Download as pdf or txt
Download as pdf or txt
You are on page 1of 48

Current Problems in Surgery 53 (2016) 107–154

Contents lists available at ScienceDirect

Current Problems in Surgery

journal homepage: www.elsevier.com/locate/cpsurg

Pancreatic adenocarcinoma
Mariam F. Eskander, MD, Lindsay A. Bliss, MD, MPH,
n
Jennifer F. Tseng, MD, MPH

Introduction

Pancreatic cancer is the ninth most common cancer in the United States, yet it is the fourth
leading cause of cancer death.1 Of the 42,000 cases diagnosed in 2014, approximately 35,000
will likely die in the next 5 years. By 2030, with a projected 55% increase in the incidence of the
disease, the number of new annual cases could be as high as 65,000.
The only potential cure for pancreatic cancer is surgery, although few patients undergo
surgery because of advanced stage, and almost all resected patients typically experience
recurrence within 10 years. Optimal treatment strategies are multimodal and should be
customized to the individual patient. Acquiring the skills to overcome this formidable disease
requires an understanding of its unique pathophysiology, risk factors, and precursors, as well as
current paradigms of therapy. Pancreatic adenocarcinoma, the most common type of pancreatic
cancer, will be referred to as pancreatic cancer in this review.

Historical considerations

Cancer of the pancreas has long fascinated and challenged the medical community. The
pathologist Giovanni Battista Morgagni first described the disease in 1769, although limited to
external examination of the specimen, he could not be sure whether it was cancer and not
chronic inflammation.2 The late 1800s ushered in a flurry of attempts at surgical removal (the
first operation for pancreatic cancer was performed in 1882) making risky surgery the only
therapeutic option and placing surgeons at the forefront of pancreatic cancer treatment. In
addition to gaps in knowledge, there were technical barriers to effective treatment. Reflecting on
these early difficulties facing surgeons, a textbook from 1902 states, “The treatment of malignant

From the Department of Surgery, Surgical Outcomes Analysis & Research, Beth Israel Deaconess Medical Center, Harvard
Medical School, Boston, MA; n Address reprint requests to Jennifer F. Tseng, MD, MPH, Division of Surgical Oncology,
BIDMC Cancer Center, Harvard Medical School, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Stoneman 9,
Boston, MA 02215. E-mail address: jftseng@bidmc.harvard.edu (J.F. Tseng)

http://dx.doi.org/10.1067/j.cpsurg.2016.01.001
0011-3840/& 2016 Elsevier Inc. All rights reserved.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
108 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

disease of the pancreas by the surgeon can hardly be said to exist…the mechanical difficulties of
the operation are well-nigh insuperable, and that if boldness and good fortune are the operator’s
gifts, the result to the patient hardly justifies the means.”3 With morbidity and mortality rates
still as high as 60% and 25%, respectively, into the 1960s, operative safety has only recently
radically improved.4,5 Although profound changes have occurred in the last 40 years in the way
we treat patients with the disease, which is now understood to be systemic, these changes
have not yet resulted in an improved overall life expectancy. Pancreatic cancer has just an 8%
5-year survival rate, only a 5% improvement since 1975.1,6,7 In the modern era of multimodal,
multidisciplinary treatment, the importance of surgery has not diminished, although it is now
seen as a tool in a larger toolbox of therapies.

Etiologic factors

Several risk factors for pancreatic cancer, some genetic and some acquired, have been
identified. Age is a substantial risk factor; as it is unusual to develop pancreatic cancer before the
age of 40, the risk increases 40-fold by the age of 80 years.8 Other basic demographic factors that
confer an increased (but less than 5-fold) risk for pancreatic cancer are male sex, Ashkenazi
Jewish descent, and black race.9

Hereditary

Family history has long been known to be a risk factor for pancreatic cancer; 10% of
pancreatic cancers are proposed to be hereditary. A case control study from Canada found that
7.8% of patients with pancreatic cancer had a positive family history compared with 0.6% of
controls.10 A similar study performed in the United states found that having a first-degree family
relative with pancreatic cancer increases the risk by 3.2-fold compared with the general
population.11 Although patients with one or more first-degree relatives with the disease have a
higher rate of pancreatic cancers outside the head of the pancreas, they show no difference in
survival after resection than do patients with sporadic pancreatic cancer.12
Up to 20% of hereditary pancreatic cancers are associated with a known cancer syndrome,
including familial breast and ovarian cancer syndrome (BRCA 2 mutations), Lynch syndrome
(mismatch repair gene mutations), atypical multiple mole melanoma (p16 mutations), Peutz-
Jeghers (STK11 mutations), and hereditary pancreatitis (PRSS1 mutations).13,14 Mutations in
PALB2, a partner and localizer of BRCA 2, also increased susceptibility.15 There is another large
group of patients with familial pancreatic cancer who have 2 or more first-degree relatives with
the disease but whose underlying genetic defect is unknown, although the mechanism of
inheritance is most likely autosomal dominant.16,17

Other risk factors

Smoking
The major modifiable risk factor for pancreatic cancer is cigarette smoking, which increases
the risk by 1.5 to 5.5-fold.18,19 Approximately a quarter of the 27,000 pancreatic cancer deaths
that occur annually could be avoided by cessation of smoking according to a 1996 prospective
study.18 If added to another established risk factor, smoking can be even more detrimental. It
increases the cancer risk of individuals with hereditary pancreatitis from 54-fold to 154-fold and
results in the diagnosis of cancer at a younger age.20 Furthermore, risk increases with longer
length of exposure and amount smoked.21 Nicotine has been shown to induce pancreatic
carcinogenesis in mice, but the exact mechanism is unknown.22

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 109

Obesity
There is also evidence of a weak association between obesity and pancreatic cancer. A recent
case control study identified a positive association between increasing body mass index (BMI)
and pancreatic cancer risk, and an earlier meta-analysis found a slightly increased relative risk of
1.19 per unit increase of BMI in obese people vs people of normal weight.23 There seems to be an
interaction between obesity and physical activity, with greatest risk in those with low physical
activity.24

Diabetes
Type II diabetes mellitus increases pancreatic cancer risk by 80% with higher relative risk if
the duration of diabetes is greater than 5 years; 25,26 hyperinsulinemia alone can increase risk by
up to 100%.27,28

Pancreatitis
Chronic pancreatitis, a progressive irreversible inflammatory condition most commonly
caused by chronic alcoholism affecting between 0.04% and 5% of the general population, confers
up to a 26-fold risk of development of pancreatic cancer, especially if the pancreatitis is
hereditary.29,30
Additional studies investigating everything from coffee to sugar have found that Helicobacter
pylori and cystic fibrosis are associated with an above-normal risk of pancreatic cancer, and
higher dietary selenium, folate, and methionine intake are associated with a lower risk.31-34 The
increased risk associated with a recent history of peptic ulcer disease is likely because of
increased surveillance.35

Pathology

Pancreatic cancer develops either from the endocrine (islet cell) or exocrine (ductal or acinar
cell) systems of the pancreas. Pancreatic adenocarcinoma, which accounts for more than 85% of
pancreatic neoplasms, primarily arises from the ductal epithelial cells of the exocrine pancreas,
yet ductal cells make up only 10% of pancreatic cells and 4% of pancreatic volume.36,37 New
evidence shows that adenocarcinoma might even arise from an alternate route, via trans-
differentiation from acinar cells in genetic mouse models.38
The genetics of pancreatic carcinogenesis is becoming more well understood. The earliest
mutations in pancreatic carcinogenesis appear to be k-ras mutations and telomeric short-
ening.39,40 The sheer number of acquired mutations is impressive; an average of 63 genetic
mutations per tumor were found in a genetic analysis of more than 20 pancreatic cancers.41 The
most commonly activated oncogene is k-ras, whereas inactivated tumor suppressors include
p16, p53, and MADH4.42 A total of 95% of tumors are k-ras positive, most of which have
mutations in k-ras2.43
Studies have shown that it takes at least 10 years between a mutation initiation and tumor
formation and another 5 years before metastatic potential.44 The Notch signaling pathway has
been implicated in neurovascular progression, the epidermal growth factor (EGF) in mitotic
activity and cell adhesion, the vascular EGF pathway in angiogenesis, and the hedgehog pathway
in cellular proliferation and fibroblast stimulation.45-48 Downregulation of e-cadherins may
contribute to the invasive tumor spread and early metastasis in pancreatic cancer and correlates
with poor survival after resection.49 Another cell adhesion molecule called Ep-CAM is an
independent prognostic factor for overall survival and could be related to suppression of
pancreatic cancer cell activity.50 The role of mast cells in angiogenesis and tumor growth has
also been described.51
Several reasons for the poor prognosis in pancreatic cancer have been postulated. In addition
to being a biologically aggressive tumor, there are many opportunities for local invasion as
follows: a rich vascular supply (allowing for vascular invasion) and nerve supply (for perineural
invasion), multidirectional lymph drainage (contributing to lymphatic invasion), and the

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
110 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

relationship of the pancreas to the retroperitoneum and peritoneum (allowing continuous local
spread).52 The venous drainage of the pancreas is via the portal vein (PV), so the liver is often the
first hematogenous metastatic site. The most common site of involved lymph nodes is the
retropancreatic region, with a prevalence of nodal metastases as high as 78.6% in one study.53
The prevalence of diabetes in patients with pancreatic cancer is approximately 40%, and it is
often newly diagnosed.54 This could be because of altered peripheral insulin resistance, the
increased risk for cancer in patients with preoperative diabetes, or possibly a paraneoplastic
phenomenon.55 A peptide called adrenomedullin that causes insulin resistance in beta cells in
mice has been found to be upregulated at the gene level and protein level as well as in the
plasma of patients with pancreatic cancer.56 Insulin resistance improves after pancreatic cancer
resection.
There have been some developments in the identification of pancreatic cancer stem cells, a
subset of cells resistant to traditional therapy that may mediate metastasis and could be a special
target for cancer treatment.57,58 Additional research to identify marker proteins for pancreatic
ductal adenocarcinoma utilizing cell lines is underway.59 For example, DPC4 immunolabeling for
genetic status at the time of autopsy for pancreatic cancer is correlated with the presence of
widespread metastasis but not with locally destructive tumors.60 Adenocarcinoma of the
exocrine pancreas is also carcinoembryonic antigen positive.61 Although tissue plasminogen
activator is absent from the normal human pancreas, it is expressed in 95% of pancreatic
cancers.62 A monoclonal antibody called PAM4 also shows high specificity for pancreatic cancer
and its precursors.63

Premalignant lesions

In this review, we describe 3 types of preinvasive neoplasias—2 that are clinically detectable
and another that is not. Although there is some overlap between them, especially on the
microscopic level, they are seen as distinct entities on different pathways to carcinogenesis.
Premalignant cystic lesions of the pancreas can manifest with concurrent cancer or transform
into malignancies over time. The key to effective management is balancing the risks of watchful
waiting with the morbidity of operative intervention.64 The 2 major premalignant cystic lesions
of the pancreas are intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic
neoplasms (MCNs).
First described in 1982 in Japan with an increase in relative frequency over time, IPMNs account
for up to 20% of pancreatic resections at some centers. They are premalignant lesions characterized
by the intraductal proliferation of neoplastic mucinous cells, leading to the formation of papillary
structures or cystic masses and often to mucin (MUC) production. They involve the native ducts by
definition (main, branch, or mixed), as opposed to MCNs, which are a different entity. More than
80% are located in the head of the pancreas with a mean diameter of 4.5 cm.65 Male to female
ratios tend to be higher in Asia; most are diagnosed during the seventh decade of life.66 Although
IPMNs have been associated with extrapancreatic neoplasms including gastric and colorectal
cancer, it is unknown how much of this is a result of surveillance bias.67,68 A recent multicenter
European study found no increase in frequency of extrapancreatic malignancies.69
IPMNs vary in level of aggressiveness and lie on a spectrum from benign adenoma to
carcinoma. They exhibit less genetic alterations than conventional ductal adenocarcinomas and
are less likely to exhibit k-ras, p16, or p53 genetic mutations, although one half of IPMNs are k-ras
positive.70 Invasive carcinoma may develop anywhere in the gland, although rates of malignancy
differ based on location, with main-branch IPMNs more likely than side-branch IPMNs to lead to
malignancy. The reported prevalence of malignancy for main-branch IPMNs ranges from 57%-
92% vs 6% to 46% in branch type, so smaller branch-type lesions without suspicious radiologic
features are usually watched.71-73 Studies have found that 3 cm marks the point of increased risk
of malignant change.74 Thus, per Sendai Consensus Guidelines, stable, asymptomatic side-
branch IPMNs less than 3 cm in diameter can be observed, and all main-branch IPMNs should be
resected.75,76 Recently, the criteria for branch-types have become controversial, as there are

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 111

reports of Sendai-negative IPMNs that have turned malignant.77 The literature reports that the
sensitivity of these criteria ranges from just 75%-97%, allowing for a potentially high false-
negative rate.78,79 More recently in 2012, a set of international consensus guidelines was
published that advocates for the resection of branch-type cysts that are symptomatic or have a
diameter 4 3 cm, have enhanced walls, mural nodules, main pancreatic duct dilation 45 mm,
an abrupt change in main pancreatic duct diameter with distal pancreatic atrophy, or positive
cytology or lymphadenopathy.80 Other investigators have identified the presence of a mural
nodule 4 5 mm and pancreatic juice carcinoembryonic antigen level of 4 30 ng/ML as clinical
factors that help identify high-risk malignant transformation in branch-duct type IPMN.81
The malignant potential of mixed-type IPMNs appears to be similar to that of main-duct.82,83
Levy and colleagues84 reported the risk of development of high-grade dysplasia or invasive
carcinoma at 2 years and 5 years for the branch-duct type at 9% and 15% respectively, and the
risks for the main-duct type and mixed-type at 58% and 63%, respectively. In patients who do
undergo surveillance for mixed-type IPMN, diffuse main pancreatic duct dilation, elevated
serum CA 19-9 levels, elevated serum alkaline phosphatase, and lack of extrapancreatic cysts
predict with 98% accuracy the likelihood of malignant progression over 5 years.
There are 4 subtypes of IPMN as follows: gastric, intestinal, pancreaticobiliary, and oncotic.
Intestinal type is the most common, and its invasive form is similar to colloid carcinoma, which
is notably less aggressive than conventional ductal adenocarcinoma.85 MUC staining of
pancreatic juice cytology has been used to differentiate the gastric (low malignant potential)
and intestinal (high malignant potential) subtypes.86
Ductal adenocarcinomas that do develop in the setting of IPMN appear to have a different
natural history—one with a better prognosis. In a study of patients who had surgery for invasive
adenocarcinoma arising in association with IPMN, patients presented at an earlier stage than
classical adenocarcinoma.87 The cancers may also differ on a molecular basis, which could be
related to better outcomes; there is near universal expression of a nuclear transcription factor
called DPC4 in IPMNs, which is only found in one half of ductal adenocarcinomas.88
Surgical resection for noninvasive IPMN has been associated with 5-year disease-free survival
rates of up to 100% compared with 40%-60% survival for the invasive type.74,89 However, IPMN
frequently recurs even after resection.90 A prospective cohort study with a median follow-up of
44.4 months found that an overall recurrence rate of 10.7%.91 Even after resection for
noninvasive IPMN, a study found that the risk of developing a new IPMN was 25%, of requiring
surgery was 14%, and of developing invasive pancreatic cancer of 7% (all at 5 years).92 Therefore,
close surveillance is recommended even after resection for noninvasive types.
MCNs, which occur predominantly in women and contain an ovarian-type stroma, are less
common and have been less genetically characterized.93 However, they too have malignant
potential and should be referred for surgical resection.94 These solitary lesions are typically
found in the body and tail of the pancreas and lack communication with the pancreatic
duct.95 Noninvasive lesions have minimal risk of recurrence and do not need surveillance
after resection, but those which are invasive have a recurrence rate between 37% and 83% at
5 years.96,97 Resected noninvasive MCN portends a 5-year survival rate of 94.7%-100% and
invasive MCN of 57%-62.5%.97,98 Imaging and pathology from a large MCN are shown in
Figure 1.
The microscopic form of dysplasia called pancreatic intraepithelial neoplasia (PanIN) is
considered the pancreatic counterpart to the epithelial dysplasia seen in other disease
systems.70 They represent stages in neoplastic growth, cyto-architectural changes, and
accumulation of genetic mutations from low grade (PanIN1A) to carcinoma in situ or PanIN3
(Fig 2).99,100 PanIN1 lesions are present in approximately one-third of the elderly population and
rarely become malignant, whereas PanIN3 lesions are direct precursors to cancer and are
commonly found in association with existing invasive carcinoma.101 Certain subpopulations of
PanINs are seen as morphologically distinct and may be able to provide molecular targets for
therapy.102 Assessment of pancreaticoduodenectomy (PD) specimens shows that adenocarcino-
mas more often arise in association with PanINs than from MCNs or IPMNs, possibly because of
higher likelihood of discovery of the latter lesions at a noninvasive stage.103

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
112 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

Fig. 1. (A) Axial CT scan image showing large infected mucinous cystic neoplasm of the pancreas causing diffuse
abdominal pain, fever, and hypotension. (B) Coronal CT scan image showing septations. (C) Pathology specimen after en
bloc distal pancreatectomy and splenectomy. (D) Pathology specimen of cystic componenents. Found to have high-grade
dysplasia. (Color version of figure is available online.)

There have been experimental attempts to interfere with the process of neoplastic
transformation in vivo. Fendrich and colleagues104 used genetically engineered mouse models
to show that aspirin and enalapril could delay the progression of PANIns. Interestingly, the
presence of PANINs at an R0 resection margin does not affect survival.105

Diagnosis

The early forefathers of pancreatic surgery recognized the importance of timely diagnosis.
William McCune, who performed the first of endoscopic retrograde cholangiopancreatography
(ERCP), acknowledged in 1968, “the notoriously poor results of surgical treatment of malignant
lesions of the pancreas and the extreme importance of early detection of cancer in this deep-
seated organ.”2 Early detection remains a challenge up to the present day.

Presentation

Symptoms of pancreatic cancer differ based on the location of the tumor (head, body, or tail)
as well as its size and stage. Most cancers result in obstruction of the pancreatic duct distal to the
tumor, often resulting in pancreatitis, and some in the head of the pancreas also result in
common bile duct obstruction and obstructive jaundice with corresponding elevations in
alkaline phosphatase and bilirubin levels. Approximately one half of patients with tumors in the
head of the pancreas present with upper abdominal pain or back pain.106 The abdominal pain is
caused by invasion of the celiac and superior mesenteric plexuses and becomes more localized
in later stages; radiation of pain to the back is associated with retroperitoneal invasion of the

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 113

Fig. 2. Progression of precursor PanIn stages to invasive cancer. (Reprinted with permission from Chang DK, Merrett ND,
Biankin AV, NSW Pancreatic Cancer Network. Improving outcomes for operable pancreatic cancer: is access to safer
surgery the problem? J Gastro Hepatol 2008;23:1036-45.) (Color version of figure is available online.)

splanchnic nerve plexus.107 Other symptoms include nausea, anorexia, weight loss, and new-
onset diabetes. Choluria (dark urine) and hypocholia (clay-colored stool) occur in 60% and 54% of
patients, respectively. Classic signs of pancreatic cancer, Courvoisier’s sign (palpable gallbladder)
and migratory thrombophlebitis, are only seen in 13% and 3% of patients with pancreatic
cancer, respectively.108 The ambiguity and absence of early symptoms contributes to delay in
diagnosis.

Screening

Owing to low lifetime risk of pancreatic cancer (1.3%), routine screening is not recommended
in an asymptomatic person in the general population.109 However, regular surveillance is
recommended for patients with at least a 5-fold increase in pancreatic risk; this includes
patients with a strong family history, patients with known genetic mutations, and combinations
of the 2. Patients with at least 3 first-degree family members with pancreatic cancer (who have
an estimated 32-fold increased risk) or 2 first-degree relatives (a 6.4-fold increased risk) should
be screened.110 An individual with 2 affected blood relatives, one of whom is a first-degree
relative, is also at increased risk and is a candidate for screening at the discretion of the provider.
Patients with Peutz-Jeghers syndrome have up to a 132-fold increased risk and should be
screened regardless of family history.111 BRCA2 carriers with at least 1 first-degree relative or 2
or more affected blood relatives should also be screened. Patients with PALB2/p16 mutations or
Lynch syndrome who have 1 or more affected first-degree relatives should be screened.9
Most experts agree that screening should start at the age of 50 years for most high-risk
individuals, except patients with hereditary pancreatitis who should begin at the age of
40 years.112 Endoscopic ultrasound (EUS), magnetic resonance imaging (MRI), or magnetic
resonance cholangiopancreatography (MRCP) are the most recommend imaging modalities. In a

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
114 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

Dutch prospective multicenter study of high-risk individuals undergoing first time screening,
EUS or MRI or both detected clinically relevant lesions in 6% of the population with 55%
agreement in detection of clinically relevant lesions.113 EUS was particularly sensitive to the
detection of small solid tumors and MRI to cystic lesions. The study implies that the modalities
are complementary rather than interchangeable in the screening setting. The optimal interval
for surveillance remains unknown, although 73.5% of participants in the International Cancer of
the Pancreas Screening Consortium in 2013 reported yearly surveillance.112 The reported rate of
cystic lesions in patients undergoing screening because of suspicion of hereditary pancreatic
cancer is as high as 40%.

Serum markers

As the only serum biomarker for pancreatic cancer approved by the Food and Drug
Administration, carbohydrate antigen 19-9 (CA 19-9, also known as sialyl Lewis-a) is useful for
detection of disease burden but is not utilized independently for diagnosis or screening because
of suboptimal sensitivity and specificity (approximately 80% and 73%, respectively.)114-116 First
identified in 1979, it is an aberrant glycoprotein expressed by cancer cells that contains 1 fewer
sialyl acid residues than its normal counterpart.
The prognostic value of CA 19-9 is widespread over the course of the disease. Its level is
associated with resectability—a median level of o100 U/mL has a positive predictive value (PPV)
of 60%-80% and 4100 U/mL a PPV for unresectability of 88%-91%.117,118 A low preoperative CA
19-9 has been linked with improved survival (levels o37 U/mL with a median survival of 22-40
months vs 7-30 months). Drops in postoperative levels and levels 437 U/mL after resection are
also associated with prolonged survival.119 Even a fall of 20% from baseline levels after 1 year in
unresectable cancer is an independent predictor for overall survival.120 Conversely, elevations in
the postoperative period are associated with likely recurrence.121
However, CA 19-9 has several limitations as a biomarker. It is only expressed in patients with
positive Lewis phenotypes and can therefore result in a false-negative in the 5%-10% of the
population who do not express the Lewis phenotype.122 It can also be falsely positive (10%-60%
of the time) in the setting of obstructive jaundice as well as in other gastrointestinal (GI) cancers
(colorectal, esophageal, and gastric) and benign hepato-pancreatico-biliary illnesses, including
pancreatitis, hepatitis, and cholangitis.118
Attempts to identify a superior serum biomarker for pancreatic cancer are underway. A 2015
study detected levels of a protein called Glypican-1 exclusively in exosomes derived from cancer
cell lines. Although it can also be elevated in breast cancer, this serum marker had a sensitivity
and specificity of 100% in detecting pancreatic cancer of all stages in humans and was able to
distinguish cancer from benign pancreatic disease. Furthermore, serum detection of the
biomarker occurred before detection of the cancerous lesion by MRI in genetically engineered
mouse models, presenting the prospect of early detection.123

Diagnostic imaging

Most pancreatic cancers are diagnosed via helical computed tomography (CT) scan, although
MRI has a near equal ability to detect tumors.124 Because the normal pancreas enhances better
than the cancer, tumors appear hypodense compared with the adjacent normal pancreas. The
pancreas distal to tumor can also appear atrophic.125 CT scans have increased difficulty detecting
lesions smaller than 2 cm, although a recent study found a sensitivity of 77% and specificity of
100% for the detection of pancreatic masses 2 cm in size by triple-phase helical CT.126

Endoscopy

The general need for a pretreatment tissue diagnosis is in general controversial but is
mandatory before the initiation of neoadjuvant therapy.125 EUS-guided fine-needle aspiration

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 115

(FNA) has the highest overall diagnostic accuracy of any modality, especially for smaller tumors,
and has a complication rate of only 2%.127,128 A meta-analysis of 33 studies assessing the
accuracy of EUS-guided FNA found a pooled sensitivity of 85% and specificity of 98% for
malignant cytology.129 When combined with a nondilated pancreatic duct, the negative
predictive value of FNA can be as high as 100%.130 The sensitivity and specificity of ERCP in the
diagnosis of pancreatic cancer are 70% and 94%, respectively.

Staging and resectability

The ultimate purpose of staging is to determine the surgical resectability of the cancer. Therefore,
it is critical to accurately assess the tumor’s size and location within the pancreas, potential vascular
involvement or the presence of any aberrant vascular anatomy, and the presence of metastasis. The
most commonly used staging system in the United States was developed by the American Joint
Committee on Cancer and utilizes a TNM-based classification (Table 1), most recently updated in
2010 with the seventh edition.131 These stages have been found to correlate with prognosis and
survival, although the addition of a grade component may improve prognostication.132
Resectability criteria for pancreatic cancer divide nonmetastatic resectability status into
3 groups as follows: (potentially) resectable, borderline resectable, and locally advanced. There is
still variable language and some controversy regarding the definition of borderline resectable
disease, but the level of disagreement has abated over time. In Table 2, we present the now
subtle differences between the 2015 National Comprehensive Cancer Network (NCCN)

Table 1
AJCC pancreatic cancer staging, 7th edition. (Adapted from AJCC Cancer Staging Manual, S.B. Edge, D.R. Byrd, C.C.
Compton, A.G. Fritz, F.L. Greene, A. Trotti (eds.) (7th ed) Springer, New York (2010).)

