carcinoma pancreático aspectos gerais pancreatic ... · carcinoma pancreático – aspectos gerais...

77
Sofia Eduarda Tavares Sousa Castro Carcinoma Pancreático Aspectos Gerais Pancreatic Adenocarcinoma An overview 2009/2010 Abril, 2010

Upload: others

Post on 25-Oct-2019

6 views

Category:

Documents


0 download

TRANSCRIPT

Sofia Eduarda Tavares Sousa Castro

Carcinoma Pancreático – Aspectos Gerais

Pancreatic Adenocarcinoma – An overview

2009/2010

Abril, 2010

Sofia Eduarda Tavares Sousa Castro

Carcinoma Pancreático – Aspectos Gerais

Pancreatic Adenocarcinoma – An overview

Mestrado Integrado em Medicina

Área: Cirurgia Geral

Trabalho efectuado sob a Orientação de:

Prof. Doutor António Taveira Gomes

Abril, 2010

Revista:

World Journal of Surgical Oncology

Projecto de Opção do 6º ano - DECLARAÇÃO DE INTEGRIDADE

Eu, _ Sofia Eduarda Tavares Sousa Castro ____________, abaixo assinado, nº

mecanográfico_040801148____, aluno do 6º ano do Mestrado Integrado em Medicina, na

Faculdade de Medicina da Universidade do Porto, declaro ter actuado com absoluta integridade

na elaboração deste projecto de opção.

Neste sentido, confirmo que NÃO incorri em plágio (acto pelo qual um indivíduo, mesmo por

omissão, assume a autoria de um determinado trabalho intelectual, ou partes dele). Mais

declaro que todas as frases que retirei de trabalhos anteriores pertencentes a outros autores,

foram referenciadas, ou redigidas com novas palavras, tendo colocado, neste caso, a citação da

fonte bibliográfica.

Faculdade de Medicina da Universidade do Porto, 19 / 04 / 2010__

Assinatura: ________________________________________________

Projecto de Opção do 6º ano - DECLARAÇÃO DE REPRODUÇÃO

Nome: Sofia Eduarda Tavares Sousa Castro

Endereço electrónico: [email protected] Telefone ou Telemóvel: 917032226

Número do Bilhete de Identidade: 12942726

Título da Dissertação/Monografia/Relatório de Estágio:

Carcinoma Pancreático – Aspectos Gerais / Pancreatic Adenocarcinoma – An Overview

Nome completo do Orientador:

Professor Doutor António Taveira Gomes

Nome completo do Co-Orientador:

_____________________________________________________________________________

Ano de conclusão: _2010______

Designação da área do projecto de opção:

Cirurgia Geral

É autorizada a reprodução integral desta Dissertação/Monografia/Relatório de Estágio (cortar o

que não interessar) apenas para efeitos de investigação, mediante declaração escrita do

interessado, que a tal se compromete.

Faculdade de Medicina da Universidade do Porto, 19 / 04 / 2010_

Assinatura: ________________________________________________

1

Carcinoma Pancreático – Aspectos gerais

Pancreatic Adenocarcinoma – An Overview

Sofia Eduarda Tavares Sousa Castro1§

1Student from Faculty of Medicine University of Porto, Porto, Portugal

§Corresponding author:

Email Address: [email protected]; [email protected].

2

INDEX

Abstract 4

Resumo 5

Introduction 6

Epidemiology 7

Risk Factors 9

Environmental Factors 9

Smoking 9

Dietary Factors 10

Pre-Existing Diseases 11

Chronic Pancreatitis and other Digestive Diseases 11

Diabetes 12

Genetic Factors 13

Percursor Lesions 16

Mucinous Cystic Neoplasms (MCNs) 16

Intraductal Papillary Mucinous Neoplasms (IPMNs) 17

Pancreatic Intraepithelial Neoplasias (PanINs) 19

Ductal Adenocarcinoma 20

Diagnosis 21

Clinical Findings 21

Laboratory Studies 22

Imaging Studies 25

Transabdominal Ultrasound 26

Computed Tomography (CT) 26

Magnetic Resonance Imaging (MRI) 27

Endoscopic Ultrasound (EUS) 29

Biopsy 30

Endoscopic Retrograde Cholangiopancreatography 31

3

Laparoscopy 32

Positron Emission Tomography (PET) 32

Staging and Resectability 34

Distinction between Resectable, Locally Advanced and Metastatic disease 35

Borderline Resectable Disease 37

Surgical Treatment 38

Resection for Tumors of the Head of the Pancreas 39

Pancreaticoduodenectomy (Kausch-Whipple procedure) 39

Pylorus-Preserving Pancreaticoduodenectomy (PPPD) 40

Pancreatic Anastomosis and Reconstruction 40

Resection for Tumors of the Body/Tail of the Pancreas 42

Distal Pancreatectomy 42

Central Pancreatectomy 42

Total Pancreatectomy 43

Extended Lymphadenectomy – Extended Lymph Node Dissection 43

Arterial/Venous Resections 44

Short-Term Outcome 44

Long-Term Outcome 45

Chemoradiotherapy 47

Palliation Treatment 52

Biliary Obstruction 52

Gastric Outlet Obstruction 52

Severe Abdominal Pain 53

Pancreatic Insufficiency 53

Conclusions 54

Acknowledgements 55

References 56

Figures 70

Tables 71

4

ABSTRACT

Pancreatic cancer remains a fearsome disease. This article focuses on the epidemiology,

pathogenesis, diagnosis, staging, and treatment of pancreatic adenocarcinoma.

Smoking has been clearly established as a major risk factor affecting the carcinogenesis

of pancreatic cancer. Other risk factors are still to be confirmed.

Different genetic alterations have been observed in pancreatic adenocarcinoma and new

developments have been made in our understanding on progression from benign lesion to

invasive cancer.

Imaging studies play a crucial role in the diagnosis, staging and management of patients

with pancreatic adenocarcinoma and computed tomography is the most widely used and best-

validated imaging method.

Surgery remains the single most important modality for curative treatment of pancreatic

adenocarcinoma, however, it can only be performed in about 10-15% of the patients. Surgery

for this disease is a complex procedure associated with considerable perioperative morbidity and

mortality. Many aspects of surgery for pancreatic adenocarcinoma, such as the extent of

resection and limphadenectomy, the value of vascular resection, and the importance of treatment

by experienced surgeons, are currently under debate.

Adjuvant treatment improves survival after surgical resection and each patient should be

offered this treatment option. However, a standard treatment protocol has not been established

yet.

For the majority of patients, treatment is palliative and may include surgery or endoscopic

or percutaneous stenting to relieve obstructive jaundice or gastric obstruction, other

interventional radiological techniques, chemotherapy or radiotherapy. Adequate pain relief and

treatment of pancreatic insufficiency are also important components of treatment.

5

RESUMO

O cancro do pâncreas continua a ser uma doença temível. Este artigo aborda a

epidemiologia, patogénese, diagnóstico, estadiamento e tratamento do adenocarcinoma

pancreático.

O tabaco é um factor de risco claramente estabelecido na carcinogénese desta neoplasia.

Outros factores de risco estão ainda por confirmar.

Diferentes alterações genéticas foram observadas nas neoplasias pancreáticas e novos

desenvolvimentos têm sido feitos na compreensão da progressão de uma lesão benigna para

carcinoma invasivo.

Os estudos imagiológicos são cruciais no diagnóstico, estadiamento e abordagem de

doentes com adenocarcinoma pancreático e a tomografia computorizada é o método de imagem

mais usado e validado.

A cirurgia mantém-se como a estratégia terapêutica curativa mais importante, contudo,

esta só pode ser realizada em cerca de 10-15% dos doentes. A cirurgia para esta patologia é

complexa e está associada a uma morbilidade e mortalidade perioperatória consideráveis.

Diversos aspectos desta continuam em discussão, como a extensão de ressecção e da

linfadenectomia, o valor da ressecção vascular e a importância da realização do tratamento por

cirurgiões experientes.

O tratamento adjuvante melhora a sobrevida após ressecção e esta opção deve ser

oferecida aos doentes. Contudo, ainda não foi estabelecido um protocolo terapêutico standard.

Para a maioria dos doentes, o tratamento paliativo pode incluir procedimentos cirúrgicos

ou colocação de stents via endoscópica ou percutânea para aliviar icterícia obstrutiva ou

obstrução gástrica; outras técnicas radiológicas de intervenção, quimioterapia ou radioterapia.

Alívio adequado da dor e tratamento da insuficiência pancreática são também componentes

importantes do tratamento.

6

INTRODUCTION

Pancreatic adenocarcinoma remains one of the most feared gastrointestinal tract

malignancies. It has the highest ratio of diagnosis to mortality.[1-2] Most patients die within a

year after establishment of the diagnosis[3], and the overall prognosis is dismal, with a 5-year

survival rate of <5% [2, 4]. The reasons for this low survival are related to late diagnosis and to

the aggressive biology of this disease: early development of retroperitoneal and perineural

infiltration, angioinvasion, peritoneal, lymphogenic, and hematogenic dissemination, and

resistance to most of the available treatment regimens.[5-6] Thus, this makes patient

management a complex and challenging task.

Despite this grim picture, significant advances have been made in recent years, both in

our understanding of the molecular behavior and pathogenesis of this disease and in its clinical

management.[6-7] Better prognosis could be achieved by combining earlier diagnose (with

markers and modern imaging techniques), surgery, and chemotherapy/radiotherapy with

targeted molecular and immune therapies. [6] Therefore, the main focus of academic medicine

in the field of pancreatic adenocarcinoma will be translational research, transferring advances

from basic research to clinical studies for the benefit of patients.[6]

This article has the objective of reviewing the current knowledge on many aspects

concerning pancreatic adenocarcinoma, like epidemiology and risk factors, as well as the recent

developments in molecular genetics and disease progression. Moreover, it has the purpose of

updating the current thinking on management of this malignant disease, including diagnosis,

staging, surgical resection, adjuvant therapy, and palliation.

7

EPIDEMIOLOGY

Over the last few decades, the study of pancreatic cancer has assumed a position of

growing importance because of its increasing incidence and poor prognosis.[8] Ductal

adenocarcinoma is the most common form of pancreatic cancer.[9]

In Europe, pancreatic cancer is the tenth most frequent cancer, accounting for 3% of

cancer in both sexes and in the year 2000 an estimated 74 000 new cases were diagnosed.[10]

In the United States pancreatic cancer develops in approximately 30,000 patients per

year, and about 20,000 annual cases in Japan. Compared with more common tumors such as

lung, breast, colon, or prostate tumors, it is a rare cancer.[11-12] However, because it is so

lethal, it ranks fourth as cause of death from cancer [11], with 80–90% of cases diagnosed in the

non-resectable stage. Consequently, the survival rate is extremely low.[8] For all stages

combined, the 1-year survival rate is 19% and the 5-year survival rate is 4%. Survival is low

because of the late development of clinical symptoms. Surgical resection (when margin negative

and node negative) offers the best possibility for cure in pancreatic cancer, with the 5-year

survival rate improving to 40%, when performed at specialized medical institutions.[9]

With the rapid advancement of effective screening and therapeutic regimes for breast and

colon cancer resulting in decreased deaths from these diseases, pancreatic cancer is likely to

become an increasingly frequent cause of death from cancer.[11]

In North America, the incidence rate of pancreatic cancer has remained constant or has

declined slightly during the last 25 years, while its frequency tend to rise in Japan and in

virtually all European countries.[7-9]

As with nearly all other types of digestive tract cancer, pancreatic cancer exhibits an

uneven world distribution. Incidence rates in high-risk countries are about 5–7 times higher than

incidence rates in low-risk countries, implying that environmental factors play an important

role.[13] There is substantial geographical variation in incidence of pancreatic cancer with the

highest rates being seen in the USA (particularly in black men), Europe and in other western,

industrialized countries.[7-8, 10-11, 14] The lowest rates are generally found in Africa and some

8

Asian countries, although Japan, which has seen a substantial increase in rates in recent decades,

now has rates similar to those seen in the USA.[10]

There may be also racial differences in survival patterns after diagnosis of pancreatic

cancer, perhaps related to racial differences in aggressiveness of tumor type.[11] Asian patients

tended to have less aggressive tumors.[13] In Europe, the highest mortality rates occur in

Austria and Sweden and in southern Europe (Spain, Portugal and Greece) the corresponding

rates are generally lower.[10]

Moreover, in all cancer registries, pancreatic cancer is slightly more common in males

than in females.[8-9, 12]

Pancreatic cancer is predominately a disease of older individuals.[13] Incidence rates

increase steadily with age, with over 80% of cases occurring between the ages of 60 and 80[7-

8], with the average age at diagnosis being 60 to 65 years[9]. The diagnosis of pancreatic

cancer in individuals younger than age 40 is uncommon and is rare in people under 25 years of

age.[7-8]

9

RISK FACTORS

The causes of all types of disease fall along a continuum ranging from diseases where a

genetic mutation causes all cases, to a purely environmental disease. Environmental factors are

believed to be the most important cause of most types of cancer, including pancreatic cancer,

although there are subgroups of patients where genetic factors are important.[12]

Environmental Factors

Smoking

For more than 30 years, we have known that exposure to tobacco smoke is the most

important environmental factor leading to pancreatic cancer.[11-12]

Pancreatic cancer is unquestionably one of the major smoking related tumors. Unlike the

lung, where tobacco smoke and tobacco degradation products are in direct contact with

pulmonary tissue, the pancreas is exposed to tobacco products indirectly. Tobacco-related

carcinogens reach the pancreas either via the blood stream, or perhaps through exposure to

either duodenal contents or to bile. Most pancreatic cancers occur in the head of the gland and

this region is where exposure to tobacco carcinogens contained in the duodenal juice or bile

could occur.[11]

Since exposure to tobacco products is lower in the pancreas than in the lung, it is

understandable that the smoking-related risk of pancreatic cancer is not as high as that of lung

cancer.[11] Most studies have found that smoking results in about a two-fold increased risk of

pancreatic cancer. [9-14] Also, it has been demonstrated a graded dose response, with heavy

smokers having a substantially higher risk.[13]

Based on current smoking prevalence rates it has been estimated that it contributes to the

development of almost 30% of all pancreatic cancers.[9-10, 12-13] Differential smoking rates

10

cause much of the gender-specific and country-specific differences in pancreatic cancer

rates.[12]

As a public health measure avoiding smoking will substantially reduce the risk of

pancreatic cancer.[12] Smoking cessation is also beneficial, but it takes about 15 years after

quitting cigarette smoking, among the heaviest smokers, for the risk to fall to a level comparable

with that in never-smokers.[8]

Dietary Factors

Some of the strongest evidence for the importance of dietary factors comes from the

several-fold differences observed in incidence rates in various countries. Dietary differences

would explain some of the wide variation in incidence rates for pancreatic cancer. However,

attempts to associate specific dietary items with an increased or decreased risk have been

frustrating. Nearly 500 published articles have examined the link between pancreatic cancer and

various dietary items. In most of these reports there is a possibility for recall bias, which would

explain why the results are often contradictory.[12]

Several studies show that caloric consumption and/or obesity can be risk factors for

pancreatic cancer. [12] Diet high in vegetables and fruits are probably protective against

pancreatic cancer, and the diets containing substantial amounts of red meat and cholesterol

possibly increase risk.[10]

Alcohol is a major risk factor for pancreatitis, but does it cause pancreatic cancer? Nearly

all studies fail to support this association. Coffee, another widely consumed beverage is also

unlikely to cause pancreatic cancer.[13]

