A semicentennial of pancreatic pathology: the genetic revolution is here, but don’t throw the baby out with the bath water!

A semicentennial of pancreatic pathology: the genetic revolution is here, but don’t throw the baby out with the bath water!

Journal Pre-proof A Semicentennial of Pancreatic Pathology: The Genetic Revolution Is Here, But Don’t Throw the Baby Out With The Bath Water! Ralph H...

28MB Sizes 0 Downloads 25 Views

Journal Pre-proof A Semicentennial of Pancreatic Pathology: The Genetic Revolution Is Here, But Don’t Throw the Baby Out With The Bath Water!

Ralph H. Hruban, David S. Klimstra, Guiseppe Zamboni, Günter Klöppel PII:

S0046-8177(19)30164-9

DOI:

https://doi.org/10.1016/j.humpath.2019.08.024

Reference:

YHUPA 4918

To appear in:

Human Pathology

Received date:

26 August 2019

Accepted date:

28 August 2019

Please cite this article as: R.H. Hruban, D.S. Klimstra, G. Zamboni, et al., A Semicentennial of Pancreatic Pathology: The Genetic Revolution Is Here, But Don’t Throw the Baby Out With The Bath Water!, Human Pathology(2019), https://doi.org/ 10.1016/j.humpath.2019.08.024

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

© 2019 Published by Elsevier.

Journal Pre-proof A Semicentennial of Pancreatic Pathology: The Genetic Revolution Is Here, But Don’t Throw the Baby Out With The Bath Water! *

Ralph H. Hruban, M.D.1, David S. Klimstra, M.D. 2, Guiseppe Zamboni, M.D. 3, and Günter Klöppel, M.D. 4

The Sol Goldman Pancreatic Cancer Research Center, Departments of Pathology and Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA; 2The Department of Pathology, Memorial Sloan Kettering Cancer Center, NY, USA; 3Sacro Cuore Don Calabria Hospital, Italy; and 4Technical University of Munich, Munich, Germany

ro

of

1

Address Correspondence to:

-p

Ralph H. Hruban, M.D.

re

The Sol Goldman Pancreatic Cancer Research Center Department of Pathology

lP

The Johns Hopkins University School of Medicine

600 North Wolfe Street Baltimore, MD 21287

na

Carnegie Room 415

ur

Email: [email protected]

Jo

Phone: (410) 955-9790 Fax: (410) 955-0394

*

Based, in part, on the Maude Abbott Lecture given by Ralph Hruban at the 2014 Annual Meeting of the United States and Canadian Academy of Pathology.

1

Journal Pre-proof Abstract

The last fifty years have witnessed an explosion in our understanding of the pathology of pancreatic diseases. Entities known to exist 50 years ago have been defined more precisely and are now better classified. New entities, previously not recognized, have been discovered and can now be treated. Importantly, new tools have been developed that have unraveled the

of

fundamental biological drivers of a number of pancreatic diseases. Many of these same tools have also been applied clinically, supplementing the tried and true hematoxylin and eosin

ro

stained slide with a plethora of new, highly sensitive and specific tests that improve diagnostic

-p

accuracy and delineate best treatments. As exciting as these many advances are, our

na

lP

re

knowledge of pancreatic pathology remains incomplete, and there is much to be learned.

Jo

ur

Key words: Pancreas, pancreatic, pathology, history

2

Journal Pre-proof Introduction

Before we dive into the dramatic changes that have occurred over the last 50 years in pancreatic pathology, we thought it would be illustrative to reflect on the >2,000 year history of modern (western) medicine, as in doing so it becomes clear that the one constant in medicine is change. Simply put, the history of pancreatic pathology, like all medical history, has been

of

marked by transformative waves of innovation that fundamentally change our concept of entire

ro

fields.(1)

-p

Modern western medicine began when Hippocrates (460-370 BCE) and his followers on the island of Kos separated that which is natural from that which is supernatural. Shortly

re

thereafter, Herophilus (335- 255 BCE), who was educated by Praxagoras of Kos, is credited with

lP

the first description of the pancreas.(1-3) The revolution created by the separation of medicine from superstition was quickly overtaken by another revolution, led by Galen of Pergamon (129

na

ACE-c.200/c.216 ACE). Galen brought science to medicine. He based his writings on dissection

ur

and experimentation on animals, proving, for example, that voice is generated in the larynx.

Jo

Unfortunately, Galen believed that the pancreas was just padding to protect the abdominal vasculature, that “Nature created ‘so-called pancreas’ and spread it beneath all vessels.”(1, 4)

Galen’s teachings prevailed for more than a thousand years, until the Dutch anatomist Andreas Vesalius (1514-1564) brought on the next disruptive wave of innovation. In his classic book, De Humani Corporis Fabrica Vesalius showed that several keystones of Galenic anatomy were incorrect. As Vesalius famously wrote, Galen “was fooled by his monkeys.”(1, 5) Although Vesalius revolutionized the understanding of the anatomy of a number of key structures, the pancreas was once again left behind as he chose to illustrate the abdominal organs with the pancreas dissected away in order highlight the splenic and superior mesenteric veins.

3

Journal Pre-proof Importantly, Vesalius promulgated the idea that dogmas should be challenged, and that students should dissect and study for themselves. Johann Georg Wirsüng (1589-1643) did just that, and in Padua in 1642 discovered that the pancreas has a duct.(1, 6) With this discovery, the study of the pancreas forever changed; if it has a duct, the pancreas must be producing something!

of

Giovanni Battista Morgagni (1682-1771), whose classic book De Sedibus et Causis Morborum per Anatomen Indigatis established the field of morbid anatomy, brought on the next

ro

disruptive revolution in medicine by showing that disease has its origins in the organs. As he

-p

beautifully put it, “symptoms are the cry of the suffering organs.”(1, 7) Indeed, in what may

re

have been one of the earliest clinical descriptions of pancreatic cancer, he described the pain

lP

experienced by a patient with pancreatic tumor as being just as great as if he were being torn to pieces by dogs.(1, 8) The next transformation in medicine was initiated by Rudolf Virchow (1821-

na

1902) with his book Die Cellularpathologie in which he brilliantly showed that disease occurs at the level of the cell.(9) Today, more than 100 years later, the microscope remains the

Jo

ur

foundation of pancreatic pathology.

Diagnosis after death became common practice; diagnosis during life was another matter. Physicians in the French schools promulgated careful observation and physical examination, and soon diseases could be diagnosed clinically. Ludwig Courvoisier (1843-1918) found that a palpable gallbladder is a sign of pancreatic cancer, and Armand Trousseau (18011867) reported that spontaneous venous thromboses are associated with pancreatic cancer.(1) In a remarkable twist of fate, Trousseau diagnosed his own fatal gastric cancer when he developed venous thromboses. By contrast, the diagnosis of diabetes mellitus was way ahead

4

Journal Pre-proof of the game. Diabetes mellitus (honey sweet) was recognized over three thousand years ago, and in 1776 Dobson showed that excess glucose caused the sweetness of the urine.(10)

The final challenge remained treatment, and this long road, on which we are just beginning our journey, started with Allen Oldfather Whipple (1881-1963). Successful pancreatic resections were first performed by Walther Kausch (1867-1928), but it was Whipple, a surgeon

of

at Columbia University, who, in the 1930s and 1940s, promulgated the surgical resection of pancreatic masses, in a procedure that now bears his name, the “Whipple operation.” (1, 11)

ro

With safe surgery, pancreatic resections grew exponentially at expert centers, providing

-p

pathologists with the materials they would need to create the revolution that has marked the

re

last 50 years of pancreatic pathology.(11, 12)

lP

Pancreatic Pathology in 1970

na

With little surgical volume to guide the classification of diseases of the pancreas, the classification of neoplasms of the pancreas was rudimentary in 1970 (Figure 1).(1) A clinically

ur

meaningful classification was hindered by the lack of significant clinical information; computed