Primary tumor (T)

TX Primary tumor cannot be assessed


T0 No evidence of primary tumor
Tis Carcinoma in situ (including PanIN3)
T1 Tumor r2 cm in diameter
T2 4 2 cm in diameter
T3 Extends beyond pancreas without involvement of celiac axis or SMA
T4 Involves celiac axis or SMA

Regional lymph nodes (N)

NX Cannot be assessed
N0 No metastasis
N1 Metastasis

Distant metastasis (M)

M0 No distant metastasis
M1 Distant metastasis

Stage T N M

Stage 0 Tis N0 M0
Stage IA T1 M0 M0
Stage IB T2 N0 M0
Stage IIA T3 N0 M0
Stage IIB T1 N1 M0
T2 N1 M0
T3 N1 M0
Stage III T4 Any N M0
Stage IV Any T Any N M1

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
116 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

Table 2
Comparison between National Comprehensive Cancer Network (NCCN) version 2.2015 and the Intergroup Trial
definitions of borderline resectable pancreatic adenocarcinoma in the head or uncinate process of the pancreas.
(Reprinted with permission from Katz et al.134)

National Comprehensive Cancer Network Intergroup Trial 2013


(NCCN) Version 2.2015

SMV-PV Solid tumor contact of 4 1801 or contact of Interface between tumor and vessel of Z1801
r 1801 with venous contour irregularity or around vessel circumference and/or
thrombosis but with suitable vessel proximal reconstructable occlusion
and distal, allowing for safe resection and vein
reconstruction
SMA Solid tumor contact of r1801 Interface between tumor and vessel of o1801
around vessel circumference
CHA Solid tumor contact without extension into Reconstructable, short-segment interface
celiac axis or hepatic artery bifurcation
allowing for safe/complete resection and
reconstruction
Celiac Trunk — Interface between tumor and vessel of o1801
around vessel circumference
IVC Solid tumor contact —

Adapted with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guideliness) for Pancreatic
Adenocarcinoma V.2.2015. s 2015 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN
Guideliness and illustrations herein may not be reproduced in any form for any purpose without the express written
permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines, go online to NCCN.org.
NATIONAL COMPREHENSIVE CANCER NETWORKs, NCCNs, NCCN GUIDELINESs, and all other NCCN Content are
trademarks owned by the National Comprehensive Cancer Network, Inc.
SMV, superior mesenteric vein; PV, portal vein; SMA, superior mesenteric artery; CHA, common hepatic artery; IVC,
inferior vena cava.

guidelines and the Intergroup trial guidelines (from 2013) for borderline resectable pancreatic
cancer.133,134 Although NCCN guidelines previously used subjective terms such as “abutment” and
“distortion,” the most recent guidelines specify the degree of contact between tumor and vessel.
Per the NCCN, in addition to no distant metastasis, clearly resectable tumors should have no
radiographic evidence of superior mesenteric vein (SMV) or PV distortion or less than 1801 of
contact without vein contour irregularity and have no contact with the celiac axis, common
hepatic artery (CHA), and superior mesenteric artery (SMA) (Fig 3). Similarly, the Intergroup
guidelines specify that the interface between tumor and veins should be less than 1801 around
the vessel to be considered resectable but make no specifications regarding venous contour.
According to the NCCN, borderline resectable tumors (Fig 4) can have solid tumor contact with
the SMV or PV of greater than 1801 or less than or equal to 1801 with contour irregularity or
thrombosis but with suitable remaining vessel for safe vein reconstruction. Inferior vena cava
contact is also considered borderline resectable. The Intergoup guidelines specify as borderline
resectable an SMV or PV interface of greater than 1801 between tumor and vessel or a
reconstructable occlusion. Both paradigms allow for solid tumor contact with the SMA of less
than 1801 or “short-segment” CHA contact that the NCCN specifies as not extending into the
celiac or hepatic artery bifurcation. An interface with the celiac axis of less than a 1801
circumference of the vessel wall is considered borderline resectable by the Intergroup guidelines;
this is not explicitly stated in the NCCN guidelines. Although the NCCN outlines criteria for
unresectability based on tumor location within the pancreas, these criteria align for the most part
with the Intergroup definitions of locally advanced disease: any solid tumor contact with the SMA
or celiac of 4 1801 makes the tumor unresectable. The NCCN notes that for tumors of the head or
uncinate process, tumor contact with the first jejunal branch of the SMA or the most proximal
jejunal branch to the SMV also makes tumors unresectable, as does tumor contact with the CA
and aortic involvment for tumors of the distal pancreas. Attainment of negative margins is rarely
feasible in the setting of locally advanced disease (Fig 5), so the great majority are unresectable
even after neoadjuvant treatment. Metastatic (stage IV) disease also precludes resection.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 117

Fig. 3. CT scan demonstrating resectable pancreatic cancer with a clear fat plane between the tumor and the portal vein.
(Courtesy of Quyen D. Chu, MD, MBA.)

Radiologic

Staging is primarily accomplished by CT although centers vary in their algorithms and may
include positron emission tomography (PET), MRI, or MRCP in addition, although they have no
clear advantage over CT.135
Dedicated pancreas protocol multidetector CT (MDCT) with submillimeter section thickness
and image reconstruction is recommended and can assess the extent of vessel involvement, nodal
involvement, and distant metastasis.136 Nonionic iodinated contrast is used, and patients are
scanned both in the arterial phase (for opacification of celiac axis and SMA) and the venous phase
(for PV, SMV, and splenic vein) in which the cancerous lesion appears hypodense. In addition,
low-density oral contrast agent is given for adequate distension of the GI tract.125 Single-center
studies have estimated the sensitivity and specificity of MDCT for predicting resectability at 96%
and 33%, respectively.137 Although CT has a high predictive value of unresectability of 90%-100%,

Fig. 4. CT scans demonstrating borderline pancreatic cancer with reconstructable involvement of the porticomesenteric
axis. On the left, the tumor adheres to and partially occludes the portal vein. On the right, the tumor encases less than
1801 of the superior mesenteric vein. Both patients received a successful pancreaticoduodenectomy following
neoadjuvant therapy. (Courtesy of Quyen D. Chu, MD, MBA.)

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
118 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

Fig. 5. CT scans showing locally advanced pancreatic cancer encasing the common hepatic artery and the celiac trunk.
This patient was unresectable and received chemoradiation. (Courtesy of Quyen D. Chu, MD, MBA.)

it has a lower predictive value of resectability of 76%-90%, which is usually because of its inability
to detect tiny liver metastasis or minimal peritoneal spread.125 When compared with
intraoperative findings, CT has an accurate prediction of margin-negative resection rate 73% of
the time.138 Of note, CT only has an accuracy of 73% in the assessment of peripancreatic lymph
nodes in resectable pancreatic cancer compared to detailed histopathologic assessment.139
Does the accuracy of MDCT change after neoadjuvant therapy? A 2006 study from MD
Anderson assessed accuracy of MDCT for staging of pancreatic cancer in the setting of
neoadjuvant therapy and found that the negative predictive value (74%) for identifying
unresectable pancreatic cancer was comparable with previously reported data (45%-79%) in the
absence of neoadjuvant therapy.140
Additional imaging is of varying utility. Diffusion-weighted MRI was shown to change manage-
ment in 12.9% of patients and allowed for better detection of liver metastasis.141 Studies examining the
ability of fluorodeoxyglucose (FDG)-PET to enhance staging for patients deemed resectable by routine
methods have found that management was altered by the discovery of unsuspected distant disease in
13% to 16% of patients, respectively.142,143 However, the cost and benefit ratio of FDG-PET for pancreatic
cancer staging has been questioned, and the routine use of this modality remains controversial.144
Standardized reporting of radiology findings has been advocated by several professional
societies, with specific guidelines for terminology. A consensus statement released in 2014
suggested that radiologists specifically denote the presence of venous thrombosis, degree of
extension into the CHA, SMA, and SMV regarding branches, presence of attenuation in vessel
contact, and the presence of arterial variants.136

Surgical

Bernheim145 first performed an early “abdominal organoscopy” before an exploratory


laparotomy for a carcinoma in the head of the pancreas in a patient of William Halsted to

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 119

corroborate the absence of liver metastasis. As recent improvements in the quality of diagnostic
imaging have caught up with laparoscopic techniques, the role of diagnostic laparoscopy for
pancreatic cancer staging has been contentious.
We know that approximately 10%-25% of patients undergo unnecessary laparotomy owing to
the discovery of unresectable disease upon surgery.146 In a study performed at MGH, CT-occult
metastasis was discovered in almost 24% of patients undergoing laparoscopic staging, although
there were a substantial number of patients with locally advanced disease included.147 Another
study suggested that laparoscopic staging could change surgical management in up to 35% of
patients already assessed by high-resolution helical CT but also had a 14.3% false-negative
rate.148 Similar experiences have been reported at other centers, with laparoscopic examination
detecting approximately 82% of unsuspected distant metastasis.149 Some lesions are still missed
on laparoscopy, however, including those on the posterior liver surface, the paraduodenal
retroperitoneum, the jejunal mesentery, and the lesser sac.150 In one study, occult peritoneal
metastases were actually more likely to be detected in patients with potentially resectable
tumors that were explored via laparoscopy than via laparotomy (32% vs 10%, P ¼ 0.018).151
A Cochrane review of 15 studies published in 2013 found a 40% probability that a patient’s
pancreatic cancer would be unresectable after diagnostic laparoscopy and CT scan, compared
with a 17% probability for those receiving CT alone. The study estimates that on average, using
diagnostic laparoscopy with histological confirmation of suspicious lesions would avoid 23
unnecessary laparotomies for every 100 patients.152 In the case of staging laparoscopy after MRI,
given the low rate of missed metastases on imaging, the addition of laparoscopy has poor cost-
effectiveness and low marginal yield.153
Aside from the well-known risks of general anesthesia and laparoscopic surgery, staging
laparoscopy has no additional risks specific to the patient with cancer. It has not been found to
increase the occurrence of trocar-site disease or peritoneal disease progression of pancreatic
cancer.154 It is usually performed at the start of a planned resection, instead of as a separate
procedure. Routine use of peritoneal lavage is not currently recommended for determination of
resectability, as studies have found that patients with positive peritoneal cytology alone can still
achieve favorable long-term survival.155,156
Laparoscopic ultrasound has been suggested as a useful adjunct to laparoscopy. Without
significantly increasing morbidity or mortality, laparoscopic staging of pancreatic tumors with
the addition of ultrasound can reveal intraparenchymal hepatic metastases, small peritoneal
metastases, and critical retroperitoneal tumor-vessel relationships with an accuracy approach-
ing that of an open approach.157 However, much of the data on the utility of laparoscopic
ultrasound are derived from single-center studies in variable populations before the era before
high-quality MDCT.158 It is not currently recommended for routine use and should be limited to
cases with equivocal resectability or suspected but unconfirmed metastasis.
CA 19-9 can also be used as a guide. Levels below 100 U/mL predict low probability of
metastatic disease; in those patients, laparoscopy can be spared. In contrast, patients with
elevated CA 19-9 have an increased probability of metastatic disease and may benefit from
diagnostic laparoscopy.159 CA 19-9 levels have also been strongly associated with subradio-
graphic unresectable disease.
Summarized in an expert consensus statement written by Callery and colleagues in 2009,
staging laparoscopy should be used selectively. High-yield predictors include pancreatic head
tumors of at least 3 cm, tumors of the pancreatic body and tail, equivocal CT findings, and CA 19-
9 levels higher than 100 U/mL. Finally, staging laparoscopy may be used to rule out undetected
metastatic disease to optimize treatment selection for patients with locally advanced
unresectable cancer.125

Endoscopic

EUS can provide information to aid staging, including detection of involvement of mesenteric
vasculature, nearby organs, or lymph nodes.160 A 2013 meta-analysis of performance

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
120 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

characteristics for EUS found a 69% sensitivity and 81% specificity for nodal staging, 85%
sensitivity and 91% specificity for vascular invasion, and a 90% sensitivity and 86% specificity for
resectability.161 However, the sensitivity of EUS declines to 73% in the setting of chronic
pancreatitis.162

Primary therapies

Although surgical resection is the only management option leading to a potential cure, the
grim median survival of 11-20 months after resection alone demonstrates the necessity of
optimally sequenced multimodal treatment.163 The rates of multimodal treatment are still lower
than they should be, but they are rising. In Medicare patients, the use of multimodal therapy has
nearly doubled from 7% in the 1990s to 14% in the 2000s, with chemotherapy delivered
postoperatively in more than 90% of cases and preoperatively in less than 10%.164

Neoadjuvant therapy

As pancreatic cancer is believed to be a systemic disease with a narrow therapeutic window,


the prompt use of systemic therapy has theoretical benefits.165 Neoadjuvant (or preoperative)
therapy is increasingly being utilized to treat occult disease as early as possible, downstage
advanced tumors, and optimize patient selection for surgery.
Critics of the neoadjuvant strategy express concerns regarding tumor progression, given that
a decade ago only 45%-74% of initially resectable patients were subsequently resected after
induction chemoradiation and 38%-70% after induction chemotherapy.166 However, the
avoidance of ineffective surgical resection in patients with aggressive tumors who are at high
likelihood of early metastasis or recurrence can be seen as an advantage. A more recent meta-
analysis from 2010 showed that pooled resectability was 73.6% in initially resectable patients
after neoadjuvant therapy; a reasonable resectability rate of 33.2% was also achieved in initially
borderline or nonresectable patients.167
Chemoradiation delivered before surgery may be more effective than that delivered after. In a
recent report of 40 patients with borderline resectable disease receiving neoadjuvant therapy,
only 10% experienced local recurrence and 9% peritoneal recurrence.168 A prospective study in
locally advanced, unresectable patients showed that a neoadjuvant regimen of gemcitabine,
docetaxel, and capecitabine for those with venous involvement followed by gemcitabine and
capecitabine or radiation therapy (RT) in those with arterial involvement resulted in higher rates
of R0 resections and prolonged overall survival.169
There has been no reported increase in surgical complications following neoadjuvant
therapy.166,170,171 In a retrospective investigation of completion of multimodal therapy at a high-
volume center, although the rates of postoperative complications were similar for patients
receiving neoadjuvant therapy vs up-front surgery, these complications affected overall survival
only for the up-front surgery group.172
Neoadjuvant therapy is currently recommended for the treatment of borderline resectable
pancreatic cancer, but this strategy is not universally practiced; moreover, its benefit for
resectable patients remains a highly controversial question.173 Accordingly, most patients with
resectable disease who receive neoadjuvant therapy are enrolled in a clinical trial or are seen at a
number of specific cancer centers.
Only a randomized control trial (RCT) attempting to evaluate neoadjuvant vs up-front surgery
in resectable patients has been conducted thus far, and in addition to suffering from problems
with standardization of staging and surgery, it was underpowered to detect a difference in
survival.174 Therefore, the evidence in favor of neoadjuvant therapy is derived from
retrospective, prospective, and nonrandomized trials, as well as decision analysis models. A
summary of select nonrandomized phase II trials of neoadjuvant therapy is provided in Table 3.
Studies comparing survival between neoadjuvant treatment and up-front surgery are difficult
to interpret because of inherent selection bias, but patients (regardless of initial stage) receiving

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 121

Table 3
Selected nonrandomized phase II trials of neoadjuvant chemotherapy. (Adapted with permission from de Geus, et al.
Neoadjuvant therapy vs up-front surgical strategies in localized pancreatic cancer: a Markov decision analysis.
(unpublished).

Study Patients Inclusion Institution, Country Treatment regimen Median overall


period survival (months)

Talamonti et al450 20 2002-2003 Feinberg School of EBRT and 26 (Resected)


Medicine and gemcitabine
Northwestern University,
USA
Evans et al171 86 1998-2001 University of Texas M.D. EBRT and 34 (Resected)/
Anderson Cancer Center, gemcitabine 7 (unresected)
USA
Heinrich et al451 25n 2001-2007 Swiss Hepato-Pancreato- EBRT, gemcitabine 19.1 (Resected)
Biliary Center, Switzerland and cisplatin
Le Scodan et al452 41 1998-2003 Centre Hospitalier Lyon- EBRT, 5-FU, and 9.4 (all)/
Sud, France cisplatin 11.7 (surgery†)
Turrini et al453 34 2003-2005 Insitute Paoli-Calmettes EBRT and docetaxel 32 (Resected)/11
and Universite de la (unresected)
Mediterrannee, France
454
Pipas et al 33 2005-2008 Darmouth-Hitchcock IMRT, cetuximab, 24.3 (Resected)/10
Medical Center, USA and gemcitabine (unresected)
Takahashi et al455 110 2002-2009 Osaka Medical Center for EBRT and Overall 5-year
Cancer and Cardio- gemcitabine survival 56%
vascular Disease, Japan
Van Buren et al456 59 2007-2011 University of Pittsburg EBRT, gemcitabine 16.8 (All)/19.7
School of Medicine, USA and bevacizumab (resected)
457
Kim et al 68 2007-2010 University of Michigan, EBRT, gemcitabine, 27.1 (Resected)/
USA and oxaliplatin 10.9 (unresected)
Motoi et al458 35 2008-2010 Tohoku University, Japan Gemcitabine and S1 34.7 (Resected)/10
(unresected‡)
459
Hong et al 48§ 2007 - 2014 Massachusetts General EBRT and 17.3 (All)/27
Hospital, USA capacitabine (resected)
460
O'Reilly et al 38 2007-2011 Memorial Sloan-Kettering Gemcitabine and 27.2 (All)
Cancer Center, USA oxaliplatin
Takeda et al461 35 2002-2006 Kansai Rosai Hospital, EBRT ║ and 41.2 (Resected)/
Japan gemcitabine 10 (unresected)

n
25 with ductal adenocarcinoma of the pancreas of a total of 28.

Operated on with curative intent.

Resected with distant metastasis.
§
A total of 48 with pancreatic cancer of a total of 50.

Accelerated hyperfractionated radiotherapy.
EBRT, external beam radiation therapy; FU, fluorouracil; IMRT, intensity modulated radiation therapy.

neoadjvuant treatment usually survive longer. For resectable patients, the median survival after
completion of neoadjuvant therapy before resection is approximately 3 years, compared with
2 years for patients who complete adjuvant therapy after resection, and 1 year for patients who
receive resection alone.170,171 A retrospective single-center analysis found longer disease-free
survival and lower rates of local and hepatic recurrence after neoadjuvant therapy compared
with up-front surgery but no difference in overall survival from date of surgery.175
Decision analyses models have consistently found a slight survival benefit for neoadjuvant
therapy over up-front surgery.176 Unpublished results from a decision analysis in 2016
(including only resectable patients) show that compared to up-front surgery, neoadjuvant
therapy offered an improved life expectancy of 32.2 vs 26.7 months and quality-adjusted life
expectancy (QALE) of 25.5 vs 20.8 quality-adjusted life months. The neoadjuvant approach
provided a benefit in LE and QALE when the probability of resectability at surgery after
neoadjuvant therapy exceeded 57% and 55%, a resectability rate that is becoming increasingly
likely with developments in the effectiveness of chemotherapy and radiation. Another decision
analysis from 2015 that used data from prospective studies of patients with resectable or

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
122 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

borderline resectable cancer also favored the neoadjuvant approach with a median overall
survival of 22 months vs 20 months and QALE of 19.8 vs 18.4 months, respectively.177
Neoadjuvant chemoradiation followed by surgery may also be more cost-effective than a
surgery-first approach with a net of $10,247 saved per patient.178
Lessons from centers that provide neoadjuvant therapy include the importance of durable
stents (especially if regimen is prolonged), attention to nutritional support and hydration,
management of diabetes, and supportive care.179 Metal stents are superior to plastic stents for
patients undergoing neoadjuvant therapy, with a complication rate almost 7 times lower.180 PET
may be more accurate than CT in detecting histopathologic response after neoadjuvant therapy
in tumors with prior vascular involvement.181

Adjuvant therapy

Adjuvant chemotherapy (after resection) provides a substantial survival benefit over surgery
only.182,183 Even for localized disease, studies have shown improved survival (hazard ratio ¼
o 0.5) after surgery and adjuvant therapy vs surgery alone.184 However, there is a discrepancy
between treatment recommendations and practice. Although adjuvant therapy is recommended
for pancreatic cancer at all stages, one-third to two-thirds of stage I and II patients who
underwent resection in 2010 did not get adjuvant therapy.185 Lower rates of adjuvant therapy
are also seen in elderly individuals.186
The addition of adjuvant chemoradiation (CRT) in adjuvant therapy is hotly debated. RCTs
have shown conflicting results. The GITSG trial (only 43 patients) found that 5-FU-based CRT
followed by maintenance 5-FU provided a significant survival benefit over observation after
surgery, whereas the EORTC trial found no difference in survival between 5-FU-based CRT and
observation after surgery.187,188 Finally, the ESPAC-1 trial reported no short-term survival benefit
and poorer long-term survival for CRT vs chemotherapy alone.189 These RCTs evaluating the role
of CRT in adjuvant therapy have been criticized for design and methodological flaws as well as
problems with accrual, standardization of therapy, and the use of inferior radiation treatment
schedules.190 Overall, the American experience has shown a benefit to CRT in certain
populations (such as those with lymph node involvement); in contrast, CRT is not widely used
in Europe.191,192 At this time, the benefit of adjuvant chemoradiation remains unclear, and
further studies are needed to further elucidate the populations in which CRT may be beneficial. A
summary of select randomized trials of adjuvant therapy are shown in Table 4.
Planned adjuvant therapy may not take place because of a myriad of barriers including
patient preference, the presence of comorbidities, poor performance status, surgical complica-
tions, and early recurrence. Postoperative complications have been reported to prevent or delay
the receipt of adjuvant therapy in 25%-30% of eligible patients.193
The exact timing of adjuvant therapy does not seem to matter; there was no difference in
survival when therapy was started within 8 weeks of surgery vs up to 12 weeks later, according
to ESPAC-3 data.194 However, completion of adjuvant therapy confers a substantial survival
benefit. The 68% of patients who completed the full 6 cycles of chemotherapy had the best
overall and recurrence-free survival. Current guidelines recommend that adjuvant therapy take
place within 4-8 weeks of surgery, and restaging is recommended after each treatment modality

Chemotherapy

Gemcitabine, a pyrimidine analog introduced in 1996, is the primary chemotherapeutic agent


used in both the neoadjuvant and adjuvant setting. The NCCN Practice Guidelines for adjuvant
treatment recommend systemic gemcitabine, continuous 5-FU, 5-FU/leucovorin, or capecitabine
after resection or gemcitabine, 5-FU, or 5-FU/leucovorin either preceded or followed by
fluoropyrimidine- or gemcitabine-based chemoradiation.133 The CRT route is endorsed
particularly for patients who are at high likelihood of local recurrence.195

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.

Table 4
Randomized control trials of adjuvant therapy in pancreatic cancer. (Adapted with permission from de Geus, et al. Neoadjuvant therapy vs up-front surgical strategies in localized pancreatic
cancer: a Markov decision analysis. (unpublished).)

Study Patients Inclusion Institution, Country Phase Treatment arms Overall median Survival in
period months (P value)

Kalser et al187 43 1974-1982 Gastrointestinal Tumor Study Group (GITSG), III Obs. vs EBRT and 5-FU 10.9 vs 21 (0.03)
USA

M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154


Klinkenbijl 114n 1987-1995 European Organization for Research and III Obs. vs EBRT and 5-FU 12.6 vs 17.1 (NS)
et al188 Treatment of Cancer (EORTC)
Neoptolemos 541 1994-2000 European Study Group for Pancreatic Cancer III 5-FU and folinic acid vs EBRT and 5-FU 16.1 vs 15.5 (NS)
et al189 (ESPAC– 1), EU
Demols et al462 30 2001-2003 Erasme University Hospital, USA II EBRT and gemcitabine 19
Blackstock 47 1999-2003 Wake Forest University Comprehensive Cancer II EBRT and gemcitabine 18.3
et al463 Center, USA
Kosuge et al464 89 1992-2000 Japanese Study Group of Adjuvant Therapy for II Obs. vs Cisplatin and 5-FU 15.8 vs 12.5 (NS)
Pancreatic cancer (JSAP-1)
Regine et al182 388† 1998-2002 Radiation Therapy Oncology Group (RTOG-97-04) III EBRT and gemcitabine and 5-FU vs EBRT 20.5 vs 16.9 (NS)
and 5-FU
Yoshitomi 100 2002-2005 Chiba University Hospital, Japan II Gemcatabine vs Uracil/tegafur and 29.8 vs 21.2 (NS)
et al465 gemcitabine
Ueno et al466 118 2002-2005 Japanese Study Group of Adjuvant Therapy for II Obs vs gemcitabine 18.4 vs 22.3 (NS)
Pancreatic cancer (JSAP-2)
Van Laethem 90 2004-2007 EORTC-40013-22012/FFCD-9203/GERCOR, EU II Gemcitabine vs EBRT and gemcitabine 24.4 vs 24.3 (NS)
et al467
Neoptolemos 1088 2000-2007 European Study Group for Pancreatic Cancer III 5-FU and folinic acid vs gemcitabine 23.0 vs 23.6 (NS)
et al468 (ESPAC– 3), EU
Reni et al469 102 2003-2008 S. Raffaele Scientific Institute, Italy II Gemcitabine vs Cisplatin, Epirubicin, 5-FU 24.8 vs 28.9
and Gemcitabine
Schmidt 132 2004-2007 Ruprecht-Karls-University, Germany III 5-FU vs EBRT, 5-FU, cisplatin and 28.5 vs 26.5 (NS)
et al470 interferon-α-2B
Fenster et al471 73 2006-2008 Philipps—University of Marburg, Germany II Gemcitabine and cetuximab 22.4
Herman 48 2006-2012 Johns Hopkins University School of Medicine, USA II IMRT, erlontib, capecitabine 24.4
et al472
Oettle et al358 354 1998-2004 Charite Onkologie (CONKO–001), EU III Obs. vs gemcitabine 20.2 vs 22.8 (0.005)
Valle et al194 1088 2000-2007 European Study Group for Pancreatic Cancer III 5-FU and folinic acid vs gemcitabine 23.0 vs 23.6 (NS)
(ESPAC– 3), EU

5-FU, 5-fluorouracil; Obs, observation; NS, non-significant; EBRT, external beam radiation therapy; IMRT, intensity modulated radiation therapy.
n
114 patients with cancer of the pancreatic head of total 218 randomized.