11

Pre-Existing Diseases

Are there pre-existing diseases that increase the risk of pancreatic cancer? For other

digestive tract organs, there are established links between the occurrence of a nonmalignant

disease and the eventual appearance of cancer in the same organs. Therefore, it is reasonable to

assume that non-malignant pancreatic disorders would increase the risk of pancreatic

cancer.[12]

Chronic Pancreatitis and other Digestive Diseases

Chronic pancreatitis develops in people who have had repeated attacks of acute

pancreatitis.[11] Several studies have now linked chronic pancreatitis with an increased risk of

pancreatic cancer.[12-13, 15-16] Most patients are males who smoke and drink heavily,

although „„idiopathic‟‟ pancreatitis can develop in persons without any history of heavy

drinking.[11]

Hereditary pancreatitis is a rare inherited disease with symptoms that mimic other types

of chronic pancreatitis. It is an autosomal dominant disease with an onset in childhood or early

adulthood [12] and with a penetrance of 80% [8, 11]. Cystic fibrosis affects the digestive tract,

including the pancreas: a few patients with pancreatic cancer have been reported with this

disease.[13] Finally, there is a type of chronic pancreatitis occurring in persons living in the

southern parts of India or Africa. This disease of unknown etiology has been termed tropical

pancreatitis.[11]

In all types of pancreatitis, the risk of pancreatic cancer is elevated. For example, patients

with idiopathic or alcoholic pancreatitis have a 15-fold increased risk of pancreatic cancer. For

those patients with hereditary pancreatitis, the lifetime risk of pancreatic cancer is

approximately 30%–40%; for patients with tropical pancreatitis the risk of pancreatic cancer is

also high.[11]

12

The link between chronic pancreatitis and pancreatic cancer is similar to the well known

increased cancer in other digestive organs where there is a background inflammatory

disease.[11] Thus, increased cell turnover and defected DNA could increase the possibility of

coding errors leading to deleterious mutations.[11-12]

Both gallstone disease and peptic ulcer disease have been suggested as possible

preexisting diseases that might cause pancreatic cancer, but the evidence is weaker than for

chronic pancreatitis.[7, 13]

Diabetes

Diabetes is a common disorder found in 5–10% of the general population [12-13] and it

has been associated with pancreatic cancer for a long time.[15]

The possible role of diabetes in the etiology of pancreatic cancer has been examined with

conflicting results by several investigators. There is an increased frequency of pancreatic cancer

among individuals with a long history of diabetes. Diabetes is an early manifestation of

pancreatic dysfunction, and it may play a causative role in the development of pancreas cancer

in subjects with a long history of diabetes.[8]

As pancreatic cancer is a rapidly progressing disorder with high mortality within 1 year of

diagnosis, it is unlikely to lead to the development of diabetes many years prior to its diagnosis.

Approximately 50% of individuals with diabetes are not aware of their disease, and non-insulin

dependent diabetic patients can live with their undiagnosed condition for extended periods of

time.[8]

A significant proportion of the cases simultaneously diagnosed with diabetes and

pancreatic cancer probably had diabetes for several years before their cancer was detected, but

the diabetes was only discovered during the exploration of pancreas dysfunction and cancer.[8]

As yet, there is no evidence that screening recent onset diabetics would reduce the

mortality from pancreatic cancer.[12]

13

Genetic Factors

The accumulation of multiple nonrandom genetic changes over time is a hallmark of

pancreatic cancer. Genetic abnormalities include alterations in chromosome or gene copy

number, microsatellite instability, epigenetic silencing, intragenic point mutations, and gene

overexpression secondary to increased transcription.[17-18]

Chromosome losses are more common than chromosome gains in pancreatic cancer.

Some of the most common regions of genomic loss in pancreatic cancers contain known tumor

suppressor genes such as CDKN2A/P16/MTS1 (9p21), p53 (17p13), and

MADH4/SMAD4/DPC4 (18q).[17, 19] Frequent gains of DNA have been observed in

pancreatic cancers in several chromosomes (for example in 12p – location of KRAS2

oncogene).[17]

Medullary carcinoma, a rare subtype of pancreatic adenocarcinoma, often contains a

defective DNA mismatch repair mechanism which gives rise to microsatellite instability.[17,

20]

Epigenetic phenomena are DNA modifications that do not involve alterations in DNA

sequence [17, 21] (chromatin structural changes, such as histone modifications and nucleosome

rearrangements, and deregulation of cytosine methylation at the promoter site) [17]. The

expression of many tumor suppressor genes and oncogenes appears to be influenced through

this mechanism.[17]

Many tumor suppressors regulate cell proliferation. Therefore, biallelic inactivation of

these genes can provide a growth advantage for tumors. Conversely, oncogenes promote cell

growth.[17]

CDKN2A/p16/MTS1 is the most frequently inactivated tumor suppressor gene in

pancreatic cancer (95%).[17, 22-23] The gene can be inactivated through homozygous deletion

(40%), single allele loss combined with an intragenic mutation in the second allele (40%), or

promoter hypermethylation (15%). When p16 is inactivated, progression through the G1/S cell

cycle checkpoint is facilitated.[17, 24]

14

p53 is the second most commonly inactivated tumor suppressor gene in pancreatic cancer

(50-75%). Usually, one p53 allele is deleted while the other allele acquires an intragenic

mutation.[17, 24] Activated p53 causes the cell to arrest in G1 or G2 of the cell cycle and

responds to genetic injury by activating apoptotic pathways. Thus, p53 inactivation allows the

tumor to proliferate despite the continued accumulation of genetic defects.[17]

MADH4/SMAD4/DPC4 is the third most frequently inactivated tumor suppressor gene in

pancreatic cancer (55%). SMAD4 is a downstream component of the transforming growth

factor (TGF) and activin signaling pathways. Loss of SMAD4 impairs the cell cycle check

mechanism during the G1/S transition. SMAD4 loss is also believed to affect pro-apoptotic

signaling.[17]

BRCA2 is altered in 7% of pancreatic cancers, but germline mutations can be seen in a

larger proportion of patients (17%) that cluster in pancreatic cancer families.[17]

KRAS2 is the most commonly mutated oncogene in pancreatic cancer (90%). Gain-of-

function mutations that impair KRAS2 GTPase activity augment downstream signaling and

increase cell proliferation.[17]

Individuals with a strong family history of pancreatic cancer have a significantly

increased risk of developing the disease. For example, individuals with a first-degree relative

with pancreatic cancer have a 2,3-fold increased risk.[25]

Five hereditary tumor predisposition syndromes associated with pancreatic cancer have

been identified so far, although these known syndromes account for only a minority (<20%) of

familial cases.

Hereditary nonpolyposis colorectal cancer (HNPCC) is an autosomal dominant disorder

responsible for roughly 3% of colorectal cancers, and has an estimated population prevalence of

1:740. Individuals with HNPCC have a 1-5% lifetime risk of developing pancreatic cancer.[17]

The pancreatic carcinomas that arise in these patients often have a “medullary histology”.[9]

Peutz-Jeghers syndrome (PJS) is an autosomal dominant disorder with an incidence of

1:25 000. The disease is characterized by mucocutaneous pigmentations and hamartomatous

15

intestinal polyps, due to mutations in the STK11/LKB1 gene.[17] Patients with PJS have a 132

fold increased risk of developing pancreatic cancer.[9, 17]

Familial atypical multiple mole-melanoma syndrome (FAMMM) is associated with

multiple nevi, melanomas, and extracutaneous tumors. The disease is caused by a mutation in

CDKN2A/p16 and accounts for approximately 12% of familial pancreatic cancers.[17] Affected

individuals have a 20-fold increased risk for developing pancreatic cancer.[9, 17]

Familial breast and ovarian cancer syndrome is due to mutations in the BRCA1 or

BRCA2 genes. Individuals with germline BRCA1 mutations have a twofold risk increase of

pancreatic cancer; those with BRCA2 mutations have between a 4 and 13-fold risk increase.[17]

Inherited BRCA2 mutations are responsible for roughly 17% of familial pancreatic cancer

cases, which makes BRCA2 the most common inherited defect contributing to pancreatic cancer

identified to date.[9, 17] Of interest, not all patients with pancreatic cancer and a germline

BRCA2 mutation come from classical BRCA2 families. In fact, some have no family history of

breast cancer.[9]

In addition to the above described familial cancer predisposition syndromes, familial

pancreatic cancer can be associated with hereditary pancreatitis. It is an autosomal dominant

disease caused in 70% of the cases by a mutation in PRSS1 gene, leading to pancreatic auto-

digestion. Affected individuals have a 50-fold increased risk of developing pancreatic

cancer.[17] Patients with hereditary pancreatitis have development of severe pancreatitis at a

young age, may have pancreatic pseudocysts and diabetes [9], and have a 50-fold increased risk

for development of pancreatic cancer [9, 17].

In the overwhelming majority (~80%) of familial pancreatic cancer cases, the underlying

genetic predisposition remains unknown.[17]

At present, screening patients with germ line mutations known to be associated with

pancreatic cancer has not been widely used. The patients most likely to benefit from screening

would be patients with a strong family history of pancreatic cancer and patients with hereditary

pancreatitis.[12]

16

PRECURSOR LESIONS

One of the most exciting developments has been a dramatically improved understanding

of the non-invasive precursor lesions that give rise to invasive pancreatic cancer. [26] The early

detection and treatment of noninvasive precursor lesions provide the best hope for reducing

mortality.[27]

Our current understanding of pancreatic neoplasia suggests that invasive pancreatic

carcinoma can arise from noninvasive mucinous cystic neoplasms (MCNs), Intraductal papillary

mucinous neoplasms (IPMNs), and Pancreatic intraepithelial neoplasia (PanINs).[28-29]

Mucinous Cystic Neoplasms (MCNs)

The MCN is a cystic neoplasm composed of mucin-producing epithelial cells.[27, 30-31]

By definition, MCNs are associated with an ovarian type of stroma and the vast majority does

not communicate with the larger pancreatic ducts. [17, 25, 31-32]

The degree of dysplasia can range from minimal dysplasia (MCN with low-grade

dysplasia – adenoma), to moderate cytological and architectural atypia (MCN with moderate

dysplasia – borderline), to significant architectural and cytological atypia (MCN with high-

grade dysplasia – carcinoma in situ).[27] One-third of MCNs have an associated invasive

carcinoma.[27, 30] The invasive carcinoma associated with MCN is usually a ductal type of

adenocarcinoma.[27, 30, 33]

Of note is that a spectrum of dysplasia can be present in a single MCN (mild dysplasia

adjacent to epithelium with marked dysplasia). Similarly, invasive carcinoma can arise focally

in an otherwise benign-appearing MCN.[27, 30] Therefore, because invasive carcinoma can be

focal, MCNs need to be completely resected surgically to rule out a focal malignancy; biopsy of

a MCN is likely to underestimate the degree of dysplasia, or even miss an invasive

carcinoma.[27]

17

Clinical, pathological, and molecular observations have established that a MCN with mild

dysplasia can progress to moderate dysplasia, and from there to carcinoma in situ. It is clear that

if left untreated, noninvasive MCN can progress to invasive carcinoma.[27, 32, 34-35] This

progression is associated with the accumulation of genetic alterations in cancer-associated

genes, including KRAS, TP53, and SMAD4/DPC4. [27, 31-32, 36-39]

The good news is that noninvasive MCNs are curable. MCNs are localized lesions;

multifocality is rare, and therefore the surgical resection of a single neoplasm can be

curative.[27]

Intraductal Papillary Mucinous Neoplasms (IPMNs)

IPMNs are papillary mucin-producing pancreatic neoplasms with prominent intraductal

growth.[27, 31] Noninvasive IPMNs are classified into: IPMN with low-grade dysplasia

(adenoma), IPMN with moderate dysplasia, and IPMN with high-grade dysplasia (carcinoma in

situ).[27] Approximately one-third of IPMNs have an associated invasive carcinoma.[27, 40]

The distinction between a noninvasive IPMN and an IPMN with an associated invasive

carcinoma is clinically critical because the presence or absence of invasion is the most important

clinical prognostic factor. Like for MCN, the invasive carcinoma in IPMNs can be very focal,

and therefore a benign diagnosis cannot be established on biopsy alone.[27]

In half of IPMNs with an associated invasive carcinoma, it has a colloid or

“muconodular” pattern of invasion, and the other half has a “tubular” or conventional ductal

pattern.[39-41] This distinction is important because the prognosis is significantly better for

patients with an IPMN with an associated invasive colloid carcinoma.[31, 41-42]

Noninvasive IPMNs can progress from IPMN with mild dysplasia, to IPMN with

moderate dysplasia, to IPMN carcinoma in situ, and to invasive carcinoma.[43] At the

molecular level, the frequency of KRAS gene mutations in IPMNs increase with increasing

grades of dysplasia.[44-45] It suggests that it takes years for noninvasive IPMNs to progress to

18

an invasive cancer, and that the treatment of a noninvasive IPMN can save lives by interrupting

the progression.[27]

IPMNs that arise in the main pancreatic duct are classified as “main-duct-type” IPMNs,

while those that arise in the secondary branches are referred to as “branch-duct-type”

IPMNs.[25, 43, 46-48] “Combined-type” IPMNs involve both the main- and branch ducts.

Branch-duct IPMNs are less likely to harbor an invasive carcinoma [27, 48] and less likely to

progress to invasive cancer[27].

A number of histologic subtypes of IPMNs have also been identified based on the

direction of differentiation of the neoplastic epithelial cells:

IPMNs “intestinal-type” because they resemble villous adenomas of the large

intestine, they express an intestinal pattern of mucins (MUC2 and MUC5AC-positive,

and MUC1-negative), and they express CDX2, a transcription factor and determinant

of intestinal differentiation.[28, 31-32, 42, 49-53]

“Gastric foveolar” type of IPMN, often seen in branch-duct-type IPMNs, and usually

express MUC5AC, and are MUC1 and MUC2 negative.[28, 31-32, 53]

“Pancreatobiliary type” of IPMNs are usually MUC2 negative, and MUC1 and

MUC5AC positive.[28, 31-32, 50, 53]

Intraductal oncocytic papillary neoplasm (IOPN) usually express MUC1 and

MUC5AC, but are MUC2 negative.[28, 31-32, 53-54]

The morphological classification of IPMNs has clinical significance. Intestinal-type,

when with invasive carcinoma, is often associated with invasive colloid carcinomas (that also

strongly express MUC2 and are MUC1 negative) (Figure 1).[41, 49, 53] Pancreatobiliary-type

IPMNs, when with invasive cancer, are usually associated with a tubular type of ductal

adenocarcinoma (that are also usually MUC1 positive and MUC2 negative) (Figure 1).[49-50,

53] These associations are important because invasive colloid carcinomas appear to have a

significantly better prognosis than invasive tubular-type adenocarcinomas of the pancreas.[41] It

should be noted that there is a significant overlap in the groups, and that a single IPMN can

contain more than one type of epithelium. The prognosis of patients with a noninvasive IPMNs

19

is significantly better than for patients with an IPMN with an associated invasive

carcinoma.[27]

Although MCNs are almost always unifocal, the same is not true for IPMNs. As many as

30% of IPMNs are grossly multifocal.[42, 55-56] It suggests that patients with one IPMN are at

risk of developing additional IPMNs, and therefore they need to be followed carefully.[40, 42,

57-58] Most IPMNs are large enough to be detected clinically. We have the opportunity to save

patients by treating noninvasive IPMNs before they progress to invasive cancer.[27]

Pancreatic Intraepithelial Neoplasia (PanINs)

PanIN is defined as a microscopic noninvasive neoplastic epithelial proliferation in the

pancreatic duct system.[59-60] PanIN lesions have been classified into three grades: PanIN-1

has only mild dysplasia, PanIN-2 has moderate cytological and architectural atypia, and PanIN-