Jo

tomography (CAT scanning) was not invented until 1972. Much of the focus was on pseudocysts, and knowledge of other entities was, in retrospect, crude. For example, cystic neoplasms of the pancreas were broadly lumped together under the designations “cystadenoma” and “cystadenocarcinoma.”(13-16) The classification of solid neoplasms similarly suffered. It wasn’t until the late 1970s and early 1980s that classifications based on phenotype began to pave the way for studies that correlated phenotype with clinical outcome.(17, 18) Furthermore, although syndromic neuroendocrine neoplasms (“functional islet cell tumors”) were recognized and wellcharacterized because of their associated distinctive symptomatology, nonsyndromic neuroendocrine neoplasms were rare and were poorly described.(19-21) It was not recognized that all neuroendocrine neoplasms are malignant, and much effort was expended attempting to distinguish “benign” from “malignant” islet cell tumors. At the “molecular” level, none of the genes

5

Journal Pre-proof that drive pancreatic neoplasia were known. Similarly, little was known about the underlying

etiologies of the two most important non-neoplastic diseases of the pancreas, diabetes mellitus and pancreatitis. Diabetes was simply classified into a juvenile and adult form, and key findings

such as insulitis and amyloidosis had been ascribed to these two forms; pancreatitis was wellrecognized, but the focus was almost entirely on alcoholic and gallstone pancreatitis and their

of

treatment.(22)

In the following sections, we trace the unimaginable progress that has been made over

-p

ro

the past 50 years in several areas of pancreas pathology (Figure 1).(23)

re

Genetics of Pancreatic Ductal Adenocarcinoma

lP

Perhaps the greatest advances in the past fifty years have come from the discovery of the genetic drivers of pancreatic diseases, and the subsequent application of genetic tools to

(Table 1).

na

dissect out disease causing pathways and to define entities based on these genetic drivers

ur

The first gene found to be targeted in pancreatic cancer, by which we mean invasive

Jo

ductal adenocarcinoma, was KRAS.(24) Oncogenes were first described by Varmus, Bishop and Vogt in 1976 when they demonstrated that viral src oncogene is a normal chicken gene transduced by viruses, thereby converting a normal gene into a potent oncogene.(25) In 1987, Murray Korc, using a primitive polymerase chain reaction (PCR) machine built by Paul Meltzer analyzed RNA from human pancreatic cancers and they observed a faint abnormal band suggestive of KRAS gene mutations (personal communication). They did not publish this finding as it wasn’t definitive, and in 1988 Almoguera, Shibata and Perucho were the first to publish definitive KRAS gene mutations in human pancreatic cancer.(24)

6

Journal Pre-proof The first tumor suppressor gene found to be genetically inactivated in pancreatic cancer was TP53. The TP53 gene was discovered in 1979, but it was initially misclassified as an oncogene.(26) It wasn’t until 1989 that TP53 was correctly classified as a tumor suppressor gene, and in 1991 C. Barton, J. Neoptolemos, and G. Klöppel first reported TP53 gene mutations in human pancreatic cancer.(27) Thus, in the early days of the cancer genetic revolution, discoveries in pancreatic cancer

of

followed discoveries in other cancer types by many years if not by decades. This changed

ro

dramatically in the 1990s. Slowly but surely, discoveries in pancreatic cancer started to precede

-p

those made in other cancer types. This pivot occurred because of a confluence of events. First, a handful of surgeons, John Cameron at Johns Hopkins, Andrew Warshaw at the

re

Massachusetts General Hospital, Murray Brennan at Memorial Sloan Kettering, and, in Europe,

lP

Franz Kümmerle, Michael Trede, Hans Beger and Marcus Büchler dedicated their careers to improving pancreas surgery.(28, 29) For example, at Johns Hopkins John Cameron personally

na

performed more than 2,400 pancreatoduodenectomies.(28) As these surgeons developed more

ur

and more experience, their teams were able to reduce dramatically the mortality rates of

Jo

pancreatic surgery.(30, 31) Patients flocked to specialized centers of excellence, and suddenly scientists had access to large numbers of surgically resected pancreatic cancers to study. At Johns Hopkins, the dramatic increase in pancreas surgery fed into a genetics laboratory run by a young, extremely creative scientist, Scott Kern. The molecular revolution was beginning and Kern, who had trained in Bert Vogelstein’s lab, was looking for a cancer to study. Kern brought tireless dedication, extraordinary creativity, a willingness to go out on a limb, and an insistence that his lab focus on big, important questions to pancreatic cancer research.(32, 33) He was also a trained gastrointestinal pathologist and he quickly recognized that the low neoplastic cellularity of pancreatic cancer would hinder the discovery of cancer

7

Journal Pre-proof causing genes. Kern therefore xenografted human pancreatic cancers into nude mice, creating highly cellular lesions that formed the basis for gene discovery.(34) The impact of a creative scientist having access to large numbers of biosamples can be seen in the rapid pace of gene discovery that followed. The p16/CDKN2A gene was discovered by Kamb and colleagues in 1994, and later that same year, Kern and colleagues reported that p16/CDKN2A was also targeted in pancreatic cancer.(35, 36) That next year, in 1995, Kern, using

of

the technique of “representational difference analysis” developed by M. Wigler at the Cold

ro

Spring Harbor Laboratories, discovered a homozygous deletion in a pancreatic cancer that

-p

proved to be the BRCA2 gene.(37) Immediately after BRCA2 was discovered, in 1996, Kern carefully mapped overlapping homozygous deletions on chromosome 18q in a series of

re

pancreatic cancers and discovered the SMAD4 gene.(38) Pancreatic cancer scientists were no

lP

longer followers; they were now leading the field of cancer research. Searching the entire genome, one gene at a time, for pancreatic cancer genes was

na

laborious and time consuming. In an audacious effort, Vogelstein and colleagues, in 2008, used

ur

Sanger sequencing to sequence the entire exome of a series of pathologically and clinically well-

Jo

annotated pancreatic cancers.(39) The core signaling pathways targeted in pancreatic cancer were defined across the entire exome (Figure 2). Four years later, the International Cancer Genome Consortium (ICGC), led by Andrew Biankin and Sean Grimonds, reported the sequencing the genomes of a series of pancreatic cancers, and in 2017 The Cancer Genome Atlas program (TCGA), led by Ben Raphael and Ralph Hruban, reported an in depth integrated characterization of the genetic alterations and gene expression changes in pancreatic cancer.(40, 41) In 2016, Nicholas Roberts and colleagues reported the sequencing of the germline genomes of 638 individuals with familial pancreatic cancer.(42) In the flash of slightly less than a decade, pancreatic cancer became one of the best-understood cancers at the genetic

8

Journal Pre-proof level. Large-scale team science was required for these efforts to succeed, and the pancreatic cancer field, with its collaborative spirit, was uniquely suited to succeed (Figure 3).

Applying genetics to patient care

Pathologist have taken advantage of the genetic advances described above and have

of

used genetics as a tool to improve the classification and treatment of pancreatic neoplasms.

Genetics has helped classify pancreatic neoplasms. For example, it was unclear which

ro

cells in undifferentiated carcinomas with osteoclast-like giant cells were neoplastic; the

-p

prominent giant cells, or the scattered atypical mononuclear cells. Westra and colleagues used

re

KRAS and TP53 mutations to show that it is the atypical mononuclear cells that are neoplastic

lP

and share genetic features with conventional ductal adenocarcinoma, and that the osteoclastlike giant cells are reactive, supporting the classification of these neoplasms as true

na

carcinomas.(43) In another example, Agaimy and colleagues used SMARCB1 (INI1) loss to define a previously unrecognized rhabdoid subtype of undifferentiated carcinoma of the pancreas.(44)

ur

Genetics has not replaced morphology, instead, pancreatic pathologists have used it as a tool to

Jo

advance morphology-based tumor classification.(45)

Genetics has also improved our clinical ability to make diagnoses. The SMAD4 gene is inactivated in 55% of pancreatic cancers, and immunolabeling for the Smad4 protein reflects SMAD4 gene status.(46) One can therefore use Smad4 immunolabeling to determine, for example, whether an atypical gland in a small pancreatic biopsy is likely malignant.

