index better with

gemcitabine-based chemoradiation than with5-?uorouracil-based chemoradiation in locally advanced pancreatic cancer?Int J Radiat Oncol,Biol,Phys

2002;52:1293–302.

38.Small W Jr,Berlin J,Freedman GM,et al.Full-dose gemcitabine with concurrent

radiation therapy in patients with nonmetastatic pancreatic cancer:a multicenter

phase II trial.J Clin Oncol2008;26:942–7.

39.Gillen S,Schuster T,Meyer Zum Buschenfelde C,et al.Preoperative/neoadjuvant

therapy in pancreatic cancer:a systematic review and meta-analysis of response

and resection percentages.PLoS Med2010;7:e1000267.

40.Assi?MM,Lu X,Eibl G,et al.Neoadjuvant therapy in pancreatic adenocarcinoma:

a meta-analysis of phase II trials.Surgery2011;150:466–73.

41.Habermehl D,Kessel K,Welzel T,et al.Neoadjuvant chemoradiation with

gemcitabine for locally advanced pancreatic cancer.Radiat Oncol2012;7:28.

42.Olson P,Hanahan D.Cancer.Breaching the cancer fortress.Science

2009;324:1400–1.

43.Von Hoff DD,Ramanathan RK,Borad MJ,et al.Gemcitabine plus nab-paclitaxel is

an active regimen in patients with advanced pancreatic cancer:a phase I/II trial.J Clin Oncol2011;29:4548–54.

44.Infante JR,Matsubayashi H,Sato N,et al.Peritumoral?broblast SPARC expression

and patient outcome with resectable pancreatic adenocarcinoma.J Clin Oncol

2007;25:319–25.

45.Frese KK,Neesse A,Cook N,et al.nab-Paclitaxel potentiates gemcitabine activity

by reducing cytidine deaminase levels in a mouse model of pancreatic cancer.

Cancer Discov2012;2:260–9.

46.Fasol U,Frost A,Buchert M,et al.Vascular and pharmacokinetic effects of

EndoTAG-1in patients with advanced cancer and liver metastasis.Ann Oncol

2012;23:1030–6.

47.Loehr M,Bodoky G,F?lsch UA,et al.Cationic liposomal paclitaxel in combination

with gemcitabine in patients with advanced pancreatic cancer:a phase II trial.

J Clin Oncol2009;27:abstract4526.

48.Ko AH,Tempero MA,Shan Y,et al.A multinational phase II study of PEP02

(liposome irinotecan)for patients with gemcitabine-refractory metastatic pancreatic cancer.J Clin Oncol2011;29:abstract4069.

49.Segal NH,Saltz LB.Evolving treatment of advanced colon cancer.Ann Rev Med

2009;60:207–19.

50.Kindler HL,Ioka T,Richel DJ,et al.Axitinib plus gemcitabine versus placebo plus

gemcitabine in patients with advanced pancreatic adenocarcinoma:a double-blind randomised https://www.wendangku.net/doc/7f11408523.html,ncet Oncol2011;12:256–62.

51.Gon?alves A,Gilabert M,Fran?ois E,et al.BAYPAN study:a double-blind phase III

randomized trial comparing gemcitabine plus sorafenib and gemcitabine plus

placebo in patients with advanced pancreatic cancer.Ann Oncol Published Online

First:5Jul2012.doi:10.1093/annonc/mds135

52.Gaya A,Tse V.A preclinical and clinical review of a?ibercept for the management

of cancer.Cancer Treat Rev2012;38:484–93.

53.Olive KP,Jacobetz MA,Davidson CJ,et al.Inhibition of Hedgehog signaling

enhances delivery of chemotherapy in a mouse model of pancreatic cancer.Science 2009;324:1457–61.

54.Momose I,Kunimoto S,Osono M,et al.Inhibitors of insulin-like growth factor-1

receptor tyrosine kinase are preferentially cytotoxic to nutrient-deprived pancreatic cancer cells.Biochem Biophys Res Commun2009;380:171–6.

55.Philip PA,Goldman BH,Ramanathan RK,et al.Phase I randomized phase II trial of

gemcitabine,erlotinib,and cixutumumab versus gemcitabine plus erlotinib as?rst-line treatment in patients with metastatic pancreatic cancer(SWOG-0727).J Clin Oncol2012;30(suppl4):abstract198.