3 has marked dysplasia. As with MCNs and IPMNs, PanINs can progress from PanIN-1 to

PanIN-2 to PanIN-3 and to infiltrating ductal adenocarcinoma (Figure 1).[25, 31-32, 61-63]

PanINs do not express MUC2 regardless of the grade of atypia, but generally express

MUC5AC.[28, 41] MUC1 immunoreactivity is identified in the majority of high-grade PanINs

and invasive adenocarcinoma.[27]

The progression of noninvasive PanIN lesions to invasive ductal adenocarcinomas of the

pancreas suggests a huge opportunity to treat and cure pancreatic neoplasia before it progresses

to an incurable invasive cancer. However, most PanINs are too small to be detected using

available imaging technologies. Remarkably, PanINs, even the low-grade PanIN-1 lesions, were

associated with lobular parenchymal atrophy.[64]

The larger lesions, IPMNs and MCNs, can readily be detected using available imaging

techniques, and new findings suggest that even the smaller PanIN lesions may be detectable

because of the lobular parenchymal atrophy they may produce. Once detected, precursor lesions

in the pancreas can be treated, thereby preventing the development of a lethal cancer.[27]

20

DUCTAL ADENOCARCINOMA

Pancreatic ductal adenocarcinoma remains a formidable challenge because of the lack of

early diagnostic tests and effective therapies.[65]

Ductal adenocarcinoma accounts for >80% of pancreatic cancers. Grossly, they are

white/yellow and firm masses. Sixty percent of the cases arise in the pancreatic head, 15% in the

body or tail, and 20% involve the gland diffusely. They are characterized microscopically by

infiltrating small glands that are lined with low-columnar, mucin-containing cells. Typically, a

strong desmoplastic reaction occurs around the cancer. [17, 66]

Ductal adenocarcinomas are highly aggressive cancers with frequent invasion of vascular,

lymphatic, and perineural tissue. Approximately 80% of surgical specimens show disease in

regional lymph nodes. There is clinically evident disease at distant organ sites in up to 80% of

all patients who are discovered to have pancreatic adenocarcinoma. The most common sites for

distant metastases are the liver (80%), peritoneum (60%), lung and pleura (50-70%), and

adrenal glands (25%).[17, 66]

Complete surgical resection offers the best hope for long-term control of the disease,

although only approximately 30% of patients present with potentially resectable disease by

imaging studies, and less than 20% of all patients ultimately undergo resection. [65] The 5-year

survival rate for all patients with pancreatic ductal adenocarcinoma is <5%. The 5-year survival

rate is 15-25% in patients who undergo surgery, and 30-40% in patients who undergo surgery

with small tumors and node-negative disease.[17]

Other rare primary non-endocrine tumors of the pancreas include adenosquamous

carcinoma, acinar cell carcinoma, giant cell carcinoma, giant cell carcinoma with osteoclast-like

giant cells, pancreatoblastoma, serous cystadenoma/cystadenocarcinoma, and solid

pseudopapillary (Hamoudi) neoplasm.[17, 66]

21

DIAGNOSIS

Clinical Findings

The early symptoms of pancreatic adenocarcinoma include anorexia, weight loss,

abdominal discomfort, and nausea. Unfortunately, the nonspecific nature of these symptoms

often contributes to a delay in diagnosis. Specific symptoms usually only develop after invasion

or obstruction of a nearby structure.[7, 9]

As most pancreatic adenocarcinomas arise in the head of the pancreas, obstruction of the

biliary tree (mainly in the intrapancreatic portion of the common bile duct) resulting in jaundice

is the hallmark presentation. Jaundice is progressive and often associated with dark urine, light

stools and significant pruritus.[7, 9]

Pain is a common symptom of pancreatic adenocarcinoma. The most common pain

pattern is described by patients as a dull epigastric pain often accompanied by back pain, worse

in the supine position, and relieved by sitting forward. Pain can be caused by invasion of the

tumor into the splanchnic plexus and retroperitoneum, as well as by obstruction of the

pancreatic duct. Although intractable pain is frequently associated with pancreatic

adenocarcinoma, it is seldom an early manifestation, with fewer than one-third of patients

presenting with moderate to severe pain.[7]

Other symptoms found in a small percentage of patients include nausea and vomiting

related to gastro-duodenal obstruction. Mechanical obstructions of the proximal duodenum by

right-sided neoplasms, or at the ligament of Treitz by cancers of the midbody of the pancreas

are often later findings of pancreatic adenocarcinoma and suggest relatively advanced

disease.[7, 9]

At times, pancreatic adenocarcinoma may present in an unusual manner. New-onset

diabetes may be the first clinical feature in approximately 10% to 20% of patients.[7, 9-10]

Occasionally pancreatitis may also be the first signal, especially in the elderly when there is no

obvious cause such as gallstones or alcohol abuse.[9-10]

22

The most common physical findings at the time of initial examination are scleral icterus

and jaundice in patients with cancer of the head of the pancreas, while patients with tail and

body tumors may complain only of pain without any other specific sign. [7, 9-10] Often,

patients with deep jaundice will exhibit cutaneous signs of scratching, related to the pruritis.

Hepatomegaly and a palpable gallbladder may also be found.[9-10] In cases of advanced

disease, there may be evidence of cachexia, muscle wasting, or an enlarged, nodular liver

consistent with metastatic disease. In patients with advanced cancer, ascites, left supraclavicular

adenopathy (Virchow‟s node), periumbilical lymphadenopathy (Sister Mary Joseph‟s nodes) or

findings of dropped metastasis in the pelvis encircling the perirectal region (Blumer‟s shelf)

may be present.[7, 9]

Laboratory Studies

Results of laboratory studies in patients with adenocarcinoma of the head of the pancreas

typically are marked by elevated serum total bilirubin, alkaline phosphatase, and -glutamyl

transpeptidase, with mild elevations of the hepatic aminotransferases.[7, 9] Hepatitis serologic

study results are often assessed as part of the workup for jaundice, and they are typically

negative.[9] In patients with localized cancer of the body and tail of the pancreas, standard

laboratory values are usually normal.[7, 9] For these tumors, when liver function test

abnormalities do occur, they typically indicate diffuse metastatic disease with involvement of

the liver or porta hepatis.[9]

Normochromic anemia and hypoalbuminemia may reflect a chronic nature of the

neoplastic process and its nutritional sequelae.

It is uncommon for patients with standard ductal adenocarcinoma of the pancreas to have

either hyperamylasemia or hyperlipasemia.[7, 9]

In patients with deep jaundice, the coagulation parameters should be checked because

prolonged exclusion of bile from the gastrointestinal tract leads to malabsorption of the fat-

23

soluble vitamins and decreased the hepatic production of vitamin K-dependent clotting

factors.[7, 9] This can result in prologation of the prothrombin time.[9]

The development of biomarkers for screening holds enormous promise for increasing

early detection and impacting mortality.[67] A wide variety of serum tumor markers have been

proposed for use in the diagnosis and follow-up of pancreatic adenocarcinoma.

One of the first systematically investigated markers is carcinoembrionary antigen (CEA).

This test identifies nearly half of patients with pancreatic adenocarcinoma, and may tell

pancreatic malignancies from benign conditions in more than 90% of the patients. However,

CEA is not a valid marker for diagnosis or follow-up.

The carbohydrate antigen 125 (CA-125) is a marker of ovarian epithelial malignancies

that has been studied with regard to other gastrointestinal tract tumors. In patients with

pancreatic adenocarcinoma, CA-125 is detected in fewer than 50% of cases. CA-125 is

unsatisfactory as a single test for pancreatic adenocarcinoma.[16]

The most widely used and best validated marker for pancreatic adenocarcinoma is the

carbohydrate antigen 19-9 (CA 19-9).[7, 9, 68] CA 19-9 has been associated with both

pancreatic neoplasms and other abdominal malignancies. Approximately 80% of patients with

pancreatic adenocarcinoma may be correctly diagnosed using marker. Steinberg et al.[69]

reported that CA 19-9 was statistically more specific than CEA (86.5 versus 48.4%) but only

slightly more sensitive than CEA (92.5 versus 87.3%).[69] Therefore, CA 19-9 may be a useful

clinical marker to detect pancreatic adenocarcinoma progression in patients with either recurrent

or advanced disease.[16, 69]

CA19-9 is a Lewis blood group–related mucin that has been extensively studied in the

diagnosis, prognosis, and monitoring of pancreatic cancer. [9]

Although CA 19-9 is not accurate enough to be used in screening asymptomatic subjects

for pancreatic adenocarcinoma, it is currently the single most useful blood test in differentiating

benign from malignant pancreatic disorders. CA 19-9 has limited value in the diagnosis,

especially for early forms of the disease. However, it may complement radiological procedures

(such as computed tomography – CT – or endoscopic ultrasound – EUS), particularly in non-

24

jaundiced patients. Moreover, appropriately interpreted CA 19-9 results can guide further

invasive testing such as endoscopic retrograde cholangiopancreatography (ERCP), laparoscopy

or EUS fine-needle aspiration. [68]

Based on the above, most expert groups cautiously recommend measurement of CA 19-9

in the initial work-up of patients presenting with suspected pancreatic adenocarcinoma.[68]

Moreover, it should be borne in mind that: benign diseases (such as pancreatitis, liver

cirrhosis, cholangitis and obstructive jaundice) may have elevated CA 19-9 levels; it can be also

increased in other types of adenocarcinoma [68, 70]; CA 19-9 lacks sensitivity for small lesions;

and poorly differentiated pancreatic cancers also appear to produce less CA 19-9 than well

differentiated ones[68].

Multiple studies showed that newly presenting patients with elevated levels of CA 19-9

had a worse prognosis than those with low levels.[68, 71-72] Significant prognostic factors for

good outcome are, among others, a postoperative CA 19-9 level <200 kU/L, a decrease in CA

19-9 levels following surgical resection as well as negative lymph nodes and low tumor

stage.[68, 72] Serum CA 19-9 levels should be considered for risk stratification in patients with

pancreatic adenocarcinoma. Although high concentrations are indicative of poor outcome, CA

19-9 is only one of the multiple factors that affect prognosis and treatment planning.[68]

One of the most frequent uses of tumor markers is in postoperative surveillance following

curative surgery for a primary cancer. The aim of this surveillance is to detect

recurrences/metastases as early as possible. This practice is based on the assumption that the

early detection and the initiation of therapy enhance the chance of cure or results in a better

outcome. A number of studies have shown that serial determinations of CA 19-9 can detect

recurrent/metastatic disease several months before finding clinical or radiological evidence of

disease and CA 19-9 should be used in the follow-up of patients after surgery.[68]

In patients with advanced inoperable pancreatic adenocarcinoma, the aim of systemic

therapy is palliative. Evaluating response to systemic therapy in patients with locally advanced

pancreatic cancer may be difficult using imaging procedures due to extensive desmoplasia and

surrounding inflammatory changes. Because of these difficulties, a number of investigators have

25

attempted to use serial CA 19-9 measurements to assess response and/or determine prognosis in

patients with advanced pancreatic adenocarcinoma undergoing systemic treatment. Patients with

declining marker levels, following initiation of chemotherapy, had a better outcome than those

showing no decrease. Serial measurements of CA 19-9 should be used along with imaging to

monitor response to therapy. If CA 19-9 increases, this may indicate disease progression.[68]

Several other serum markers have been proposed for pancreatic adenocarcinoma. None of

these have been shown to be superior to CA 19-9 and none are widely used for clinical

purposes. [68] CA 19-9 remains the most useful molecular marker for the diagnosis and follow-

up of clinically and radiological evident pancreatic adenocarcinoma.[73]

One hope for the future involves new developments in the area of early detection, by use

of molecular strategies. With gene expression data and data from other molecular strategies,

earlier detection of pancreatic cancer may be possible. [9]

Imaging Studies

The early diagnosis of pancreatic adenocarcinoma requires a high index of suspicion and

appropriate aggressiveness in pursuing the diagnosis.[7] Imaging has an important role in the

diagnosis and management of patients with pancreatic adenocarcinoma. Early tumor detection

and accurate radiologic staging are crucial for identifying patients with potentially resectable

disease and avoiding unnecessary surgery in patients with unresectable disease. To ensure that

the correct imaging study is requested, physicians must have an understanding of the strengths

and limitations of the imaging modalities available. [74]

Many diagnostic modalities have been used in patients with suspected pancreatic tumors.

These modalities include transabdominal ultrasound (US), CT, magnetic resonance imaging

(MRI), ERCP, EUS and positron emission tomography (PET).[10]

26

Transabdominal Ultrasound (US)

Transabdominal US is often the initial investigation performed in patients with jaundice

or upper abdominal pain.[74-75] It is extremely sensitive in distinguishing non-obstructive

jaundice and obstructive jaundice – the presenting feature in majority of patients with pancreatic

cancer.[75] The presence of biliary dilatation in pancreatic head cancer facilitates tumor

detection. For assessing the pancreatic body and tail, the oral administration of water or other

contrast agents may help delineate the organ.[4]

Masses as small as 2cm can be detected, as well as secondary features, such as atrophy,

pancreatic duct dilatation, hepatic metastases and ascites. [16, 75] However, transabdominal US

is not a reliable method for confident diagnosis or exclusion of small pancreatic tumors.[4]

Vascular invasion and local extension are poorly depicted which often prompts further

investigations.[75]

Although it has been reported that US performed by experienced operators using proper

equipment is equivalent to or better than CT for detection and staging of pancreatic

adenocarcinoma, US is likely to remain a tool for initial assessment of patients with suspected

pancreatic disease that will be followed up by additional imaging procedures.[74]

Computed Tomography (CT)

CT remains the premier imaging test for diagnosing and staging of suspected pancreatic

adenocarcinoma because it is a robust and reliable technique.[2, 4, 75] Dual phase, contrast

enhanced thin section (spiral or multidetector) CT has greatly increased our ability to diagnose

and stage this neoplasm and to determine patient suitability for resection. Multidetector CT

(MDCT) acquisition technology was introduced in the late 1990s. Before this, high-quality

spiral CT was the preferred noninvasive imaging modality.[9]

MDCT incorporates dual-phase imaging in both the arterial and venous phases of

enhancement. Water is used as the oral contrast agent of choice and slices through the pancreas

are obtained every 1.25 mm.[9] With the development of MDCT, the acquisition of the third

27

dimension has stimulated pancreatic imaging[4] and improved the detection, staging, and

surgical planning.[9]

CT features of pancreatic adenocarcinoma are variable, depending on the size and extent

of the disease.[75] The most common abnormality is a small hypodense/hypovascular mass

within the pancreas.[2, 4, 9, 74-75] Rarely they can be isodense to the normal pancreatic

parenchyma, and difficult to detect. [2, 4] In most cases, the presence and location of a

pancreatic mass can be inferred from secondary signs, including interruption and dilation of the

pancreatic duct and/or the common bile duct, mass effect, convex abnormality of the contour,

and atrophy of the pancreas.[2, 4, 74-75]

Although radiologic signs used to diagnose pancreatic adenocarcinoma are not

pathognomonic of the condition, biopsy is usually not necessary before surgical intervention in

patients with potentially resectable disease. When the diagnosis is in doubt, or if chemotherapy

and/or radiation therapy is planned, confirmation by biopsy is necessary before treatment.[74]

The reported sensitivity of CT for detecting pancreatic adenocarcinoma is high, ranging

from 89% to 97%.[74] Not surprisingly, the sensitivity of CT is higher for larger lesions than

smaller ones.[4, 74]

Ideally, CT should be performed before biliary stenting because the stent may cause

artifact in the pancreatic head that can mask the lesion, and the trauma of stent insertion often

produces inflammatory changes that can be indistinguishable from tumor.[74]