123

388 patients with cancer of the pancreatic head of total 451 randomized.
124 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

No specific neoadjuvant regimens are recommended by the NCCN, but the most commonly
used agents are FOLFIRINOX (5-fluorouracil, oxaliplatin, irinotecan, and leucovorin), gemcitabine,
5-FU, mitomycin C, and platinum compounds. Almost one half of the neoadjuvant studies in a
recent meta-analysis used gemcitabine-based therapy, and radiotherapy with doses ranging from
24-63 Gy was applied in 93.7%.167 One strategy is the use of “induction chemotherapy” before
CRT, usually gemcitabine alone or in combination with other agents. Induction chemotherapy can
be used to “select” patients with no evidence of disease progression as suitable candidates for
subsequent CRT. 5-FU or gemcitabine are also often used as radiosensitizers, given at intervals
during radiotherapy.196
The efficacy of FOLFIRINOX in the setting of metastatic disease has led to its use as part of
neoadjuvant regimens for borderline resectable cases as well. For borderline resectable patients,
the period of induction neoadjuvant chemotherapy is longer (and is often followed by
chemoradiotherapy especially if vascular reconstruction is planned). In one high-volume center
experience, systemic therapy (FOLFIRINOX or nab-paclitaxel/gemcitabine) is initiated first (2
months) and is then followed by chemoradiation (gemcitabine-based, intensity-modulated RT)
in the setting of stable or responsive disease.197 At another center, systemic FOLFIRINOX therapy
can last up to 4 months.168 The median length of neoadjuvant therapy can be as long as
7 months at other centers.198,199 A meta-analysis of studies of FOLFIRINOX-based neoadjuvant
therapy in borderline resectable or unresectable patients found a 43% resectability rate and an
R0 resection rate of 39.4%. Particularly among the borderline resectable patients, a R0 resection
rate of 63.5% was achieved.200
Unfortunately, chemoresistance is often a problem. Although gemcitabine appears to be quite
effective as a cytotoxic agent in vitro, the same results are not seen in vivo; predictive models
have been created to enhance understanding of tumor cell behavior.201

Radiation therapy

RT is thought to promote locoregional control, as an association between RT and decreased


incidence of local recurrence has been observed.202 Radiation in various forms can be used both
in neoadjuvant and adjuvant regimens. The NCCN guidelines for radiation in adjuvant therapy
are 45-46 Gy in 1.8-2 Gy fractions to the tumor bed, anastomoses, and adjacent lymph nodes,
followed by another 5-9 Gy to the tumor bed and anastomoses. Recommended dose limitations
in the preoperative vs postoperative setting by organ are also provided.133
Recent studies have shown the safety of dose escalation of external beam RT (EBRT) up to
55 Gy, but several techniques have been developed that allow for the focused treatment of
diseased tissue with reduced dose exposure to normal tissue.203,204 Intraoperative radiotherapy
allows for the delivery of RT to the tumor bed under direct vision. This can be accomplished via
implantation of iodine-125 seeds, brachytherapy, or the more popular method of intraoperative
electron beam radiotherapy. Phase II trials of intraoperative electron beam radiotherapy in
locally advanced pancreatic cancer have shown median survivals between 9 and 13 months.
Intensity-modulated radiotherapy (IMRT) is a type of radiation delivered with varying intensities
within a field. A small study from Germany showed comparable outcomes for IMRT and
conventional 3D radiotherapy in the neoadjuvant setting.205 However, studies evaluating IMRT
in the adjuvant setting have shown varying levels of GI toxicity.206–208 Furthermore, IMRT
treatments are typically delivered daily for 6 weeks, and limits to the maximal dose may be
necessary because of tumor movement with respiration or intestinal distension.209 Stereotactic
body radiotherapy (SBRT) delivers intensified treatment to the tumor and surrounding
vasculature (in 1-5 high-dose fractions) with submillimeter precision and rapid falloff, thereby
sparing normal tissue.210 Because of the short treatment length (usually under a week), it is
convenient for patients and interferes minimally with timing of systemic therapy.211 It can be
used as a boost for conventional EBRT or IMRT or as primary radiation treatment. CyberKnife
(Accuray, Sunnyvale, CA) is a real-time image-guided robotic radiosurgery system used for
delivery of SBRT that tracks the tumor during respiration and movement using a combination of

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 125

previously implanted fiducial markers and an external camera. In sequence with induction
chemotherapy, SBRT facilitates negative resection margins in borderline resectable patients and
local control in unresectable patients.212 For resected patients, adjuvant SBRT can allow for
quicker initiation of chemotherapy.213
The toxicity for conventional photon-based radiotherapy is not trivial. The RTOG 9704 trial
reported a 59% incidence of grade 3 nonhematologic toxicity in patients receiving a dose of
50.40 Gy with 5-FU as adjuvant therapy.182 Similar rates were reported in the FFCD or SFRO trial
for unresectable patients receiving 60 Gy with 5-FU and cisplatin.214 Given these risks, the
potential risks vs benefits of chemoradiation must be weighed and decisions made for the
individual patient.
Some studies have suggested a reduction in normal tissue exposure for protons (ionizing
radiation) as opposed to photons.215 A phase I study demonstrated the feasibility of neoadjuvant
chemoradiation with 1 week of proton beam therapy and capecitabine followed by early surgery,
but a second series was halted because of unexpected intraoperative fibrosis.216,217

Targeted therapy and immunotherapy

On the horizon are new therapeutic strategies including “targeted therapy” aimed at specific
biologic pathways of tumor growth and immunotherapy, the stimulation of the immune system
to eradicate tumors.218 Several molecular targeted therapies, many in oral form, are being tested,
although their efficacy in pancreatic cancer is yet to be proven; cetuximab, an EGFR-targeted
therapy, as well as axitinib and bevacizumab, vascular EGFR inhibitors, and matrix metal-
loproteinase (MMP) inhibitors such as Marimastat have shown similar survival to Gem
monotherapy in advanced pancreatic cancer.219,220 Erlotibib, an EGFR inhibitor used in advanced
pancreatic cancer, is also being tested as a component of adjuvant therapy.221
Immunotherapy includes direct methods where immune-competent cells are stimulated to
attack cancer cells or indirect methods such as immunization where an immunogen stimulates
the body to inhibit the growth of tumors.222 Because many patients with pancreatic cancer have
known mutations, vaccines targeted against these tumor-specific mutations may provide a
benefit. Studies demonstrate that adjuvant vaccination with mutant K-ras, MUC1, or telomerase
peptides is safe and tolerable but their efficacy as immunogenic agents is not consistent in
human subjects.223-225 In particular, a combination of whole-cell vaccine-based immunotherapy
and chemotherapy shows promise.226 A single-center phase II study testing the efficacy of
granulocyte-macrophage colony-stimulating factor adjuvant immunotherapy with subsequent
chemoradiation in resected patients revealed a favorable median survival of 24.8 months.227 In a
Phase II RCT of an immunogen called Gastrimmune (G17DT) in patients with advanced
pancreatic cancer, responders to G17DT survived significantly longer.228 Other methods to
reverse immune suppression, including CD-40 antibodies, have shown tumoricidal properties
both in vitro and in vivo.229 The use of immunotherapy in cases of chemoresistance is also a
topic of increased interest.

Surgery

Preoperative considerations and preparation


Surgery is the cornerstone of therapy for pancreatic cancer and is the only hope for cure.
Efforts should be made to optimize patients for surgery to attain the best possible outcome, as
several preoperative factors such as malnutrition are independent risk factors for poor outcomes
following pancreatic surgery for cancer.230 Early identification and intervention is vital and
should be performed in a multidisciplinary setting. Screening tools including the subjective
global assessment and malnutrition screening, as well as preoperative measurement of albumin
and prealbumin, can help the provider identify patients who may benefit from additional
nutritional support.231

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
126 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

Evidence suggests that preoperative biliary drainage not be performed routinely and instead
be reserved for patients with coagulation disorders or those who would undergo neoadjuvant
therapy. Although the delay in surgery for preoperative biliary drainage makes no difference for
survival, there is also no discernable benefit to drainage seen on multiple studies.232 A
multicenter European trial (in which plastic stents were used) showed higher complication rates
with no improvement in perioperative outcomes, and a review showed no benefit (but increased
costs) with biliary drainage before surgery.233,234 However, another multicenter study found
increased odds of severe postoperative complications and decreased survival when preoperative
serum bilirubin levels exceeded 300 mmol/L, suggesting a benefit to biliary stenting in this
subset of patients with extreme jaundice.235
Finally, close attention to preoperative imaging also assists with operative planning. CT scans
can show the presence of anatomic anomalies, such as a replaced right hepatic artery. The need
for venous reconstruction should also be anticipated preoperatively to avoid the complications
associated with “technical misadventures” and delays in the operating room.236

Patient selection and risk calculators


Patient selection is paramount in surgery for pancreatic cancer, as patients are often elderly,
malnourished, and coagulopathic.237 Pancreatic resection in elderly adults can result in higher
rates of morbidity compared to younger individuals, but the survival benefit of resection does
not decrease with increasing age.238-241
Performance status can be described according to the Eastern Cooperative Oncology Group
scale as follows: 0 ¼ fully ambulatory, symptom free; 1 ¼ fully ambulatory but with symptoms;
2 ¼ in bed less than 50% of the day; 3 ¼ in bed more than 50% of the day; and 4 ¼ confined to
bed all day. Patients with poor performance status are typically not offered surgery because of
poor outcomes.242
Several prediction rules and risk calculators using preoperative characteristics have been
designed for pancreatic surgery. For stratifying risk of in-hospital mortality after pancreatic
resection for cancer, there is an integer-based risk score incorporating age group, Charlson score,
sex, type of pancreatectomy, and hospital volume status.243 Using the American College of
Surgeons National Surgical Quality Improvement Program (ACS NSQIP) database, there is a
prediction tool for probability of 30-day morbidity and mortality for patients undergoing PD.244
Variables include age, sex, body mass index, functional health status, comorbidities, as well as
laboratory values such as creatinine, albumin, platelets, and white blood cell count. A similar
tool using NSQIP for distal pancreatectomy (DP) has also been created.245 These calculators can
be used for preoperative counseling and for the optimization of perioperative care in high-risk
patients. Other risk calculators such as the Physiologic and Operative Severity Score for the
Enumeration of Mortality and Morbidity (POSSUM) and the Surgical Apgar score involve
operative variables and therefore cannot be used for preoperative risk stratification.246,247

Technique

Surgical strategies
A classic PD, also known as the Whipple procedure, is typically performed for
adenocarcinoma located in the pancreatic head, neck, or uncinate process. In its original form
as described by Whipple and colleagues,248 it was a 2-stage procedure beginning with biliary
and gastric outlet decompression followed a few weeks later by resection of the second part of
the duodenum and head of the pancreas. Along with technical advancements, the operation has
undergone various modifications that have improved its safety and widened the boundaries of
resectability.
Exclusion of metastasis via thorough examination of the abdominal cavity is first performed,
often via laparoscopy. Special attention is paid to the liver, omentum, and peritoneum.
Suspicious lesions are sent for frozen section. Resectability is determined by examination of the
extent of local and nodal tumor involvement. A Kocher maneuver is performed to mobilize the

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 127

Fig. 6. Reconstructions in a standard pancreaticoduodenectomy (A) and pylorus-sparing pancreaticoduodenectomy (B).


(Courtesy of Douglas B. Evans, MD.) (Color version of figure is available online.)

duodenum and head of the pancreas, exposing the underlying inferior vena cava and aorta. The
surgeon can then palpate the relationship to the SMA.249 SMV and PV involvement is no longer
prohibitive for resection, and certain levels of arterial reconstruction can be attempted on an
individual basis. Many centers that perform surgery within the context of multimodal therapy
do not consider nodal involvement an absolute contraindication to surgery and do not routinely
perform frozen sections of nodes before PD.236
In the classic Whipple procedure, the organs removed en bloc are the gastric antrum, the
duodenum, the proximal 10 cm of the jejunum, the head of the pancreas, the gallbladder, and
the distal common bile duct. After reconstruction, 3 anastomoses are created as follows: a
pancreaticojejunostomy (PJ), hepaticojejunostomy (HJ), and gastrojejunostomy (GJ).250 Figure 6
shows the resections completed in a classic Whipple and Figure 7 a typical pathologic specimen.

Fig. 7. Pathologic specimen from a standard pancreaticoduodenectomy. (Courtesy of Douglas B. Evans, MD.) (Color
version of figure is available online.)

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
128 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

An open PD is typically performed via upper midline or bilateral subcostal incision. For a
classic Whipple, the antrum is transected, and the gastroduodenal artery is divided near its
origin. The pancreatic neck is transected from the body of the pancreas anterior to the level of
the SMV or portal confluence.251 The pancreatic head is then dissected from the retroperitoneal
soft tissue and from the vascular bed of the SMV or portal confluence and SMA. The proximal
jejunum approximately 5-10 cm distal to the Ligament of Treitz is divided and the end brought
through the transverse mesocolon in a retrocolic manner to form the PJ. The anastomosis can be
created via the duct to mucosa technique that entails an inner layer of absorbable monofilament
suture connecting the duct to a small enterotomy in the jejunal mucosa and an outer layer of silk
suture joining the pancreatic capsule to the jejunum. Alternatively, a running anastomosis can
be performed connecting the whole gland to a larger jejunal enterotomy; the neck can be
invaginated 1-2 cm into the lumen of the bowel.249 The distal one-third of the CBD is transected,
and the remaining proximal segment is anastomosed to the jejunum about 10 cm distal to the PJ
in an end-to-side orientation to form the HJ anastomosis. Fifteen centimeters distally to the HJ,
an end-to-side GJ is performed. The GJ can be performed more distally in an antecolic fashion
as well.
In the pylorus-preserving PD (PpPD), the entire stomach as well as 2-3 cm of the proximal
duodenum are preserved, resulting in the creation of a duodenojejunostomy. Reductions in
complications including delayed gastric emptying, dumping, and anastomotic ulcers have been
reported, although a meta-analysis showed no difference in morbidity or mortality.252,253 Some
studies have also found reduced operative time, blood loss, and hospital stay for the PpPD.253,254
A Japanese RCT comparing resection of the pylorus ring with no resection of the pylorus ring
and preservation of the entire stomach found higher rates of delayed gastric emptying with
preservation of the pylorus.255 A lower rate of delayed gastric emptying has been reported for
antecolic (as opposed to retrocolic) reconstruction of the duodenojejunostomy in the PpPD.256
Frozen sections of the pancreatic and bile duct margins are performed with further resection
until negative. The surgeon should clearly document in the operative report the presence or
absence of a grossly complete resection. Avoidance of a positive margin at the peripancreatic
retroperitoneal tissue near the origin of the SMA is important, and it is precisely this location
where an R1 margin is most likely.257 The SMV-PV confluence must be mobilized for maximum
visualization of the SMA, and although sharp, ultrasonic, or thermal means of skeletonization are
acceptable, the use of a stapler to divide the uncinate from the SMA is not recommended,
as it leaves behind too much tissue.258 The “uncinate process first” (retrograde dissection of
the pancreatic head) and the “hanging maneuver” are two methods to attain negative
retroperitoneal margins.259,260
Because of its location, cancer of the body or tail of the pancreas is less likely to be
symptomatic in its earlier stages and therefore less likely to be resectable at presentation.261 In a
DP, the spleen and distal pancreas are mobilized and removed en bloc; the remnant pancreas is
oversewn proximal to any evidence of gross disease.249
No survival benefit has been reported for routine total pancreatectomy (additional removal of
entire pancreas, spleen, and margin of peripancreatic tissue) over the classic Whipple operation,
and it should only be performed if oncologically indicated.262–264 Likewise, an extended PD
(which involves dissection of the retroperitoneal and aortocaval lymph nodes) increases
morbidity without providing a survival benefit and is also not recommended.265,266

Extent of lymphadenectomy
Lymph node involvement is an important negative prognosticator for survival. Because
lymph nodes are embedded in the retroperitoneal adipose tissue, the assessment of lymph
nodes in pancreatic cancer is a joint venture between surgeons and pathologists.
Although current American Joint Committee on Cancer guidelines recommend the
examination of 12 nodes, some studies suggested that 15 lymph nodes should be examined
during PD for accurate staging of node-negative pancreatic cancer.267 A study using the
Surveillance, Epidemiology, and End Results (SEER) database found that many patients had an

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 129

inadequate number of lymph nodes examined and could therefore have been understaged
following pancreatic surgery; the median number of lymph nodes examined was 7, and
approximately 10% of patients resected between 1988 and 2003 had no lymph nodes examined.
A subsequent study in the National Cancer Database (NCDB) from 2003-2004 found that only
16.4% of patients with pancreatic cancer had at least 15 nodes examined.268
Regional lymph nodes of interest for standard lymphadenectomy during PD are located along
the gallbladder or common bile duct, anterior CHA, PV, the head of the pancreas, the SMV, and
the right side of the SMA; celiac, splenic, and left gastric artery nodes should not be
included.131,269 For distal splenectomy, nodes from the splenic hilum, splenic artery, and
superior margins of the pancreas should be examined.
A total of 5 prospective, randomized trials of extended vs standard lymphadenectomy have
shown no long-term survival benefit for extended resection.270,271 Current evidence supports a
standard lymphadenectomy as opposed to an extended lymphadenectomy with retroperitoneal
soft tissue clearance.272
For DP, some have advocated for the use of a radical antegrade modular pancreatosplenec-
tomy (RAMPS) for more extensive lymph node dissection. A retrospective comparison of
conventional DP and RAMPS found that although RAMPS allowed for a greater number of
retrieved lymph nodes, it did not lead to improved overall survival.273

Operative techniques and outcomes


Several methods of pancreatic anastomosis have been promoted but evidence for the
superiority of any single method has so far been inconclusive. RCTs comparing end-to-side, end-
to-end, or duct to mucosa and invagination techniques reveal a range of rates of fistula and are
difficult to compare owing to different definitions. A randomized trial comparing a duct with
mucosa anastomotic technique with end-to-side for creation of the PJ found no significant
difference in complication rate, whereas a prospective randomized trial found fewer pancreatic
fistulas using the invagination technique compared with duct to mucosa.274,275 Pancreaticogas-
trostomy, as an alternative to PJ, is associated with a lower risk of postoperative pancreatic fistula
as well as decreased severity of fistula but no difference in overall morbidity or mortality.276 The
use of fibrin sealants or glues to high-risk PJ anastomoses has not proven beneficial.
A meta-analysis including both internal and external stents found no benefit to intra-
operative pancreatic duct stent placement; however, those with external pancreatic duct stents
were found to have reduced fistula rates in a subgroup analysis and a subsequent randomized
trial.277,278
Almost one-third of patients who receive a DP develop pancreatic fistula. The optimal
technique for closure of the pancreatic remnant remains unknown. Options include hand-sewn
vs stapled closure as well as the use of fibrin glue or a jejunal patch.279,280 Selective duct ligation
or prophylactic transpapillary pancreatic stent insertion or both can also be performed.281

Minimally invasive vs open


Minimally invasive techniques including laparoscopy and robotic surgery have the potential
to provide improved visualization, contribute to decreased postoperative narcotic use, and lead
to shorter hospital stays.282 The first laparoscopic pancreatectomy (a pylorus-preserving PD) was
performed in 1994 for chronic pancreatitis and since then, surgeon expertise in this technique
has rapidly improved.283 More recently, the draw of high-definition graphics, increased degrees
of freedom for movement, and ergonomic advantages have led to the rise of robotic systems.284
In particular, minimally invasive DP is emerging as a viable alternative to open DP.
Retrospective single-center series (mostly in benign or borderline tumors) have shown lower
morbidity rates and shorter hospital stays after receipt of laparoscopic surgery.285,286 A national
study found equivalent adjusted odds of complete resection, number of lymph nodes removed,
and 30-day readmission and a shorter length of stay for laparoscopic or robotic DP compared
with open pancreatectomy.287 Equivalent median survival has been reported among experi-
enced surgeons.288,289 A reduction in operative time, blood loss, and conversion rate is seen after

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
130 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

the completion of only 10 laparoscopic distal PDs.290 Adequate volume remains a challenge—
between 2010 and 2011, 85% of Committee on Cancer-accredited cancer centers performed
fewer than 10 cases.287
In comparison, the uptake of laparoscopic pancreaticoduodenecomy has been less wide-
spread: only a few surgeons perform purely laparoscopic PDs. Some centers have achieved
decreased blood loss, hospital stay, and complication rates compared to open PD using the Da
Vinci robotic system.291 National data on the topic of minimally invasive PD (MIPD) is lacking.
MIPD use for pancreatic cancer has increased by 45% between 2010 and 2011 but 92% of
hospitals performing MIPD during that time period had a 2-year volume of less than 10 cases.
A total of 2 articles examining perioperative outcomes after MIPD using the National Cancer
Database found that although 30-day mortality did not significantly differ (5.1-5.2% for MIPD vs
3.1-3.7% for open surgery), MIPD was associated with almost double the odds of perioperative
death on multivariate analysis.292,293 However, when limited to hospitals with high hospital
volume (more than 10 cases in 2 years), operative technique was no longer independently
associated with mortality.293 An investigation into the learning curve for robotic PD
demonstrated statistical improvements in estimated blood loss and conversion to open surgery
after 20 cases, the incidence of pancreatic fistula after 40 cases, and operative time after 80
cases.294
There are limited data on the cost-effectiveness of minimally invasive techniques. A small
single-center study found that although upfront operative cost is higher for robotic PDs, once
follow-up hospitalization costs are included, costs became equivalent.
Minimally invasive techniques for the resection of pancreatic cancer are feasible and safe if
performed by experienced surgeons in a high-volume setting. Larger series with longer follow-
up times are needed for perioperative and oncologic outcomes after minimally invasive
pancreatic surgery for cancer.

Vascular resections and extended pancreatectomy


Initially described in the 1970s, mesentericoportal vein resection with the goal of negative
margins is performed today at many high-volume centers. Indeed, the International Study Group
of Pancreatic Surgery (ISGPS) supports the up-front operative resection of “reconstructable” SMV
or PV involvement.295 To be reconstructable, these lesions should allow for adequate inflow and
outflow in the reconstructed veins and lack of involvement of the SMA and CHA.296
Vascular resection can be performed via direct anastomosis, patch, or interposition graft that
can be either prosthetic or autogenous in nature (Fig 8).297-300 The concern for infection has
restricted the use of prosthetic grafts although good results have been demonstrated at some
centers. A series of 33 patients (85% with pancreatic cancer) receiving segmental vein resection
via interposition PTFE graft reconstruction discovered no cases of graft infection over a median
survival of 12 months for the patients with cancer.301 Figure 9 illustrates the use of autogenous
vein patches and interposition grafts. Size matching for autogenous veins can be challenging,
and veins from various locations including the gonadal veins, superficial femoral vein, and left
renal vein have been harvested.302-304 A study of 34 patients receiving PV or SMV reconstruction
using lower extremity vein (saphenous for patching and femoral for replacement) found an
overall patency rate of 88% at a mean follow-up of 5 months.305 Another study (mostly using
vein patches) achieved an exceptional 97% patency rate within 6 months postoperatively,
attributed by the authors to the assistance of a vascular surgeon during vascular resection.306 For
tension-free anastomosis, bovine patch is also a reasonably priced option that is durable and
biocompatible with no need for vein harvesting, but little is known about its role in venous
repair.307
Because operative time and blood loss tend to be greater in PDs with vascular resection,
surgical candidates should be carefully selected.308,309 However, the overall safety of venous
vascular resection has been established. A total of 2 meta-analyses of retrospective studies have
found that morbidity, mortality, and survival outcomes do not differ between pancreatic
resections with and without venous vascular resection.310,311

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 131

Fig. 8. Techniques for reconstruction of the superior mesenteric vein/portal vein. (Reprinted with permission from
Tseng JF et al., Pancreaticoduodenectomy with vascular resection: margin status and survival duration. J Gastroint Surg
2004;8:935.)

In contrast, arterial resection during pancreatectomy is associated with a 5-fold increased


odds of perioperative mortality compared with no arterial resection during pancreatectomy but
improved survival compared with those who do not undergo resection.312 This may be because
of the theory that venous involvement is merely related to tumor location, whereas arterial
involvement is due to aggressive tumor biology.313 For celiac reconstruction, the common or
proper hepatic artery must be preserved and re-anastomosed to ensure hepatic perfusion.314

Fig. 9. Intraoperative examples of venous reconstruction include (A) a saphenous vein patch on the portal vein, (B) a
saphenous vein graft from the proximal to distal replaced right hepatic artery (RRHA), and (C) an IJ interposition graft of
the SMV. (Courtesy of the Medical College of Wisconsin Pancreas Program and Douglas B. Evans, MD.) LHA, left hepatic
artery; GDA, gastroduodenal artery; RRHA, replaced right hepatic artery; HA, hepatic artery. (Color version of figure is
available online.)