56.Remmers N,Bailey JM,Mohr AM,et al.Molecular pathology of early pancreatic

cancer.Cancer Biomarkers:Sect A Dis Markers2011;9:421–40.

Recent advances in clinical practice

57.Michl P,Downward J.Mechanisms of disease:PI3K/AKT signaling in

gastrointestinal cancers.Z Gastroenterol2005;43:1133–9.

58.Yamamoto S,Tomita Y,Hoshida Y,et al.Prognostic signi?cance of activated Akt

expression in pancreatic ductal adenocarcinoma.Clin Cancer Res2004;10:2846–50.

59.Okami K,Wu L,Riggins G,et al.Analysis of PTEN/MMAC1alterations in

aerodigestive tract tumors.Cancer Res1998;58:509–11.

60.Pal SK,Reckamp K,Yu H,et al.Akt inhibitors in clinical development for the

treatment of cancer.Expert Opin Investig Drugs2010;19:1355–66.

61.Wolpin BM,Hezel AF,Abrams T,et al.Oral mTOR inhibitor everolimus in patients

with gemcitabine-refractory metastatic pancreatic cancer.J Clin Oncol

2009;27:193–8.

62.Pollak M.Metformin and pancreatic cancer:a clue requiring investigation.Clin

Cancer Res2012;18:2723–5.

63.Evans JM,Donnelly LA,Emslie-Smith AM,et al.Metformin and reduced risk of

cancer in diabetic patients.BMJ2005;330:1304–5.

64.Bodmer M,Becker C,Meier C,et https://www.wendangku.net/doc/7f11408523.html,e of antidiabetic agents and the risk of

pancreatic cancer:a case–control analysis.Am J Gastroenterol2012;107:620–6. 65.Regel I,Kong B,Raulefs S,et al.Energy metabolism and proliferation in pancreatic

https://www.wendangku.net/doc/7f11408523.html,ngenbeck’s Archives ofSsurgery/Deutsche Gesellschaft fur

Chirurgie2012;397:507–12.

66.Pardoll DM.The blockade of immune checkpoints in cancer immunotherapy.

Nature reviews.Cancer2012;12:252–64.

67.Vanneman M,Dranoff https://www.wendangku.net/doc/7f11408523.html,bining immunotherapy and targeted therapies in

cancer treatment.Nature reviews.Cancer2012;12:237–51.

68.Dodson LF,Hawkins WG,Goedegebuure P.Potential targets for pancreatic cancer

immunotherapeutics.Immunotherapy2011;3:517–37.

69.Beatty GL,Chiorean EG,Fishman MP,et al.CD40agonists alter tumor stroma and

show ef?cacy against pancreatic carcinoma in mice and humans.Science

2011;331:1612–16.

70.Wagner K,Schulz P,Scholz A,et al.The targeted immunocytokine L19-IL2

ef?ciently inhibits the growth of orthotopic pancreatic cancer.Clin Cancer Res

2008;14:4951–60.

71.Bubenik J,Den Otter W,Huland E.Local cytokine therapy of cancer:interleukin-2,

interferons and related cytokines.Cancer Immunol Immunother2000;49:116–22. 72.Michl P,Gress TM.Improving drug delivery to pancreatic cancer:breaching the

stromal fortress by targeting hyaluronic acid.Gut2012;61:1377–9.

73.Neesse A,Michl P,Frese KK,et al.Stromal biology and therapy in pancreatic

cancer.Gut2011;60:861–8.

74.Hilbig A,Oettle H.Transforming growth factor beta in pancreatic cancer.Curr

Pharm Biotechnol2011;12:2158–64.

75.Fuxe J,Karlsson MC.TGF-beta-induced epithelial-mesenchymal transition:A link

between cancer and in?ammation.Semin Cancer Biol2012;22:455–61.76.Oettle H,Seufferlein T,et al.Phase I/II study with trabedersen(AP12009)

monotherapy for the treatment of patients with advanced pancreatic cancer,

malignant melanoma,and colorectal carcinoma.J Clin Oncol2011;29:

abstract2513.