Magnetic Resonance Imaging (MRI)

Recent advances in technology allowed MRI to improve its ability to diagnose and stage

pancreatic adenocarcinoma (high-resolution and fast imaging, volume acquisitions, magnetic

resonance cholangiopancreatography, and functional imaging).[9]

The normal pancreas is high in signal intensity on noncontrast T1-weighted fat-

suppressed images. Regarding gadolinium enhancement, it demonstrates a uniform capillary

blush on immediate postcontrast images and fades to isointense signal to the liver on interstitial

28

phase images.[76] Conversely, pancreatic adenocarcinoma appears as a low-signal intensity

mass on noncontrast T1-weighted fat-suppressed images and enhances to a lesser extent than the

surrounding normal pancreatic tissue on immediate postcontrast images.[2, 76] These MRI

features are related to the tumor sparse vascularity and dense cellularity.[9, 76] T2 weighted

sequences are employed to detect liver metastasis; vascular invasion can be evaluated with T1

weighted images post-contrast.[75]MRI may be helpful for differentiating an inflammatory

pancreatic mass from a pancreatic adenocarcinoma.[74]

There have been several studies in the past comparing MRI with CT in terms of

diagnosing and staging and all of them concluded that there is no significant benefit of MRI

over CT in its ability to predict resectability.[9, 16, 75] Because of the increased cost, room time

and potential inability of MRI in assessment of lung metastases, CT remains the primary

imaging modality of choice.[75] Moreover, some recent studies have shown slight superiority

for MDCT, in part due to the recent technical improvements.[2]

Currently, contrast-enhanced MRI has a role as a problem-solving tool if MDCT reveals

equivocal results or is not possible.[4] MRI is of value in patients with impaired renal function

or patients sensitive to iodinated contrast material.[74, 77] In selected cases, MRI may be useful

as an adjunct to CT, as for example, in characterization of small (<1 cm) hepatic lesions (when

CT findings are indeterminate)[74-75] and when there is clear biliary dilatation with no obvious

mass seen on CT.[74]

Nowadays, the MRI-cholangiopancreatography has shown promise as a noninvasive

technique with the ability to visualize both the bile duct and the pancreatic duct with images

similar to those obtained by ERCP. MRI-cholangiopancreatography may also provide important

information about the level and degree of obstruction. [75]

29

Endoscopic Ultrasound (EUS)

Another tool used for the diagnosis and staging of patients with pancreatic cancer is EUS.

It produces high-frequency images of the pancreatic parenchyma using the wall of the stomach

and duodenum as an acoustic window, thereby improving image resolution. [9-10, 74]

Therefore, EUS can evaluate peripancreatic structures, detect distinct parenchymal changes, and

it also enables the detection and evaluation of focal lesions with a minimum size of 2–3

mm.[78] For small tumors EUS has been reported to be superior to CT.[2, 4, 10, 16, 78]

Another advantage of EUS is that it can observe the whole pancreas, especially the uncinate

process and farthest tail near the splenic hilum, which is difficult to scan using transabdominal

US.[78]

An additional aspect of the EUS is the ability to perform EUS-guided fine-needle

aspiration (EUS–FNA) biopsy.[10, 16, 74, 78] Compared with other imaging modalities, the

results of EUS–FNA biopsy of pancreatic masses are excellent, with sensitivity of around 90%

and specificity of virtually 100% with a low frequency of complication. [78]

Notable disadvantages of EUS include its limited availability, marked operator

dependence, and inability to evaluate for distant metastases.[74-75]

A recent study by DeWitt et al.[79] has shown that EUS had a sensitivity of 98%

compared to 86% for MDCT. [2, 4, 74, 79-80] EUS has also been shown to have a high

negative predictive value for excluding pancreatic cancer (almost 100%), and may therefore

play a role in screening for pancreatic malignancies.[2, 4, 80]

EUS is indicated when there is high suspicion of pancreatic malignancy with no definite

mass seen on CT or MRI and also when there is some doubt regarding the local extent of the

disease (vascular invasion) and to assess involvement of normal size lymph nodes. [74-75]

However, despite these facts, CT remains the imaging modality of choice for patients

with suspected pancreatic adenocarcinoma.[74]

30

Biopsy

Biopsies can be performed under US or CT (percutaneously) or under EUS guidance,

using an aspiration technique.[10] With the advent of EUS-FNA, it became a viable and useful

alternative procedure for acquiring a tissue diagnosis. For diagnostic purposes, the sensitivity of

EUS-FNA varies from 75% to > 90%, the specificity being 82%-100%.[10, 81]

If a pancreatic mass is clearly unresectable based on CT or MRI results or if metastatic

disease is identified, either percutaneous image-guided or EUS-guided FNA can be performed

for a tissue diagnosis to confirm the presence of cancer and to offer chemotherapy or

radiation.[9, 16, 81] EUS-FNA offers some advantages over other techniques in cases where a

tissue diagnosis of pancreatic adenocarcinoma is required before treatment.[9]

Multiple factors favoring EUS-FNA over percutaneous FNA of pancreatic cancer are: (1)

it is the most cost-effective approach as a diagnostic modality; (2) it has a smaller risk when

compared with percutaneous FNA; and (3) EUS-FNA uses a short needle track. Indeed, the

aspiration needle travels from the gut lumen to the lesion, a pathway that usually does not cross

peritoneal or pleural surfaces. The exception to this is in EUS-FNA of liver lesions and of

pancreatic body/tail masses where the lesser sac of the peritoneum is breached.[81]

Moreover, EUS-FNA is more accurate than percutaneous techniques for masses < 3 cm

and is the only preoperative procedure which can demonstrate invasion of lymph nodes located

in the celiac, lumboaortic, retroduodenopancreatic or superior mesenteric regions. Also,

aspiration of ascitic fluid with a cytological study done by EUS can validate a carcinomatosis

that could not be revealed using conventional imaging. Small metastases of the left liver lobe

can be found and are easily accessible. The finding of such lesions modifies considerably the

management of supposed resectable cancer.[81]

In case of a resectable tumor, a histological diagnosis is not necessary and of little use

because it does not change the ultimate need for surgery. However, because some institutions

have a policy of giving preoperative neoadjuvant chemotherapy or radiation in resectable

pancreatic adenocarcinoma, tissue diagnosis would be a pre-requisite for that.[9, 74, 81] Others

31

argue that pre-operative diagnosis can exclude the occasional patients with unusual histology

found in 5% to 10% of pancreatic tumors (lymphoma, endocrine tumors and metastases) who

would not benefit from surgery.[81]

If CT or MRI results show a pancreatic mass with equivocal resectability, EUS is

generally the next staging procedure. If it reveals that the mass is clearly unresectable, one can

proceed with EUS-guided FNA for tissue diagnosis. If the EUS results show that the mass is

potentially resectable, then EUS-FNA should be reconsidered.[81]

Endoscopic Retrograde Cholangiopancreatography (ERCP)

Traditionally, in the evaluation of the jaundiced patient we have the cholangiography,

either by the endoscopic or percutaneous approach. The choice of technique depends primarily

on local expertise. Using the endoscopic approach, ampullary and duodenal carcinomas can be

visualized and biopsied. In addition, a pancreatogram may be obtained, which may be important

in differential diagnosis. In most cases of pancreatic adenocarcinoma, the ductal system is

obstructed, with no distal filling of the duct.[7] ERCP allows direct imaging of the pancreatic

duct, the site of origin of most pancreatic adenocarcinomas. [9-10] The sensitivity of ERCP for

diagnosis is quite high, with the finding of long, irregular stricture in an otherwise normal

pancreatic duct being virtually pathognomonic in the appropriate clinical setting.[9]

The percutaneous cholangiography is usually technically easier with a dilated biliary tree

and is useful in defining the proximal biliary system.[7]

Although there is no question that ERCP is reliable in confirming the clinical suspicion of

pancreatic adenocarcinoma, it is rarely necessary and should not be routinely used.[9-10] With

the current sophistication of CT scanning and MRI, the routine practice of diagnostic ERCP is

unsupported.[7, 9] The ERCP should be reserved for: the evaluation of a patient with presumed

pancreatic cancer and obstructive jaundice in whom no mass is evident on CT; the symptomatic

but nonjaundiced patient without an obvious pancreatic mass; or the patient with chronic

pancreatitis in whom the development of a pancreatic neoplasm is suspected.[7]

32

In addition, a biliary stent can be placed through the obstructing lesion by either the

endoscopic or percutaneous approach to decompress of the biliary tree and to alleviate

jaundice.[7, 10]

Laparoscopy

Laparoscopy has been recently introduced as a diagnostic and staging technique

potentially allowing palliative therapy.[16]

Staging laparoscopy is often performed in patients who appear to have resectable disease

on imaging studies with the purpose of avoiding unnecessary laparotomy. Many surgeons

routinely perform diagnostic laparoscopy in all patients being considered for surgical resection,

but recent studies suggest this may be unnecessary. Among the recommended indications for

diagnostic laparoscopy are small hypodense hepatic lesions that are not amenable to

percutaneous biopsy and findings that are equivocal for peritoneal carcinomatosis.[74]

Positron Emission Tomography (PET)

Recently, PET scanning has been evaluated as a tool for the diagnosis and staging of

pancreatic tumors.[4, 10, 74] At the present time, the role of PET scanning in the management

of patients with pancreatic adenocarcinoma is under development. [10, 74]

PET uses the increased metabolism of glucose by pancreatic cancer cells as the basis of

imaging. Current PET scanning for pancreatic cancer uses fluorine-18 (a positron-emitting

tracer) as a glucose-like substrate. This substance is rapidly taken up by malignant tumor cells.

However, it localizes not only tumor sites, but also sites of inflammation and infection.[9]

Therefore, PET scan has a high sensitivity (89%) but suffers from a moderate specificity

(69%).[4] Moreover, PET may have the biggest impact for detection of distant metastases and

its sensitivity may be superior to that of standard staging procedures.[74] PET has been shown

33

to give information relevant to prognosis, and to add diagnostic accuracy to CT and ERCP in

detecting tumor dissemination.[9]

The diagnosis of pancreatic adenocarcinoma still represents a difficult task and multiple

imaging tests have been proposed over the years, including transabdominal US, CT, PET, MRI,

EUS, and ERCP. Taken individually, these methods have variable sensitivity for the diagnosis

of specific pancreatic disorders. All these imaging methods improve the specificity of the

diagnosis substantially, usually providing complementary information that determines the best

treatment option. [80]

After the diagnosis of pancreatic adenocarcinoma is made, the key to management is

determining whether or not a patient has potentially resectable disease.[74]

34

STAGING AND RESECTABILITY

The tumor-node-metastasis (TNM) staging system facilitates the objective description

and classification of the anatomic extent of malignant disease in a simple, reproducible, site-

specific way. Fundamental to this staging system is the premise that cancers of the same

anatomic site and histology, with a similar extent of disease, share a common natural history.

This system provides physicians a language with which to estimate and communicate prognosis,

allows for the development and selection of stage-specific treatment strategies, and permits the

evaluation of similar groups of patients in clinical trials.[82]

For pancreatic adenocarcinoma, the definition of a resectable tumor has become more

clearly defined anatomically based on the availability of high-quality CT scans.[82] Such

imaging techniques provide more accurate staging, which has an impact on preoperative

decision making regarding tumor resectability.[16]

The current Sixth Edition of the AJCC staging system [82] evaluates pancreatic exocrine

malignancies in terms of the size and anatomic extent of the primary tumor (T), the presence or

absence of regional lymph node metastases (N), and the presence or absence of distant

metastases (M) (Table 1). [82]

The T-stage of pancreatic cancer is defined by the tumor size and the local spread.[2, 4]

T1 and T2 describe tumors confined to the pancreas, with T1 tumors being ≤2 cm in size (rarely

found) and with T2 tumors being >2 cm in size [2, 4, 82]; T3 and T4 indicate extrapancreatic

extension by the primary tumor. T4 tumors are those that are unresectable due to tumor

extension to the celiac axis (CA) or superior mesenteric artery (SMA). In contrast,

extrapancreatic tumor extension that does not involve the CA or SMA does not typically

influence the local resectability status of the tumor and, therefore, carries the T3

designation.[82]

The N-stage is dependent on the presence of nodal metastasis, with N1 representing

peripancreatic nodal metastases. [2, 4]

35

Metastasis to more distant nodes such as para-aortic nodes is defined as M1 disease.

Other common sites of distant metastases are the liver and peritoneum. [2, 4]

The combination of T, N and M is made into Stage groups (Table 1). Stage 1 disease is

tumor limited to pancreas with no nodal involvement and it is potentially resectable. In Stage 2

disease there may be extension into adjacent tissue (duodenum, bile duct, venous structures, and

regional lymph nodes) with no involvement of SMA or CA and it is usually potentially

resectable. Stage 3 is classified as unresectable locally advanced disease with invasion of SMA

and/or CA. Stage 4 disease is unresectable with distant metastasis. [75, 82]

This system now divides patients with pancreatic adenocarcinoma into distinct prognostic

and clinical groups based on contemporary definitions of resectability (Stage I/II) and

unresectability (local-regional, Stage III; metastatic, Stage IV).[82]

Distinction between Resectable, Locally Advanced and

Metastatic Disease

At present, it is accepted that surgical resection offers the only chance of cure for patients

with pancreatic adenocarcinoma.[74, 82] Whether or not the primary tumor can be removed

represents the strongest prognostic factor for patients with this disease and underlies the

distinction between Stages I and II (resectable) and Stages III and IV (unresectable due to

locally advanced or metastatic disease).