Genetics has also been used to guide therapy. Medullary carcinomas of the pancreas often have microsatellite instability, and cancers with microsatellite instability are exquisitely sensitive to immunotherapy.(47) Similarly, pancreatic cancers arising in individuals with a 9

Journal Pre-proof deleterious variant in a germline Fanconi anemia pathway gene (BRCA1, BRCA2, or PALB2) are sensitive to poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors.(48) We foresee a future in which pathologists, integrating tumor histology with genetics, will be the ones determining the best treatment for our patients.

A personalized treatment approach made possible by the revolution in cancer genetics

of

has been tested in the IMPACT and COMPASS trials, and one-quarter to a half of pancreatic cancers harbor mutations that are potentially therapeutically targetable.(49, 50) The challenge

ro

is to identify these therapeutic targets and treat the patients in the face of a rapidly progressing

re

Pancreatic Intraepithelial Neoplasia

-p

disease.(49, 50)

lP

The lesions that we today call pancreatic intraepithelial neoplasia (PanIN) were described more than 100 years ago by S.P.L. Hulst in 1905 when he described

na

“Zwischenformen;” lesions in between normal ducts and invasive cancer, in the pancreas.(51)

ur

Sommers et al., and Cubilla and Fitzgerald drew attention to an association of PanIN with

Jo

pancreatic cancer, and Klöppel et al. reported on the relationship of human PanIN to experimental pancreatic carcinogenesis in Syrian hamsters.(52-54) Klimstra and Longnecker then introduced the term PanIN in an editorial 1994, and genetic analyses, spearheaded by Wilentz and Hruban, helped establish PanINs as bona fide precancers (Figure 4).(55-62) The recognition that PanINs are precancers has formed a scientific basis for early detection and chemo/immune prevention research.

Cystic neoplasms

10

Journal Pre-proof The first real break in our understanding of cystic neoplasms of the pancreas came in 1978 when J.E. Oertel and J. Compagno, and independently D.J. Hodgkinson and colleagues, separated serous from mucin-producing cystic neoplasms of the pancreas (63, 64). These groups recognized that serous cystic neoplasms composed of glycogen-rich cuboidal cells were benign, while the mucin-producing cystic neoplasms had significant malignant potential (Figure 5). This was a critical advance as it identified a distinct cystic neoplasm that was almost

of

uniformly benign, and therefore did not require aggressive management.

ro

Soon thereafter, in 1982, K. Ohhashi and colleagues in Japan described four mucin-

-p

producing tumors of the pancreas that had led to duct dilatation and that are today known as intraductal papillary mucinous neoplasms (IPMNs) (Figure 5), a term introduced in 1994 by Sessa

re

and colleagues.(65, 66) As IPMNs had several features in common with mucinous cystic

lP

neoplasms (MCNs), the clear separation of MCNs from IPMNs by Zamboni and colleagues in 1999 allowed follow-up studies showing that non-invasive mucinous cystic neoplasms, defined

na

by the presence of ovarian stroma, are cured when surgically resected. (67, 68) By contrast,

ur

there is a significant risk of synchronous and even metachronous disease with IPMNs.(69)

Jo

Again, careful pathologic studies helped define clinically significant groups of patients. The report by K. Ohhashi and colleagues began what would prove to be a long series of noteworthy contributions by investigators in Japan characterizing IPMNs.(70) Grossly and clinically, it was recognized that IPMNs involving the main pancreatic duct are more likely to harbor high-grade dysplasia or an associated invasive carcinoma than are IPMNs that arise in branches off the main duct.(71, 72) This finding helped prioritize which patients needed surgery, and which had a low risk of developing invasive cancer and could therefore be clinically observed – an important contribution given the high frequency with which IPMNs are now being detected on cross-sectional imaging (73).

11

Journal Pre-proof At the microscopic level, it quickly became clear that the neoplastic cells of IPMNs can have a variety of directions of differentiation. Biliary, intestinal, and gastric directions of differentiation were identified, as were intraductal neoplasms with tubulopapillary architecture and intraductal neoplasms with oncocytic differentiation.(66, 74-76) This histologic classification of intraductal neoplasms was useful for pathologists as it helped define the observed patterns of progression. For example, it was shown that invasive colloid carcinomas of the pancreas almost

of

always arise in association with IPMNs with intestinal differentiation.(77) The grading of

ro

dysplasia was also improved over time.(70) The term “adenoma” was replaced by low-grade

-p

dysplasia, and “in situ carcinoma” was replaced with high-grade dysplasia, reflecting the continuous spectrum of neoplastic evolution that occurs in IPMNs and reducing the confusion

re

generated by the use of the term “malignant IPMN.”(78)

lP

As experience correlating preoperative clinical findings with pathological examination of resected lesions grew, consensus criteria were developed to guide the clinical management of

na

patients with IPMNs. The “Sendai criteria” were developed by M. Tanaka and colleagues in

ur

Sendai Japan in 2004 and published in 2006, and these criteria were subsequently revised as the

Jo

“Fukuoka criteria” in 2012.(79, 80) These criteria have proven valuable in the preoperative management of the growing number of patients being diagnosed with a pancreatic cyst, and the development of these criteria highlights the impact pathologists and clinicians can have when they work closely together. The Fukuoka criteria, however, are not perfect. They have high sensitivity in identifying high-risk cysts, but poor specificity.(81) Pancreas pathologists have risen to the challenge of improving the management of pancreas cysts by correlating cyst histopathology with the results of genetic sequencing. Indeed, the most recent advances in the management of cystic neoplasms of the pancreas have come from genetics. The exomes of a series of surgically

12

Journal Pre-proof resected and pathologically well-characterized cystic neoplasms of the pancreas have been sequenced, and the results directly correlated with cyst pathology.(82, 83) Remarkably, the genetic alterations identified closely match the histologic cyst type, some genetic alterations correlate with grade of dysplasia, and the genetic changes in the neoplastic cells can be detected in cyst fluid.(82-84) The close correlation of genetic alterations with cyst pathology allows for the development of new clinical tests based on cyst fluid sequencing to aid in the

of

preoperative diagnosis and management of cysts in the pancreas.(82, 85)

ro

These same genetic tools have been used to improve the classification of cystic lesions

-p

in the pancreas. Genetic analyses have shown that many intraductal lesions intermediate in size between PanINs and IPMNs are, in fact, small (“incipient”) IPMNs; that IPMNs can be truly

re

multifocal; and that some IPMNs progress to invasive cancer.(45, 86, 87) More recently, O.

lP

Basturk and colleagues showed that intraductal tubulopapillary neoplasms (ITPNs) lack the common alterations of IPMNs and represent a distinct neoplastic entity.(88) Similarly, A. Singhi

na

has shown that, unlike other intraductal neoplasms, gene fusions cause intraductal papillary

ur

oncocytic neoplasms (IOPNs), further helping to establish IOPNs as a unique tumor type

Jo

(personal communication).

Acinar neoplasms and solid pseudopapillary neoplasm Our understanding of non-ductal pancreatic neoplasms, such as the family of pancreatic neoplasms with acinar differentiation (acinar cell carcinoma, mixed acinar carcinomas with neuroendocrine and/or ductal differentiation, and pancreatoblastoma) along with solid pseudopapillary neoplasm, has also increased dramatically in the past 50 years. The distinction of acinar cell carcinoma from other types of pancreatic cancer was initially based upon the occasional occurrence of a dramatic syndrome of subcutaneous fat

13

Journal Pre-proof necrosis and polyarthralgia, first reported by Berner in 1908.(89) Additional case reports subsequently characterized the histologic features of acinar cell carcinoma, which was defined by evidence that the neoplastic cells produce exocrine enzymes such as trypsin, chymotrypsin, and lipase, the last responsible for the classic paraneoplastic syndrome now designated the lipase hypersecretion syndrome, or pancreatic panniculitis.(90) Demonstration of enzyme production by the neoplastic cells, now regarded as the sine qua non of the diagnosis, was first

of

based on electronic microscopy, and in 1987 Morohoshi et al. first used immunohistochemical

ro

labeling for trypsinogen, chymotrypsinogen, and lipase to define acinar differentiation in