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
132 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

Table 5
Grades of delayed gastric emptying. (Adapted with permission from Wente MN, et al. Delayed gastric emptying (DGE)
after pancreatic surgery: a suggested definition by the International Study Group of Pancreatic Surgery (ISGPS). Surgery
2007;142(5):761-8.)

Grade NGT required Unable to tolerate solid Vomiting or gastric Use of


oral intake by POD distension prokinetics

A 4-7 Days or reinsertion 4 POD 3 7 Yes or no Yes or no


B 8-14 Days or reinsertion 4POD 7 14 Yes Yes
C 4 14 Days or reinsertion 4POD 14 21 Yes Yes

NGT, nasogastric tube; POD, postoperative day.

DP with celiac resection (DP-CAR, also called the modified Appleby procedure) for tumors of
the body and tail is also possible in select patients. Here, collateral flow through the
gastroduodenal artery provides perfusion to the liver.315 Compared to a DP with no celiac
resection, DP-CAR is associated with longer operative time, increased rates of postoperative
kidney injury, and a 30-day postoperative mortality rate of 10%.316 In a Japanese study, DP with en
bloc celiac axis resection for locally advanced cancer of the pancreatic body resulted in a 91% R0
resection rate and a 5-year survival of 42%.317 A case series from one high-volume center showed
favorable outcomes for patients with arterial encasement who responded to neoadjuvant therapy
and underwent planned arterial resection, most often an Appleby procedure.318
Multivisceral resections (involving colon, stomach, kidney, adrenals, liver, etc.) are associated
with increased short-term morbidity but still provide a survival benefit compared with palliative
procedures.319,320 One-fifth of patients receiving multivisceral resections also have a venous
resection.
Vascular resection is not without its risks. A population-based analysis found that the
frequency of vascular resection during PD has increased between 2000 and 2009 and that the
postoperative complication rate with vascular resection is higher than without, even at high-
volume hospitals.321 In a large multicenter retrospective cohort study from the UK comparing
PD with vein resection, PD only, and palliative bypass in patients with T3 borderline resectable
pancreatic cancer, higher complication rates were found in the PD groups but no difference
between PD and PD with vascular resection. Perioperative mortality was similar in all 3 groups;
no difference in overall survival between the PD only and PD with vascular resection groups was
identified.322
There are no guidelines regarding the need for type and length of anticoagulation after a
venous reconstruction during PD. This is based on studies showing no difference in thrombosis
rates after venous reconstruction between those receiving and not receiving anticoagulation
(aspirin, warfarin, or clopidogrel.)323
More well-designed studies are needed to compare survival for increasing degrees of
vascular resection in locally advanced pancreatic cancer with nonsurgical chemotherapy or
chemoradiation treatments.

Perioperative outcomes—Morbidity and mortality


Based on NSQIP data, the 30-day mortality rate is approximately 2.6% for PD, 1.2% for DP, and
5.4% for total pancreatectomy.244,245,324 Although mortality after pancreatic resection has
improved, morbidity remains high at 30%-40%.325,326 Postoperative complications delay the
receipt of adjuvant therapy.327 Furthermore, their effective management contributes to lower
overall mortality rates.328 Sepsis, surgical site infection, and respiratory complications are the
most common postoperative complications.244,245 Pancreas-specific complications include
bleeding, anastomotic leaks, intra-abdominal infection, and delayed gastric emptying. Grades
of delayed gastric emptying are presented in Table 5. Small pancreatic duct diameter and soft
pancreatic tissue are two risk factors for anastomotic leak.329 Every effort should be made to
prevent leaks from pancreaticoenteric anastomoses, as these increase the risk of abscess
formation, postpancreatectomy hemorrhage, and delayed gastric emptying.330 Late

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 133

Table 6
Grades of postoperative pancreatic fistula with main parameters. (Adapted with permission from Bassi et al.331)

Grade A Grade B Grade C

Clinical condition Well Often well Ill-Appearing


n
Specific treatment No Yes or no Yes
US or CT if obtained Negative Negative or positive Positive
Persistent drainage after 3 weeks No Usually yes Yes
Reoperation No No Yes
Cause-specific death No No Possibly yes
Signs of infection or sepsis No Yes Yes
Readmission No Yes or no Yes or no

US, ultrasound; CT, computed tomography.


n
Peripheral or total parenteral nutrition, antibiotics, enteral nutrition, somatostatin analog and/or drainage.

complications include dumping and endocrine or exocrine insufficiency. An estimated 20%-30%


of all complications are life-threatening or require intervention (Clavien-Dindo grade III-IV.)235
Interventional drainage or stenting for leaks, fistulas, and abscesses, endoscopic dilation of
stenosis, and angiography or endovascular management of delayed hemorrhage are important
minimally invasive advances in the management of complications after PD that have reduced
the rate of reoperation.4
The International Study Group on Pancreatic Fistula published a standardized definition of
postoperative pancreatic fistula in 2005: “drain output of any measurable volume of fluid on or
after postoperative day 3 with an amylase content greater than 3 times the serum amylase
activity.” They created 3 grades (A, B, and C) of postoperative pancreatic fistula with increasing
severity (Table 6).331 A recent study highlighted the burden of Grade C fistulas; they develop in
approximately 2% of patients receiving PD.332 More than 85% have an additional complication.
Approximately 70% require reoperation and nearly two-thirds do not receive adjuvant therapy. A
10-point clinical risk score for pancreatic fistula (CS-POPF) is presented in Table 7.333
Somatostatin, an octreotide analog that inhibits pancreatic enzyme secretion, can be given as
prophylaxis postoperatively to decrease the rate of pancreatic fistulas, although there is no clear
evidence that somatostatin enhances closure for preexisting fistulas.334
As patients with cancer undergoing surgery, patients after PD are at above-average risk for
venous thromboembolic events. A study using data from NSQIP found an incidence of deep vein
thrombosis of 2.3% and pulmonary embolism of 1.5% within 30 days after pancreatectomy for
cancer.335
Approximately 1 of every 5 patients undergoing PD is readmitted. An analysis of 30-day
readmissions using SEER data found that more than 95% of the variation in readmissions was
attributable to patient-level factors such as comorbidity, 4% to hospital factors, and only 0.3% to
surgeon-level factors.336

Volume or outcome relationships


There is now widespread agreement that the experience and volume of a surgeon and
hospital are important features influencing successful pancreatic resections and long-term
results.337 Owing to advancements in anesthesia, operative selection, and surgical technique as
well as the regionalization of pancreatic cancer care to high-volume centers, mortality after PD
has dropped from approximately 25% in the 1960s to less than 3% at some high-volume
centers.338 In a study using Medicare data, higher hospital volume explained 67% of the decrease
in operative mortality for pancreatectomy from 1999-2008.339 High hospital volume has even
been linked to improved long-term survival.340
The effect of volume is seen on intermediate outcomes as well. Patients with localized
pancreatic cancer are more likely to receive multimodal treatment at high-volume centers.184 In
a study from the NCDB, patients undergoing PD at low-volume centers were more likely to have
margin-positive resections.341 They also had fewer lymph nodes examined during PD than at

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
134 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

Table 7
Risk score for prediction of postoperative pancreatic fistula. (Reprinted with permission from Callery et al.333)

Risk factor Points

Gland texture
Firm (6-10) 0
Soft (1-5) 2

Pathology
Pancreatic adenocarcinoma or chronic pancreatitis 0
All others 2

Pancreatic duct diameter (mm)


Z5 0
4-4.9 1
3-3.9 2
2-2.9 3
r1.9 4

Intraoperative blood loss (mL)


r400 0
401-700 1
701-1000 2
4 1000 3

Negligible risk, 0 points; low risk, 1-2 points; intermediate risk, 3-6 points; high risk, 7-10 points.

high-volume hospitals.342 High-volume centers are more equipped to deal with complications of
treatment and have the multidisciplinary teams necessary to manage multimodal therapy.330 As
an illustration, the presence of interventional radiology at a hospital is associated with lower
perioperative mortality after pancreatic surgery.343 In a landmark “failure to rescue” article,
Medicare patients receiving pancreatectomy at very-low-volume hospitals were 1.7 times more
likely to suffer a complication and 3.2 times more likely to die after that complication than
patients at very-high-volume hospitals.344
The belief that high-volume centers are safer locations for the performance of pancreatic
surgery has led to the regionalization of pancreatic cancer care. From 2007-2011, National Cancer
Institute-designated Cancer Centers (NCI-CCs) performed 53.5% of pancreatic operations for
cancer.345 A Dutch study in the Netherlands found improved survival after centralization of
pancreatic surgery.346

Oncological outcomes

Survival

Resection is critically important for survival—patients with unresected pancreatic cancer


have a 5-year survival of 8.7% if locally advanced and 1.8% if metastatic.7 In contrast, studies of
patients with resected pancreatic cancer (most of whom have adjuvant therapy) have shown 5-
year survival rates between 18% and 20%, a number that has not meaningfully changed in the last
25 years.347,348 Figure 10 shows Kaplan-Meier curves for adjusted cancer-specific survival by
stage and resection status.
Positive margins, lymph node metastasis, larger tumor size, and poor differentiation have all
been found to predict poor survival after resection.347,349 In total, 3 or more units of blood
transfused during pancreaticoduodenecomy is also predictive of poor survival as is perineural
invasion.348,350 Patients who develop a thromboembolism, especially early on in their illness,
have worsened survival.351
Although the prognostic value of microscopically negative (R0) resection margins has
differed in some studies, this can be attributed to pathology evaluation and the definition of R1
resection.352 When R1 resection is defined as cancer within 1 mm of the resection border, R0

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 135

Fig. 10. Cancer-specific survival for (A) resected and (B) unresected patients. Survival is adjusted for age, sex, race, tumor
site, tumor grade, and receipt of radiotherapy and stratified by disease stage for patients diagnosed with pancreatic
adenocarcinoma from 1998 to 2005 in the Surveillance Epidemiology End Results cancer registry. The survival curves for
patients with unresected stage II and III cancers are superimposed. Overall, 47% of people who underwent resection
received radiotherapy vs 19% among those who did not. The use of systemic chemotherapy was not available. (Adapted
with permission from Katz MH, Hu CY, Fleming JB, Pisters PW, Lee JE, Chang GJ. Clinical calculator of conditional survival
estimates for resected and unresected survivors of pancreatic cancer. Arch Surg 2012;147(6):513-9.) (Color version of
figure is available online.)

resection is superior.353,354 It remains a key objective in the resection of adenocarcinoma of the


pancreas, as it influences both local recurrence and survival.
In an analysis of patients with pancreatic cancer who received surgical resection between
1998 and 2002, 3.9% survived at least 10 years. Predictors of long-term survival included tumor
characteristics (lymph node positivity ratio, pathologic T or M stage, size, grade, and surgical
margin), treatment variables (receipt of adjuvant chemotherapy), and sociodemographic factors
(age, education level, and insurance status.)355

Recurrence

Early recurrence of invasive pancreatic cancer is common even after curative resection: 80%
of patients have a recurrence within 2 years.356 Most patients develop a recurrence, with highest
likelihood in the abdominal cavity, and subsequently die from metastatic disease. There is a

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
136 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

significant amount of drug resistance in pancreatic cancer, and the cause is not well
understood.357
Several studies have addressed patterns of recurrence. Extrapancreatic recurrence appears to
be more common than isolated local recurrence. One study found a 69.5% recurrence rate over a
mean follow-up time of 32.4 months. The first site of recurrence was local only in 23.7%
(estimated at 10%-35% in other studies), local plus distant in 21.2%, and only distant in 55.1%.358-360
In another study, 75.9% of patients experienced a recurrence over a median follow-up of 18.5
months. Extrapancreatic recurrence most commonly occurred in the liver.361 There is a vast
difference seen in postoperative survival between patients with isolated local recurrence (16.4
months) vs patients with metastasis (9.4 months.)356
The guidelines for postoperative surveillance from the NCCN call for a physical examination,
CA 19-9 level, and CT scan at 3-6 month intervals for the first 2 years and then annually.362
However, a study using a large administrative database of Medicare patients found that 23% of
patients did not receive a CT scan in the year following surgery, and that there was no temporal
pattern to CT use, suggesting that scans were obtained for symptoms rather than regular
surveillance.363
A total of 3 factors together have been found to predict local early recurrence with a 90.4%
accuracy: pre-op CA 19-9 level 4 228 U/mL, tumor diameter 4 3.1 cm, and pathologic lymph
nodes preoperatively.364 Positive surgical margins and an elevated postoperative CA19-9 predict
early development of distant metastases but not locoregional failure.365 Longer progression-free
survival has been independently associated with neoadjuvant chemoradiation, lower estimated
blood loss, lack of positive posterior margin, lack of nodal microinvolvement, and adjuvant
therapy.183,358,360,366,367 A study found that the accuracy of MDCT in detecting recurrence was
85%, for FDG-PET the accuracy was 85%, and for CA 19-9 the accuracy was 95%. The authors
conclude that a combination of CA 19-9 with either multidetector CT or FDG-PET CT is 100%
accurate.368
There does appear to be a response to chemoradiation in patients with recurrent pancreatic
cancer without distant metastasis.369 In a small Japanese study, 30 patients treated with
radiotherapy at a median dose of 54 Gy experienced encouraging local control and overall
survival.370 However, surgical reoperation for local recurrence may not be feasible because of
dense adhesions and the extent of tumor invasion. A study found that patients who had an
interval of greater than 9 months between resection and recurrence were more likely to benefit
from re-resection.371 Encouraging results were reported in Germany in a small study of patients
with local recurrence who were surgically explored (and some grossly resected or debulked),
then treated with both intraoperative RT and external beam radiation.372

Quality of life

The data on quality of life (QOL) in pancreatic cancer are lacking. Almost one half of the
number of patients with pancreatic cancer experience pain.373,374 Among newly diagnosed
patients, 29% reported severe pain and 37% moderate pain.373 Relief of pain is associated with
enhanced quality of life, including improved mood and decreased interference with activities.375
Methods for pain relief in unresectable patients range from nonsteroidal anti-inflammatory
drugs (NSAIDs) to narcotics to epidurals to celiac plexus blocks. Neurolytic celiac axis block
provides effective pain relief for 74% of patients and is especially effective for tumors in the head
of the pancreas.376 Multimodal pain control is recommended, as pain from pancreatic cancer is
multifocal, with visceral, somatic, and neuropathic components.377 Early symptom control for
anxiety, nausea, fatigue, and anorexia is also important.378
A diagnosis of pancreatic cancer can have severe psychological effects. A total of 38% of
patients were found to have elevated depression scores after a new diagnosis with pancreatic
cancer.373 Men with pancreatic cancer are 11 times more likely to commit suicide than the
general population.379 Fear of recurrence is also common: more than one-third of patients with

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 137

pancreatic cancer had a fear of cancer recurrence score that indicated “frequent fearful thoughts,
emotional disturbance, and functional impairment.”380
The receipt of surgery for pancreatic cancer appears to improve QOL in a small study from
Mexico.381 Another study evaluating quality of life in the elderly after PD in Israel found
improvement in functional, symptom-based, and global QOL scores in the months after
surgery.382 It takes 3-6 months after surgery for QOL to return to preoperative levels.383 The
receipt of adjuvant chemotherapy after macroscopically complete resection has been associated
with improved QOL in a prospective randomized study.384

Palliation

Given the 6-10 month median survival time for patients with advanced pancreatic cancer,
palliative efforts should focus on increasing comfort and improving quality of life.385,386 In
addition to addressing pain control and biliary or intestinal obstruction, palliation should also
manage malnutrition, thromboembolism, and depression.387 Currently, an RCT is underway to
evaluate the benefits of integration of early palliative care with standard oncologic care for
advanced lung and noncolorectal GI malignancies.388 Although hospice use at end of life has
increased over time for Medicare patients, there has been a decrease in early enrollment and
an increase in ICU admissions and chemotherapy use in the last month of life.389 This
suggests that hospice services may not be used optimally although they increase QOL and
lower cost.

Surgical

The need for surgical palliation is usually a result of obstructive jaundice, gastric outlet
obstruction, or pain unresponsive to other modalities. When proximal GI tract obstruction
(which is more commonly gastric than duodenal in patients with pancreatic cancer) cannot be
treated endoscopically, a bypass or double bypass operation can be undertaken.390 Roux-en-y HJ
is generally preferable to choledochoduodenostomy or cholecystojejunostomy owing to superior
outcomes.391,392 A meta-analysis from 2009 found that prophylactic gastroenterostomy during
surgical exploration of unresectable patients reduced the incidence of gastric outlet obstruction
without influencing morbidity and mortality.393 A total of 38% of patients with unresectable
cancer and biliary stents who receive chemoradiation, however, do develop a symptomatic
duodenal obstruction at a mean of 11.4 months after diagnosis requiring intervention, and that
rate has risen over time.394
Palliative bypass is preferable to planned palliative resection, as studies comparing the 2 have
found higher morbidity and mortality in the resection group as well as longer operating times
and longer hospital lengths of stay.395
There are several methods of surgical pain control, including celiac axis block and
splanchnicectomy. Neurolytic celiac axis plexus block (now done laparoscopically or endoscopi-
cally) is safe and decreases the need for pain medication.396,397 A study evaluating the efficacy of
laparoscopic celiac axis block in patients with unresectable cancer found that it resulted in
improved pain scores with minimal complications and short operative time.398 An RCT found
that celiac axis block even improves survival.375 Thoracoscopic splanchnicectomy is a minimally
invasive interruption of the fibers of the splanchnic nerve, allowing for relief of abdominal pain
while avoiding some of the potential side effects of a celiac plexus neurolysis. It has shown good
short-term pain relief and improved QOL for patients with unresectable cancer in single-center
studies.399,400

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
138 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

Endoscopic

The reality is that 80% of patients with pancreatic cancer who have progressed to biliary
obstruction are not operative candidates.401 In addition to being performed therapeutically for
symptomatic patients with obstructive jaundice who are awaiting surgery, ERCP can also be
performed for unresectable patients in need of biliary decompression. In the case of long-term
use (43 months), expandable metal stents are preferable to plastic stents, which occlude sooner
and at higher rates.402
Studies have shown mixed outcomes for biliary bypass vs stenting for palliation, likely
because of the variety of outcomes measured. Early RCTs found no difference in complication
rates.403 Other RCTs found more complications in the surgery group.404 However, these were
performed before advances in expandable metal stents. A meta-analysis from the year 2000
found that 3% (0%-16%) of patients required endoscopic re-intervention in the biliary (all
plastic) stent group vs 35% (28%-43%) requiring surgical re-intervention in the surgical bypass
group.405 The most recent meta-analysis on the topic, published in 2014, did not differentiate
between metal and plastic stents and found no difference in technical success rates or
morbidity or mortality but that recurrent biliary obstruction was 9 times more likely after
stent placement.406
Given that the short-term outcomes of surgical bypass may be inferior to stenting, current
recommendations call for endoscopic stenting in poor surgical candidates with a short life
expectancy and biliary bypass for patients with good performance status and longer life
expectancy.407,408 A recent study of palliative bypass for pancreatic cancer cites a 30-day
mortality rate of 6.5% and major complication rate of 20%.409 For some patients, these short-term
risks may be prohibitive. The decision of which path to pursue in cases of biliary obstruction
should be undertaken on an individual basis with the input of a multidisciplinary team and
should take into account life expectancy.410 This paradigm should be revisited as data on
minimally invasive bypass become available, which has the potential of changing the short-term
risk profile of surgical bypass.

Chemotherapy

Gemcitabine has been the preferred method of palliative chemotherapy, having been
shown to prolong survival and improve QOL for patients with unresectable pancreatic
cancer.386,411 Its side effects include nausea, vomiting, lethargy, and bone marrow suppression.
Recent phase III trials have supported the addition of second cytotoxic agents, although the
benefits are small.412 Albumin-bound paclitaxel (nab-paclitaxel) plus gemcitabine lead to
significantly improved progression-free and overall survival (by only 1.8 months for both), but
with increased rates of peripheral neuropathy and myelosuppression. Gemcitabine plus
capecitabine (GEM-CAP) vs gemcitabine alone shows a significant survival benefit on meta-
analysis.413
In 2011, the PRODIGE 11 study found that FOLFIRINOX (oxaliplatinin, irinotecan, fluorouracil,
and leucovorin) was superior to gemcitabine monotherapy for metastatic pancreatic cancer in
patients with good performance status, with a notable survival advantage over gemcitabine but
increased toxicity (neutropenia, thrombocytopenia, diarrhea, and neuropathy.)414 Notably,
locally advanced patients were excluded from this study. FOLFIRINOX is now preferentially used
for patients with high performance status and good hepatobiliary function. If gemcitabine or
FOLFIRINOX are unavailable, capecitabine with or without oxaliplatinin or 5-FU or leucovorin or
oxaliplatinin (OFF) are also viable options.415

Radiation therapy

The combination of radiotherapy with chemotherapy for palliation remains controversial.416


The LAP 07 study found no benefit to the addition of radiation to chemotherapy for locally

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 139

advanced pancreatic cancer; however, 2 efficacy trials have demonstrated the safety of
concurrent radiotherapy with fixed-dose gemcitabine, and another study found improved
survival with chemoradiotherapy vs radiation alone.208,385,417
Newer therapies have focused on reducing systemic toxicity and increasing local control.
Some hospitals have shown good results for unresectable locally advanced patients treated with
EBRT, chemotherapy, and intraoperative radiotherapy.418 SBRT using Cyberknife is also an
emerging technique for palliation.419 EUS-guided fiducial placement for SBRT can be performed
to minimize the need for surgery.420 SBRT has been reported to relieve pain and improve quality
of life in patients with unresectable and metastatic disease.421,422 Although earlier studies using
a single fraction of 25 Gy showed high rates of late GI toxicity, later studies have achieved over a
50% decrease in toxicity using 5 fractions of 6.6 Gy.423–425 Survival outcomes appear to be
improving over time; whereas early studies showed poor survival, more recent reports show
results comparable to conventional radiotherapy (ERBT.)426,427 Importantly, SBRT with or
without chemotherapy is safe and effective in patients older than 80 years age who are either
unresectable or cannot have surgery owing to comorbid conditions.428 More studies are needed
comparing EBRT and SBRT for unresectable pancreatic cancer.

Other therapies

Erlotinib, an EGFR tyrosine kinase inhibitor, is Food and Drug Administration-approved for
unresectable pancreatic cancer in combination with gemcitabine.429 The addition of other agents
is also being tested: an efficacy study of the addition of upamostat (a urokinase plasminogen
activator) to Gem showed tolerability but similar survival for nonresectable locally advanced
patients.430 Recent studies have demonstrated the safety of ablative therapies such as
radiofrequency ablation (RFA) for liver metastasis, microwave ablation, high-intensity–focused
ultrasound (HIFU), carbon ion radiotherapy, and photodynamic therapy for locally advanced
cancer.431–436

Future directions

Detection of the pancreatic cancer at earlier stages is critical. Translational research should
continue to identify biomarkers that can be used for early diagnosis and prognostic
prediction.437 A systematic review of 78 studies revealed 76 potential molecular markers, of
which only 11 were confirmed in 2 or more studies. None could yet be recommended for clinical
use. A total of 2 micro-RNA panels in whole blood as well as an exosomal protein have been
identified that distinguish patients with pancreatic cancer from healthy controls, but more
research is needed to understand their utility.438
We should focus on improving the modalities that we do have, including the application of
image-guided surgery and SBRT, as well as the development of new methods and paradigms for
effective treatment.204,439-441 Enhancing the sensitivity of pancreatic cancer cells to chemo-
therapy is an important undertaking—methods involving small interfering RNA (siRNA) have
been proposed. Novel methods for delivery of chemotherapeutic agents are also being
developed, including nanoparticle-mediated drug-eluding scaffolds.442 Gene transfer and
immunotherapy are quickly advancing fields that may allow for inhibition of tumor growth or
systemic antitumor immunity.443 A better understanding of genetics as well as medication
transport and metabolism would permit us to investigate variation in response to therapy and
allow for more personalized medicine. The optimal sequencing of evolving therapies with
surgery should also be examined.
The use of multiple therapeutic modalities has created a need for increasing coordination
between specialties, leading to a move toward multidisciplinary care. A single-day pancreatic
multidisciplinary clinic provided a comprehensive and co-ordinated evaluation of patients that
led to changes in therapeutic recommendations in close to a quarter, according to a 2008
study.444

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
140 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

Finally, numerous studies have pointed to the underuse of surgery, different referral patterns,
and overall inadequate care for certain populations.445-449 We must ensure that everyone
eligible for potentially life-saving treatment receives it.

Conclusions

Although pancreatic cancer remains a fatal disease, pancreatic surgery is rapidly evolving,
and therapeutic breakthroughs are on the horizon. Efforts to improve outcomes for pancreatic
cancer should focus on early diagnosis and referral, reducing and managing operative
complications, and the development of novel and optimally sequenced therapeutic techniques.

Funding

Howard Hughes Medical Institute Early Career Award, American Surgical Association
Foundation Fellowship, American Cancer Society MSRG 10-003-01 (to JFT).
Pyrtek Fund Research Fellowship (to LAB).