Specific oncologic and anatomic findings distinguish resectable (Stages I and II) from

unresectable (Stages III and IV) pancreatic cancer. Surgical resection of the pancreatic tumor is

generally considered to be inappropriate in patients with metastatic disease (Stage IV), as the

metastases are virtually always multifocal and associated with survival duration of

approximately 6 months. Patients with unresectable disease together account for the

overwhelming majority of patients who present with newly diagnosed pancreatic cancer. These

36

patients are generally treated with a nonsurgical strategy (chemoradiotherapy and/or

chemotherapy alone). [82]

One of the most common causes of non-resectability is vascular infiltration.[16, 75]

Pancreatic cancer has a great propensity to involve the adjacent vessels including the main

portal vein, the superior mesenteric vein (SMV), the CA and its branches, and the SMA. [76]

Nowadays, the terms “abutment” (involvement of 180 degrees or less of the

circumference of the vessel) and “encasement” (involvement of greater than 180 degrees of the

vessel) are more precise to describe tumor-vessel relationships. There is general consensus that

a tumor mass encasing the SMA or CA is considered to represent locally advanced, surgically

unresectable, Stage III disease. Arterial resection and reconstruction is technically difficult and

is associated with increased risk of perioperative morbidity and mortality. Moreover, resection

of tumors involving the CA or SMA is unlikely to be complete, and the majority of patients

with such locally advanced disease also have synchronous systemic metastases.[82]

Nowadays, the T3 category includes all forms of nonarterial tumor extension beyond the

pancreas, including extension to the SMV and portal vein. Venous resection and reconstruction

can be performed safely with no additional morbidity or mortality.[74, 82] Therefore, it is no

longer viewed as a contraindication to surgery.[4, 74, 82]

The presence of metastases to regional lymph nodes is difficult to determine

radiographically because small, benign-appearing lymph nodes frequently harbor occult

micrometastatic disease, whereas many enlarged lymph nodes are reactive and benign.[74, 82]

Moreover, metastatic involvement in normal sized lymph nodes is not detected. [75] In a patient

with pancreatic adenocarcinoma, the finding of enlarged peripancreatic lymph nodes on CT

should not preclude attempted resection.[74]

Metastatic disease is most commonly seen in the peritoneum and liver and in these

patients surgical resection is of no benefit.[74-76, 83] When the liver lesions are large, the

diagnosis of metastatic disease is usually straightforward. However, in the presence of small

lesions (<10 mm), the diagnosis of metastatic disease is difficult and biopsy may yield a

definitive role. Findings of peritoneal carcinomatosis on CT include ascites, peritoneal

37

thickening, nodular bowel wall thickening (serosal implants), and soft-tissue infiltration of the

omentum. Peritoneal carcinomatosis is diagnosed readily when advanced, but the sensitivity of

imaging studies for small peritoneal implants is limited.[74-75] Other common sites of

metastases include lung, pleura and adrenal glands. [75]

Current criteria for resectability include: (1) no evidence of tumor involvement of major

arteries (such as SMA or CA); (2) (if there is venous invasion) a suitable segment of SMV

below and portal vein above the site of venous involvement to allow for venous reconstruction;

and (3) absence of distant metastases.[74, 82] It should be also remembered that the definition

of resectability is evolving, and what was once considered unresectable disease may now be

resectable.[74]

Borderline Resectable Disease

Even though CT and MRI can provide very accurate assessments of the relationship

between the pancreatic adenocarcinoma and adjacent structures, the Stage III category includes

a wide range of tumor-vessel involvement. Tumors that demonstrate arterial abutment may be

considered for surgery as part of a multimodality approach to the disease. Patients with

borderline resectable pancreatic cancer include those whose tumors exhibit abutment or

encasement of a short segment of the hepatic artery, without evidence of tumor extension to the

CA; tumor abutment of the SMA involving 180 degrees or less of the circumference of the

artery; or short-segment occlusion of the SMV, portal vein, or their confluence, with a suitable

option available for vascular reconstruction. [82]

38

SURGICAL TREATMENT

Ductal pancreatic adenocarcinoma, the most frequent malignancy of the pancreas, is

characterized by retroperitoneal and perineural infiltration, early formation of multiple

metastases, and resistance to most of the treatment regimens currently available. Handling the

aggressive growth of this disease represents a complex and challenging task.[84]

Surgical resection, the patient‟s only hope for cure, offers a significantly improved

prognosis. Pancreatic surgery is considered one of the most technically demanding and

challenging surgical disciplines. Steady improvement in surgical techniques and advances in

perioperative supportive care have reduced the mortality.[85] However, only a minority of

patients (10–20%) present with resectable disease at the time of diagnosis.[85-86]

The anatomic location of the tumor within the pancreas dictates the type of resection.[87]

A lesion confined to the pancreatic head/uncinate process requires pancreaticoduoedenectomy

(PD).[5, 84-85, 87] Given that 60%–70% of pancreatic cancers arise in the head, PD is by far

the most common operation performed.

Because of the late presentation of symptoms, most patients with adenocarcinoma of the

body and tail present with locally advanced disease or distant metastases, thus precluding

surgical therapy.[87] However, for patients with clinically localized disease, a distal

pancreatectomy is the appropriate surgical resection. [5, 84, 87] Central pancreatic tumors of the

neck and body are rarely resectable, again because of either the presence of metastatic disease or

extension to the SMA. If they are nearer to the head of the gland, an extended PD may be

performed. For lesions nearer the tail, a distal subtotal pancreatectomy is performed.[87] Central

pancreatectomy, which is now often used to resect premalignant and low-grade lesions of the

neck and mid body [5, 87], has not been adopted for the treatment of pancreatic adenocarcinoma

by most surgeons because of concerns regarding adequate lymph node and retroperitoneal soft

tissue clearance.[87]

39

Resection for Tumors of the Head of the Pancreas

Pancreaticoduodenectomy (Kausch-Whipple procedure)

The first successful en bloc resection of part of the pancreatic head, distal common bile

duct, and duodenum was actually performed by Kausch in 1909. Twenty-five years later,

Whipple performed his first PD, also as a two-stage procedure.[4-5] It took another 7 years

before the one-stage resection was perfected, much as we know it today.[4] It consists of the

resection of the pancreatic head and duodenum along with a distal gastrectomy,

cholecystectomy, removal of the common bile duct and proximal jejunum, and enbloc resection

of regional lymph nodes.[4, 85] For a long time, this technically demanding procedure was

associated with high morbidity and mortality and a poor long-term outcome.[5, 84] However,

remarkable advances have been made in (peri)operative management and especially in the

surgical techniques used in the operation and PD has become the standard operative procedure

for tumors of the pancreatic head.[84]

The PD is divided into three parts: exploration, resection, and reconstruction.[5, 84] After

examination of the peritoneal cavity and the liver for metastatic disease, the gastrocolic ligament

is opened, the transverse and right colon are mobilized, and the duodenum is exposed.[87]

Then, a wide Kocher maneuver is performed to examine the retroperitoneum and the SMA, the

CA, the portal vein and the SMV for potential tumor infiltration.[5, 84-85] Furthermore, the size

of the tumor, and its relation to the retroperitoneal margin of the pancreas should be evaluated,

and infiltration of the retroperitoneal space needs to be ruled out.[5]

Following the gastrocolic venous trunk distally, the SMV is localized and a tunnel is

dissected between the neck of the pancreas anteriorly and the SMV-portal vein trunk

posteriorly, while at the same time the portal vein is exposed at the superior part of the pancreas.

The gallbladder and the common bile duct are then removed, and a transaction is performed

1cm from the tumor at the neck of the pancreas. The (distal) stomach and duodenum are

dissected, enabling complete removal of the specimen.[5, 84-85]

40

The reconstruction is then completed, beginning with a pancreaticojejunostomy (PJ) or

pancreaticogastrostomy (PG) and followed by a choledochojejunostomy and gastrojejunostomy

or duodenojejunostomy.[87]

Pylorus-preserving Pancreaticoduodenectomy (PPPD)

Complications seen following the Kausch-Whipple procedure, such as gastric dumping

syndromes, gastritis, and ulcerations due to bile reflux, led to the introduction of a pylorus-

preserving modification of the classical PD.[84] Introduced by Watson in the 1940s, the PPPD

was not frequently used until Traverso and Longmire re-introduced it in the late 1970s.[5, 84-

85, 87] The rationale behind preservation of the stomach is to improve gastrointestinal

function. To retain a functioning pylorus, the entire stomach and 2 cm of the first part of the

duodenum are preserved along with their neurovascular supply.[85]

There has been controversy over whether PPPD is a sufficiently radical treatment for

pancreatic cancer with respect to tumor outgrowth to the pylorus and spread to lymph nodes

along the greater and lesser curvatures of the stomach.[85, 87] Some retrospective studies

showed benefits in digestive function and quality of life for the PPPD.[5, 84] However,

eventually, many studies showed that there are no significant differences in postoperative rates

of delayed gastric emptying (DGE) between PD and PPPD.[5, 84, 87-88] Both classic and

PPPD are thus recommended for patients with resectable pancreatic adenocarcinomas.[84-85]

Therefore, surgeon‟s preference and experience should dictate the type of pancreatic resection

and reconstruction.[87]

Pancreatic Anastomosis and Reconstruction

Several prognostic factors have been identified for pancreatic anastomosis failure,

including the pancreatic tissue texture, the surgical technique, the extent of resection, and the

41

extent of dilatation of the pancreatic duct. The two most common pancreatic anastomosis

techniques are PJ and PG.[85]

The PJ can be performed by invaginating the transected pancreas into the jejunum.

Another possibility is the duct-to-mucosa technique, anastomosing the pancreatic duct directly

to the jejunum. Separate duct-to-mucosa adaptation helps to keep the duct orifice open, allowing

the unobstructed flow of pancreatic juice through the anastomosis. Using the end-to-side

anastomosis, the jejunal opening can be adapted specifically to the requirements of the

pancreatic remnant. [85]

Another technique (PG) often used is to anastomose the pancreatic remnant to the

stomach.[85] PG is technically easier to perform because of the proximity of the stomach and

the pancreatic remnant, which usually assures that the anastomosis is well perfused. Moreover,

pancreatic enzymes are not directly activated in the acidic milieu of the stomach.[5]

Large series were not able to show superiority of any of the techniques, suggesting equal

results for PG and PJ.[5, 84-85, 88]

As a third option, the pancreatic duct can be occluded by sutures, glue, or biologic

material. This practice, however, has been associated with higher fistula rates [84-85] and an

increased risk of pancreatic insufficiency[85]. As a result, ductal occlusion has largely been

abandoned.

Based on current data, there is no evidence in favor of one particular technique.[85] As

long as the three basic requirements of a safe anastomosis are met – a tension-free adaptation,

well-perfused tissues, and no distal obstruction – any anastomotic technique can result in good

outcome.[5, 84-85]

Prophylactic pharmacological approaches using somatostatin analogs (octreotide) have

been used in Europe and the United States with conflicting results.[88]

For re-establishment of gastrointestinal continuity, a variety of techniques may be

performed, such as retrocolic reconstruction with a single limb for all anastomoses, antecolic or

infracolic reconstructions, but there is no definitive evidence in favor of any one of them.[84]

42

However, some recent studies suggest that antecolic reconstruction may have some superiority

over retrocolic duodenojejunostomy in terms of decreased rates of DGE.[5]

Resection for Tumors of the Body/Tail of the Pancreas

Distal pancreatectomy

A distal pancreatectomy is performed for tumors of the pancreatic body and/or tail.[5, 84,

89] In most cases, a splenectomy has to be performed when tumor localization and surgical

technique do not permit preservation of the splenic vessels.[5] Conventionally, this was

believed to be necessary, because it was thought that splenic preservation might compromise the

oncological resection. However, it has been demonstrated that splenic preservation could be

accomplished with no increase in complication rate, operative time, or length of postoperative

stay. It is recommended that a spleen-preserving procedure should be performed for patients

with pancreatic adenocarcinoma at any intrapancreatic location unless the splenic capsule is

directly involved with or adherent to the tumor. Furthermore, the splenic vessels must be

unaffected by the tumor. [85]

Numerous techniques have been described for the subsequent division of the pancreas:

linear stapling, duct ligation and fibrin glue sealing.[5] The conventional method is ligation of

the main pancreatic duct and additional suturing of the stump to approximate the anterior and

posterior capsule. With the advent of surgical stapling devices, a new tool became available for

sealing the pancreatic stump, joining the harmonic scalpel, fibrin glue, and prolamine injection.

However, the use of a stapling device for closure of the pancreatic remnant was associated with

a significantly higher fistula rate.[85]

Central pancreatectomy

Central pancreatectomy (CP) has been proposed as an alternative for patients with small,

benign or low-grade malignant tumors such as endocrine and cystic neoplasms located in the

43

neck of the pancreas. The rationale for CP is to remove the neoplasm with preservation of the

functional parenchyma, and thereby avoiding a major resection. This method reduces the risk of

diabetes and exocrine insufficiency, and maintains the upper digestive and biliary anatomy.

Therefore, CP might be a safe and effective treatment option for small and/or low-grade

malignant neoplasms as well as for pancreatic metastases. However, it is questionable whether

this technique will also be applicable in patients with ductal adenocarcinoma, because this

tumor has a more aggressive biology. [89]

Total pancreatectomy

Total pancreatectomy (TP) is a combination of PD and distal pancreatectomy together

with a local lymphadenectomy. It is reserved for patients with multilocular or large tumors of

the pancreas. However, because the procedure is accompanied by postoperative exocrine and

endocrine insufficiency, and is associated with the highest mortality rates from all pancreatic

surgeries, it is rarely used. [84]

In the last decades, however, TP has become a possible treatment option, since

remarkable improvements in both surgery and postoperative management of the apancreatic

patient, with successful management of endocrine and exocrine insufficiency, were achieved.

However, there has not been shown any long-term survival benefit. [89]

Extended Lymphadenectomy – Extended Lymph Node

Dissection (ELND)

Up to 30% of patients with pancreatic cancer have retroperitoneal lymph node disease

that would not be resected by standard PD. It was hypothesized that survival could be improved

by using an ELND to help achieve node negativity. ELND involves, in addition to removal of

the pancreaticoduodenal nodes, removal of the lymph nodes along the hepatic artery, SMA, CA,

44

and between the aorta and the inferior pancreaticoduodenal artery. Furthermore, the

anterolateral aspect of the aorta and the inferior vena cava are also dissected. The earliest

proponents of ELND were the Japanese, who demonstrated improved survival rates with

extended surgery.[90] However, several studies, including for example Yeo et al.[91] and

Michalsky et al.[92], confirmed that ELND increased morbidity and offered no survival benefit

over standard lymphadenectomy with PD and concluded that ELND should only be performed

within controlled trials, if at all.[89-92]

Arterial/Venous Resections with Reconstruction

Vascular resections can be performed based on two rationales: firstly, to achieve negative

resection margins in case of vessel invasion by the tumor, making separation impossible;

secondly, it can be performed as a part of an extended pancreactectomy. Vein resections include

the portal vein, SMV, or the SMV-portal vein confluence.[90] Venous resections followed by

graft reconstruction can be performed without increased morbidity and mortality and may be

performed to achieve negative resection margins. [84, 90] Therefore, venous infiltration should

not be viewed as a contraindication for resection.[5]

In contrast, arterial resections of the SMA, celiac, and hepatic arteries are rarely

performed and they are considered by most as contraindicated in PD due to the greatly increased

morbidity and mortality.[90]

Short-Term Outcome

The main complications of PD are pancreatic fistulas, DGE, intra-abdominal abscesses,

and hemorrhage.[5]

A review article by Alexakis et al.[93] showed that the incidence of fistula or leakage was

2–24%.[5, 88, 93] Importantly, pancreatic fistula is closely associated with subsequent

mortality, mainly because of retroperitoneal sepsis and bleeding.[93-94] Thus, meticulous

45

surgical technique during construction of the pancreaticoenteric anastomosis is of high

importance. The value of octreotide is still debated in the literature: some studies show a

decrease in the occurrence of pancreatic fistula with octreotide, whereas others show no benefit.

Intra-abdominal abscesses can frequently occur following pancreatic fistula

development.[93, 95-96] These abscesses can usually be drained percutaneously and are most

commonly found sub-hepatically or at the left diaphragm.[5] The usual cause is a leak from the

pancreatic-enteric anastomosis. A CT scan will usually diagnose the abscess, and image-guided

drainage is usually successful.[88]

DGE is usually related to intra-abdominal complications.[93, 95] The incidence of DGE

reported by various studies has varied considerably (14–30%), because there is no standard

definition.[5, 95-96]

Bleeding can be classified roughly into early and late hemorrhage. Bleeding within the

first 24 hours postoperatively is usually a result of inadequate intraoperative hemostasis or

hemorrhage from one of the enteric anastomoses, and it can usually be treated conservatively.