77.Jacobetz MA,Chan DS,Neesse A,et al.Hyaluronan impairs vascular function and

drug delivery in a mouse model of pancreatic cancer.Gut Published Online First:30 Mar2012.doi:10.1136/gutjnl-2012-302529

78.Provenzano PP,Cuevas C,Chang AE,et al.Enzymatic targeting of the stroma

ablates physical barriers to treatment of pancreatic ductal adenocarcinoma.

Cancer Cell2012;21:418–29.

79.Heiser PW,Hebrok M.Development and cancer:lessons learned in the pancreas.

Cell Cycle2004;3:270–2.

80.Jones S,Zhang X,Parsons DW,et al.Core signaling pathways in human pancreatic

cancers revealed by global genomic analyses.Science2008;321:1801–6.

81.Ristorcelli E,Lombardo D.Targeting Notch signaling in pancreatic cancer.Expert

Opin Ther Targets2010;14:541–52.

82.Thayer SP,di Magliano MP,Heiser PW,et al.Hedgehog is an early and late

mediator of pancreatic cancer tumorigenesis.Nature2003;425:851–6.

83.Sjolund J,Manetopoulos C,Stockhausen MT,et al.The Notch pathway in cancer:

differentiation gone awry.Eur J Cancer2005;41:2620–9.

84.Mazur PK,Einwachter H,Lee M,et al.Notch2is required for progression of

pancreatic intraepithelial neoplasia and development of pancreatic ductal

adenocarcinoma.Proc Natl Acad Sci U S A2010;107:13438–43.

85.Omura N,Goggins M.Epigenetics and epigenetic alterations in pancreatic cancer.

Int J Clin Exp Pathol2009;2:310–26.

86.Fritsche P,Seidler B,Schuler S,et al.HDAC2mediates therapeutic resistance of

pancreatic cancer cells via the BH3-only protein NOXA.Gut2009;58:1399–409. 87.Nawrocki ST,Carew JS,Pino MS,et al.Bortezomib sensitizes pancreatic cancer

cells to endoplasmic reticulum stress-mediated apoptosis.Cancer Res

2005;65:11658–66.

88.Marks PA,Breslow R.Dimethyl sulfoxide to vorinostat:development of this

histone deacetylase inhibitor as an anticancer drug.Nature Biotechnol

2007;25:84–90.

89.Lopez-Serra P,Esteller M.DNA methylation-associated silencing of

tumor-suppressor microRNAs in cancer.Oncogene2012;31:1609–22.

90.Missiaglia E,Donadelli M,Palmieri M,et al.Growth delay of human pancreatic

cancer cells by methylase inhibitor5-aza-20-deoxycytidine treatment is

associated with activation of the interferon signalling pathway.Oncogene

2005;24:199–211.

91.Collisson EA,Sadanandam A,Olson P,et al.Subtypes of pancreatic

ductal adenocarcinoma and their differing responses to therapy.Nat Med

2011;17:500–3.

Recent advances in clinical practice

doi: 10.1136/gutjnl-2012-303588

2013 62: 317-326 originally published online October 30, 2012

Gut

Patrick Michl and Thomas M Gress

advanced pancreatic cancer

Current concepts and novel targets in

https://www.wendangku.net/doc/7f11408523.html,/content/62/2/317.full.html Updated information and services can be found at: These include:

References

https://www.wendangku.net/doc/7f11408523.html,/content/62/2/317.full.html#ref-list-1This article cites 84 articles, 39 of which can be accessed free at:service

Email alerting

the box at the top right corner of the online article.

Receive free email alerts when new articles cite this article. Sign up in Collections

Topic

(586 articles)

Pancreatic cancer (1817 articles)Pancreas and biliary tract (61 articles)GUT Recent advances in clinical practice

Articles on similar topics can be found in the following collections Notes

https://www.wendangku.net/doc/7f11408523.html,/group/rights-licensing/permissions To request permissions go to:

https://www.wendangku.net/doc/7f11408523.html,/cgi/reprintform To order reprints go to:

https://www.wendangku.net/doc/7f11408523.html,/subscribe/To subscribe to BMJ go to:

相关文档
相关文档 最新文档