References

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA: a cancer journal for clinicians. 2016;66(1):7–30.
2. McClusky 3rd DA, Skandalakis LJ, Colborn GL, Skandalakis JE. Harbinger or hermit? Pancreatic anatomy and surgery
through the ages—part 3. World J Surg. 2002;26:1512–1524.
3. Moynihan B. Neoplasms of the pancreas. In: Moynihan B, ed. Abdominal Operations. Philadelphia: WB Saunders;
1906.
4. Buchler MW, Wagner M, Schmied BM, Uhl W, Friess H, Z’Graggen K. Changes in morbidity after pancreatic
resection: toward the end of completion pancreatectomy. Arch Surgery. 2003;138:1310–1314. [discussion 5].
5. Crile Jr. G. The advantages of bypass operations over radical pancreatoduodenectomy in the treatment of pancreatic
carcinoma. Surg Gynecol Obstet. 1970;130:1049–1053.
6. Muniraj T, Jamidar PA, Aslanian HR. Pancreatic cancer: a comprehensive review and update. Dis Mon. 2013;59:
368–402.
7. Howlader NNA, Kraphco M, et al. SEER Cancer Statistics Review, 1975-2008. In: Institute NC, ed. Bethesda, MD;
2011.
8. Anderson KE PJ, Mack TM. In: F.J. Schottenfeld D, ed. Cancer Epidemiology and Prevention. New York: Oxford
University Press; 1996:725–771.
9. Brand RE, Lerch MM, Rubinstein WS, et al. Advances in counselling and surveillance of patients at risk for
pancreatic cancer. Gut. 2007;56:1460–1469.
10. Ghadirian P, Boyle P, Simard A, Baillargeon J, Maisonneuve P, Perret C. Reported family aggregation of pancreatic
cancer within a population-based case-control study in the Francophone community in Montreal, Canada. Int J
Pancreatol. 1991;10:183–196.
11. Silverman DT. Risk factors for pancreatic cancer: a case-control study based on direct interviews. Teratog Carcinog
Mutagen. 2001;21:7–25.
12. Barton JG, Schnelldorfer T, Lohse CM, et al. Patterns of pancreatic resection differ between patients with familial
and sporadic pancreatic cancer. J Gastrointest Surg. 2011;15:836–842.
13. Hruban RH, Canto MI, Goggins M, Schulick R, Klein AP. Update on familial pancreatic cancer. Adv Surg. 2010;44:
293–311.
14. Amin S, McBride RB, Kline JK, et al. Incidence of subsequent pancreatic adenocarcinoma in patients with a history
of nonpancreatic primary cancers. Cancer. 2012;118:1244–1251.
15. Jones S, Hruban RH, Kamiyama M, et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility
gene. Science. 2009;324:217.
16. Bartsch DK. Familial pancreatic cancer. Br J Surg. 2003;90:386–387.
17. Bartsch DK, Gress TM, Langer P. Familial pancreatic cancer–current knowledge. Nat Rev Gastroenterol Hepatol.
2012;9:445–453.
18. Fuchs CS, Colditz GA, Stampfer MJ, et al. A prospective study of cigarette smoking and the risk of pancreatic cancer.
Arch Intern Med. 1996;156:2255–2260.
19. Gold EB, Goldin SB. Epidemiology of and risk factors for pancreatic cancer. Surg Oncol Clin N Am. 1998;7:67–91.
20. Lowenfels AB MP, Whitcomb DC. Risk factors for cancer in hereditary pancreatitis. In: Whitcomb CJ, Ulrich CD, eds.
The Medical Clinics of North America: Inherited Diseases of the Pancreas. Philadelphia: W.B. Saunders Co; 2000:
3565–3573.
21. Hassan MM, Bondy ML, Wolff RA, et al. Risk factors for pancreatic cancer: case-control study. Am J Gastroenterol.
2007;102:2696–2707.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 141

22. Hermann PC, Sancho P, Canamero M, et al. Nicotine promotes initiation and progression of KRAS-induced
pancreatic cancer via Gata6-dependent dedifferentiation of acinar cells in mice. Gastroenterology. 2014;147. 1119-
33.e4.
23. Arslan AA, Helzlsouer KJ, Kooperberg C, et al. Anthropometric measures, body mass index, and pancreatic cancer: a
pooled analysis from the Pancreatic Cancer Cohort Consortium (PanScan). Arch Intern Med. 2010;170:791–802.
24. Michaud DS, Giovannucci E, Willett WC, Colditz GA, Stampfer MJ, Fuchs CS. Physical activity, obesity, height, and
the risk of pancreatic cancer. J Am Med Assoc. 2001;286:921–929.
25. Huxley R, Ansary-Moghaddam A, Berrington de Gonzalez A, Barzi F, Woodward M. Type-II diabetes and pancreatic
cancer: a meta-analysis of 36 studies. Br J Cancer. 2005;92:2076–2083.
26. Everhart J, Wright D. Diabetes mellitus as a risk factor for pancreatic cancer. A meta-analysis. J Am Med Assoc.
1995;273:1605–1609.
27. Muntoni S, Muntoni S, Draznin B. Effects of chronic hyperinsulinemia in insulin-resistant patients. Curr Diab Rep.
2008;8:233–238.
28. Stolzenberg-Solomon RZ, Graubard BI, Chari S, et al. Insulin, glucose, insulin resistance, and pancreatic cancer in
male smokers. J Am Med Assoc. 2005;294:2872–2878.
29. Tsirambidis JV, Conwell DL, Zuccaro G. Chronic pancreatitis. MedGenMed. 2003;5:17.
30. Lowenfels AB, Maisonneuve P, Cavallini G, et al. Pancreatitis and the risk of pancreatic cancer. International
Pancreatitis Study Group. N Engl J Med. 1993;328:1433–1437.
31. Stolzenberg-Solomon RZ, Blaser MJ, Limburg PJ, et al. Helicobacter pylori seropositivity as a risk factor for
pancreatic cancer. J Natl Cancer Inst. 2001;93:937–941.
32. Maisonneuve P, FitzSimmons SC, Neglia JP, Campbell 3rd PW, Lowenfels AB. Cancer risk in nontransplanted and
transplanted cystic fibrosis patients: a 10-year study. J Natl Cancer Inst. 2003;95:381–387.
33. Han X, Li J, Brasky TM, et al. Antioxidant intake and pancreatic cancer risk: the Vitamins and Lifestyle (VITAL)
Study. Cancer. 2013;119:1314–1320.
34. Larsson SC, Giovannucci E, Wolk A. Folate intake, MTHFR polymorphisms, and risk of esophageal, gastric, and
pancreatic cancer: a meta-analysis. Gastroenterology. 2006;131:1271–1283.
35. Bosetti C, Lucenteforte E, Bracci PM, et al. Ulcer, gastric surgery and pancreatic cancer risk: an analysis from the
International Pancreatic Cancer Case-Control Consortium (PanC4). Ann Oncol. 2013;24:2903–2910.
36. Bardeesy N, DePinho RA. Pancreatic cancer biology and genetics. Nat Rev Cancer. 2002;2:897–909.
37. Hingorani SR, Petricoin EF, Maitra A, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection
in the mouse. Cancer Cell. 2003;4:437–450.
38. Rooman I, Real FX. Pancreatic ductal adenocarcinoma and acinar cells: a matter of differentiation and
development? Gut. 2012;61:449–458.
39. Hruban RH, Goggins M, Parsons J, Kern SE. Progression model for pancreatic cancer. Clin Cancer Res. 2000;6:
2969–2972.
40. van Heek NT, Meeker AK, Kern SE, et al. Telomere shortening is nearly universal in pancreatic intraepithelial
neoplasia. Am J Pathol. 2002;161:1541–1547.
41. Jones S, Zhang X, Parsons DW, et al. Core signaling pathways in human pancreatic cancers revealed by global
genomic analyses. Science. 2008;321:1801–1806.
42. Brand R. Pancreatic cancer. Disease-a-month: DM. 2004;50:545–555.
43. Hidalgo M. Pancreatic cancer. N Engl J Med. 2010;362:1605–1617.
44. Yachida S, Jones S, Bozic I, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer.
Nature. 2010;467:1114–1117.
45. Buchler P, Gazdhar A, Schubert M, et al. The Notch signaling pathway is related to neurovascular progression of
pancreatic cancer. Ann Surg. 2005;242:791–800. [Discussion-1].
46. Handra-Luca A, Hammel P, Sauvanet A, Lesty C, Ruszniewski P, Couvelard A. EGFR expression in pancreatic
adenocarcinoma. Relationship to tumour morphology and cell adhesion proteins. J Clin Pathol. 2014;67:295–300.
47. Buchler P, Reber HA, Buchler MW, Friess H, Hines OJ. VEGF-RII influences the prognosis of pancreatic cancer. Ann
Surg. 2002;236:738–749. [discussion 49].
48. Hidalgo M, Maitra A. The hedgehog pathway and pancreatic cancer. N Engl J Med. 2009;361:2094–2096.
49. Aghdassi A, Sendler M, Guenther A, et al. Recruitment of histone deacetylases HDAC1 and HDAC2 by the
transcriptional repressor ZEB1 downregulates E-cadherin expression in pancreatic cancer. Gut. 2012;61:439–448.
50. Akita H, Nagano H, Takeda Y, et al. Ep-CAM is a significant prognostic factor in pancreatic cancer patients by
suppressing cell activity. Oncogene. 2011;30:3468–3476.
51. Theoharides TC. Mast cells and pancreatic cancer. N Engl J Med. 2008;358:1860–1861.
52. Hermanek P. Pathology and biology of pancreatic ductal adenocarcinoma. Langenbecks Arch Surg. 1998;383:
116–120.
53. Nagakawa T, Kobayashi H, Ueno K, et al. The pattern of lymph node involvement in carcinoma of the head of the
pancreas. A histologic study of the surgical findings in patients undergoing extensive nodal dissections. Int J
Pancreatol. 1993;13:15–22.
54. Chari ST, Leibson CL, Rabe KG, et al. Pancreatic cancer-associated diabetes mellitus: prevalence and temporal
association with diagnosis of cancer. Gastroenterology. 2008;134:95–101.
55. Chen N, Unnikrishnan IR, Anjana RM, Mohan V, Pitchumoni CS. The complex exocrine-endocrine relationship and
secondary diabetes in exocrine pancreatic disorders. J Clin Gastroenterol. 2011;45:850–861.
56. Aggarwal G, Ramachandran V, Javeed N, et al. Adrenomedullin is up-regulated in patients with pancreatic cancer
and causes insulin resistance in beta cells and mice. Gastroenterology. 2012;143(1510-7):e1.
57. Abel EV, Simeone DM. Biology and clinical applications of pancreatic cancer stem cells. Gastroenterology. 2013;144:
1241–1248.
58. Adikrisna R, Tanaka S, Muramatsu S, et al. Identification of pancreatic cancer stem cells and selective toxicity of
chemotherapeutic agents. Gastroenterology. 2012;143(234-45):e7.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
142 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

59. Alhamdani MS, Youns M, Buchholz M, et al. Immunoassay-based proteome profiling of 24 pancreatic cancer cell
lines. J Proteomics. 2012;75:3747–3759.
60. Iacobuzio-Donahue CA, Fu B, Yachida S, et al. DPC4 gene status of the primary carcinoma correlates with patterns of
failure in patients with pancreatic cancer. J Clin Oncol. 2009;27:1806–1813.
61. Albers GH, Fleuren G, Escribano MJ, Nap M. Immunohistochemistry of CEA in the human pancreas
during development, in the adult, chronic pancreatitis, and pancreatic adenocarcinoma. Am J Clin Pathol. 1988;90:
17–22.
62. Aguilar S, Corominas JM, Malats N, et al. Tissue plasminogen activator in murine exocrine pancreas cancer:
selective expression in ductal tumors and contribution to cancer progression. Am J Pathol. 2004;165:
1129–1139.
63. Shi C, Merchant N, Newsome G, Goldenberg DM, Gold DV. Differentiation of pancreatic ductal adenocarcinoma
from chronic pancreatitis by PAM4 immunohistochemistry. Arch Pathol Lab Med. 2014;138:220–228.
64. Dudeja V, Allen PJ. Premalignant cystic neoplasms of the pancreas. Semin Oncol. 2015;42:70–85.
65. Adsay NV, Conlon KC, Zee SY, Brennan MF, Klimstra DS. Intraductal papillary-mucinous neoplasms of the pancreas:
an analysis of in situ and invasive carcinomas in 28 patients. Cancer. 2002;94:62–77.
66. Balsarkar D, Takahata S, Tanaka M. Intraductal papillary mucinous neoplasms of the pancreas: is the puzzle solved?
Indian J Gastroenterol. 2013;32:213–221.
67. Choi MG, Kim SW, Han SS, Jang JY, Park YH. High incidence of extrapancreatic neoplasms in patients with
intraductal papillary mucinous neoplasms. Arch Surg. 2006;141:51–56. [discussion 6].
68. Eguchi H, Ishikawa O, Ohigashi H, et al. Patients with pancreatic intraductal papillary mucinous neoplasms are at
high risk of colorectal cancer development. Surgery. 2006;139:749–754.
69. Marchegiani G, Malleo G, D’Haese JG, et al. Association between pancreatic intraductal papillary mucinous
neoplasms and extrapancreatic malignancies. Clin Gastroenterol Hepatol. 2015;13:1162–1169.
70. Adsay NV. The “new kid on the block”: Intraductal papillary mucinous neoplasms of the pancreas: current concepts
and controversies. Surgery. 2003;133:459–463.
71. Doi R, Fujimoto K, Wada M, Imamura M. Surgical management of intraductal papillary mucinous tumor of the
pancreas. Surgery. 2002;132:80–85.
72. Terris B, Ponsot P, Paye F, et al. Intraductal papillary mucinous tumors of the pancreas confined to secondary ducts
show less aggressive pathologic features as compared with those involving the main pancreatic duct. Am J Surg
Pathol. 2000;24:1372–1377.
73. Matsumoto T, Aramaki M, Yada K, et al. Optimal management of the branch duct type intraductal papillary
mucinous neoplasms of the pancreas. J Clin Gastroenterol. 2003;36:261–265.
74. Rodriguez JR, Salvia R, Crippa S, et al. Branch-duct intraductal papillary mucinous neoplasms: observations in 145
patients who underwent resection. Gastroenterology. 2007;133:72–79. [quiz 309-10].
75. Tanaka M, Chari S, Adsay V, et al. International consensus guidelines for management of intraductal papillary
mucinous neoplasms and mucinous cystic neoplasms of the pancreas. Pancreatology. 2006;6:17–32.
76. Werner J, Fritz S, Buchler MW. Intraductal papillary mucinous neoplasms of the pancreas—a surgical disease. Nat
Rev Gastroenterol Hepatol. 2012;9:253–259.
77. Fritz S, Klauss M, Bergmann F, et al. Small (Sendai negative) branch-duct IPMNs: not harmless. Ann Surg. 2012;256:
313–320.
78. Lee CJ, Scheiman J, Anderson MA, et al. Risk of malignancy in resected cystic tumors of the pancreas o or ¼3 cm in
size: is it safe to observe asymptomatic patients? A multi-institutional report. J Gastrointest Surg. 2008;12:234–242.
79. Nagai K, Doi R, Ito T, et al. Single-institution validation of the international consensus guidelines for treatment of
branch duct intraductal papillary mucinous neoplasms of the pancreas. J Hepatobiliary Pancreat Surg. 2009;16:
353–358.
80. Tanaka M, Fernandez-del Castillo C, Adsay V, et al. International consensus guidelines 2012 for the management of
IPMN and MCN of the pancreas. Pancreatology. 2012;12:183–197.
81. Sugiyama M, Izumisato Y, Abe N, Masaki T, Mori T, Atomi Y. Predictive factors for malignancy in intraductal
papillary-mucinous tumours of the pancreas. Br J Surg. 2003;90:1244–1249.
82. Furukawa T, Takahashi T, Kobari M, Matsuno S. The mucus-hypersecreting tumor of the pancreas. Development
and extension visualized by three-dimensional computerized mapping. Cancer. 1992;70:1505–1513.
83. Serikawa M, Sasaki T, Fujimoto Y, Kuwahara K, Chayama K. Management of intraductal papillary-mucinous
neoplasm of the pancreas: treatment strategy based on morphologic classification. J Clin Gastroenterol. 2006;40:
856–862.
84. Levy P, Jouannaud V, O’Toole D, et al. Natural history of intraductal papillary mucinous tumors of the pancreas:
actuarial risk of malignancy. Clin Gastroenterol Hepatol. 2006;4:460–468.
85. Chari ST, Yadav D, Smyrk TC, et al. Study of recurrence after surgical resection of intraductal papillary mucinous
neoplasm of the pancreas. Gastroenterology. 2002;123:1500–1507.
86. Hara T, Ikebe D, Odaka A, et al. Preoperative histological subtype classification of intraductal papillary mucinous
neoplasms (IPMN) by pancreatic juice cytology with MUC stain. Ann Surg. 2013;257:1103–1111.
87. Alexander BM, Fernandez-Del Castillo C, Ryan DP, et al. Intraductal papillary mucinous adenocarcinoma of the
pancreas: clinical outcomes, prognostic factors, and the role of adjuvant therapy. Gastrointest Cancer Res. 2011;4:
116–121.
88. Iacobuzio-Donahue CA, Klimstra DS, Adsay NV, et al. Dpc-4 protein is expressed in virtually all human intraductal
papillary mucinous neoplasms of the pancreas: comparison with conventional ductal adenocarcinomas. Am J
Pathol. 2000;157:755–761.
89. Grutzmann R, Post S, Saeger HD, Niedergethmann M. Intraductal papillary mucinous neoplasia (IPMN) of the
pancreas: its diagnosis, treatment, and prognosis. Dtsch Arztebl Int. 2011;108:788–794.
90. Traverso LW, Moriya T, Hashimoto Y. Intraductal papillary mucinous neoplasms of the pancreas: making a
disposition using the natural history. Curr Gastroenterol Rep. 2012;14:106–111.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 143

91. Kang MJ, Jang JY, Lee KB, Chang YR, Kwon W, Kim SW. Long-term prospective cohort study of patients undergoing
pancreatectomy for intraductal papillary mucinous neoplasm of the pancreas: implications for postoperative
surveillance. Ann Surg. 2014;260:356–363.
92. He J, Cameron JL, Ahuja N, et al. Is it necessary to follow patients after resection of a benign pancreatic intraductal
papillary mucinous neoplasm? J Am Coll Surg. 2013;216:657–665. [discussion 65-7].
93. Hong SM, Park JY, Hruban RH, Goggins M. Molecular signatures of pancreatic cancer. Arch Pathol Lab Med.
2011;135:716–727.
94. Kim TS, Fernandez-Del Castillo C. Diagnosis and management of pancreatic cystic neoplasms. Hematol Oncol Clin
North Am. 2015;29:655–674.
95. Hart PA, Chari ST. Management of pancreatic mucinous cystic neoplasms: surgery or surveillance? Pancreatology.
2015;15:97–98.
96. Robinson SM, Scott J, Oppong KW, White SA. What to do for the incidental pancreatic cystic lesion? Surg Oncol.
2014;23:117–125.
97. Crippa S, Salvia R, Warshaw AL, et al. Mucinous cystic neoplasm of the pancreas is not an aggressive entity: lessons
from 163 resected patients. Ann Surg. 2008;247:571–579.
98. Yamao K, Yanagisawa A, Takahashi K, et al. Clinicopathological features and prognosis of mucinous cystic neoplasm
with ovarian-type stroma: a multi-institutional study of the Japan pancreas society. Pancreas. 2011;40:67–71.
99. Haugk B. Pancreatic intraepithelial neoplasia—can we detect early pancreatic cancer? Histopathology. 2010;57:
503–514.
100. Corbo V, Tortora G, Scarpa A. Molecular pathology of pancreatic cancer: from bench-to-bedside translation. Curr
Drug Targets. 2012;13:744–752.
101. Hristov AC, Cope L, Di Cello F, et al. HMGA1 correlates with advanced tumor grade and decreased survival in
pancreatic ductal adenocarcinoma. Mod Pathol. 2010;23:98–104.
102. Bailey JM, Alsina J, Rasheed ZA, et al. DCLK1 marks a morphologically distinct subpopulation of cells with stem cell
properties in preinvasive pancreatic cancer. Gastroenterology. 2014;146:245–256.
103. Iacobuzio-Donahue CA. Genetic evolution of pancreatic cancer: lessons learnt from the pancreatic cancer genome
sequencing project. Gut. 2012;61:1085–1094.
104. Fendrich V, Chen NM, Neef M, et al. The angiotensin-I-converting enzyme inhibitor enalapril and aspirin delay
progression of pancreatic intraepithelial neoplasia and cancer formation in a genetically engineered mouse model
of pancreatic cancer. Gut. 2010;59:630–637.
105. Matthaei H, Hong SM, Mayo SC, et al. Presence of pancreatic intraepithelial neoplasia in the pancreatic transection
margin does not influence outcome in patients with R0 resected pancreatic cancer. Ann Surg Oncol. 2011;18:
3493–3499.
106. Watanabe I, Sasaki S, Konishi M, et al. Onset symptoms and tumor locations as prognostic factors of pancreatic
cancer. Pancreas. 2004;28:160–165.
107. Takhar AS, Palaniappan P, Dhingsa R, Lobo DN. Recent developments in diagnosis of pancreatic cancer. BMJ.
2004;329:668–673.
108. Porta M, Fabregat X, Malats N, et al. Exocrine pancreatic cancer: symptoms at presentation and their relation to
tumour site and stage. Clin Transl Oncol. 2005;7:189–197.
109. Bussom S, Saif MW. Methods and rationale for the early detection of pancreatic cancer. Highlights from the "2010
ASCO Gastrointestinal Cancers Symposium". Orlando, FL, USA. January 22-24, 2010. JOP. 2010;11:128–130.
110. Grover S, Syngal S. Hereditary pancreatic cancer. Gastroenterology. 2010;139:1076–1080. 80.e1-2.
111. Giardiello FM, Brensinger JD, Tersmette AC, et al. Very high risk of cancer in familial Peutz-Jeghers syndrome.
Gastroenterology. 2000;119:1447–1453.
112. Canto MI, Harinck F, Hruban RH, et al. International Cancer of the Pancreas Screening (CAPS) Consortium summit on
the management of patients with increased risk for familial pancreatic cancer. Gut. 2013;62:339–347.
113. Harinck F, Konings IC, Kluijt I, et al. A multicentre comparative prospective blinded analysis of EUS and MRI for
screening of pancreatic cancer in high-risk individuals. Gut. 2015, doi: 10.1136/gutjnl-2014-308008.
114. Goggins M. Molecular markers of early pancreatic cancer. J Clin Oncol. 2005;23:4524–4531.
115. Pleskow DK, Berger HJ, Gyves J, Allen E, McLean A, Podolsky DK. Evaluation of a serologic marker, CA19-9, in the
diagnosis of pancreatic cancer. Ann Intern Med. 1989;110:704–709.
116. Steinberg W. The clinical utility of the CA 19-9 tumor-associated antigen. Am J Gastroenterol. 1990;85:350–355.
117. Hartwig W, Strobel O, Hinz U, et al. CA19-9 in potentially resectable pancreatic cancer: perspective to adjust
surgical and perioperative therapy. Ann Surg Oncol. 2013;20:2188–2196.
118. Ballehaninna UK, Chamberlain RS. The clinical utility of serum CA 19-9 in the diagnosis, prognosis and management
of pancreatic adenocarcinoma: An evidence based appraisal. J Gastrointest Oncol. 2012;3:105–119.
119. Hata S, Sakamoto Y, Yamamoto Y, et al. Prognostic impact of postoperative serum CA 19-9 levels in patients with
resectable pancreatic cancer. Ann Surg Oncol. 2012;19:636–641.
120. Maisey NR, Norman AR, Hill A, Massey A, Oates J, Cunningham D. CA19-9 as a prognostic factor in inoperable
pancreatic cancer: the implication for clinical trials. Br J Cancer. 2005;93:740–743.
121. Sperti C, Pasquali C, Catalini S, et al. CA 19-9 as a prognostic index after resection for pancreatic cancer. J Surg Oncol.
1993;52:137–141.
122. Ballehaninna UK, Chamberlain RS, Serum CA. 19-9 as a Biomarker for Pancreatic Cancer-A Comprehensive Review.
Indian J Surg Oncol. 2011;2:88–100.
123. Melo SA, Luecke LB, Kahlert C, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer.
Nature. 2015;523:177–182.
124. Shrikhande SV, Barreto SG, Goel M, Arya S. Multimodality imaging of pancreatic ductal adenocarcinoma: a review
of the literature. HPB (Oxford)n. 2012;14:658–668.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
144 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