Late hemorrhage is a severe complication, with considerable mortality (up to 60%), and occurs

mainly after leakage of the pancreatic anastomosis.[5] Its management is still controversial:

radiological embolization of the bleeding vessel and surgical hemostasis are both valid

treatment options.[5, 88, 93, 95, 97]

In pancreatic resection, perioperative mortality is closely associated with the level of

experience of the team providing therapy, thus it should be performed by experienced

surgeons.[5, 88]

Long-Term Outcome

Pancreatic cancer is commonly viewed as a disease uniformly leading to death. The long-

term prognosis, however, has improved over the past decade.[5] Recent studies demonstrated 5

year survival rates of approximately 20% after curative resection.[5, 98-99] In a subgroup of

patients with lymph-node and margin-negative tumors, 5-year survival of up to 40% has been

46

achieved.[5, 100] Recently, it has been shown that patients who survive 5 years after resection

have a subsequent 5-year survival rate of approximately 55%.[5, 101]

The most important factors influencing outcome after PD are curative resection, absence

of lymph node metastases, tumor size (<2cm), well or moderately differentiated tumors, absence

of perineural or vascular invasion, and the surgeons‟ experience. [5, 88] Wagner et al.

concluded that “curative resection is the single most important factor determining the outcome

in patients with pancreatic cancer”.[99]

CA19-9 levels also seem to predict long-term outcome. Preoperative CA19-9 levels

correlate with disease stage, and a postoperative decrease are strong independent predictors of

survival.[5]

Recently, quality of life after PD was evaluated, and shown to be impaired only in the

first 3-6 months after surgery, following this period quality of life was comparable to

preoperative levels.[5]

47

CHEMORADIOTHERAPY

Despite attempted curative resection of localized pancreatic adenocarcinoma, most

patients recur and die of their disease.[102] The high risk of local and systemic disease

recurrence, as well as overall poor prognosis, laid down the rationale for adjuvant therapy after

resection of pancreatic adenocarcinoma.[103-105] There is no consensus on what constitutes

“standard” adjuvant therapy. This controversy derives from several studies, each one with its

own limitations.[105]

The Gastro-Intestinal Study Group (GITSG) trial was the first prospective randomized

trial suggesting survival advantage with postoperative chemo-radiotherapy using bolus 5-

Fluorouracil (5-FU).[106] However, this study was criticized for poor patient selection, early

termination, small patient number, and suboptimal radiotherapy dose.[102]

The European Organization of Research and Treatment of Cancer (EORTC) compared 5-

FU plus radiotherapy with observation only in patients with resected pancreatic

adenocarcinoma.[107] The study showed a trend toward benefit in median survival (17.1

months vs. 12.6 months; P=0.099) of the 5-FU plus radiotherapy group.[107] This study was

also criticized for suboptimal dose of radiotherapy and split courses.[102]

ESPAC-1 trial sparked a new debate over the role of radiotherapy in the adjuvant therapy

of pancreatic cancer.[108] ESPAC-1 trial was designed to compare adjuvant concurrent

chemoradiotherapy (5-FU plus radiotherapy), chemotherapy alone (5-FU), chemoradiotherapy

followed by chemotherapy, and observation. Chemotherapy only arm had statistically

significant benefit over observation arm in median survival (20.1 months vs. 15.5 months;

P=0.009). However, chemoradiotherapy arm showed worse median survival compared with

patients who did not receive chemo-radiotherapy (15.9 months vs. 17.9 months; P=0.05).[108]

Major criticism was made on this study for possible selection bias, suboptimal radiotherapy, and

for allowing the final radiotherapy dose to be left to the judgment of the treating

physicians.[102]

48

CONKO-001 study randomized patients with resected pancreatic cancer to gemcitabine

or observation.[109] This trial showed statistically significant disease free survival benefit (13.4

months vs. 6.9 months; P<0.001) of gemcitabine over observation. This benefit of

chemotherapy in CONKO-001 study was consistent with the result from ESPAC-1 trial which

showed benefit of 5-FU over no adjuvant therapy.[108-109]

Radiation Therapy Oncology Group (RTOG) 97-04 was the first American cooperative

group study since the GITSG trial. The study had no observation alone arm. RTOG 97-04 study

randomized resected pancreatic cancer patients to evaluate benefit of adding gemcitabine to 5-

FU combined with radiotherapy.[109] One arm received 5-FU plus radiotherapy and the other

arm was treated with gemcitabine before and after 5-FU plus radiotherapy.[105] The addition of

gemcitabine to adjuvant fluorouracil-based chemoradiation was associated with a survival

benefit for patients, although this improvement was not statistically significant.[110]

Neoptolemos et al. presented the results of ESPAC-3 study.[102] This is a multicenter,

international, open-label, randomized, controlled, phase III trial that aimed at comparing of

adjuvant 5-FU/leucovorin versus gemcitabine in patients with resected pancreatic ductal

adenocarcinoma. The primary outcome measure was overall survival; the secondary measures

were toxicity, progression free survival and quality of life. The study confirmed the role of

adjuvant chemotherapy. However, overall survival was similar on both arms, hence showing

that gemcitabine is not superior to 5-FU in adjuvant setting. Median overall survival was 23.0

months with 5-FU and it was 23.6 months with gemcitabine. However, safety and dose intensity

favors gemcitabine. This study is very important because there has been a tendency to reject 5-

FU in pancreatic cancer and now it is very much back on the stage.[102]

Pancreatic adenocarcinoma remains a dismal disease with poor prognosis, even after

curative resection and most aggressive combine-modality approaches. This serves as a reminder

that we need to push the envelope even further.

The varying results of these randomized trials make it difficult to establish a standard

adjuvant therapy for resected pancreatic adenocarcinoma.[105]

49

The National Comprehensive Cancer Network (NCCN) recommends that investigational

options should be considered in all phases of disease management.[105, 111] Additionally, until

further data are available, the NCCN recommends postoperative radiotherapy plus 5-FU with or

without additional chemotherapy (gemcitabine), or chemotherapy alone (gemcitabine based) for

all patients after curative resection for pancreatic cancer, regardless of nodal status.[105, 111]

One way of eliminating potential treatment delays is to consider neoadjuvant

chemoradiotherapy or chemotherapy. This approach leads to increased survival, down-staging

marginal lesions, and sparing patients with rapidly progressive disease unnecessary

surgery.[105, 112] Single institution studies including neoadjuvant chemoradiotherapy or

chemotherapy have shown promise and provide the framework for larger controlled trials

evaluating the role of neoadjuvant therapy in the management of both resectable and marginally

resectable lesions.[105]

In patients with locally advanced disease local control remains an important issue in

terms of palliation (discussed later). There is promise that the therapeutic ratio using modern

radiotherapy techniques will improve and patients should be offered trials employing these

techniques. Furthermore, it is still to be proven whether chemotherapy alone can provide to

these patients tumor control, symptom palliation and survival with less toxicity.[1]

In patients with metastatic disease improvements in supportive care have dramatically

altered their quality of life: the use of anabolic agents such as Megestrol acetate, dexamethasone

and pancreatic supplements.[1]

Until 1995 there was little evidence that chemotherapy provided any benefit, and it was

recognized that the agents used showed no major apparent gains in survival and were toxic.

However, some small studies, all showing improved survival over best supportive care, have

altered perceptions. The first study using gemcitabine in 5-FU refractory patients showed a

response rate of 9.5%, a median survival of 3.9 months and a 6-month survival rate of 31%.[1,

113] Gemcitabine has subsequently been studied in a large number of studies involving

alternative comparisons including both 5-FU and novel therapies.[1]

50

Nowadays, single agent gemcitabine remains the treatment of choice for metastatic

pancreatic cancer patients of reasonable performance status, with the promise of both improved

symptom control, survival and tolerable toxicity.[1]

In patients with confirmed failure of gemcitabine first-line therapy one should consider a

second-line strategy. Some regimens showed activity in second-line therapy like paclitaxel,

gemcitabine and capecitabine or oxaliplatin combinations, raltitrexed, irinotecan, erlotinib.

However, these results were never confirmed. The available data suggest that some selected

patients can benefit from second-line therapy, but there is no standard option at this time.[114]

Novel targeted agents have received extensive attention because of the relative

insensitivity to conventional therapy. Recent attention has been directed at epidermal growth

factor (EGF) inhibition.[1] Human epidermal growth factor receptor (EGFR) type 1 is

overexpressed in many pancreatic tumors and is associated with poor prognosis and disease

progression. In experimental models, blocking EGFR improves the anticancer effects of

gemcitabine. Ongoing studies are incorporating inhibitors of the EGFR (e.g., erlotinib) as well

as monoclonal antibodies (e.g., cetuximab). A trial compared gemcitabine with and without

erlotinib in patients with locally advanced or metastatic pancreatic cancer and it showed an

overall survival significantly prolonged on the erlotinib plus gemcitabine arm. One-year

survival and progression free survival were also greater with this combination. However, there

was a higher incidence of some adverse events. Although the gain in survival was statistically

significant, the modest 2-week improvement in median survival hardly has a clinical

impact.[114]

Cetuximab presented promising results in early studies in combination with gemcitabine.

Those results led to a trial (gemcitabine alone or plus cetuximab).[112] Unfortunately, in a

preliminary report, the addition of cetuximab did not improve any of the planned

outcomes.[114]

As many other tumors, pancreatic cancers also frequently overexpress vascular

endothelial growth factor (VEGF) and its receptor.[114] Therefore, there has been substantial

interest in the role of bevacizumab (a VEGF ligand inhibitor). Other VEGF receptor inhibitors

51

are also likely to be studied given the high expression of VEGF receptor on pancreatic cancer

cells.[1]

Systemic therapy benefits pancreatic cancer patients who are not suitable for surgical

therapy. The regimens that proved benefit in survival always involved gemcitabine, establishing

such drug as the backbone of the treatment. The association of a second agent like capecitabine

or erlotinib helps some patients, especially those with better performance, but the cost-benefit

analysis has to be considered. For patients with a good performance status after failing initial

gemcitabine-based therapy, second-line therapy can be considered. The optimal regimen is

unknown, but options include fluoropyrimidines alone or oxaliplatin plus either 5-FU,

capecitabine, irinotecan or single-agent paclitaxel.[114]

52

PALLIATION TREATMENT

Treatment depends upon the probable length of survival, presence of metastatic disease or

advanced local disease as well as symptoms. Common symptoms include jaundice, weight loss,

pain, and vomiting from gastric outlet obstruction.[83]

Biliary Obstruction

Patients with metastatic disease and biliary obstruction are candidates for endoscopic or

percutaneous placement of biliary stent.[114] Perforation and pancreatitis are unusual

complications and antibiotic coverage of biliary flora is required to prevent cholangitis.

Pancreatitis, hemorrhage and biliary peritonitis have all been described as complications of

percutaneous stent placement.[83]

Surgical options to relieve obstructive jaundice are preferably performed in patients with

locally advanced disease and when stent placement is impracticable for technical reasons. A

bypass between the gallbladder and jejunum or, preferably, the common bile duct and jejunum

are the main options available. One advantage of surgical bypass is to perform a prophylactic

gastrojejunostomy to prevent a later duodenal obstruction. The high incidence of jaundice

recurrence following endoscopic stenting and the occurrence of tumor-associated duodenal

obstruction are points favoring operative palliative management in selected patients.[114]

Gastric Outlet Obstruction

Duodenal obstruction leading to gastric outlet obstruction occurs in 15–20% of patients

with advanced disease.[114] Although routine prophylactic gastrojejunostomy for operative

palliation of pancreatic cancer has been proposed by many authors [114-116], it has been

53

contested by others due to the high incidence of postoperative DGE [114, 117]. Some reports

show an incidence of DGE after duodenum bypass surgery ranging from 24 to 29%.[114, 117-

118] Endoscopically placed stents are a more favorable option in patients with metastatic

disease, when a shorter survival is expected. They offer palliative benefit with less

morbidity.[83, 114]

Severe Abdominal Pain

Pharmacologic therapy with opioids and neurolytic celiac plexus block are the most

commonly used techniques for palliation of pain associated with unresectable pancreatic

adenocarcinoma. Existing studies suggest that celiac plexus block is a well-tolerated

intervention that may improve analgesia, decrease opioid requirements, and minimize

deterioration of quality of life. Celiac plexus block can be performed percutaneously, surgically,

or under endosonographic guidance.[114] Pain relief lasting for up 24 weeks has been observed

in 70% of patients. The main complications include transient diarrhea and hypotension because

of sympathetic blockade and may be present in about 40% of patients. Severe adverse effects

are uncommon.[114, 119] Radiotherapy can be used to relieve pain due local invasion of

pancreatic cancer and is successful in 35–65% of patients.[114]

Pancreatic insufficiency

Obstruction of the pancreatic duct may lead to symptoms of pancreatic insufficiency such

as steatorrhoea and weight loss. The addition of oral pancreatic exocrine supplements will

usually control these symptoms. Pancreatic endocrine insufficiency (diabetes mellitus) should

be treated as type 2 diabetes.[83]

54

CONCLUSIONS

Pancreatic adenocarcinoma still remains a disease with a dismal prognosis, despite all of

the advances made in its management. The etiology of this neoplasia is still unknown, but

smoking is strongly associated with it. Also, in the last decades we have witnessed an explosion

in our understanding of the genetic modifications underlining the pathogenesis and progression

to invasive carcinoma. Moreover, recent advances in imaging techniques and tumor markers

allowed a better and earlier diagnose and staging as well as more accurate follow-up. Therefore,

aggressive evaluation should be performed.

Surgical resection is still the best hope for cure for pancreatic adenocarcinoma. However,

only a small number of patients have indication for curative resection. Between the different

surgical techniques, it has been proved that there are no significant differences in outcome.

Therefore, surgeon‟s preference and experience should dictate the type and procedure.

Moreover, surgical management (either resection for cure or operative palliation) should be

performed by experienced surgeons to minimize morbidity and mortality.

Following resection, postoperative adjuvant radiation and chemotherapy should be

offered to most patients for improved long-term survival. However, a standard treatment is not

yet available.

55

ACKNOWLEDGEMENTS

The author would like to thank Prof. António Taveira Gomes for his support and

contribution in this article.

56

REFERENCES

1. Goldstein D, Carroll S, Apte M, Keogh G: Modern management of pancreatic

carcinoma. Intern Med J, 2004. 34(8): p. 475-81.

2. Francis IR: Pancreatic adenocarcinoma: diagnosis and staging using multidetector-

row computed tomography (MDCT) and magnetic resonance imaging (MRI). Cancer

Imaging, 2007. 7 Spec No A: p. S160-5.

3. Beger HG, Rau B, Gansauge F, Poch B, Link KH: Treatment of pancreatic cancer:

challenge of the facts. World J Surg, 2003. 27(10): p. 1075-84.

4. Schima W, Ba-Ssalamah A, Kolblinger C, Kulinna-Cosentini C, Puespoek A, Gotzinger P:

Pancreatic adenocarcinoma. Eur Radiol, 2007. 17(3): p. 638-49.

5. Michalski CW, Weitz J, Buchler MW: Surgery insight: surgical management of

pancreatic cancer. Nat Clin Pract Oncol, 2007. 4(9): p. 526-35.

6. Kleeff J, Michalski C, Friess H, Buchler MW: Pancreatic cancer: from bench to 5-year

survival. Pancreas, 2006. 33(2): p. 111-8.

7. Lillemoe KD, Yeo CJ, Cameron JL: Pancreatic cancer: state-of-the-art care. CA Cancer J

Clin, 2000. 50(4): p. 241-68.

8. Ghadirian P, Lynch HT, Krewski D: Epidemiology of pancreatic cancer: an overview.

Cancer Detect Prev, 2003. 27(2): p. 87-93.

9. Yeo TP, Hruban RH, Leach SD, Wilentz RE, Sohn TA, Kern SE, Iacobuzio-Donahue CA,

Maitra A, Goggins M, Canto MI, Abrams RA, Laheru D, Jaffee EM, Hidalgo M, Yeo CJ:

Pancreatic cancer. Curr Probl Cancer, 2002. 26(4): p. 176-275.

10. de Braud F, Cascinu S, Gatta G: Cancer of pancreas. Crit Rev Oncol Hematol, 2004.

50(2): p. 147-55.

11. Lowenfels AB, Maisonneuve P: Risk factors for pancreatic cancer. J Cell Biochem, 2005.

95(4): p. 649-56.