-p

pancreatic neoplasms.(91, 92) The first modern clinicopathologic studies of acinar cell carcinoma were published in the early 1990s by Klimstra et al. and Hoorens and colleagues.(93,

re

94) With the morphologic features more fully delineated, the genetic alterations that drive

lP

acinar cell carcinoma and related mixed acinar neuroendocrine, mixed acinar ductal, and mixed acinar neuroendocrine ductal carcinomas could then be characterized.(95-100) The mutations

na

commonly found in ductal adenocarcinomas, KRAS in particular, were rarely detected; instead,

ur

alterations in the wnt pathway (APC primarily), mismatch repair genes, and DNA repair genes

Jo

such as BRCA1/2 were found. In 2014, Chmielecki et al. first demonstrated BRAF and RAF1 fusions in acinar cell carcinomas; these alterations and other potentially targetable genetic changes have now been shown in 35-65% of these carcinomas.(101) Pancreatoblastoma was first reported by Becker in 1957 and the term pancreatoblastoma was proposed by Horie at al. in 1977.(102, 103) Immunohistochemical studies demonstrating consistent acinar differentiation, along with common less abundant elements showing neuroendocrine and ductal differentiation, paralleled the work in acinar cell carcinomas, as did molecular studies that similarly showed alterations in the wnt pathway mostly involving CTNNB1 rather than APC.(104) The other alterations found in acinar cell

14

Journal Pre-proof carcinomas are lacking in pancreatoblastoma, however, as are mutations in the genes altered in ductal adenocarcinomas. Solid pseudopapillary neoplasm was first illustrated by Frantz in the Armed Forces Institute of Pathology Fascicle on Tumors of the Pancreas in 1959(Figures 1 and 5).(105) Over the past decades, numerous studies using electron microscopy, immunohistochemistry, and molecular analyses have failed to fully define the direction of differentiation for this enigmatic

of

neoplasm, and no normal cellular counterpart exists. Abraham et al.’s finding of nearly universal

ro

mutations in CTNNB1 led to a helpful immunohistochemical marker, -catenin, which shows

-p

diffuse abnormal nuclear labeling.(106) Many other markers are typically expressed but lack specificity, and the immunohistochemical evaluation is largely based on excluding acinar and

re

specific neuroendocrine differentiation. Despite the lack of clarity about its differentiation, solid

lP

pseudopapillary neoplasm has highly reproducible clinical features.(107) It largely affects young women and has an outstanding prognosis, with long-term survival even in the 10-15% of

ur

na

patients with metastatic disease.

Jo

Pancreatic neuroendocrine tumors The past fifty years have also witnessed dramatic developments in our understanding of the genetic drivers of pancreatic neuroendocrine tumors (PanNETs), as well as significant improvements in the classification of these neoplasms. Fifty years ago, the “grading” of PanNETs was an unusual combination of staging and histologic grading. Today, thanks in many ways to the work of the European NeuroEndocrine Tumor Society (ENETS), a reproducible grading system based on histomorphology and proliferation rate has been developed that accurately predicts patient prognosis.(108, 109) Most recently, the introduction of a four tier grading system, one that separates well-

15

Journal Pre-proof differentiated PanNETs grades 1-3 from poorly-differentiated neuroendocrine carcinomas (NECs) has allowed the development of distinct treatment protocols for patients with very high risk lesions.(110) Just as the sequencing of pancreatic cancer provided deep insights into the fundamental biology of pancreatic cancer, so too has the sequencing of the exomes of a series of wellcharacterized PanNETs profoundly advanced our understanding of PanNETs.(111, 112) In sharp

of

contrast to the genetic profile of ductal adenocarcinomas, the genes targeted in PanNETs

ro

include MEN1, DAXX, ATRX and several mTOR pathway genes including TSC2.(111, 112) The

-p

alterations in DAXX and ATRX in PanNETs were discovered to be tightly linked to alternative lengthening of telomeres (ALT), a unique mechanism of telomere maintenance.(111, 112) Thus,

re

the study of tumors of the pancreas led to the discovery of the genetic basis of a novel

lP

mechanism of telomere maintenance. Pancreas cancer research is again leading the field! This new understanding of the genetics of PanNETs has been used to support the

na

separation of poorly-differentiated NECs from grade 3 well-differentiated PanNETs, as MEN1,

ur

DAXX, ATRX and mTOR pathway genes are not typically targeted in NECs. Instead, NECs typically

Jo

harbor TP53 and RB1 gene alterations.(113) Immunohistochemical markers for most of these genomic alterations allow simple application in routine diagnosis.(114) This separation of PanNETs from NECs is another example of how the integration of genetics has added to, and not replaced, morphologic diagnoses in the pancreas. Similarly, particular tumor morphologies have been discovered to characterize two recently described inherited tumor syndromes, familial insulinomatosis and glucagon cell hyperplasia and neoplasia (GCHN). (115-117) These syndromes were found to be linked to germ line mutations in the genes MAFA1 and GR, respectively. Pathologists have shown that the insulin producing PanNETs in insulinomatosis all have a trabecular histology and develop from

16

Journal Pre-proof islets without any preceding insulin cell hyperplasia, and that the glucagon-producing PanNETs in GCHN emerge from islets with conspicuous glucagon cell hyperplasia. (118, 119)

Non-neoplastic Beta Cell Disorders

Our understanding of diabetes mellitus has also advanced dramatically. Diabetes is no longer considered a single disease, but instead is now recognized to be a group of disorders

of

whose major types are juvenile onset diabetes, now called type 1, and adult onset or type 2

ro

diabetes. The insulin producing beta cells of the pancreatic islets have an essential role in both

-p

forms of diabetes, although their histological changes are often not eye-catching. It is therefore not surprising that the changes in the beta cells have only been defined in the last 50 years.

re

Meticulous histopathological studies in type 1 diabetics revealed that the cause of the

lP

insulin deficiency in these patients is an autoimmune process with beta cell destruction by Tlymphocytes, a finding called insulitis.(120-122) Type 2 diabetes remains to be a complex and

na

enigmatic disease in which the beta cells are involved, but are only one player in a combination

ur

of pathogenetic factors that include obesity, insulin resistance and strong genetic susceptibility.

Jo

In sharp contrast to type 1 diabetes, the beta cells in type 2 diabetes are always present in the pancreas, although their number may be reduced, particularly if they are affected by deposition of islet amyloid polypeptide in the islets. The conclusion from these observations is that the beta cells in type 2 diabetes develop an increasing (inborn?) inability to deliver enough insulin, mainly because of excessive demand due to long-lasting insulin resistance related to obesity. The last fifty years have witnessed dramatic improvements in our understanding of other diseases that disturb the regulation of glucose. For example, the morphology of congenital hyperinsulinism in infancy (CHI), was described in its various forms in the 70s, 80s and 90s under the term nesidioblastosis, and the first underlying genetic changes causing CHI were discovered 17

Journal Pre-proof at the end of the 90s.(123-127) These genetic studies revealed a number of abnormalities in genes whose proteins are all involved in the process of insulin secretion.(128) Diffuse disease is most frequently associated with recessively inherited loss-of-function mutations of ABCC8 (SUR1) or KCNJ11 (Kir6.2), both of which code for proteins regulating the K(ATP)-channel in the beta cell membrane that controls insulin secretion by beta cell depolarization, increased Ca2+ entry, and elevated intracellular Ca2+. In focal disease there is a paternally inherited KCNJ11 or

of

ABCC8 mutation and the loss of the corresponding maternal allele in β cells of a group of

ro

islets.(129) Recently a morphological variant has been genetically defined, which is a kind of

-p

mixture (“mosaicism”) of diffuse with focal disease, in which hyperactive islets are located in one or in a few adjacent lobules, whereas hypoactive islets are present in the whole

lP

re

pancreas.(130, 131)

Pancreatitis

na

Our understanding of pancreatitis has similarly undergone significant change over the

ur

past five decades, again, thanks to significant contributions from pathologists.(132) Acute and