125. Callery MP, Chang KJ, Fishman EK, Talamonti MS, William Traverso L, Linehan DC. Pretreatment assessment of
resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16:
1727–1733.
126. Bronstein YL, Loyer EM, Kaur H, et al. Detection of small pancreatic tumors with multiphasic helical CT. AJR Am J
Roentgenol. 2004;182:619–623.
127. Boujaoude J. Role of endoscopic ultrasound in diagnosis and therapy of pancreatic adenocarcinoma. World J
Gastrointest Oncol. 2007;13:3662–3666.
128. Eloubeidi MA, Tamhane A, Varadarajulu S, Wilcox CM. Frequency of major complications after EUS-guided FNA of
solid pancreatic masses: a prospective evaluation. Gastrointest Endosc. 2006;63:622–629.
129. Hewitt MJ, McPhail MJ, Possamai L, Dhar A, Vlavianos P, Monahan KJ. EUS-guided FNA for diagnosis of solid
pancreatic neoplasms: a meta-analysis. Gastrointest Endosc. 2012;75:319–331.
130. Rodriguez S, Faigel D. Absence of a dilated duct predicts benign disease in suspected pancreas cancer: a simple
clinical rule. Dig Dis Sci. 2010;55:1161–1166.
131. Edge S. AJCC Cancer Staging Manual. 7th ed. New York, NY: Springer; 2010.
132. Rochefort MM, Ankeny JS, Kadera BE, et al. Impact of tumor grade on pancreatic cancer prognosis: validation of a
novel TNMG staging system. Ann Surg Oncol. 2013;20:4322–4329.
133. Pancreatic Adenocarcinoma: NCCN Clinical Practice Guidelines in Oncology Version 2. 〈http://www.nccn.org/
professionals/physician_gls/pdf/pancreatic.pdf〉; 2015 Accessed 31.08.15.
134. Katz MH, Marsh R, Herman JM, et al. Borderline resectable pancreatic cancer: need for standardization and
methods for optimal clinical trial design. Ann Surg Oncol. 2013;20:2787–2795.
135. Schima W, Fugger R, Schober E, et al. Diagnosis and staging of pancreatic cancer: comparison of mangafodipir
trisodium-enhanced MR imaging and contrast-enhanced helical hydro-CT. AJR Am J Roentgenol. 2002;179:
717–724.
136. Al-Hawary MM, Francis IR, Chari ST, et al. Pancreatic ductal adenocarcinoma radiology reporting template:
consensus statement of the society of abdominal radiology and the american pancreatic association. Gastro-
enterology. 2014;146:291–304. e1.
137. Ellsmere J, Mortele K, Sahani D, et al. Does multidetector-row CT eliminate the role of diagnostic laparoscopy in
assessing the resectability of pancreatic head adenocarcinoma? Surg Endosc. 2005;19:369–373.
138. House MG, Yeo CJ, Cameron JL, et al. Predicting resectability of periampullary cancer with three-dimensional
computed tomography. J Gastrointest Surg. 2004;8:280–288.
139. Roche CJ, Hughes ML, Garvey CJ, et al. CT and pathologic assessment of prospective nodal staging in patients with
ductal adenocarcinoma of the head of the pancreas. AJR Am J Roentgenol. 2003;180:475–480.
140. Tamm EP, Loyer EM, Faria S, et al. Staging of pancreatic cancer with multidetector CT in the setting of preoperative
chemoradiation therapy. Abdom Imaging. 2006;31:568–574.
141. Holzapfel K, Reiser-Erkan C, Fingerle AA, et al. Comparison of diffusion-weighted MR imaging and multidetector-
row CT in the detection of liver metastases in patients operated for pancreatic cancer. Abdom Imaging. 2011;36:
179–184.
142. Heinrich S, Goerres GW, Schafer M, et al. Positron emission tomography/computed tomography influences on the
management of resectable pancreatic cancer and its cost-effectiveness. Ann Surg. 2005;242:235–243.
143. Bang S, Chung HW, Park SW, et al. The clinical usefulness of 18-fluorodeoxyglucose positron emission tomography
in the differential diagnosis, staging, and response evaluation after concurrent chemoradiotherapy for pancreatic
cancer. J Clin Gastroenterol. 2006;40:923–929.
144. Pappas SG, Christians KK, Tolat PP, et al. Staging chest computed tomography and positron emission tomography in
patients with pancreatic adenocarcinoma: utility or futility? HPB. 2014;16:70–74.
145. Bernheim IV BM. Organoscopy: cystoscopy of the abdominal cavity. Ann Surg. 1911;53:764–767.
146. Mayo SC, Austin DF, Sheppard BC, Mori M, Shipley DK, Billingsley KG. Evolving preoperative evaluation
of patients with pancreatic cancer: does laparoscopy have a role in the current era? J Am Coll Surg. 2009;208:
87–95.
147. Jimenez RE, Warshaw AL, Rattner DW, Willett CG, McGrath D, Fernandez-del Castillo C. Impact of laparoscopic
staging in the treatment of pancreatic cancer. Arch Surg. 2000;135:409–414. [discussion 14-5].
148. Ahmed SI, Bochkarev V, Oleynikov D, Sasson AR. Patients with pancreatic adenocarcinoma benefit from staging
laparoscopy. J Laparoendosc Adv Surg Tech A. 2006;16:458–463.
149. Warshaw AL, Tepper JE, Shipley WU. Laparoscopy in the staging and planning of therapy for pancreatic cancer. Am J
Surg. 1986;151:76–80.
150. Schnelldorfer T, Gagnon AI, Birkett RT, Reynolds G, Murphy KM, Jenkins RL. Staging laparoscopy in pancreatic
cancer: a potential role for advanced laparoscopic techniques. J Am Coll Surg. 2014;218:1201–1206.
151. Contreras CM, Stanelle EJ, Mansour J, et al. Staging laparoscopy enhances the detection of occult metastases in
patients with pancreatic adenocarcinoma. J Surg Oncol. 2009;100:663–669.
152. Allen VB, Gurusamy KS, Takwoingi Y, Kalia A, Davidson BR. Diagnostic accuracy of laparoscopy following computed
tomography (CT) scanning for assessing the resectability with curative intent in pancreatic and periampullary
cancer. Cochrane Database Syst Rev. 2013;11:CD009323.
153. Tapper E, Kalb B, Martin DR, Kooby D, Adsay NV, Sarmiento JM. Staging laparoscopy for proximal pancreatic cancer
in a magnetic resonance imaging-driven practice: what’s it worth? HPB. 2011;13:732–737.
154. Velanovich V. The effects of staging laparoscopy on trocar site and peritoneal recurrence of pancreatic cancer. Surg
Endosc. 2004;18:310–313.
155. Yoshioka R, Saiura A, Koga R, et al. The implications of positive peritoneal lavage cytology in potentially resectable
pancreatic cancer. World J Surg. 2012;36:2187–2191.
156. Meszoely IM, Lee JS, Watson JC, Meyers M, Wang H, Hoffman JP. Peritoneal cytology in patients with potentially
resectable adenocarcinoma of the pancreas. Am Surg. 2004;70:208–213. discussion 13-4.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 145

157. Camacho D, Reichenbach D, Duerr GD, Venema TL, Sweeney JF, Fisher WE. Value of laparoscopy in the staging of
pancreatic cancer. JOP. 2005;6:552–561.
158. Pisters PW, Lee JE, Vauthey JN, Charnsangavej C, Evans DB. Laparoscopy in the staging of pancreatic cancer. Br
J Surg. 2001;88:325–337.
159. Karachristos A, Scarmeas N, Hoffman JP. CA 19-9 levels predict results of staging laparoscopy in pancreatic cancer. J
Gastrointest Surg. 2005;9:1286–1292.
160. Lewis JJ, Kowalski TE. Endoscopic ultrasound and fine needle aspiration in pancreatic cancer. Cancer J. 2012;18:
523–529.
161. Nawaz H, Fan CY, Kloke J, et al. Performance characteristics of endoscopic ultrasound in the staging of pancreatic
cancer: a meta-analysis. JOP. 2013;14:484–497.
162. Krishna NB, Mehra M, Reddy AV, Agarwal B. EUS/EUS-FNA for suspected pancreatic cancer: influence of chronic
pancreatitis and clinical presentation with or without obstructive jaundice on performance characteristics.
Gastrointest Endosc. 2009;70:70–79.
163. Katz MH, Fleming JB, Lee JE, Pisters PW. Current status of adjuvant therapy for pancreatic cancer. Oncologist.
2010;15:1205–1213.
164. Parmar AD, Vargas GM, Tamirisa NP, Sheffield KM, Riall TS. Trajectory of care and use of multimodality therapy in
older patients with pancreatic adenocarcinoma. Surgery. 2014;156:280–289.
165. Haeno H, Gonen M, Davis MB, Herman JM, Iacobuzio-Donahue CA, Michor F. Computational modeling of pancreatic
cancer reveals kinetics of metastasis suggesting optimum treatment strategies. Cell. 2012;148:362–375.
166. Palmer DH, Stocken DD, Hewitt H, et al. A randomized phase 2 trial of neoadjuvant chemotherapy in resectable
pancreatic cancer: gemcitabine alone versus gemcitabine combined with cisplatin. Ann Surg Oncol. 2007;14:
2088–2096.
167. Gillen S, Schuster T, Meyer Zum Buschenfelde C, Friess H, Kleeff J. Preoperative/neoadjuvant therapy in pancreatic
cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med. 2010;7:e1000267.
168. Ferrone CR, Marchegiani G, Hong TS, et al. Radiological and surgical implications of neoadjuvant treatment with
FOLFIRINOX for locally advanced and borderline resectable pancreatic cancer. Ann Surg. 2015;261:12–17.
169. Sherman WH, Chu K, Chabot J, et al. Neoadjuvant gemcitabine, docetaxel, and capecitabine followed by
gemcitabine and capecitabine/radiation therapy and surgery in locally advanced, unresectable pancreatic
adenocarcinoma. Cancer. 2015;121:673–680.
170. Varadhachary GR, Wolff RA, Crane CH, et al. Preoperative gemcitabine and cisplatin followed by gemcitabine-based
chemoradiation for resectable adenocarcinoma of the pancreatic head. J Clin Oncol. 2008;26:3487–3495.
171. Evans DB, Varadhachary GR, Crane CH, et al. Preoperative gemcitabine-based chemoradiation for patients with
resectable adenocarcinoma of the pancreatic head. J Clin Oncol. 2008;26:3496–3502.
172. Tzeng CW, Tran Cao HS, Lee JE, et al. Treatment sequencing for resectable pancreatic cancer: influence of early
metastases and surgical complications on multimodality therapy completion and survival. J Gastrointest Surg.
2014;18:16–24. [discussion-5].
173. Tempero MA, Malafa MP, Behrman SW, et al. Pancreatic adenocarcinoma, version 2.2014: featured updates to the
NCCN guidelines. J Natl Compr Canc Netw. 2014;12:1083–1093.
174. Golcher H, Brunner TB, Witzigmann H, et al. Neoadjuvant chemoradiation therapy with gemcitabine/cisplatin and
surgery versus immediate surgery in resectable pancreatic cancer: results of the first prospective randomized phase
II trial. Strahlenther Onkol. 2015;191:7–16.
175. Papavasiliou P, Hoffman JP, Cohen SJ, Meyer JE, Watson JC, Chun YS. Impact of preoperative therapy on patterns of
recurrence in pancreatic cancer. HPB. 2014;16:34–39.
176. VanHouten JP, White RR, Jackson GP. A decision model of therapy for potentially resectable pancreatic cancer. J Surg
Res. 2012;174:222–230.
177. Sharma G, Whang EE, Ruan DT, Ito H. Efficacy of neoadjuvant versus adjuvant therapy for resectable pancreatic
adenocarcinoma: a decision analysis. Ann Surg Oncol. 2015;22(Suppl 3):1229–1237.
178. Abbott DE, Tzeng CW, Merkow RP, et al. The cost-effectiveness of neoadjuvant chemoradiation is superior to a
surgery-first approach in the treatment of pancreatic head adenocarcinoma. Ann Surg Oncol. 2013;20(suppl 3):
S500–S508.
179. Evans DB, Ritch PS, Erickson BA. Neoadjuvant therapy for localized pancreatic cancer: support is growing? Ann Surg.
2015;261:18–20.
180. Adams MA, Anderson MA, Myles JD, Khalatbari S, Scheiman JM. Self-expanding metal stents (SEMS) provide
superior outcomes compared to plastic stents for pancreatic cancer patients undergoing neoadjuvant therapy.
J Gastrointest Oncol. 2012;3:309–313.
181. Heinrich S, Schafer M, Weber A, et al. Neoadjuvant chemotherapy generates a significant tumor response in
resectable pancreatic cancer without increasing morbidity: results of a prospective phase II trial. Ann Surg.
2008;248:1014–1022.
182. Regine WF, Winter KA, Abrams RA, et al. Fluorouracil vs gemcitabine chemotherapy before and after fluorouracil-
based chemoradiation following resection of pancreatic adenocarcinoma: a randomized controlled trial. J Am Med
Assoc. 2008;299:1019–1026.
183. Oettle H, Post S, Neuhaus P, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing
curative-intent resection of pancreatic cancer: a randomized controlled trial. J Am Med Assoc. 2007;297:267–277.
184. Bilimoria KY, Bentrem DJ, Ko CY, et al. Multimodality therapy for pancreatic cancer in the U.S.: utilization,
outcomes, and the effect of hospital volume. Cancer. 2007;110:1227–1234.
185. Raigani S, Ammori J, Kim J, Hardacre JM. Trends in the treatment of resectable pancreatic adenocarcinoma. J
Gastrointest Surg. 2014;18:113–123.
186. Barbas AS, Turley RS, Ceppa EP, et al. Comparison of outcomes and the use of multimodality therapy in young and
elderly people undergoing surgical resection of pancreatic cancer. J Am Geriatr Soc. 2012;60:344–350.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
146 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

187. Kalser MH, Ellenberg SS. Pancreatic cancer. Adjuvant combined radiation and chemotherapy following curative
resection. Arch Surg. 1985;120:899–903.
188. Klinkenbijl JH, Jeekel J, Sahmoud T, et al. 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;230:776–782. [discussion 82-4].
189. Neoptolemos JP, Dunn JA, Stocken DD, et al. Adjuvant chemoradiotherapy and chemotherapy in resectable
pancreatic cancer: a randomised controlled trial. Lancet. 2001;358:1576–1585.
190. Kimple RJ, Russo S, Monjazeb A, Blackstock AW. The role of chemoradiation for patients with resectable or
potentially resectable pancreatic cancer. Expert Rev Anticancer Ther. 2012;12:469–480.
191. Corsini MM, Miller RC, Haddock MG, et al. Adjuvant radiotherapy and chemotherapy for pancreatic carcinoma: the
Mayo Clinic experience (1975-2005). J Clin Oncol. 2008;26:3511–3516.
192. Herman JM, Swartz MJ, Hsu CC, et al. Analysis of fluorouracil-based adjuvant chemotherapy and radiation after
pancreaticoduodenectomy for ductal adenocarcinoma of the pancreas: results of a large, prospectively collected
database at the Johns Hopkins Hospital. J Clin Oncol. 2008;26:3503–3510.
193. Spitz FR, Abbruzzese JL, Lee JE, et al. Preoperative and postoperative chemoradiation strategies in patients treated
with pancreaticoduodenectomy for adenocarcinoma of the pancreas. J Clin Oncol. 1997;15:928–937.
194. Valle JW, Palmer D, Jackson R, et al. Optimal duration and timing of adjuvant chemotherapy after definitive
surgery for ductal adenocarcinoma of the pancreas: ongoing lessons from the ESPAC-3 Study. J Clin Oncol. 2014;32:
504–512.
195. Abrams RA, Lowy AM, O’Reilly EM, Wolff RA, Picozzi VJ, Pisters PW. Combined modality treatment of resectable and
borderline resectable pancreas cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1751–1756.
196. Gillmore R, Laurence V, Raouf S, et al. Chemoradiotherapy with or without induction chemotherapy for locally
advanced pancreatic cancer: a UK multi-institutional experience. Clin Oncol (R Coll Radiol). 2010;22:564–569.
197. Christians KK, Tsai S, Mahmoud A, et al. Neoadjuvant FOLFIRINOX for borderline resectable pancreas cancer: a new
treatment paradigm? Oncologist. 2014;19(3):266–274.
198. Rose JB, Rocha FG, Alseidi A, et al. Extended neoadjuvant chemotherapy for borderline resectable pancreatic cancer
demonstrates promising postoperative outcomes and survival. Ann Surg Oncol. 2014;21(5):1530–1537.
199. Kadera BE, Sunjaya DB, Isacoff WH, et al. Locally advanced pancreatic cancer: association between prolonged
preoperative treatment and lymph-node negativity and overall survival. JAMA Surg. 2014;149:145–153.
200. Petrelli F, Coinu A, Borgonovo K, et al. FOLFIRINOX-based neoadjuvant therapy in borderline resectable or
unresectable pancreatic cancer: a meta-analytical review of published studies. Pancreas. 2015;44:515–521.
201. Lee JJ, Huang J, England CG, McNally LR, Frieboes HB. Predictive modeling of in vivo response to gemcitabine in
pancreatic cancer. PLoS Comput Biol. 2013;9:e1003231.
202. Valentini V, Morganti AG, Macchia G, et al. Intraoperative radiation therapy in resected pancreatic carcinoma: long-
term analysis. Int J Radiat Oncol Biol Phys. 2008;70:1094–1099.
203. Morganti AG, Picardi V, Ippolito E, et al. Capecitabine based postoperative accelerated chemoradiation of pancreatic
carcinoma. A dose-escalation study. Acta Oncol. 2010;49:418–422.
204. Berber B, Sanabria JR, Braun K, et al. Emerging role of stereotactic body radiotherapy in the treatment of pancreatic
cancer. Expert Rev Anticancer Ther. 2013;13:481–487.
205. Combs SE, Habermehl D, Kessel K, et al. Intensity modulated radiotherapy as neoadjuvant chemoradiation for the
treatment of patients with locally advanced pancreatic cancer. Outcome analysis and comparison with a 3D-treated
patient cohort. Strahlenther Onkol. 2013;189:738–744.
206. Milano MT, Chmura SJ, Garofalo MC, et al. Intensity-modulated radiotherapy in treatment of pancreatic and bile
duct malignancies: toxicity and clinical outcome. Int J Radiat Oncol Biol Phys. 2004;59:445–453.
207. Abelson JA, Murphy JD, Minn AY, et al. Intensity-modulated radiotherapy for pancreatic adenocarcinoma. Int J
Radiat Oncol Biol Phys. 2012;82:e595–e601.
208. Ben-Josef E, Schipper M, Francis IR, et al. A phase I/II trial of intensity modulated radiation (IMRT) dose escalation
with concurrent fixed-dose rate gemcitabine (FDR-G) in patients with unresectable pancreatic cancer. Int J Radiat
Oncol Biol Phys. 2012;84:1166–1171.
209. Murphy JD, Chang DT, Abelson J, et al. Cost-effectiveness of modern radiotherapy techniques in locally advanced
pancreatic cancer. Cancer. 2012;118:1119–1129.
210. Goyal K, Einstein D, Ibarra RA, et al. Stereotactic body radiation therapy for nonresectable tumors of the pancreas.
J Surg Res. 2012;174:319–325.
211. Chang DT, Schellenberg D, Shen J, et al. Stereotactic radiotherapy for unresectable adenocarcinoma of the pancreas.
Cancer. 2009;115:665–672.
212. Chuong MD, Springett GM, Freilich JM, et al. Stereotactic body radiation therapy for locally advanced and borderline
resectable pancreatic cancer is effective and well tolerated. Int J Radiat Oncol Biol Phys. 2013;86:516–522.
213. Rwigema JC, Heron DE, Parikh SD, et al. Adjuvant stereotactic body radiotherapy for resected pancreatic
adenocarcinoma with close or positive margins. J Gastrointest Cancer. 2012;43:70–76.
214. Chauffert B, Mornex F, Bonnetain F, et al. Phase III trial comparing intensive induction chemoradiotherapy (60 Gy,
infusional 5-FU and intermittent cisplatin) followed by maintenance gemcitabine with gemcitabine alone for
locally advanced unresectable pancreatic cancer. Definitive results of the 2000-01 FFCD/SFRO study. Ann Oncol.
2008;19:1592–1599.
215. Nichols Jr. RC, Huh SN, Prado KL, et al. Protons offer reduced normal-tissue exposure for patients receiving
postoperative radiotherapy for resected pancreatic head cancer. Int J Radiat Oncol Biol Phys. 2012;83:158–163.
216. Hong TS, Ryan DP, Blaszkowsky LS, et al. Phase I study of preoperative short-course chemoradiation with proton
beam therapy and capecitabine for resectable pancreatic ductal adenocarcinoma of the head. Int J Radiat Oncol Biol
Phys. 2011;79:151–157.
217. Wo JY, Mamon HJ, Ferrone CR, et al. Phase I study of neoadjuvant accelerated short course radiation therapy with
photons and capecitabine for resectable pancreatic cancer. Radiother Oncol. 2013;110(1):160–164.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 147

218. Saif MW. Advancements in the management of pancreatic cancer: 2013. JOP. 2013;14:112–118.
219. Guo XZ, Cui ZM, Liu X. Current developments, problems and solutions in the non-surgical treatment of pancreatic
cancer. World J Gastrointest Oncol. 2013;5:20–28.
220. Spano JP, Chodkiewicz C, Maurel J, et al. Efficacy of gemcitabine plus axitinib compared with gemcitabine
alone in patients with advanced pancreatic cancer: an open-label randomised phase II study. Lancet. 2008;371:
2101–2108.
221. Ma WW, Herman JM, Jimeno A, et al. A tolerability and pharmacokinetic study of adjuvant erlotinib and
capecitabine with concurrent radiation in resected pancreatic cancer. Transl Oncol. 2010;3:373–379.
222. Brett BT, Caplin ME. Towards immunotherapy for pancreatic cancer. Gut. 2000;46:582–583.
223. Abou-Alfa GK, Chapman PB, Feilchenfeldt J, et al. Targeting mutated K-ras in pancreatic adenocarcinoma using an
adjuvant vaccine. Am J Clin Oncol. 2011;34:321–325.
224. Ramanathan RK, Lee KM, McKolanis J, et al. Phase I study of a MUC1 vaccine composed of different doses of MUC1
peptide with SB-AS2 adjuvant in resected and locally advanced pancreatic cancer. Cancer Immunol Immunother.
2005;54:254–264.
225. Bernhardt SL, Gjertsen MK, Trachsel S, et al. Telomerase peptide vaccination of patients with non-resectable
pancreatic cancer: A dose escalating phase I/II study. Br J Cancer. 2006;95:1474–1482.
226. Koido S, Homma S, Takahara A, et al. Current immunotherapeutic approaches in pancreatic cancer. Clin Dev
Immunol. 2011;2011:267539.
227. Lutz E, Yeo CJ, Lillemoe KD, et al. A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-
secreting tumor vaccine for pancreatic adenocarcinoma. A Phase II trial of safety, efficacy, and immune activation.
Ann Surg. 2011;253:328–335.
228. Gilliam AD, Broome P, Topuzov EG, et al. An international multicenter randomized controlled trial of G17DT in
patients with pancreatic cancer. Pancreas. 2012;41:374–379.
229. Beatty GL, Chiorean EG, Fishman MP, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic
carcinoma in mice and humans. Science. 2011;331:1612–1616.
230. La Torre M, Ziparo V, Nigri G, Cavallini M, Balducci G, Ramacciato G. Malnutrition and pancreatic surgery:
prevalence and outcomes. J Surg Oncol. 2013;107:702–708.
231. Afaneh C, Gerszberg D, Slattery E, Seres DS, Chabot JA, Kluger MD. Pancreatic cancer surgery and nutrition
management: a review of the current literature. Hepatobiliary Surg Nutr. 2015;4:59–71.
232. Eshuis WJ, van der Gaag NA, Rauws EA, et al. Therapeutic delay and survival after surgery for cancer of the
pancreatic head with or without preoperative biliary drainage. Ann Surg. 2010;252:840–849.
233. van der Gaag NA, Rauws EA, van Eijck CH, et al. Preoperative biliary drainage for cancer of the head of the pancreas.
N Engl J Med. 2010;362:129–137.
234. Fang Y, Gurusamy KS, Wang Q, et al. Pre-operative biliary drainage for obstructive jaundice. Cochrane Database Syst
Rev. 2012;9:CD005444.
235. Sauvanet A, Boher JM, Paye F, et al. Severe jaundice increases early severe morbidity and decreases long-term
survival after pancreaticoduodenectomy for pancreatic adenocarcinoma. J Am Coll Surg. 2015;221:380–389.
236. Christians KK, Tsai S, Tolat PP, Evans DB. Critical steps for pancreaticoduodenectomy in the setting of pancreatic
adenocarcinoma. J Surg Oncol. 2013;107:33–38.
237. Myatra S, Divatia JV, Jibhkate B, Barreto GS, Shrikhande SV. Preoperative assessment and optimization in
periampullary and pancreatic cancer. Indian J Cancer. 2011;48:86–93.
238. Hodul P, Tansey J, Golts E, Oh D, Pickleman J, Aranha GV. Age is not a contraindication to pancreaticoduodenectomy.
Am Surg. 2001;67:270–275. [discussion 5-6].
239. Hatzaras I, Schmidt C, Klemanski D, et al. Pancreatic resection in the octogenarian: a safe option for pancreatic
malignancy. J Am Coll Surg. 2011;212:373–377.
240. Riall TS, Sheffield KM, Kuo YF, Townsend Jr. CM, Goodwin JS. Resection benefits older adults with locoregional
pancreatic cancer despite greater short-term morbidity and mortality. J Am Geriatr Soc. 2011;59:647–654.
241. Melis M, Marcon F, Masi A, et al. The safety of a pancreaticoduodenectomy in patients older than 80 years: risk vs.
benefits. HPB. 2012;14:583–588.
242. Oken MM, Creech RH, Tormey DC, et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group.
Am J Clin Oncol. 1982;5:649–655.
243. Hill JS, Zhou Z, Simons JP, et al. A simple risk score to predict in-hospital mortality after pancreatic resection for
cancer. Ann Surg Oncol. 2010;17:1802–1807.
244. Greenblatt DY, Kelly KJ, Rajamanickam V, et al. Preoperative factors predict perioperative morbidity and mortality
after pancreaticoduodenectomy. Ann Surg Oncol. 2011;18:2126–2135.
245. Kelly KJ, Greenblatt DY, Wan Y, et al. Risk stratification for distal pancreatectomy utilizing ACS-NSQIP: preoperative
factors predict morbidity and mortality. J Gastrointest Surg. 2011;15:250–259. [discussion 9-61].
246. Pratt W, Joseph S, Callery MP, Vollmer Jr. CM. POSSUM accurately predicts morbidity for pancreatic resection.
Surgery. 2008;143:8–19.
247. Gawande AA, Kwaan MR, Regenbogen SE, Lipsitz SA, Zinner MJ. An Apgar score for surgery. J Am Coll Surg.
2007;204:201–208.
248. Whipple AO, Parsons WB, Mullins CR. Treatment of carcinoma of the ampulla of vater. Ann Surg. 1935;102:
763–779.
249. D.K. NAaL. Neoplasms of the exocrine pancreas. In: Michael Mulholland KL, Gerard Doherty, Ronald Maier, Diane
Simeone, Gilbert Upchurch, ed. Greenfield’s Surgery: Scientific Principles and Practice. 5th ed. Philadelphia:
Lippincott Williams & Watkins.
250. Murr MM, Sarr MG, Oishi AJ, van Heerden JA. Pancreatic cancer. CA Cancer J Clin. 1994;44:304–318.
251. Adsay NV, Basturk O, Saka B, et al. Whipple made simple for surgical pathologists: orientation, dissection, and
sampling of pancreaticoduodenectomy specimens for a more practical and accurate evaluation of pancreatic, distal
common bile duct, and ampullary tumors. The American journal of surgical pathology. 2014;38(4):480–493.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
148 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