57

12. Lowenfels AB, Maisonneuve P: Epidemiology and risk factors for pancreatic cancer.

Best Pract Res Clin Gastroenterol, 2006. 20(2): p. 197-209.

13. Lowenfels AB, Maisonneuve P: Epidemiology and prevention of pancreatic cancer. Jpn

J Clin Oncol, 2004. 34(5): p. 238-44.

14. Fraumeni JF Jr.: Cancers of the pancreas and biliary tract: epidemiological

considerations. Cancer Res, 1975. 35(11 Pt. 2): p. 3437-46.

15. Flanders TY, Foulkes WD: Pancreatic adenocarcinoma: epidemiology and genetics. J

Med Genet, 1996. 33(11): p. 889-98.

16. Hidalgo Pascual M, Ferrero Herrero E, Castillo Fe MJ, Guadarrama Gonzalez FJ, Pelaez

Torres P, Botella Ballesteros F: Epidemiology and diagnosis of the pancreatic cancer.

Rev Esp Enferm Dig, 2004. 96(10): p. 714-8; 719-22.

17. Winter JM, Maitra A, Yeo CJ: Genetics and pathology of pancreatic cancer. HPB

(Oxford), 2006. 8(5): p. 324-36.

18. Hansel DE, Kern SE, Hruban RH: Molecular pathogenesis of pancreatic cancer. Annu

Rev Genomics Hum Genet, 2003. 4: p. 237-56.

19. Iacobuzio-Donahue CA, van der Heijden MS, Baumgartner MR, Troup WJ, Romm JM,

Doheny K, Pugh E, Yeo CJ, Goggins MG, Hruban RH, Kern SE: Large-scale allelotype of

pancreaticobiliary carcinoma provides quantitative estimates of genome-wide allelic

loss. Cancer Res, 2004. 64(3): p. 871-5.

20. Wilentz RE, Goggins M, Redston M, Marcus VA, Adsay NV, Sohn TA, Kadkol SS, Yeo CJ,

Choti M, Zahurak M, Johnson K, Tascilar M, Offerhaus GJ, Hruban RH, Kern SE: Genetic,

immunohistochemical, and clinical features of medullary carcinoma of the pancreas:

A newly described and characterized entity. Am J Pathol, 2000. 156(5): p. 1641-51.

21. Feinberg AP: The epigenetics of cancer etiology. Semin Cancer Biol, 2004. 14(6): p.

427-32.

58

22. Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, Seymour AB, Weinstein CL,

Hruban RH, Yeo CJ, Kern SE: Frequent somatic mutations and homozygous deletions

of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nat Genet, 1994. 8(1): p. 27-

32.

23. Schutte M, Hruban RH, Geradts J, Maynard R, Hilgers W, Rabindran SK, Moskaluk CA,

Hahn SA, Schwarte-Waldhoff I, Schmiegel W, Baylin SB, Kern SE, Herman JG:

Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic

carcinomas. Cancer Res, 1997. 57(15): p. 3126-30.

24. Feldmann G, Maitra A: Molecular genetics of pancreatic ductal adenocarcinomas and

recent implications for translational efforts. J Mol Diagn, 2008. 10(2): p. 111-22.

25. Maitra A, Hruban RH: Pancreatic cancer. Annu Rev Pathol, 2008. 3: p. 157-88.

26. Maitra A, Kern SE, HrubanRH: Molecular pathogenesis of pancreatic cancer. Best Pract

Res Clin Gastroenterol, 2006. 20(2): p. 211-26.

27. Hruban RH, Takaori K, Canto M, Fishman EK, Campbell K, Brune K, Kern SE, Goggins M:

Clinical importance of precursor lesions in the pancreas. J Hepatobiliary Pancreat

Surg, 2007. 14(3): p. 255-63.

28. Takaori K: Current understanding of precursors to pancreatic cancer. J Hepatobiliary

Pancreat Surg, 2007. 14(3): p. 217-23.

29. Feldmann G, Beaty R, Hruban RH, Maitra A: Molecular genetics of pancreatic

intraepithelial neoplasia. J Hepatobiliary Pancreat Surg, 2007. 14(3): p. 224-32.

30. Wilentz RE, Albores-Saavedra J, Hruban RH: Mucinous cystic neoplasms of the

pancreas. Semin Diagn Pathol, 2000. 17(1): p. 31-42.

31. Maitra A, Fukushima N, Takaori K, Hruban RH: Precursors to invasive pancreatic

cancer. Adv Anat Pathol, 2005. 12(2): p. 81-91.

32. Singh M, Maitra A: Precursor lesions of pancreatic cancer: molecular pathology and

clinical implications. Pancreatology, 2007. 7(1): p. 9-19.

59

33. van den Berg W, Tascilar M, Offerhaus GJ, Albores-Saavedra J, Wenig BM, Hruban RH,

Gabrielson E: Pancreatic mucinous cystic neoplasms with sarcomatous stroma:

molecular evidence for monoclonal origin with subsequent divergence of the

epithelial and sarcomatous components. Mod Pathol, 2000. 13(1): p. 86-91.

34. Hyde GL, Davis JB Jr, McMillin RD, McMillin M: Mucinous cystic neoplasm of the

pancreas with latent malignancy. Am Surg, 1984. 50(4): p. 225-9.

35. Le Borgne J, de Calan L, Partensky C: Cystadenomas and cystadenocarcinomas of the

pancreas: a multiinstitutional retrospective study of 398 cases. French Surgical

Association. Ann Surg, 1999. 230(2): p. 152-61.

36. Iacobuzio-Donahue CA, Wilentz RE, Argani P, Yeo CJ, Cameron JL, Kern SE, Hruban RH:

Dpc4 protein in mucinous cystic neoplasms of the pancreas: frequent loss of

expression in invasive carcinomas suggests a role in genetic progression. Am J Surg

Pathol, 2000. 24(11): p. 1544-8.

37. Flejou JF, Boulange B, Bernades P, Belghiti J, Henin D: p53 protein expression and DNA

ploidy in cystic tumors of the pancreas. Pancreas, 1996. 13(3): p. 247-52.

38. Jimenez RE, Warshaw AL, Z'Graggen K, Hartwig W, Taylor DZ, Compton CC, Fernandez-

del Castillo C: Sequential accumulation of K-ras mutations and p53 overexpression in

the progression of pancreatic mucinous cystic neoplasms to malignancy. Ann Surg,

1999. 230(4): p. 501-9; discussion 509-11.

39. Luttges J, Feyerabend B, Buchelt T, Pacena M, Kloppel G: The mucin profile of

noninvasive and invasive mucinous cystic neoplasms of the pancreas. Am J Surg

Pathol, 2002. 26(4): p. 466-71.

40. Chari ST, Yadav D, Smyrk TC, DiMagno EP, Miller LJ, Raimondo M, Clain JE, Norton IA,

Pearson RK, Petersen BT, Wiersema MJ, Farnell MB, Sarr MG: Study of recurrence

after surgical resection of intraductal papillary mucinous neoplasm of the pancreas.

Gastroenterology, 2002. 123(5): p. 1500-7.

60

41. Adsay NV, Pierson C, Sarkar F, Abrams J, Weaver D, Conlon KC, Brennan MF, Klimstra

DS: Colloid (mucinous noncystic) carcinoma of the pancreas. Am J Surg Pathol, 2001.

25(1): p. 26-42.

42. D'Angelica M, Brennan MF, Suriawinata AA, Klimstra D, Conlon KC: Intraductal

papillary mucinous neoplasms of the pancreas: an analysis of clinicopathologic

features and outcome. Ann Surg, 2004. 239(3): p. 400-8.

43. Kobayashi G, Fujita N, Noda Y, Ito K, Horaguchi J, Takasawa O, Akaishi S, Tsuchiya T,

Kobari M: Mode of progression of intraductal papillary-mucinous tumor of the

pancreas: analysis of patients with follow-up by EUS. J Gastroenterol, 2005. 40(7): p.

744-51.

44. Yoshizawa K, Nagai H, Sakurai S, Hironaka M, Morinaga S, Saitoh K, Fukayama M:

Clonality and K-ras mutation analyses of epithelia in intraductal papillary mucinous

tumor and mucinous cystic tumor of the pancreas. Virchows Arch, 2002. 441(5): p.

437-43.

45. Z'Graggen K, Rivera JA, Compton CC, Pins M, Werner J, Fernandez-del Castillo C,

Rattner DW, Lewandrowski KB, Rustgi AK, Warshaw AL: Prevalence of activating K-ras

mutations in the evolutionary stages of neoplasia in intraductal papillary mucinous

tumors of the pancreas. Ann Surg, 1997. 226(4): p. 491-8; discussion 498-500.

46. Kobari M, Egawa S, Shibuya K, Shimamura H, Sunamura M, Takeda K, Matsuno S,

Furukawa T: Intraductal papillary mucinous tumors of the pancreas comprise 2

clinical subtypes: differences in clinical characteristics and surgical management.

Arch Surg, 1999. 134(10): p. 1131-6.

47. Nakagohri T, Kenmochi T, Kainuma O, Tokoro Y, Asano T: Intraductal papillary

mucinous tumors of the pancreas. Am J Surg, 1999. 178(4): p. 344-7.

48. Terris B, Ponsot P, Paye F, Hammel P, Sauvanet A, Molas G, Bernades P, Belghiti J,

Ruszniewski P, Flejou JF: Intraductal papillary mucinous tumors of the pancreas

61

confined to secondary ducts show less aggressive pathologic features as compared

with those involving the main pancreatic duct. Am J Surg Pathol, 2000. 24(10): p.

1372-7.

49. Adsay NV, Merati K, Andea A, Sarkar F, Hruban RH, Wilentz RE, Goggins M, Iocobuzio-

Donahue C, Longnecker DS, Klimstra DS: The dichotomy in the preinvasive neoplasia

to invasive carcinoma sequence in the pancreas: differential expression of MUC1 and

MUC2 supports the existence of two separate pathways of carcinogenesis. Mod

Pathol, 2002. 15(10): p. 1087-95.

50. Luttges J, Zamboni G, Longnecker D, Kloppel G: The immunohistochemical mucin

expression pattern distinguishes different types of intraductal papillary mucinous

neoplasms of the pancreas and determines their relationship to mucinous noncystic

carcinoma and ductal adenocarcinoma. Am J Surg Pathol, 2001. 25(7): p. 942-8.

51. Terris B, Dubois S, Buisine MP, Sauvanet A, Ruszniewski P, Aubert JP, Porchet N,

Couvelard A, Degott C, Flejou JF: Mucin gene expression in intraductal papillary-

mucinous pancreatic tumours and related lesions. J Pathol, 2002. 197(5): p. 632-7.

52. Adsay NV, Merati K, Basturk O, Iacobuzio-Donahue C, Levi E, Cheng JD, Sarkar FH,

Hruban RH, Klimstra DS: Pathologically and biologically distinct types of epithelium in

intraductal papillary mucinous neoplasms: delineation of an "intestinal" pathway of

carcinogenesis in the pancreas. Am J Surg Pathol, 2004. 28(7): p. 839-48.

53. Furukawa T, Kloppel G, Volkan Adsay N, Albores-Saavedra J, Fukushima N, Horii A,

Hruban RH, Kato Y, Klimstra DS, Longnecker DS, Luttges J, Offerhaus GJ, Shimizu M,

Sunamura M, Suriawinata A, Takaori K, Yonezawa S: Classification of types of

intraductal papillary-mucinous neoplasm of the pancreas: a consensus study.

Virchows Arch, 2005. 447(5): p. 794-9.

54. Adsay NV, Adair CF, Heffess CS, Klimstra DS: Intraductal oncocytic papillary neoplasms

of the pancreas. Am J Surg Pathol, 1996. 20(8): p. 980-94.

62

55. Cuillerier E, Cellier C, Palazzo L, Deviere J, Wind P, Rickaert F, Cugnenc PH, Cremer M,

Barbier JP: Outcome after surgical resection of intraductal papillary and mucinous

tumors of the pancreas. Am J Gastroenterol, 2000. 95(2): p. 441-5.

56. Inagaki M, Maguchi M, Kino S, Obara M, Ishizaki A, Onodera K, Yokoyama K, Makino I,

Ojima H, Tokusashi Y, Miyokawa N, Kasai S: Mucin-producing tumors of the pancreas:

clinicopathological features, surgical treatment, and outcome. J Hepatobiliary

Pancreat Surg, 1999. 6(3): p. 281-5.

57. Kitagawa Y, Unger TA, Taylor S, Kozarek RA, Traverso LW: Mucus is a predictor of

better prognosis and survival in patients with intraductal papillary mucinous tumor

of the pancreas. J Gastrointest Surg, 2003. 7(1): p. 12-8; discussion 18-9.

58. Sugiura H, Kondo S, Islam HK, Ito K, Ono K, Morikawa T, Okushiba S, Katoh H:

Clinicopathologic features and outcomes of intraductal papillary-mucinous tumors of

the pancreas. Hepatogastroenterology, 2002. 49(43): p. 263-7.

59. Hruban RH, Adsay NV, Albores-Saavedra J, Compton C, Garrett ES, Goodman SN, Kern

SE, Klimstra DS, Kloppel G, Longnecker DS, Luttges J, Offerhaus GJ: Pancreatic

intraepithelial neoplasia: a new nomenclature and classification system for

pancreatic duct lesions. Am J Surg Pathol, 2001. 25(5): p. 579-86.

60. Hruban RH, Takaori K, Klimstra DS, Adsay NV, Albores-Saavedra J, Biankin AV, Biankin

SA, Compton C, Fukushima N, Furukawa T, Goggins M, Kato Y, Kloppel G, Longnecker

DS, Luttges J, Maitra A, Offerhaus GJ, Shimizu M, Yonezawa S: An illustrated consensus

on the classification of pancreatic intraepithelial neoplasia and intraductal papillary

mucinous neoplasms. Am J Surg Pathol, 2004. 28(8): p. 977-87.

61. Hruban RH, Goggins M, Parsons J, Kern SE: Progression model for pancreatic cancer.

Clin Cancer Res, 2000. 6(8): p. 2969-72.

63

62. Brat DJ, Lillemoe KD, Yeo CJ, Warfield PB, Hruban RH: Progression of pancreatic

intraductal neoplasias to infiltrating adenocarcinoma of the pancreas. Am J Surg

Pathol, 1998. 22(2): p. 163-9.

63. Brockie E, Anand A, Albores-Saavedra J: Progression of atypical ductal

hyperplasia/carcinoma in situ of the pancreas to invasive adenocarcinoma. Ann

Diagn Pathol, 1998. 2(5): p. 286-92.

64. Brune K, Abe T, Canto M, O'Malley L, Klein AP, Maitra A, Volkan Adsay N, Fishman EK,

Cameron JL, Yeo CJ, Kern SE, Goggins M, Hruban RH: Multifocal neoplastic precursor

lesions associated with lobular atrophy of the pancreas in patients having a strong

family history of pancreatic cancer. Am J Surg Pathol, 2006. 30(9): p. 1067-76.

65. Brown KM: Multidisciplinary approach to tumors of the pancreas and biliary tree.

Surg Clin North Am, 2009. 89(1): p. 115-31, ix.

66. Wilentz RE, Hruban RH: Pathology of cancer of the pancreas. Surg Oncol Clin N Am,

1998. 7(1): p. 43-65.

67. Hoimes CJ, Moyer MT, Saif MW: Biomarkers for early detection and screening in

pancreatic cancer. Highlights from the 45th ASCO annual meeting. Orlando, FL, USA.

May 29-June 2, 2009. JOP, 2009. 10(4): p. 352-6.