Jo

chronic pancreatitis were long-regarded as two separate diseases. However in the 80s and 90s it was recognized, in studies analyzing the pancreatic changes in resection specimens of patients with a long history of recurrent pancreatitis, that chronic pancreatitis, particularly when caused by alcohol abuse, evolves from recurrent episodes of acute pancreatitis, a pathogenetic concept that was termed the necrosis-fibrosis sequence.(133-135) In other studies it was noted that the pathology of chronic pancreatitis, which was formerly considered to be uniform and “nonspecific,” varies according to the different causative factors of the disease, such as alcohol abuse, inherited gene mutations, autoimmune syndromes, metabolic disturbances, environmental conditions and anatomical abnormalities.(136) The rather vague descriptive

18

Journal Pre-proof term chronic sclerosing pancreatitis has therefore been replaced by etiologically more specific terms, such as alcoholic chronic pancreatitis, hereditary pancreatitis, autoimmune pancreatitis , paraduodenal pancreatitis and obstructive chronic pancreatitis. Among these forms of pancreatitis two have received particular attention in recent years: autoimmune pancreatitis and hereditary pancreatitis.

of

The association of pancreatitis with autoimmune diseases, including Sjögrens syndrome and sclerosing cholangitis, has been recognized for more than a half century.(137-144) Patients

ro

with autoimmune pancreatitis sometimes undergo surgery because the disease can clinically

-p

mimic a neoplasm, giving pathologists an opportunity to define the histopathology of this

re

disease and describe its lymphoplasmacytic and fibrosing features. The fact that patients with

lP

this type of pancreatitis respond rapidly to steroid therapy fostered the hypothesis that the disease has an autoimmune pathogenesis.(145, 146) In 2001 Hamano and colleagues reported

na

the association with elevated serum IgG4 levels.(147) This discovery aided diagnosis as pathologists can now use immunolabeling for IgG4 to make diagnoses on limited biopsies.(148,

ur

149) Subsequently, two forms of autoimmune pancreatitis were recognized: type I is

Jo

characterized by increased numbers of IgG4 expressing cells, periductal lymphoplasmacytic infiltrates, storiform fibrosis, obliterative venulitis and is commonly associated with a generalized disorder called IgG4 related disease, and type II, which is often associated with inflammatory bowel disease, by the presence of granulocytic epithelial lesions (GELs) in the pancreas.(148, 150-157) Deltlefsen and colleagues described the deposition of complement at the basement membrane of pancreatic ducts, and recently, M. Shiokawa and colleagues suggested that the autoantigen responsible for autoimmune pancreatitis is a cleaved form of laminin 511, and antibodies to laminin 511 are already being used to aid in clinical diagnoses.(158-160) As exciting and clinically useful as these advances are, we by no means truly 19

Journal Pre-proof understand autoimmune pancreatitis. If antibodies to cleaved laminin 511 cause the disease, why are IgG4 levels increased?

Another major revolution in pancreatitis has been the discovery of germline variants that predispose to the disease. Whitcomb and colleagues first reported that germline variants in PRSS1 cause pancreatitis in 1996, and in the ensuing decades SPINK1, CFTR, CTRC and other

of

genes have all been shown to increase the risk of pancreatitis.(132, 161) Today, many patients previously diagnosed as having idiopathic pancreatitis, are correctly classified as having a

ro

familial form of this disease.(162, 163) This obviously has significant implications for other

-p

family members, and since some forms of inherited pancreatitis are associated with a high

lP

re

lifetime risk of developing pancreatic cancer, for the patients themselves.(164)

Conclusions

na

The last 50 years have witnessed dramatic advances in pancreatic pathology.(165) Many of these advances have been driven by the revolution in cancer genetics. Unlike some

ur

other fields, genetics has largely added to, and not replaced, histology in the pancreas.

Jo

Many of the most significant of these advances in pancreatic pathology have been made possible by collaborations, by team science (Figure 3). The pancreas group has been marked by enormous collegiality and warmth (Figure 6). Cases, biosamples and ideas are freely shared, and a sense of comradery prevails over individuality. Our hope looking forward is that that this collegiality will continue, for great challenges remain ahead. As we put the dramatic changes of the past fifty years in perspective, it is clear that change and innovation have been part of medicine from the beginning. While in the past, centuries elapsed between these innovative waves in medicine, today these waves are coming faster and faster. On the crest of one of these waves it all seems clear and inevitable, only to be 20

Journal Pre-proof overturned by the next wave (Figure 7). As soon as we think we understand an entity, our paradigm is quickly overthrown by a new understanding. We should expect that our work, our current understanding of pancreatic pathology, will be overturned. One thing that the history of medicine has taught us- there will always be more waves. Indeed, new waves of innovation are sorely needed in the war against pancreatic cancer as the five year survival rate remains a dismal 8%, and pancreatic cancer is predicted to become the second leading cause of cancer

of

death in the United States by the year 2030.(166) As L. Premuda states in the Preface to John

ro

Howard and Walter Hess’ magnificent book on the history of the pancreas, “its history, centuries

-p

old, is still in its adolescence.”(1) We have lived through an amazing fifty years, but wonderfully disruptive advances are yet to come.

re

The authors should end with a disclaimer. In this review we have attributed some

lP

discoveries to individual people. We are fully aware that it is impossible to accurately attribute any discovery to a single person, and we remind the reader of Stigler’s law of eponymy, that “no

na

scientific discovery is named after its original discoverer.”(167) We apologize to the many

ur

people who contributed to advancing the understanding of pancreas pathology, but were not

Jo

recognized in this review, although in our defense we note that in an intended act of irony, Stigler was not the first to describe Stigler’s law of eponymy!

21

Journal Pre-proof Figure Legends-

Figure 1: The four American Registry of Pathology “Fascicles” that span the last 60 years. The first edition, by Virginia Frantz, was published in 1959. The second edition by Antonio Cubilla and Patrick Fitzgerald, was published in 1984. The third edition, published in 1997, was written by Enrico Solcia, Carlo Capella, and Günter Klöppel. The fourth edition, by Ralph Hruban,

of

Martha Pitman and David Klimstra came out in 2007. The dramatic changes in the classification systems described in these Fascicles nicely highlights the changes in pancreas pathology over

-p

ro

the last 50 years.

Figure 2: The genes somatically targeted in ductal adenocarcinoma of the pancreas can be

re

visualized in this plot of the chromosome location (X axis) and prevalence of alterations (Y axis).

lP

Note the four “mountains,” KRAS, TP53, SMAD4 and p16/CDKN2A. (Courtesy of Duc Nguyen)

na

Figure 3: The team approach has characterized pancreatic cancer research. The American Association for Cancer Research awarded their 2013 Team Science Award to the team that

ur

sequenced the first exomes of pancreatic cancer. (Pictured are some of the awardees. From left

Jo

to right in the first row are D Klimstra, K Kinzler, R Hruban, B Vogelstein. N Papadopolos, M Choti and V Velculescu. Back rows are C Wolfgang, J Herman, S Kern, NV Adsay, LD Wood, A Klein and C Iacobuzio-Donahue).

Figure 4: The 2004 meeting at Johns Hopkins to define the terminology for the classification of precancerous lesions in the pancreas. (Pictured from left to right; first row: Y Kato, NV Adsay, GJA Offerhaus, DS Klimstra, RH Hruban, K Takaori, D Longnecker, T Furukawa, J Lüttges. Second row: N Prasad, JA Albores-Saavedra, D Sui, AV Biankin, A Maitra, M Shimizu, A Yeh,u and N Fukushima) 22

Journal Pre-proof Figure 5: The four most common cystic neoplasms of the pancreas. Serous cystadenoma (A), mucinous cystic neoplasm (B), Intraductal papillary mucinous neoplasm (C) and solid pseudopapillary neoplasm (D). Fifty years ago these entities were broadly lumped together.

Figure 6: The authors, D. Klimstra, R. Hruban, G. Zamboni and G. Klöppel. Together, with others, their years of collaboration and friendship characterize the attitude of the field of

of

pancreas pathology.

ro

Figure 7: “Catch a wave and you can see real far.” On the crest of a disruptive wave of

-p

innovation, the field seems clear, only to be disrupted by the next innovative wave. (Illustration created by Hannah Ahn for Ralph Hruban’s 2014 Maude Abbott lecture. With permission,

Jo

ur

na

lP

re

copyright Hannah Ahn).