252. Pitt HA, Grace PA. Cancer of the pancreas. Pylorus-preserving resection of the pancreas. Baillieres Clin Gastroenterol.
1990;4:917–930.
253. Diener MK, Fitzmaurice C, Schwarzer G, et al. Pylorus-preserving pancreaticoduodenectomy (pp Whipple) versus
pancreaticoduodenectomy (classic Whipple) for surgical treatment of periampullary and pancreatic carcinoma.
Cochrane Database Syst Rev. 2014;11:CD006053.
254. Klinkenbijl JH, van der Schelling GP, Hop WC, van Pel R, Bruining HA, Jeekel J. The advantages of pylorus-preserving
pancreatoduodenectomy in malignant disease of the pancreas and periampullary region. Ann Surg. 1992;216:
142–145.
255. Kawai M, Tani M, Hirono S, et al. Pylorus ring resection reduces delayed gastric emptying in patients undergoing
pancreatoduodenectomy: a prospective, randomized, controlled trial of pylorus-resecting versus pylorus-
preserving pancreatoduodenectomy. Ann Surg. 2011;253:495–501.
256. Tani M, Terasawa H, Kawai M, et al. Improvement of delayed gastric emptying in pylorus-preserving
pancreaticoduodenectomy: results of a prospective, randomized, controlled trial. Ann Surg. 2006;243:316–320.
257. Gaedcke J, Gunawan B, Grade M, et al. The mesopancreas is the primary site for R1 resection in pancreatic head
cancer: relevance for clinical trials. Langenbecks Arch Surg. 2010;395:451–458.
258. Baque P, Iannelli A, Delotte J, de Peretti F, Bourgeon A. Division of the right posterior attachments of the head of the
pancreas with a linear stapler during pancreaticoduodenectomy: vascular and oncological considerations based on
an anatomical cadaver-based study. Surg Radiol Anat. 2009;31:13–17.
259. Hackert T, Werner J, Weitz J, Schmidt J, Buchler MW. Uncinate process first—a novel approach for pancreatic head
resection. Langenbecks Arch Surg. 2010;395:1161–1164.
260. Pessaux P, Rosso E, Panaro F, et al. Preliminary experience with the hanging maneuver for pancreaticoduode-
nectomy. Eur J Surg Oncol. 2009;35:1006–1010.
261. Artinyan A, Soriano PA, Prendergast C, Low T, Ellenhorn JD, Kim J. The anatomic location of pancreatic cancer is a
prognostic factor for survival. HPB. 2008;10:371–376.
262. van Heerden JA, McIlrath DC, Ilstrup DM, Weiland LH. Total pancreatectomy for ductal adenocarcinoma of the
pancreas: an update. World J Surg. 1988;12:658–662.
263. Schmidt CM, Glant J, Winter JM, et al. Total pancreatectomy (R0 resection) improves survival over subtotal
pancreatectomy in isolated neck margin positive pancreatic adenocarcinoma. Surgery. 2007;142:572–578.
[discussion 8-80].
264. Reddy S, Wolfgang CL, Cameron JL, et al. Total pancreatectomy for pancreatic adenocarcinoma: evaluation of
morbidity and long-term survival. Ann Surg. 2009;250:282–287.
265. Yeo CJ, Cameron JL, Lillemoe KD, et al. Pancreaticoduodenectomy with or without distal gastrectomy and extended
retroperitoneal lymphadenectomy for periampullary adenocarcinoma, part 2: randomized controlled trial
evaluating survival, morbidity, and mortality. Ann Surg. 2002;236:355–366. [discussion 66-8].
266. Riall TS, Cameron JL, Lillemoe KD, et al. Pancreaticoduodenectomy with or without distal gastrectomy and extended
retroperitoneal lymphadenectomy for periampullary adenocarcinoma—part 3: update on 5-year survival. J
Gastrointest Surg. 2005;9:1191–1204. [discussion 204-6].
267. Tomlinson JS, Jain S, Bentrem DJ, et al. Accuracy of staging node-negative pancreas cancer: a potential quality
measure. Arch Surg. 2007;142(8):767–773.
268. Slidell MB, Chang DC, Cameron JL, et al. Impact of total lymph node count and lymph node ratio on staging and
survival after pancreatectomy for pancreatic adenocarcinoma: a large, population-based analysis. Ann Surg Oncol.
2008;15:165–174.
269. Tol JA, Gouma DJ, Bassi C, et al. Definition of a standard lymphadenectomy in surgery for pancreatic ductal
adenocarcinoma: a consensus statement by the International Study Group on Pancreatic Surgery (ISGPS). Surgery.
2014;156:591–600.
270. 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:
265–273.
271. Jang JY, Kang MJ, Heo JS, et al. A prospective randomized controlled study comparing outcomes of standard
resection and extended resection, including dissection of the nerve plexus and various lymph nodes, in patients
with pancreatic head cancer. Ann Surg. 2014;259:656–664.
272. Henne-Bruns D, Vogel I, Luttges J, Kloppel G, Kremer B. Surgery for ductal adenocarcinoma of the pancreatic head:
staging, complications, and survival after regional versus extended lymphadenectomy. World J Surg. 2000;24:
595–601. [discussion-2].
273. Park HJ, You DD, Choi DW, Heo JS, Choi SH. Role of radical antegrade modular pancreatosplenectomy for
adenocarcinoma of the body and tail of the pancreas. World J Surg. 2014;38:186–193.
274. Bassi C, Falconi M, Molinari E, et al. Duct-to-mucosa versus end-to-side pancreaticojejunostomy reconstruction
after pancreaticoduodenectomy: results of a prospective randomized trial. Surgery. 2003;134:766–771.
275. Berger AC, Howard TJ, Kennedy EP, et al. Does type of pancreaticojejunostomy after pancreaticoduodenectomy
decrease rate of pancreatic fistula? A randomized, prospective, dual-institution trial. J Am Coll Surg. 2009;208:
738–747. [discussion 47-9].
276. Que W, Fang H, Yan B, et al. Pancreaticogastrostomy versus pancreaticojejunostomy after pancreaticoduodenec-
tomy: a meta-analysis of randomized controlled trials. Am J Surg. 2015;209:1074–1082.
277. Xiong JJ, Altaf K, Mukherjee R, et al. Systematic review and meta-analysis of outcomes after intraoperative
pancreatic duct stent placement during pancreaticoduodenectomy. Br J Surg. 2012;99:1050–1061.
278. Motoi F, Egawa S, Rikiyama T, Katayose Y, Unno M. Randomized clinical trial of external stent drainage of the
pancreatic duct to reduce postoperative pancreatic fistula after pancreaticojejunostomy. Br J Surg. 2012;99:
524–531.
279. Diener MK, Seiler CM, Rossion I, et al. Efficacy of stapler versus hand-sewn closure after distal pancreatectomy
(DISPACT): a randomised, controlled multicentre trial. Lancet. 2011;377:1514–1522.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 149

280. Carter TI, Fong ZV, Hyslop T, et al. A dual-institution randomized controlled trial of remnant closure after distal
pancreatectomy: does the addition of a falciform patch and fibrin glue improve outcomes? J Gastrointest Surg.
2013;17:102–109.
281. Frozanpor F, Lundell L, Segersvard R, Arnelo U. The effect of prophylactic transpapillary pancreatic stent insertion
on clinically significant leak rate following distal pancreatectomy: results of a prospective controlled clinical trial.
Ann Surg. 2012;255:1032–1036.
282. Keck T, Kusters S, Wellner UF, Hopt UT, Karcz KW. Total laparoscopic partial pancreatoduodenectomy and
reconstruction via laparoscopic pancreatogastrostomy. Langenbecks Arch Surg. 2012;397:1009–1012.
283. Gagner M, Pomp A. Laparoscopic pylorus-preserving pancreatoduodenectomy. Surg Endosc. 1994;8:408–410.
284. Baker EH, Ross SW, Seshadri R, et al. Robotic pancreaticoduodenectomy for pancreatic adenocarcinoma: role in
2014 and beyond. J Gastrointest Oncol. 2015;6:396–405.
285. Venkat R, Edil BH, Schulick RD, Lidor AO, Makary MA, Wolfgang CL. Laparoscopic distal pancreatectomy is
associated with significantly less overall morbidity compared to the open technique: a systematic review and
meta-analysis. Ann Surg. 2012;255:1048–1059.
286. Kooby DA, Hawkins WG, Schmidt CM, et al. A multicenter analysis of distal pancreatectomy for adenocarcinoma: is
laparoscopic resection appropriate? J Am Coll Surg. 2010;210(779-85):86–87.
287. Adam MA, Choudhury K, Goffredo P, et al. Minimally invasive distal pancreatectomy for cancer: short-term
oncologic outcomes in 1733 patients. World J Surg. 2015;39:2564–2572.
288. Kooby DA, Gillespie T, Bentrem D, et al. Left-sided pancreatectomy: a multicenter comparison of laparoscopic and
open approaches. Ann Surg. 2008;248:438–446.
289. Magge D, Gooding W, Choudry H, et al. Comparative effectiveness of minimally invasive and open distal
pancreatectomy for ductal adenocarcinoma. JAMA Surg. 2013;148:525–531.
290. Braga M, Ridolfi C, Balzano G, Castoldi R, Pecorelli N, Di Carlo V. Learning curve for laparoscopic distal
pancreatectomy in a high-volume hospital. Updates Surg. 2012;64:179–183.
291. Zhou NX, Chen JZ, Liu Q, et al. Outcomes of pancreatoduodenectomy with robotic surgery versus open surgery. Int J
Med Robot. 2011;7:131–137.
292. Abdelgadir Adam M, Choudhury K, Dinan MA, et al. Minimally invasive versus open pancreaticoduodenectomy for
cancer: practice patterns and short-term outcomes among 7061 patients. Ann Surg. 2015;262:372–377.
293. Sharpe SM, Talamonti MS, Wang CE, et al. Early national experience with laparoscopic pancreaticoduodenectomy
for ductal adenocarcinoma: a comparison of laparoscopic pancreaticoduodenectomy and open pancreaticoduo-
denectomy from the National Cancer Data Base. J Am Coll Surg. 2015;221:175–184.
294. Boone BA, Zenati M, Hogg ME, et al. Assessment of quality outcomes for robotic pancreaticoduodenectomy:
identification of the learning curve. JAMA Surg. 2015;150:416–422.
295. Bockhorn M, Uzunoglu FG, Adham M, et al. Borderline resectable pancreatic cancer: a consensus statement by the
International Study Group of Pancreatic Surgery (ISGPS). Surgery. 2014;155:977–988.
296. Evans DB, Farnell MB, Lillemoe KD, Vollmer Jr. C, Strasberg SM, Schulick RD. Surgical treatment of resectable and
borderline resectable pancreas cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1736–1744.
297. Sasson AR, Hoffman JP, Ross EA, Kagan SA, Pingpank JF, Eisenberg BL. En bloc resection for locally advanced cancer
of the pancreas: is it worthwhile? J Gastrointest Surg. 2002;6:147–157. discussion 57-8.
298. Weitz J, Kienle P, Schmidt J, Friess H, Buchler MW. Portal vein resection for advanced pancreatic head cancer. J Am
Coll Surg. 2007;204:712–716.
299. Shoup M, Conlon KC, Klimstra D, Brennan MF. Is extended resection for adenocarcinoma of the body or tail of the
pancreas justified? J Gastrointest Surg. 2003;7:946–952. discussion 52.
300. Yeo CJ, Cameron JL, Sohn TA, et al. Six hundred fifty consecutive pancreaticoduodenectomies in the 1990 s:
pathology, complications, and outcomes. Ann Surg. 1997;226:248–257. [discussion 57-60].
301. Chu CK, Farnell MB, Nguyen JH, et al. Prosthetic graft reconstruction after portal vein resection in
pancreaticoduodenectomy: a multicenter analysis. J Am Coll Surg. 2010;211:316–324.
302. Yamamoto Y, Sakamoto Y, Nara S, et al. Reconstruction of the portal and hepatic veins using venous grafts
customized from the bilateral gonadal veins. Langenbecks Arch Surg. 2009;394:1115–1121.
303. Fleming JB, Barnett CC, Clagett GP. Superficial femoral vein as a conduit for portal vein reconstruction during
pancreaticoduodenectomy. Arch Surg. 2005;140:698–701.
304. Suzuki T, Yoshidome H, Kimura F, et al. Renal function is well maintained after use of left renal vein graft for
vascular reconstruction in hepatobiliary-pancreatic surgery. J Am Coll Surg. 2006;202:87–92.
305. Lee DY, Mitchell EL, Jones MA, et al. Techniques and results of portal vein/superior mesenteric vein reconstruction
using femoral and saphenous vein during pancreaticoduodenectomy. J Vasc Surg. 2010;51:662–666.
306. Turley RS, Peterson K, Barbas AS, et al. Vascular surgery collaboration during pancreaticoduodenectomy with
vascular reconstruction. Ann Vasc Surg. 2012;26:685–692.
307. Kim SM, Min SK, Park D, et al. Reconstruction of portal vein and superior mesenteric vein after extensive resection
for pancreatic cancer. J Korean Surg Soc. 2013;84:346–352.
308. Bachellier P, Nakano H, Oussoultzoglou PD, et al. Is pancreaticoduodenectomy with mesentericoportal venous
resection safe and worthwhile? Am J Surg. 2001;182:120–129.
309. Aktekin A, Kucuk M, Odabasi M, et al. The importance of invasion and resection of superior mesenteric and portal
veins in adenocarcinoma of the pancreas. Hepatogastroenterology. 2013;60:1194–1198.
310. Chua TC, Saxena A. Extended pancreaticoduodenectomy with vascular resection for pancreatic cancer: a systematic
review. J Gastrointest Surg. 2010;14:1442–1452.
311. Zhou Y, Zhang Z, Liu Y, Li B, Xu D. Pancreatectomy combined with superior mesenteric vein-portal vein resection for
pancreatic cancer: a meta-analysis. World J Surg. 2012;36:884–891.
312. Mollberg N, Rahbari NN, Koch M, et al. Arterial resection during pancreatectomy for pancreatic cancer: a systematic
review and meta-analysis. Ann Surg. 2011;254:882–893.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
150 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

313. Fuhrman GM, Leach SD, Staley CA, et al. Rationale for en bloc vein resection in the treatment of pancreatic
adenocarcinoma adherent to the superior mesenteric-portal vein confluence. Pancreatic Tumor Study Group. Ann
Surg. 1996;223:154–162.
314. Hackert T, Buchler MW. Vascular resections in pancreatic cancer. Updates Surg. 2010;62:73–74.
315. Smoot RL, Donohue JH. Modified Appleby procedure for resection of tumors of the pancreatic body and tail with
celiac axis involvement. J Gastrointest Surg. 2012;16:2167–2169.
316. Beane JD, House MG, Pitt SC, et al. Distal pancreatectomy with celiac axis resection: what are the added risks? HPB.
2015;17:777–784.
317. Hirano S, Kondo S, Hara T, et al. Distal pancreatectomy with en bloc celiac axis resection for locally advanced
pancreatic body cancer: long-term results. Ann Surg. 2007;246:46–51.
318. Christians KK, Pilgrim CH, Tsai S, et al. Arterial resection at the time of pancreatectomy for cancer. Surgery.
2014;155:919–926.
319. Hartwig W, Hackert T, Hinz U, et al. Multivisceral resection for pancreatic malignancies: risk-analysis and long-term
outcome. Ann Surg. 2009;250:81–87.
320. Burdelski CM, Reeh M, Bogoevski D, et al. Multivisceral resections in pancreatic cancer: identification of risk factors.
World J Surg. 2011;35:2756–2763.
321. Worni M, Castleberry AW, Clary BM, et al. Concomitant vascular reconstruction during pancreatectomy for
malignant disease: a propensity score-adjusted, population-based trend analysis involving 10,206 patients. JAMA
Surg. 2013;148:331–338.
322. Ravikumar R, Sabin C, Abu Hilal M, et al. Portal vein resection in borderline resectable pancreatic cancer: a United
Kingdom multicenter study. J Am Coll Surg. 2014;218:401–411.
323. Smoot RL, Christein JD, Farnell MB. Durability of portal venous reconstruction following resection during
pancreaticoduodenectomy. J Gastrointest Surg. 2006;10:1371–1375.
324. Kneuertz PJ, Pitt HA, Bilimoria KY, et al. Risk of morbidity and mortality following hepato-pancreato-biliary
surgery. J Gastrointest Surg. 2012;16:1727–1735.
325. Glasgow RE, Jackson HH, Neumayer L, et al. Pancreatic resection in Veterans Affairs and selected university medical
centers: results of the patient safety in surgery study. J Am Coll Surg. 2007;204:1252–1260.
326. Parikh P, Shiloach M, Cohen ME, et al. Pancreatectomy risk calculator: an ACS-NSQIP resource. HPB. 2010;12:
488–497.
327. Xu M, Hines OJ. Long-term survival after pancreatic cancer: hope has arrived. JAMA Surg. 2015;150:710–711.
328. Ghaferi AA, Birkmeyer JD, Dimick JB. Variation in hospital mortality associated with inpatient surgery. N Engl J Med.
2009;361:1368–1375.
329. Mann CD, Palser T, Briggs CD, et al. A review of factors predicting perioperative death and early outcome in
hepatopancreaticobiliary cancer surgery. HPB. 2010;12:380–388.
330. Hartwig W, Werner J, Jager D, Debus J, Buchler MW. Improvement of surgical results for pancreatic cancer. Lancet
Oncol. 2013;14:e476–e485.
331. Bassi C, Dervenis C, Butturini G, et al. Postoperative pancreatic fistula: an international study group (ISGPF)
definition. Surgery. 2005;138:8–13.
332. McMillan MT, Vollmer Jr. CM, Asbun HJ, et al. The characterization and prediction of ISGPF Grade C fistulas
following pancreatoduodenectomy. J Gastrointest Surg. 2015.
333. Callery MP, Pratt WB, Kent TS, Chaikof EL, Vollmer Jr. CM. A prospectively validated clinical risk score accurately
predicts pancreatic fistula after pancreatoduodenectomy. J Am Coll Surg. 2013;216:1–14.
334. Gans SL, van Westreenen HL, Kiewiet JJ, Rauws EA, Gouma DJ, Boermeester MA. Systematic review and meta-
analysis of somatostatin analogues for the treatment of pancreatic fistula. Br J Surg. 2012;99:754–760.
335. De Martino RR, Goodney PP, Spangler EL, et al. Variation in thromboembolic complications among patients
undergoing commonly performed cancer operations. J Vasc Surg. 2012;55. 1035-40.e4.
336. Hyder O, Dodson RM, Nathan H, et al. Influence of patient, physician, and hospital factors on 30-day readmission
following pancreatoduodenectomy in the United States. JAMA Surg. 2013;148:1095–1102.
337. Begg CB, Cramer LD, Hoskins WJ, Brennan MF. Impact of hospital volume on operative mortality for major cancer
surgery. J Am Med Assoc. 1998;280:1747–1751.
338. Sohn TA, Yeo CJ, Cameron JL, et al. Resected adenocarcinoma of the pancreas-616 patients: results, outcomes, and
prognostic indicators. J Gastrointest Surg. 2000;4:567–579.
339. Finks JF, Osborne NH, Birkmeyer JD. Trends in hospital volume and operative mortality for high-risk surgery. N Engl
J Med. 2011;364:2128–2137.
340. Gooiker GA, van Gijn W, Wouters MW, Post PN, van de Velde CJ, Tollenaar RA. Systematic review and meta-analysis
of the volume-outcome relationship in pancreatic surgery. Br J Surg. 2011;98:485–494.
341. Bilimoria KY, Talamonti MS, Sener SF, et al. Effect of hospital volume on margin status after pancreaticoduode-
nectomy for cancer. J Am Coll Surg. 2008;207:510–519.
342. Bilimoria KY, Talamonti MS, Wayne JD, et al. Effect of hospital type and volume on lymph node evaluation for
gastric and pancreatic cancer. Arch Surg 2008;143:671-678; [discussion 8].
343. Joseph B, Morton JM, Hernandez-Boussard T, Rubinfeld I, Faraj C, Velanovich V. Relationship between
hospital volume, system clinical resources, and mortality in pancreatic resection. J Am Coll Surg. 2009;208:
520–527.
344. Ghaferi AA, Birkmeyer JD, Dimick JB. Hospital volume and failure to rescue with high-risk surgery. Med Care.
2011;49:1076–1081.
345. Merkow RP, Bentrem DJ, Chung JW, Paruch JL, Ko CY, Bilimoria KY. Differences in patients, surgical complexity, and
outcomes after cancer surgery at National Cancer Institute-designated cancer centers compared to other hospitals.
Med Care. 2013;51:606–613.
346. Lemmens VE, Bosscha K, van der Schelling G, Brenninkmeijer S, Coebergh JW, de Hingh IH. Improving outcome for
patients with pancreatic cancer through centralization. Br J Surg. 2011;98:1455–1462.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 151