68. Duffy MJ, Sturgeon C, Lamerz R, Haglund C, Holubec VL, Klapdor R, Nicolini A, Topolcan

O, Heinemann V: Tumor markers in pancreatic cancer: a European Group on Tumor

Markers (EGTM) status report. Ann Oncol, 2009.

69. Steinberg WM, Gelfand R, Anderson KK, Glenn J, Kurtzman SH, Sindelar WF, Toskes PP:

Comparison of the sensitivity and specificity of the CA19-9 and carcinoembryonic

antigen assays in detecting cancer of the pancreas. Gastroenterology, 1986. 90(2): p.

343-9.

64

70. Goonetilleke KS, Siriwardena AK: Systematic review of carbohydrate antigen (CA 19-9)

as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol, 2007.

33(3): p. 266-70.

71. Karachristos A, Scarmeas N, Hoffman JP: CA 19-9 levels predict results of staging

laparoscopy in pancreatic cancer. J Gastrointest Surg, 2005. 9(9): p. 1286-92.

72. Ferrone CR, Finkelstein DM, Thayer SP, Muzikansky A, Fernandez-delCastillo C,

Warshaw AL: Perioperative CA19-9 levels can predict stage and survival in patients

with resectable pancreatic adenocarcinoma. J Clin Oncol, 2006. 24(18): p. 2897-902.

73. Fry LC, Monkemuller K, Malfertheiner P: Molecular markers of pancreatic cancer:

development and clinical relevance. Langenbecks Arch Surg, 2008. 393(6): p. 883-90.

74. Wong JC, Lu DS: Staging of pancreatic adenocarcinoma by imaging studies. Clin

Gastroenterol Hepatol, 2008. 6(12): p. 1301-8.

75. Peddu P, Quaglia A, Kane PA, Karani JB: Role of imaging in the management of

pancreatic mass. Crit Rev Oncol Hematol, 2009. 70(1): p. 12-23.

76. Vachiranubhap B, Kim YH, Balci NC, Semelka RC: Magnetic resonance imaging of

adenocarcinoma of the pancreas. Top Magn Reson Imaging, 2009. 20(1): p. 3-9.

77. Miller FH, Rini NJ, Keppke AL: MRI of adenocarcinoma of the pancreas. AJR Am J

Roentgenol, 2006. 187(4): p. W365-74.

78. Irisawa A, Sato A, Sato M, Ikeda T, Suzuki R, Ohira H: Early diagnosis of small

pancreatic cancer: role of endoscopic ultrasonography. Dig Endosc, 2009. 21 Suppl 1:

p. S92-6.

79. DeWitt J, Devereaux B, Chriswell M, McGreevy K, Howard T, Imperiale TF, Ciaccia D,

Lane KA, Maglinte D, Kopecky K, LeBlanc J, McHenry L, Madura J, Aisen A, Cramer H,

Cummings O, Sherman S: Comparison of endoscopic ultrasonography and

multidetector computed tomography for detecting and staging pancreatic cancer.

Ann Intern Med, 2004. 141(10): p. 753-63.

65

80. Saftoiu A, Vilmann P: Role of endoscopic ultrasound in the diagnosis and staging of

pancreatic cancer. J Clin Ultrasound, 2009. 37(1): p. 1-17.

81. Boujaoude J: Role of endoscopic ultrasound in diagnosis and therapy of pancreatic

adenocarcinoma. World J Gastroenterol, 2007. 13(27): p. 3662-6.

82. Katz MH, Hwang R, Fleming JB, Evans DB: Tumor-node-metastasis staging of

pancreatic adenocarcinoma. CA Cancer J Clin, 2008. 58(2): p. 111-25.

83. Thomson BN, Banting SW, Gibbs P: Pancreatic cancer - current management. Aust

Fam Physician, 2006. 35(4): p. 212-7.

84. Bachmann J, Michalski CW, Martignoni ME, Buchler MW, Friess H: Pancreatic

resection for pancreatic cancer. HPB (Oxford), 2006. 8(5): p. 346-51.

85. Loos M, Kleeff J, Friess H, Buchler MW: Surgical treatment of pancreatic cancer. Ann N

Y Acad Sci, 2008. 1138: p. 169-80.

86. Hines OJ, Reber HA: Pancreatic surgery. Curr Opin Gastroenterol, 2006. 22(5): p. 520-6.

87. Wray CJ, Ahmad SA, Matthews JB, Lowy AM: Surgery for pancreatic cancer: recent

controversies and current practice. Gastroenterology, 2005. 128(6): p. 1626-41.

88. Ujiki MB, Talamonti MS: Surgical management of pancreatic cancer. Semin Radiat

Oncol, 2005. 15(4): p. 218-25.

89. Glanemann M, Shi B, Liang F, Sun XG, Bahra M, Jacob D, Neumann U, Neuhaus P:

Surgical strategies for treatment of malignant pancreatic tumors: extended, standard

or local surgery? World J Surg Oncol, 2008. 6: p. 123.

90. D'Souza MA, Shrikhande SV: Pancreatic resectional surgery: an evidence-based

perspective. J Cancer Res Ther, 2008. 4(2): p. 77-83.

91. Yeo CJ, Cameron JL, Lillemoe KD, Sohn TA, Campbell KA, Sauter PK, Coleman J, Abrams

RA, Hruban RH: Pancreaticoduodenectomy with or without distal gastrectomy and

extended retroperitoneal lymphadenectomy for periampullary adenocarcinoma,

66

part 2: randomized controlled trial evaluating survival, morbidity, and mortality. Ann

Surg, 2002. 236(3): p. 355-66; discussion 366-8.

92. Michalski CW, Kleeff J, Wente MN, Diener MK, Buchler MW, Friess H: Systematic

review and meta-analysis of standard and extended lymphadenectomy in

pancreaticoduodenectomy for pancreatic cancer. Br J Surg, 2007. 94(3): p. 265-73.

93. Alexakis N, Halloran C, Raraty M, Ghaneh P, Sutton R, Neoptolemos JP: Current

standards of surgery for pancreatic cancer. Br J Surg, 2004. 91(11): p. 1410-27.

94. Aranha GV, Aaron JM, Shoup M, Pickleman J:et al., Current management of pancreatic

fistula after pancreaticoduodenectomy. Surgery, 2006. 140(4): p. 561-8; discussion

568-9.

95. Halloran CM, Ghaneh P, Bosonnet L, Hartley MN, Sutton R, Neoptolemos JP:

Complications of pancreatic cancer resection. Dig Surg, 2002. 19(2): p. 138-46.

96. Yeo CJ, Cameron JL, Sohn TA, Lillemoe KD, Pitt HA, Talamini MA, Hruban RH, Ord SE,

Sauter PK, Coleman J, Zahurak ML, Grochow LB, Abrams RA: Six hundred fifty

consecutive pancreaticoduodenectomies in the 1990s: pathology, complications, and

outcomes. Ann Surg, 1997. 226(3): p. 248-57; discussion 257-60.

97. Berberat PO, Friess H, Kleeff J, Uhl W, Buchler MW: Prevention and treatment of

complications in pancreatic cancer surgery. Dig Surg, 1999. 16(4): p. 327-36.

98. Winter JM, Cameron JL, Campbell KA, Arnold MA, Chang DC, Coleman J, Hodgin MB,

Sauter PK, Hruban RH, Riall TS, Schulick RD, Choti MA, Lillemoe KD, Yeo CJ: 1423

pancreaticoduodenectomies for pancreatic cancer: A single-institution experience. J

Gastrointest Surg, 2006. 10(9): p. 1199-210; discussion 1210-1.

99. Wagner M, Redaelli C, Lietz M, Seiler CA, Friess H, Buchler MW: Curative resection is

the single most important factor determining outcome in patients with pancreatic

adenocarcinoma. Br J Surg, 2004. 91(5): p. 586-94.

67

100. Cameron JL, Riall TS, Coleman J, Belcher KA: One thousand consecutive

pancreaticoduodenectomies. Ann Surg, 2006. 244(1): p. 10-5.

101. Riall TS, Cameron JL, Lillemoe KD, Winter JM, Campbell KA, Hruban RH, Chang D, Yeo

CJ: Resected periampullary adenocarcinoma: 5-year survivors and their 6- to 10-year

follow-up. Surgery, 2006. 140(5): p. 764-72.

102. Saif MW: Adjuvant treatment of pancreatic cancer in 2009: where are we? Highlights

from the 45th ASCO annual meeting. Orlando, FL, USA. May 29-June 2, 2009. JOP,

2009. 10(4): p. 373-7.

103. Mu DQ, Peng SY, Wang GF: Risk factors influencing recurrence following resection of

pancreatic head cancer. World J Gastroenterol, 2004. 10(6): p. 906-9.

104. Shibata K, Matsumoto T, Yada K, Sasaki A, Ohta M, Kitano S: Factors predicting

recurrence after resection of pancreatic ductal carcinoma. Pancreas, 2005. 31(1): p.

69-73.

105. Saif MW: Controversies in the adjuvant treatment of pancreatic adenocarcinoma.

JOP, 2007. 8(5): p. 545-52.

106. Kalser MH, Ellenberg SS: Pancreatic cancer. Adjuvant combined radiation and

chemotherapy following curative resection. Arch Surg, 1985. 120(8): p. 899-903.

107. Klinkenbijl JH, Jeekel J, Sahmoud T, van Pel R, Couvreur ML, Veenhof CH, Arnaud JP,

Gonzalez DG, de Wit LT, Hennipman A, Wils J: Adjuvant radiotherapy and 5-

fluorouracil after curative resection of cancer of the pancreas and periampullary

region: phase III trial of the EORTC gastrointestinal tract cancer cooperative group.

Ann Surg, 1999. 230(6): p. 776-82; discussion 782-4.

108. Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, Beger H, Fernandez-

Cruz L, Dervenis C, Lacaine F, Falconi M, Pederzoli P, Pap A, Spooner D, Kerr DJ, Buchler

MW: A randomized trial of chemoradiotherapy and chemotherapy after resection of

pancreatic cancer. N Engl J Med, 2004. 350(12): p. 1200-10.

68

109. Oettle H, Post S, Neuhaus P, Gellert K, Langrehr J, Ridwelski K, Schramm H, Fahlke J,

Zuelke C, Burkart C, Gutberlet K, Kettner E, Schmalenberg H, Weigang-Koehler K,

Bechstein WO, Niedergethmann M, Schmidt-Wolf I, Roll L, Doerken B, Riess H:

Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing

curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA,

2007. 297(3): p. 267-77.

110. Regine WF, Winter KA, Abrams RA, Safran H, Hoffman JP, Konski A, Benson AB,

Macdonald JS, Kudrimoti MR, Fromm ML, Haddock MG, Schaefer P, Willett CG, Rich

TA: Fluorouracil vs gemcitabine chemotherapy before and after fluorouracil-based

chemoradiation following resection of pancreatic adenocarcinoma: a randomized

controlled trial. JAMA, 2008. 299(9): p. 1019-26.

111. Tempero MA, Behrman S, Ben-Josef E, Benson AB 3rd, Cameron JL, Casper ES,

Hoffman JP, Karl RC, Kim P, Koh WJ, Kuvshinoff BW 2nd, Melvin WS, Muscarella P 2nd,

Sasson AR, Shibata S, Shrieve DC, Talamonti MS, Tyler DS, Vickers SM, Warren RS,

Willett C, Wolff RA: Pancreatic adenocarcinoma: Clinical Practice Guidelines in

Oncology. J Natl Compr Canc Netw, 2005. 3(5): p. 598-626.

112. Kim R, Saif MW: Is there an optimal neoadjuvant therapy for locally advanced

pancreatic cancer? JOP, 2007. 8(3): p. 279-88.

113. Rothenberg ML, Moore MJ, Cripps MC, Andersen JS, Portenoy RK, Burris HA 3rd, Green

MR, Tarassoff PG, Brown TD, Casper ES, Storniolo AM, Von Hoff DD: A phase II trial of

gemcitabine in patients with 5-FU-refractory pancreas cancer. Ann Oncol, 1996. 7(4):

p. 347-53.

114. Freitas D, Fernandes Gdos S, Hoff PM, Cunha JE: Medical management of pancreatic

adenocarcinoma. Pancreatology, 2009. 9(3): p. 223-32.

69

115. van Wagensveld BA, Coene PP, van Gulik TM, Rauws EA, Obertop H, Gouma DJ:

Outcome of palliative biliary and gastric bypass surgery for pancreatic head

carcinoma in 126 patients. Br J Surg, 1997. 84(10): p. 1402-6.

116. Lillemoe KD, Cameron JL, Yeo CJ, Sohn TA, Nakeeb A, Sauter PK, Hruban RH, Abrams

RA, Pitt HA: Pancreaticoduodenectomy. Does it have a role in the palliation of

pancreatic cancer? Ann Surg, 1996. 223(6): p. 718-25; discussion 725-8.

117. van der Schelling GP, van den Bosch RP, Klinkenbij JH, Mulder PG, Jeekel J: Is there a

place for gastroenterostomy in patients with advanced cancer of the head of the

pancreas? World J Surg, 1993. 17(1): p. 128-32; discussion 132-3.

118. Konishi M, Ryu M, Kinoshita T, Kawano N, Tanizaki H, Cho A: Stomach-preserving

gastric bypass for unresectable pancreatic cancer. Surg Today, 1997. 27(5): p. 429-33.

119. Wong GY, Schroeder DR, Carns PE, Wilson JL, Martin DP, Kinney MO, Mantilla CB,

Warner DO: Effect of neurolytic celiac plexus block on pain relief, quality of life, and

survival in patients with unresectable pancreatic cancer: a randomized controlled

trial. JAMA, 2004. 291(9): p. 1092-9.

70

FIGURES

Figure 1. Hypothesis of progression from precursor lesions to invasive carcinoma.

Schematic presentation of a hypothesis for the progression pathways from pancreatic

intraepithelial neoplasias (PanINs) and various subtypes of intraductal papillary mucinous

neoplasms (IPMNs) towards invasive carcinoma. Also, the mucin expression pattern of the

different types of PanINs and IPMNs, as well as the invasive carcinomas. PB type,

pancreatobiliary type; MNCC, mucinous noncystic carcinoma. Adapted from Takaori et al. [28]

71

TABLES

Table1. Staging of Pancreatic Adenocarcinoma (6th edition of the American Joint

Association on Cancer).

Note that, in general, Stages I and II are reserved for potentially resectable disease, Stage III for

locally unresectable disease, and Stage IV for metastatic disease. LN, lymph node; CA, celiac

axis; SMA, superior mesenteric artery; CHA, hepatic artery. Adapted from Katz et al [82].

72

Definitions Sixth Edition – Description Comments

Primary tumor (T)

T0 No evidence of primary

Tis In situ

T1 Limited to the pancreas, ≤2 cm

T2 Limited to the pancreas, >2 cm

T3 Extends beyond pancreas, no

involvement of CA or SMA

T4 Involves CA or SMA

Regional lymph nodes (N)

N0 No nodal metastasis

N1 Regional lymph node metastasis No distinction between the number

of involved nodes

Distant metastasis (M)

M0 No distant metastasis

M1 Distant metastasis

Stage Groupings

Stage 0 Tis N0 M0 In situ disease

Stage I IA – T1 N0 M0

IB – T2 N0 M0

Potentially resectable disease that

is confined to the pancreas

Stage II IIA – T3 N0 M0

IIB – T1-3 N1 M0

Usually potentially resectable; may

involve venous structures, adjacent

organs, LN, or CHA, but not CA

or SMA

Stage III T4 N0-1 M0 Locally advanced; unresectable

due to CA or SMA involvement

Stage IV T1-4 N0-1 M1 Metastatic; unresectable due to

distant metastatic disease