23

Journal Pre-proof References

Jo

ur

na

lP

re

-p

ro

of

1. Howard JM, Hess W. History of the pancreas: mysteries of a hidden organ. New York: Kluwer Academic/Penum Publishers, 2002. 2. Bay NS, Bay BH. Greek anatomist herophilus: the father of anatomy. Anat Cell Biol 2010; 43, 280-283. 3. Busnardo AC, DiDio LJ, Tidrick RT, Thomford NR. History of the pancreas. Am J Surg 1983; 146, 539-550. 4. Tsuchiya R, Kuroki T, Eguchi S. The pancreas from Aristotle to Galen. Pancreatology 2015; 15, 27. 5. Mesquita ET, Souza Junior CV, Ferreira TR. Andreas Vesalius 500 years--A Renaissance that revolutionized cardiovascular knowledge. Rev Bras Cir Cardiovasc 2015; 30, 260-265. 6. Howard JM, Hess W, Traverso W. Johann Georg Wirsung (1589-1643) and the pancreatic duct: the prosector of Padua, Italy. J Am Coll Surg 1998; 187, 201-211. 7. Vale FF, Correa PC. Response to Giovanni Battista Morgagni and the Foundation of Modem Medicine. Clin Cardiol 2016; 39, 484-485. 8. Ghosh SK. Giovanni Battista Morgagni (1682-1771): father of pathologic anatomy and pioneer of modern medicine. Anat Sci Int 2017; 92, 305-312. 9. Scarani P. Rudolf Virchow (1821-1902). Virchows Arch 2003; 442, 95-98. 10. King KM, Rubin G. A history of diabetes: from antiquity to discovering insulin. Br J Nurs 2003; 12, 1091-1095. 11. Are C, Dhir M, Ravipati L. History of pancreaticoduodenectomy: early misconceptions, initial milestones and the pioneers. HPB : the official journal of the International Hepato Pancreato Biliary Association 2011; 13, 377-384. 12. He J, Ahuja N, Makary MA, Cameron JL, Eckhauser FE, Choti MA, Hruban RH, Pawlik TM, Wolfgang CL. 2564 resected periampullary adenocarcinomas at a single institution: trends over three decades. HPB : the official journal of the International Hepato Pancreato Biliary Association 2014; 16, 8390. 13. Olurin EO. Pancreatic cysts: a report of 10 cases. The British journal of surgery 1971; 58, 502508. 14. Warren KW, Hardy KJ. Cystadenocarcinoma of the pancreas. Surgery, gynecology & obstetrics 1968; 127, 734-736. 15. Sawyer RB, Sawyer KC, Spencer JR. Proliferative cysts of the pancreas. Am J Surg 1968; 116, 763767. 16. Scott-Young M. Cystadenoma of the pancreas. Discussion and case report. Med J Aust 1971; 1, 468-470. 17. Cubilla AL, Fitzgerald PJ. Classification of pancreatic cancer (nonendocrine). Mayo Clinical Proceedings 1979; 54, 449-458. 18. Morohoshi T, Held G, Klöppel G. Exocrine pancreatic tumours and their histological classification. A study based on 167 autopsy and 97 surgical cases. Histopathology 1983; 7, 645-661. 19. Greider MH, Bencosme SA, Lechago J. The human pancreatic islet cells and their tumors. I. The normal pancreatic islets. Laboratory investigation; a journal of technical methods and pathology 1970; 22, 344-354. 20. Kraft AR, Tompkins RK, Zollinger RM. Recognition and management of the diarrheal syndrome caused by nonbeta islet cell tumors of the pancreas. Am J Surg 1970; 119, 163-170.

24

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

21. Ellison EC, Johnson JA. The Zollinger-Ellison syndrome: a comprehensive review of historical, scientific, and clinical considerations. Curr Probl Surg 2009; 46, 13-106. 22. Editorial. Mechanism of pancreatic autodigestion. The New England journal of medicine 1970; 283, 487-488. 23. Fitzgerald; PJ, Morrison AB. The Pancreas. Baltimore: Williams and Wilkins, 1980. 24. Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most human carcinomas of the exocrine pancreas contain mutant c- K-ras genes. Cell 1988; 53, 549-554. 25. Stehelin D, Varmus HE, Bishop JM, Vogt PK. DNA related to the transforming gene(s) of avian sarcoma viruses is present in normal avian DNA. Nature 1976; 260, 170-173. 26. DeLeo AB, Jay G, Appella E, Dubois GC, Law LW, Old LJ. Detection of a transformation-related antigen in chemically induced sarcomas and other transformed cells of the mouse. Proc Natl Acad Sci U S A 1979; 76, 2420-2424. 27. Barton CM, Staddon SL, Hughes CM, Hall PA, O'Sullivan C, Klöppel G, Theis B, Russell RCG, Neoptolemos J, Williamson RCN, Lane DP, Lemoine NR. Abnormalities of the p53 tumor suppressor gene in human pancreatic cancer. British Journal of Cancer 1991; 64, 1076-1082. 28. Cameron JL, He J. Two thousand consecutive pancreaticoduodenectomies. J Am Coll Surg 2015; 220, 530-536. 29. Warshaw AL. Four decades fighting pancreatic cancer. The American surgeon 2010; 76, 921-924. 30. Cameron JL, Pitt HA, Yeo CJ, Lillemoe KD, Kaufman HS, Coleman J. One hundred and forty-five consecutive pancreaticoduodenectomies without mortality. Ann Surg 1993; 217, 430-435; discussion 435-438. 31. Gordon TA, Burleyson GP, Tielsch JM, Cameron JL. The effects of regionalization on cost and outcome for one general high-risk surgical procedure. Ann Surg 1995; 221, 43-49. 32. Kern SE. Where's the passion? Cancer Biol Ther 2010; 10, 655-657. 33. Brody JR, Kern SE. Stagnation and herd mentality in the biomedical sciences. Cancer Biol Ther 2004; 3, 903-910. 34. Hahn SA, Seymour AB, Hoque AT, Schutte M, da Costa LT, Redston MS, Caldas C, Weinstein CL, Fischer A, Yeo CJ, Hruban RH, Kern SE. Allelotype of pancreatic adenocarcinoma using xenograft enrichment. Cancer Research 1995; 55, 4670-4675. 35. Kamb A, Gruis NA, Weaver-Feldhaus J, Liu Q, Harshman K, Tavtigian SV, Stockert E, Day RS, III, Johnson BE, Skolnick M. A cell cycle regulator potentially involved in genesis of many tumor types. Science 1994; 264, 436-440. 36. Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, Seymour AB, Weinstein CL, Hruban RH, Yeo CJ, Kern SE. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nature Genet 1994; 8, 27-32. 37. Schutte M, da Costa LT, Hahn SA, Moskaluk CA, Hoque AT, Rozenblum E, Weinstein CL, Meltzer PS, Trent JM, Yeo CJ, Hruban RH, Kern SE. Identification by representational difference analysis of a homozygous deletion in pancreatic carcinoma that lies within the BRCA2 region. Proc Natl Acad Sci U S A 1995; 92, 5950-5954. 38. Hahn SA, Schutte M, Hoque AT, Moskaluk CA, da Costa LT, Rozenblum E, Weinstein CL, Fischer A, Yeo CJ, Hruban RH, Kern SE. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 1996; 271, 350-353. 39. Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A, Hong SM, Fu B, Lin MT, Calhoun ES, Kamiyama M, Walter K, Nikolskaya T, Nikolsky Y, Hartigan J, Smith DR, Hidalgo M, Leach SD, Klein AP, Jaffee EM, Goggins M, Maitra A, Iacobuzio-Donahue C, Eshleman JR, Kern SE, Hruban RH, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 2008; 321, 1801-1806. 25