347. Benassai G, Mastrorilli M, Quarto G, et al. Factors influencing survival after resection for ductal adenocarcinoma of
the head of the pancreas. J Surg Oncol. 2000;73:212–218.
348. Cameron JL, Crist DW, Sitzmann JV, et al. Factors influencing survival after pancreaticoduodenectomy for pancreatic
cancer. Am J Surg. 1991;161:120–124. [discussion 4-5].
349. Geer RJ, Brennan MF. Prognostic indicators for survival after resection of pancreatic adenocarcinoma. Am J Surg.
1993;165:68–72. [discussion-3].
350. Zhang JF, Hua R, Sun YW, et al. Influence of perineural invasion on survival and recurrence in patients with resected
pancreatic cancer. Asian Pac J Cancer Prev. 2013;14:5133–5139.
351. Epstein AS, Soff GA, Capanu M, et al. Analysis of incidence and clinical outcomes in patients with thromboembolic
events and invasive exocrine pancreatic cancer. Cancer. 2012;118:3053–3061.
352. Butturini G, Stocken DD, Wente MN, et al. Influence of resection margins and treatment on survival in patients with
pancreatic cancer: meta-analysis of randomized controlled trials. Arch Surg. 2008;143:75–83. [discussion].
353. Mayo SC, Nathan H, Cameron JL, et al. Conditional survival in patients with pancreatic ductal adenocarcinoma
resected with curative intent. Cancer. 2012;118:2674–2681.
354. Konstantinidis IT, Warshaw AL, Allen JN, et al. Pancreatic ductal adenocarcinoma: is there a survival difference for R1
resections versus locally advanced unresectable tumors? What is a "true" R0 resection? Ann Surg. 2013;257:731–736.
355. Paniccia A, Hosokawa P, Henderson W, et al. Characteristics of 10-year survivors of pancreatic ductal
adenocarcinoma. JAMA Surg. 2015;150:701–710.
356. Strobel O, Hartwig W, Hackert T, et al. Re-resection for isolated local recurrence of pancreatic cancer is feasible,
safe, and associated with encouraging survival. Ann Surg Oncol. 2013;20:964–972.
357. Asuthkar S, Rao JS, Gondi CS. Drugs in preclinical and early-stage clinical development for pancreatic cancer. Expert
Opin Investig Drugs. 2012;21:143–152.
358. Oettle H, Neuhaus P, Hochhaus A, et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes
among patients with resected pancreatic cancer: the CONKO-001 randomized trial. J Am Med Assoc. 2013;310:
1473–1481.
359. Hishinuma S, Ogata Y, Tomikawa M, Ozawa I, Hirabayashi K, Igarashi S. Patterns of recurrence after curative
resection of pancreatic cancer, based on autopsy findings. J Gastrointest Surg. 2006;10:511–518.
360. Gnerlich JL, Luka SR, Deshpande AD, et al. Microscopic margins and patterns of treatment failure in resected
pancreatic adenocarcinoma. Arch Surg. 2012;147:753–760.
361. Van den Broeck A, Sergeant G, Ectors N, Van Steenbergen W, Aerts R, Topal B. Patterns of recurrence after curative
resection of pancreatic ductal adenocarcinoma. European J Surg Oncol. 2009;35:600–604.
362. Castellanos JA, Merchant NB. Intensity of follow-up after pancreatic cancer resection. Ann Surg Oncol. 2014;21:
747–751.
363. Sheffield KM, Crowell KT, Lin YL, Djukom C, Goodwin JS, Riall TS. Surveillance of pancreatic cancer patients after
surgical resection. Ann Surg Oncol. 2012;19:1670–1677.
364. La Torre M, Nigri G, Lo Conte A, et al. Is a preoperative assessment of the early recurrence of pancreatic cancer
possible after complete surgical resection? Gut Liver. 2014;8:102–108.
365. Kinsella TJ, Seo Y, Willis J, et al. The impact of resection margin status and postoperative CA19-9 levels on survival
and patterns of recurrence after postoperative high-dose radiotherapy with 5-FU-based concurrent chemotherapy
for resectable pancreatic cancer. Am J Clin Oncol. 2008;31:446–453.
366. Katz MH, Wang H, Balachandran A, et al. Effect of neoadjuvant chemoradiation and surgical technique on
recurrence of localized pancreatic cancer. J Gastrointest Surg. 2012;16:68–78. [discussion-9].
367. Bogoevski D, Yekebas EF, Schurr P, et al. Mode of spread in the early phase of lymphatic metastasis in pancreatic
ductal adenocarcinoma: prognostic significance of nodal microinvolvement. Ann Surg. 2004;240:993–1000.
[discussion-1.].
368. Hamidian Jahromi A, Sangster G, Zibari G, et al. Accuracy of multi-detector computed tomography,
fluorodeoxyglucose positron emission tomography-CT, and CA 19-9 levels in detecting recurrent pancreatic
adenocarcinoma. JOP. 2013;14:466–468.
369. Habermehl D, Brecht IC, Bergmann F, et al. Chemoradiation in patients with isolated recurrent pancreatic cancer—
herapeutical efficacy and probability of re-resection. Radiat Oncol. 2013;8:27.
370. Nakamura A, Itasaka S, Takaori K, et al. Radiotherapy for patients with isolated local recurrence of primary resected
pancreatic cancer: prolonged disease-free interval associated with favorable prognosis. Strahlenther Onkol.
2014;190(5):485–490.
371. Kleeff J, Reiser C, Hinz U, et al. Surgery for recurrent pancreatic ductal adenocarcinoma. Ann Surg. 2007;245:
566–572.
372. Roeder F, Timke C, Uhl M, et al. Aggressive local treatment containing intraoperative radiation therapy (IORT) for
patients with isolated local recurrences of pancreatic cancer: a retrospective analysis. BMC Cancer. 2012;12:295.
373. Kelsen DP, Portenoy RK, Thaler HT, et al. Pain and depression in patients with newly diagnosed pancreas cancer. J
Clin Oncol. 1995;13:748–755.
374. House MG, Choti MA. Palliative therapy for pancreatic/biliary cancer. Surg Clin North Am. 2005;85:359–371.
375. Staats PS, Hekmat H, Sauter P, Lillemoe K. The effects of alcohol celiac plexus block, pain, and mood on longevity in
patients with unresectable pancreatic cancer: a double-blind, randomized, placebo-controlled study. Pain Med.
2001;2:28–34.
376. Rykowski JJ, Hilgier M. Efficacy of neurolytic celiac plexus block in varying locations of pancreatic cancer: influence
on pain relief. Anesthesiology. 2000;92:347–354.
377. Shaikh TR, Desai P. Multimodal analgesia is central to alleviating pain in pancreatic adenocarcinoma. Br Med
J. 2012;345:e4307.
378. Bukki J. Pancreatic adenocarcinoma. N Engl J Med. 2014;371:2139–2140.
379. Turaga KK, Malafa MP, Jacobsen PB, Schell MJ, Sarr MG. Suicide in patients with pancreatic cancer. Cancer. 2011;117:
642–647.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
152 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

380. Petzel MQ, Parker NH, Valentine AD, et al. Fear of cancer recurrence after curative pancreatectomy: a cross-
sectional study in survivors of pancreatic and periampullary tumors. Ann Surg Oncol. 2012;19:4078–4084.
381. Chan C, Franssen B, Dominguez I, Ramirez-Del Val A, Uscanga LF, Campuzano M. Impact on quality of life after
pancreatoduodenectomy: a prospective study comparing preoperative and postoperative scores. J Gastrointest Surg.
2012;16:1341–1346.
382. Gerstenhaber F, Grossman J, Lubezky N, et al. Pancreaticoduodenectomy in elderly adults: is it justified in terms of
mortality, long-term morbidity, and quality of life? J Am Geriatr Soc. 2013;61:1351–1357.
383. Schniewind B, Bestmann B, Henne-Bruns D, Faendrich F, Kremer B, Kuechler T. Quality of life after
pancreaticoduodenectomy for ductal adenocarcinoma of the pancreatic head. Br J Surg. 2006;93:1099–1107.
384. Morak MJ, Pek CJ, Kompanje EJ, Hop WC, Kazemier G, van Eijck CH. Quality of life after adjuvant intra-arterial
chemotherapy and radiotherapy versus surgery alone in resectable pancreatic and periampullary cancer: a
prospective randomized controlled study. Cancer. 2010;116:830–836.
385. Sultana A, Tudur Smith C, Cunningham D, et al. Systematic review, including meta-analyses, on the management of
locally advanced pancreatic cancer using radiation/combined modality therapy. Br J Cancer. 2007;96:1183–1190.
386. Burris 3rd HA, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-
line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–2413.
387. De La Cruz MS, Young AP, Ruffin MT. Diagnosis and management of pancreatic cancer. Am Fam Physician. 2014;89:
626–632.
388. Moyer MT, Gaffney RR. Pancreatic adenocarcinoma. N Engl J Med. 2014;371:2140.
389. Sheffield KM, Boyd CA, Benarroch-Gampel J, Kuo YF, Cooksley CD, Riall TS. End-of-life care in Medicare
beneficiaries dying with pancreatic cancer. Cancer. 2011;117:5003–5012.
390. Gomez-Martin C, Hidalgo M, Tabernero J, Isla D. SEOM clinical guidelines for the treatment of pancreatic cancer.
Clin Transl Oncol. 2011;13:528–535.
391. Di Fronzo LA, Cymerman J, Egrari S, O’Connell TX. Unresectable pancreatic carcinoma: correlating length of survival
with choice of palliative bypass. Am Surg. 1999;65:955–958.
392. Sarfeh IJ, Rypins EB, Jakowatz JG, Juler GL. A prospective, randomized clinical investigation of cholecystoenter-
ostomy and choledochoenterostomy. Am J Surg. 1988;155:411–414.
393. Huser N, Michalski CW, Schuster T, Friess H, Kleeff J. Systematic review and meta-analysis of prophylactic
gastroenterostomy for unresectable advanced pancreatic cancer. Br J Surg. 2009;96:711–719.
394. Shah A, Fehmi A, Savides TJ. Increased rates of duodenal obstruction in pancreatic cancer patients receiving modern
medical management. Dig Dis Sci. 2014;59:2294–2298.
395. Gillen S, Schuster T, Friess H, Kleeff J. Palliative resections versus palliative bypass procedures in pancreatic cancer-a
systematic review. Am J Surg. 2012;203:496–502.
396. Puli SR, Reddy JB, Bechtold ML, Antillon MR, Brugge WR. EUS-guided celiac plexus neurolysis for pain due to
chronic pancreatitis or pancreatic cancer pain: a meta-analysis and systematic review. Dig Dis Sci. 2009;54:
2330–2337.
397. Polati E, Finco G, Gottin L, Bassi C, Pederzoli P, Ischia S. Prospective randomized double-blind trial of neurolytic
coeliac plexus block in patients with pancreatic cancer. Br J Surg. 1998;85:199–201.
398. Allen PJ, Chou J, Janakos M, Strong VE, Coit DG, Brennan MF. Prospective evaluation of laparoscopic celiac plexus
block in patients with unresectable pancreatic adenocarcinoma. Ann Surg Oncol. 2011;18:636–641.
399. Pietrabissa A, Vistoli F, Carobbi A, Boggi U, Bisa M, Mosca F. Thoracoscopic splanchnicectomy for pain relief in
unresectable pancreatic cancer. Arch Surg. 2000;135:332–335.
400. Smigielski J, Piskorz L, Wawrzycki M, Kutwin L, Misiak P, Brocki M. Assessment of quality of life in patients with
non-operated pancreatic cancer after videothoracoscopic splanchnicectomy. Wideochir Inne Tech Maloinwazyjne.
2011;6:132–137.
401. Beger HG. Pancreatic cancer staging systems and their clinical impact. In: Beger HWA, Büchler M, eds. The pancreas:
an integrated textbook of basic science, medicine, and surgery. 2nd ed. Oxford, England: Blackwell; 2008.
402. Moss AC, Morris E, Mac Mathuna P. Palliative biliary stents for obstructing pancreatic carcinoma. Cochrane Database
Syst Rev. 2006:CD004200.
403. Shepherd HA, Royle G, Ross AP, Diba A, Arthur M, Colin-Jones D. Endoscopic biliary endoprosthesis in the palliation
of malignant obstruction of the distal common bile duct: a randomized trial. Br J Surg. 1988;75:1166–1168.
404. Smith AC, Dowsett JF, Russell RC, Hatfield AR, Cotton PB. Randomised trial of endoscopic stenting versus surgical
bypass in malignant low bileduct obstruction. Lancet. 1994;344:1655–1660.
405. Taylor MC, McLeod RS, Langer B. Biliary stenting versus bypass surgery for the palliation of malignant distal bile
duct obstruction: a meta-analysis. Liver Transpl. 2000;6:302–308.
406. Glazer ES, Hornbrook MC, Krouse RS. A meta-analysis of randomized trials: immediate stent placement vs. surgical
bypass in the palliative management of malignant biliary obstruction. J Pain Symptom Manage. 2014;47:307–314.
407. Lillemoe KD, Pitt HA. Palliation. Surgical and otherwise. Cancer. 1996;78:605–614.
408. 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;17:128–132. [discussion 32-3].
409. Bartlett EK, Wachtel H, Fraker DL, et al. Surgical palliation for pancreatic malignancy: practice patterns and
predictors of morbidity and mortality. J Gastrointest Surg. 2014;18:1292–1298.
410. Assail V, Huser N, Michalski C, Gillen S, Kleeff J, Friess H. Palliative interventional and surgical therapy for
unresectable pancreatic cancer. Cancers. 2011;3:652–661.
411. Glimelius B, Hoffman K, Sjoden PO, et al. Chemotherapy improves survival and quality of life in advanced pancreatic
and biliary cancer. Ann Oncol. 1996;7:593–600.
412. Von Hoff DD, Goldstein D, Renschler MF. Albumin-bound paclitaxel plus gemcitabine in pancreatic cancer. N Engl J
Med. 2014;370:479–480.
413. Cunningham D, Chau I, Stocken DD, et al. Phase III randomized comparison of gemcitabine versus gemcitabine plus
capecitabine in patients with advanced pancreatic cancer. J Clin Oncol. 2009;27:5513–5518.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154 153

414. Conroy T, Desseigne F, Ychou M, et al. Folfirinox versus gemcitabine for metastatic pancreatic cancer. N Engl J Med.
2011;364:1817–1825.
415. Bayraktar S, Bayraktar UD, Rocha-Lima CM. Recent developments in palliative chemotherapy for locally advanced
and metastatic pancreas cancer. World J Gastrointest Oncol. 2010;16:673–682.
416. Johung K, Saif MW, Chang BW. Treatment of locally advanced pancreatic cancer: the role of radiation therapy. Int J
Radiat Oncol Biol Phys. 2012;82:508–518.
417. Cardenes HR, Moore AM, Johnson CS, et al. A phase II study of gemcitabine in combination with radiation therapy in
patients with localized, unresectable, pancreatic cancer: a Hoosier Oncology Group study. Am J Clin Oncol. 2011;34:
460–465.
418. Cai S, Hong TS, Goldberg SI, et al. Updated long-term outcomes and prognostic factors for patients with
unresectable locally advanced pancreatic cancer treated with intraoperative radiotherapy at the Massachusetts
General Hospital, 1978 to 2010. Cancer. 2013;119:4196–4204.
419. Mahadevan A, Jain S, Goldstein M, et al. Stereotactic body radiotherapy and gemcitabine for locally advanced
pancreatic cancer. Int J Radiat Oncol Biol Phys. 2010;78:735–742.
420. Sanders MK, Moser AJ, Khalid A, et al. EUS-guided fiducial placement for stereotactic body radiotherapy in locally
advanced and recurrent pancreatic cancer. Gastrointest Endosc. 2010;71:1178–1184.
421. Su TS, Liang P, Lu HZ, et al. Stereotactic body radiotherapy using CyberKnife for locally advanced unresectable and
metastatic pancreatic cancer. World J Gastrointest Oncol. 2015;21:8156–8162.
422. Polistina F, Costantin G, Casamassima F, et al. Unresectable locally advanced pancreatic cancer: a multimodal
treatment using neoadjuvant chemoradiotherapy (gemcitabine plus stereotactic radiosurgery) and subsequent
surgical exploration. Ann Surg Oncol. 2010;17:2092–2101.
423. Schellenberg D, Kim J, Christman-Skieller C, et al. Single-fraction stereotactic body radiation therapy and sequential
gemcitabine for the treatment of locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys. 2011;81:181–188.
424. Herman JM, Chang DT, Goodman KA, et al. Phase 2 multi-institutional trial evaluating gemcitabine and stereotactic
body radiotherapy for patients with locally advanced unresectable pancreatic adenocarcinoma. Cancer. 2015;121:
1128–1137.
425. Pollom EL, Alagappan M, von Eyben R, et al. Single- versus multifraction stereotactic body radiation therapy for
pancreatic adenocarcinoma: outcomes and toxicity. Int J Radiat Oncol Biol Phys. 2014;90:918–925.
426. Hoyer M, Roed H, Sengelov L, et al. Phase-II study on stereotactic radiotherapy of locally advanced pancreatic
carcinoma. Radiother Oncol. 2005;76:48–53.
427. Gurka MK, Kim C, He AR, et al. Stereotactic body radiation therapy (SBRT) combined with chemotherapy for
unresected pancreatic adenocarcinoma. Am J Clin Oncol. 2014.
428. Kim CH, Ling DC, Wegner RE, et al. Stereotactic body radiotherapy in the treatment of pancreatic adenocarcinoma
in elderly patients. Radiat Oncol. 2013;8:240.
429. Engelhardt K, Riley C, Cooke L, Mahadevan D. Monoclonal antibody therapies targeting pancreatic ductal
adenocarcinoma. Curr Drug Discov Technol. 2006;3:231–243.
430. Heinemann V, Ebert MP, Laubender RP, Bevan P, Mala C, Boeck S. Phase II randomised proof-of-concept study of the
urokinase inhibitor upamostat (WX-671) in combination with gemcitabine compared with gemcitabine alone in
patients with non-resectable, locally advanced pancreatic cancer. Br J Cancer. 2013;108:766–770.
431. Rombouts SJ, Vogel JA, van Santvoort HC, et al. Systematic review of innovative ablative therapies for the treatment
of locally advanced pancreatic cancer. Br J Surg. 2015;102:182–193.
432. Cao C, Yan TD, Morris DL, Bester L. Radioembolization with yttrium-90 microspheres for pancreatic cancer liver
metastases: results from a pilot study. Tumori. 2010;96:955–958.
433. Carrafiello G, Ierardi AM, Fontana F, et al. Microwave ablation of pancreatic head cancer: safety and efficacy. J Vasc
Interv Radiol. 2013;24:1513–1520.
434. Lee JY, Choi BI, Ryu JK, et al. Concurrent chemotherapy and pulsed high-intensity focused ultrasound therapy for
the treatment of unresectable pancreatic cancer: initial experiences. Korean J Radiol. 2011;12:176–186.
435. Combs SE, Habermehl D, Kieser M, et al. Phase I study evaluating the treatment of patients with locally advanced
pancreatic cancer with carbon ion radiotherapy: the PHOENIX-01 trial. BMC Cancer. 2013;13:419.
436. Huggett MT, Jermyn M, Gillams A, et al. Phase I/II study of verteporfin photodynamic therapy in locally advanced
pancreatic cancer. Br J Cancer. 2014;110(7):1698–1704.
437. Ansari D, Chen BC, Dong L, Zhou MT, Andersson R. Pancreatic cancer: translational research aspects and clinical
implications. World J Gastrointest Oncol. 2012;18:1417–1424.
438. Schultz NA, Dehlendorff C, Jensen BV, et al. MicroRNA biomarkers in whole blood for detection of pancreatic cancer.
J Am Med Assoc. 2014;311:392–404.
439. Bouvet M, Hoffman RM. In vivo imaging of pancreatic cancer with fluorescent proteins in mouse models. Methods
Mol Biol. 2012;872:51–67.
440. Metildi CA, Kaushal S, Hardamon CR, et al. Fluorescence-guided surgery allows for more complete resection of
pancreatic cancer, resulting in longer disease-free survival compared with standard surgery in orthotopic mouse
models. J Am Coll Surg. 2012;215:126–135. [discussion 35-6].
441. Abe Y, Itano O, Kitago M, et al. Computer assisted surgery, preoperative planning and navigation for pancreatic
cancer. J Hepatobiliary Pancreat Sci. 2014;21(4):251–255.
442. Zhan Q, Shen B, Deng X, et al. Drug-eluting scaffold to deliver chemotherapeutic medication for management of
pancreatic cancer after surgery. Int J Nanomedicine. 2013;8:2465–2472.
443. Tseng JF, Mulligan RC. Gene therapy for pancreatic cancer. Surg Oncol Clin North Am. 2002;11:537–569.
444. Pawlik TM, Laheru D, Hruban RH, et al. Evaluating the impact of a single-day multidisciplinary clinic on the
management of pancreatic cancer. Ann Surg Oncol. 2008;15:2081–2088.
445. Chang KJ, Parasher G, Christie C, Largent J, Anton-Culver H. Risk of pancreatic adenocarcinoma: disparity between
African Americans and other race/ethnic groups. Cancer. 2005;103:349–357.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
154 M.F. Eskander et al. / Current Problems in Surgery 53 (2016) 107–154

446. Chang DC, Zhang Y, Mukherjee D, et al. Variations in referral patterns to high-volume centers for pancreatic cancer.
J Am Coll Surg. 2009;209:720–726.
447. Cheung MC, Yang R, Byrne MM, Solorzano CC, Nakeeb A, Koniaris LG. Are patients of low socioeconomic status
receiving suboptimal management for pancreatic adenocarcinoma? Cancer. 2010;116:723–733.
448. Helling TS. Pancreas cancer in Mississippi: present challenges and future directions. J Miss State Med Assoc. 2010;51:
99–103.
449. Bilimoria KY, Bentrem DJ, Ko CY, Stewart AK, Winchester DP, Talamonti MS. National failure to operate on early
stage pancreatic cancer. Ann Surg. 2007;246:173–180.
450. Talamonti MS, Small Jr. W, Mulcahy MF, et al. A multi-institutional phase II trial of preoperative full-dose
gemcitabine and concurrent radiation for patients with potentially resectable pancreatic carcinoma. Ann Surg
Oncol. 2006;13:150–158.
451. Heinrich S, Pestalozzi BC, Schafer M, et al. Prospective phase II trial of neoadjuvant chemotherapy with gemcitabine
and cisplatin for resectable adenocarcinoma of the pancreatic head. J Clin Oncol. 2008;26:2526–2531.
452. Le Scodan R, Mornex F, Girard N, et al. Preoperative chemoradiation in potentially resectable pancreatic
adenocarcinoma: feasibility, treatment effect evaluation and prognostic factors, analysis of the SFRO-FFCD 9704
trial and literature review. Ann Oncol. 2009;20:1387–1396.
453. Turrini O, Ychou M, Moureau-Zabotto L, et al. Neoadjuvant docetaxel-based chemoradiation for resectable
adenocarcinoma of the pancreas: New neoadjuvant regimen was safe and provided an interesting pathologic
response. Eur J Surg Oncol. 2010;36:987–992.
454. Pipas JM, Zaki BI, McGowan MM, et al. Neoadjuvant cetuximab, twice-weekly gemcitabine, and intensity-
modulated radiotherapy (IMRT) in patients with pancreatic adenocarcinoma. Ann Oncol. 2012;23:2820–2827.
455. Takahashi H, Ohigashi H, Ishikawa O, et al. Perineural invasion and lymph node involvement as indicators of
surgical outcome and pattern of recurrence in the setting of preoperative gemcitabine-based chemoradiation
therapy for resectable pancreatic cancer. Ann Surg. 2012;255:95–102.
456. Van Buren 2nd G, Ramanathan RK, Krasinskas AM, et al. Phase II study of induction fixed-dose rate gemcitabine and
bevacizumab followed by 30 Gy radiotherapy as preoperative treatment for potentially resectable pancreatic
adenocarcinoma. Ann Surg Oncol. 2013;20:3787–3793.
457. Kim EJ, Ben-Josef E, Herman JM, et al. A multi-institutional phase 2 study of neoadjuvant gemcitabine and
oxaliplatin with radiation therapy in patients with pancreatic cancer. Cancer. 2013;119:2692–2700.
458. Motoi F, Ishida K, Fujishima F, et al. Neoadjuvant chemotherapy with gemcitabine and S-1 for resectable and
borderline pancreatic ductal adenocarcinoma: results from a prospective multi-institutional phase 2 trial. Ann Surg
Oncol. 2013;20:3794–3801.
459. Hong TS, Ryan DP, Borger DR, et al. A phase 1/2 and biomarker study of preoperative short course chemoradiation
with proton beam therapy and capecitabine followed by early surgery for resectable pancreatic ductal
adenocarcinoma. Int J Radiat Oncol Biol Phys. 2014;89:830–838.
460. O’Reilly EM, Perelshteyn A, Jarnagin WR, et al. A single-arm, nonrandomized phase II trial of neoadjuvant
gemcitabine and oxaliplatin in patients with resectable pancreas adenocarcinoma. Ann Surg. 2014;260:142–148.
461. Takeda Y, Nakamori S, Eguchi H, et al. Neoadjuvant gemcitabine-based accelerated hyperfractionation chemo-
radiotherapy for patients with borderline resectable pancreatic adenocarcinoma. Jpn J Clin Oncol. 2014;44:
1172–1180.
462. Demols A, Peeters M, Polus M, et al. Adjuvant gemcitabine and concurrent continuous radiation (45 Gy) for
resected pancreatic head carcinoma: a multicenter Belgian Phase II study. Int J Radiat Oncol Biol Phys. 2005;62:
1351–1356.
463. Blackstock AW, Mornex F, Partensky C, et al. Adjuvant gemcitabine and concurrent radiation for patients with
resected pancreatic cancer: a phase II study. Br J Cancer. 2006;95:260–265.
464. Kosuge T, Kiuchi T, Mukai K, Kakizoe T. A multicenter randomized controlled trial to evaluate the effect of adjuvant
cisplatin and 5-fluorouracil therapy after curative resection in cases of pancreatic cancer. Jpn J Clin Oncol. 2006;36:
159–165.
465. Yoshitomi H, Togawa A, Kimura F, et al. A randomized phase II trial of adjuvant chemotherapy with uracil/tegafur
and gemcitabine versus gemcitabine alone in patients with resected pancreatic cancer. Cancer. 2008;113:
2448–2456.
466. Ueno H, Kosuge T, Matsuyama Y, et al. A randomised phase III trial comparing gemcitabine with surgery-only in
patients with resected pancreatic cancer: Japanese Study Group of Adjuvant Therapy for Pancreatic Cancer. Br J
Cancer. 2009;101:908–915.
467. Van Laethem JL, Hammel P, Mornex F, et al. Adjuvant gemcitabine alone versus gemcitabine-based chemo-
radiotherapy after curative resection for pancreatic cancer: a randomized EORTC-40013-22012/FFCD-9203/GERCOR
phase II study. J Clin Oncol. 2010;28:4450–4456.
468. Neoptolemos JP, Stocken DD, Bassi C, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs
gemcitabine following pancreatic cancer resection: a randomized controlled trial. J Am Med Assoc. 2010;304:
1073–1081.
469. Reni M, Balzano G, Aprile G, et al. Adjuvant PEFG (cisplatin, epirubicin, 5-fluorouracil, gemcitabine) or gemcitabine
followed by chemoradiation in pancreatic cancer: a randomized phase II trial. Ann Surg Oncol. 2012;19:2256–2263.
470. Schmidt J, Abel U, Debus J, et al. Open-label, multicenter, randomized phase III trial of adjuvant chemoradiation
plus interferon Alfa-2b versus fluorouracil and folinic acid for patients with resected pancreatic adenocarcinoma.
J Clin Oncol. 2012;30:4077–4083.
471. Fensterer H, Schade-Brittinger C, Muller HH, et al. Multicenter phase II trial to investigate safety and efficacy of
gemcitabine combined with cetuximab as adjuvant therapy in pancreatic cancer (ATIP). Annals of oncology.
2013;24:2576–2581.
472. Herman JM, Fan KY, Wild AT, et al. Phase 2 study of erlotinib combined with adjuvant chemoradiation and
chemotherapy in patients with resectable pancreatic cancer. Int J Radiat Oncol Biol Phys. 2013;86:678–685.

Downloaded for BOLIVAR ISEA (gamiva94@gmail.com) at ClinicalKey Global Guest Users from ClinicalKey.com by Elsevier on
October 22, 2018. For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.

You might also like