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

40. Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K, Quinn MC, Robertson AJ, Fadlullah MZ, Bruxner TJ, Christ AN, Harliwong I, Idrisoglu S, Manning S, Nourse C, Nourbakhsh E, Wani S, Wilson PJ, Markham E, Cloonan N, Anderson MJ, Fink JL, Holmes O, Kazakoff SH, Leonard C, Newell F, Poudel B, Song S, Taylor D, Waddell N, Wood S, Xu Q, Wu J, Pinese M, Cowley MJ, Lee HC, Jones MD, Nagrial AM, Humphris J, Chantrill LA, Chin V, Steinmann AM, Mawson A, Humphrey ES, Colvin EK, Chou A, Scarlett CJ, Pinho AV, Giry-Laterriere M, Rooman I, Samra JS, Kench JG, Pettitt JA, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Jamieson NB, Graham JS, Niclou SP, Bjerkvig R, Grutzmann R, Aust D, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Wolfgang CL, Morgan RA, Lawlor RT, Corbo V, Bassi C, Falconi M, Zamboni G, Tortora G, Tempero MA, Australian Pancreatic Cancer Genome I, Gill AJ, Eshleman JR, Pilarsky C, Scarpa A, Musgrove EA, Pearson JV, Biankin AV, Grimmond SM. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015; 518, 495-501. 41. Cancer Genome Atlas Research Network. Electronic address aadhe, Cancer Genome Atlas Research N. Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma. Cancer Cell 2017; 32, 185-203 e113. 42. Roberts NJ, Norris AL, Petersen GM, Bondy ML, Brand R, Gallinger S, Kurtz RC, Olson SH, Rustgi AK, Schwartz AG, Stoffel E, Syngal S, Zogopoulos G, Ali SZ, Axilbund J, Chaffee KG, Chen YC, Cote ML, Childs EJ, Douville C, Goes FS, Herman JM, Iacobuzio-Donahue C, Kramer M, Makohon-Moore A, McCombie RW, McMahon KW, Niknafs N, Parla J, Pirooznia M, Potash JB, Rhim AD, Smith AL, Wang Y, Wolfgang CL, Wood LD, Zandi PP, Goggins M, Karchin R, Eshleman JR, Papadopoulos N, Kinzler KW, Vogelstein B, Hruban RH, Klein AP. Whole Genome Sequencing Defines the Genetic Heterogeneity of Familial Pancreatic Cancer. Cancer Discov 2016; 6, 166-175. 43. Westra WH, Sturm PJ, Drillenburg P, Choti MA, Klimstra DS, Abores-Saavedra J, Montag A, Offerhaus GJ, Hruban RH. K-ras oncogene mutations in osteoclast-like giant cell tumors of the pancreas and liver: genetic evidence to support origin from the duct epithelium. Am J Surg Pathol 1998; 22, 12471254. 44. Agaimy A, Haller F, Frohnauer J, Schaefer IM, Strobel P, Hartmann A, Stoehr R, Klöppel G. Pancreatic undifferentiated rhabdoid carcinoma: KRAS alterations and SMARCB1 expression status define two subtypes. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 2015; 28, 248-260. 45. Matthaei H, Wu J, Dal Molin M, Shi C, Perner S, Kristiansen G, Lingohr P, Kalff JC, Wolfgang CL, Kinzler KW, Vogelstein B, Maitra A, Hruban RH. GNAS sequencing identifies IPMN-specific mutations in a subgroup of diminutive pancreatic cysts referred to as "incipient IPMNs". Am J Surg Pathol 2014; 38, 360-363. 46. Wilentz RE, Su GH, Dai JL, Sparks AB, Argani P, Sohn TA, Yeo CJ, Kern SE, Hruban RH. Immunohistochemical labeling for Dpc4 mirrors genetic status in pancreatic adenocarcinomas: a new marker of DPC4 inactivation. American Journal of Pathology 2000; 156, 37-43. 47. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Duffy SM, Goldberg RM, de la Chapelle A, Koshiji M, Bhaijee F, Huebner T, Hruban RH, Wood LD, Cuka N, Pardoll DM, Papadopoulos N, Kinzler KW, Zhou S, Cornish TC, Taube JM, Anders RA, Eshleman JR, Vogelstein B, Diaz LA, Jr. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. The New England journal of medicine 2015; 372, 2509-2520. 48. Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh DY, Reinacher-Schick A, Tortora G, Algul H, O'Reilly EM, McGuinness D, Cui KY, Schlienger K, Locker GY, Kindler HL. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. The New England journal of medicine 2019.

26

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

49. Lowery MA, Jordan EJ, Basturk O, Ptashkin RN, Zehir A, Berger MF, Leach T, Herbst B, Askan G, Maynard H, Glassman D, Covington C, Schultz N, Abou-Alfa GK, Harding JJ, Klimstra DS, Hechtman JF, Hyman DM, Allen PJ, Jarnagin WR, Balachandran VP, Varghese AM, Schattner MA, Yu KH, Saltz LB, Solit DB, Iacobuzio-Donahue CA, Leach SD, O'Reilly EM. Real-Time Genomic Profiling of Pancreatic Ductal Adenocarcinoma: Potential Actionability and Correlation with Clinical Phenotype. Clin Cancer Res 2017; 23, 6094-6100. 50. Aung KL, Fischer SE, Denroche RE, Jang GH, Dodd A, Creighton S, Southwood B, Liang SB, Chadwick D, Zhang A, O'Kane GM, Albaba H, Moura S, Grant RC, Miller JK, Mbabaali F, Pasternack D, Lungu IM, Bartlett JMS, Ghai S, Lemire M, Holter S, Connor AA, Moffitt RA, Yeh JJ, Timms L, Krzyzanowski PM, Dhani N, Hedley D, Notta F, Wilson JM, Moore MJ, Gallinger S, Knox JJ. Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial. Clin Cancer Res 2018; 24, 1344-1354. 51. Hulst SPL. Zur kenntnis der Genese des Adenokarzinoms und Karzinoms des Pankreas. Virchows Arch (B) 1905; 180, 288-316. 52. Sommers SC, Murphy SA, Warren S. Pancreatic duct hyperplasia and cancer. Gastroenterology 1954; 27, 629-640. 53. Cubilla AL, Fitzgerald PJ. Morphological lesions associated with human primary invasive nonendocrine pancreas cancer. Cancer Research 1976; 36, 2690-2698. 54. Klöppel G, Bommer G, Ruckert K, Seifert G. Intraductal proliferation in the pancreas and its relationship to human and experimental carcinogenesis. Virchows Arch A Pathol Anat Histol 1980; 387, 221-233. 55. Klimstra DS, Longnecker DS. K- ras mutations in pancreatic ductal proliferative lesions. American Journal of Pathology 1994; 145, 1547-1550. 56. Hosoda W, Chianchiano P, Griffin JF, Pittman ME, Brosens LA, Noe M, Yu J, Shindo K, Suenaga M, Rezaee N, Yonescu R, Ning Y, Albores-Saavedra J, Yoshizawa N, Harada K, Yoshizawa A, Hanada K, Yonehara S, Shimizu M, Uehara T, Samra JS, Gill AJ, Wolfgang CL, Goggins MG, Hruban RH, Wood LD. Genetic analyses of isolated high-grade pancreatic intraepithelial neoplasia (HG-PanIN) reveal paucity of alterations in TP53 and SMAD4. The Journal of pathology 2017; 242, 16-23. 57. Wilentz RE, Geradts J, Maynard R, Offerhaus GJ, Kang M, Goggins M, Yeo CJ, Kern SE, Hruban RH. Inactivation of the p16 (INK4A) tumor-suppressor gene in pancreatic duct lesions: loss of intranuclear expression. Cancer Res 1998; 58, 4740-4744. 58. Wilentz RE, Iacobuzio-Donahue CA, Argani P, McCarthy DM, Parsons JL, Yeo CJ, Kern SE, Hruban RH. Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Res 2000; 60, 2002-2006. 59. Hruban RH, Adsay NV, Albores-Saavedra J, Compton C, Garrett ES, Goodman SN, Kern SE, Klimstra DS, Klöppel G, Longnecker DS, Lüttges, J, Offerhaus GJ. Pancreatic intraepithelial neoplasia: a new nomenclature and classification system for pancreatic duct lesions. Am J Surg Pathol 2001; 25, 579586. 60. Lemoine NR, Jain S, Hughes CM, Staddon SL, Maillet B, Hall PA, Klöppel G. Ki- ras oncogene activation in preinvasive pancreatic cancer. Gastroenterology 1992; 102, 230-236. 61. Tabata T, Fujimori T, Maeda S, Yamamoto M, Saitoh Y. The role of Ras mutation in pancreatic cancer, precancerous lesions, and chronic pancreatitis. International Journal of Pancreatology 1993; 14, 237-244. 62. Yanagisawa A, Ohtake K, Ohashi K, Hori M, Kitagawa T, Sugano H, Kato Y. Frequent c-Ki- ras oncogene activation in mucous cell hyperplasias of pancreas suffering from chronic inflammation. Cancer Research 1993; 53, 953-956. 63. Compagno J, Oertel JE. Microcystic adenomas of the pancreas (glycogen-rich cystadenomas): a clinicopathologic study of 34 cases. Am J Clin Pathol 1978; 69, 289-298. 27

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

64. Hodgkinson DJ, ReMine WH, Weiland LH. Pancreatic cystadenoma. A clinicopathologic study of 45 cases. Arch Surg 1978; 113, 512-519. 65. Ohhashi K, Murakami Y, Takekoshi T. Four cases of "mucin producing" cancer of the pancreas on specific findings of the papilla of Vater (Abstr). Prog Diagn Endosc 1982; 20, 348-351. 66. Sessa F, Solcia E, Capella C, Bonato M, Scarpa A, Zamboni G, Pellegata NS, Ranzani GN, Rickaert F, Klöppel G. Intraductal papillary-mucinous tumours represent a distinct group of pancreatic neoplasms: an investigation of tumour cell differentiation and K-ras, p53 and c-erbB-2 abnormalities in 26 patients. Virchows Arch 1994; 425, 357-367. 67. Wilentz RE, Albores-Saavedra J, Zahurak M, Talamini MA, Yeo CJ, Cameron JL, Hruban RH. Pathologic examination accurately predicts prognosis in mucinous cystic neoplasms of the pancreas. Am J Surg Pathol 1999; 23, 1320-1327. 68. Zamboni G, Scarpa A, Bogina G, Iacono C, Bassi C, Talamini G, Sessa F, Capella C, Solcia E, Rickaert F, Mariuzzi GM, Klöppel G. Mucinous cystic tumors of the pancreas: clinicopathological features, prognosis, and relationship to other mucinous cystic tumors. Am J Surg Pathol 1999; 23, 410422. 69. Fischer CG, Guthrie VB, Braxton AM, Zheng L, Wang P, Song Q, Griffin JF, Chianchiano PE, Hosoda W, Niknafs N, Springer S, Molin MD, Masica D, Scharpf RB, Thompson ED, He J, Wolfgang CL, Hruban RH, Roberts NJ, Lennon AM, Jiao Y, Karchin R, Wood LD. Intraductal Papillary Mucinous Neoplasms Arise from Multiple Independent Clones, Each With Distinct Mutations. Gastroenterology 2019. 70. Tanaka M. Thirty years of experience with intraductal papillary mucinous neoplasm of the pancreas: from discovery to international consensus. Digestion 2014; 90, 265-272. 71. Itoh S, Ishiguchi T, Ishigaki T, Sakuma S, Maruyama K, Senda K. Mucin-producing pancreatic tumor: CT findings and histopathologic correlation. Radiology 1992; 183, 81-86. 72. Obara T, Maguchi H, Saitoh Y, Itoh A, Arisato S, Ashida T, Nishino N, Ura H, Namiki M. Mucinproducing tumor of the pancreas: natural history and serial pancreatogram changes. The American journal of gastroenterology 1993; 88, 564-569. 73. Laffan TA, Horton KM, Klein AP, Fishman EK, Johnson PT, Hruban RH. Prevelance of unsuspected pancreatic cysts on MDCT. AJRAmerican Journal of Roentgenology 2008; 191, 802-807. 74. Adsay NV, Adair CF, Heffess CS, Klimstra DS. Intraductal oncocytic papillary neoplasms of the pancreas. Am J Surg Pathol 1996; 20, 980-994. 75. Furukawa T, Klöppel G, Volkan Adsay N, Albores-Saavedra J, Fukushima N, Horii A, Hruban RH, Kato Y, Klimstra DS, Longnecker DS, Lüttges, J, Offerhaus GJ, Shimizu M, Sunamura M, Suriawinata A, Takaori K, Yonezawa S. Classification of types of intraductal papillary-mucinous neoplasm of the pancreas: a consensus study. Virchows Arch 2005; 447, 794-799. 76. Nakamura A, Horinouchi M, Goto M, Nagata K, Sakoda K, Takao S, Imai K, Kim YS, Sato E, Yonezawa S. New classification of pancreatic intraductal papillary-mucinous tumour by mucin expression: its relationship with potential for malignancy. The Journal of pathology 2002; 197, 201-210. 77. Seidel G, Zahurak M, Iacobuzio-Donahue C, Sohn TA, Adsay NV, Yeo CJ, Lillemoe KD, Cameron JL, Hruban RH, Wilentz RE. Almost all infiltrating colloid carcinomas of the pancreas and periampullary region arise from in situ papillary neoplasms: a study of 39 cases. Am J Surg Pathol 2002; 26, 56-63. 78. Basturk O, Hong SM, Wood LD, Adsay NV, Albores-Saavedra J, Biankin AV, Brosens LA, Fukushima N, Goggins M, Hruban RH, Kato Y, Klimstra DS, Klöppel G, Krasinskas A, Longnecker DS, Matthaei H, Offerhaus GJ, Shimizu M, Takaori K, Terris B, Yachida S, Esposito I, Furukawa T, Baltimore Consensus M. A Revised Classification System and Recommendations From the Baltimore Consensus Meeting for Neoplastic Precursor Lesions in the Pancreas. Am J Surg Pathol 2015; 39, 1730-1741.

28

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

79. Tanaka M, Chari S, Adsay NV, Fernandez-del Castillo C, Falconi M, Shimizu M, Yamaguchi K, Yamao K, Matsuno S. International consensus guidelines for management of intraductal papillary mucinous neoplasms and mucinous cystic neoplasms of the pancreas. Pancreatology 2006; 6 32. 80. Tanaka M, Fernandez-del Castillo C, Adsay V, Chari S, Falconi M, Jang JY, Kimura W, Levy P, Pitman MB, Schmidt CM, Shimizu M, Wolfgang CL, Yamaguchi K, Yamao K, International Association of P. International consensus guidelines 2012 for the management of IPMN and MCN of the pancreas. Pancreatology 2012; 12, 183-197. 81. Heckler M, Michalski CW, Schaefle S, Kaiser J, Buchler MW, Hackert T. The Sendai and Fukuoka consensus criteria for the management of branch duct IPMN - A meta-analysis on their accuracy. Pancreatology 2017; 17, 255-262. 82. Springer S, Masica DL, Dal Molin M, Douville C, Thoburn CJ, Afsari B, Li L, Cohen JD, Thompson E, Allen PJ, Klimstra DS, Schattner MA, Schmidt CM, Yip-Schneider M, Simpson RE, Fernandez-Del Castillo C, Mino-Kenudson M, Brugge W, Brand RE, Singhi AD, Scarpa A, Lawlor R, Salvia R, Zamboni G, Hong SM, Hwang DW, Jang JY, Kwon W, Swan N, Geoghegan J, Falconi M, Crippa S, Doglioni C, Paulino J, Schulick RD, Edil BH, Park W, Yachida S, Hijioka S, van Hooft J, He J, Weiss MJ, Burkhart R, Makary M, Canto MI, Goggins MG, Ptak J, Dobbyn L, Schaefer J, Sillman N, Popoli M, Klein AP, Tomasetti C, Karchin R, Papadopoulos N, Kinzler KW, Vogelstein B, Wolfgang CL, Hruban RH, Lennon AM. A multimodality test to guide the management of patients with a pancreatic cyst. Science translational medicine 2019; 11. 83. Wu J, Matthaei H, Maitra A, Dal Molin M, Wood LD, Eshleman JR, Goggins M, Canto MI, Schulick RD, Edil BH, Wolfgang CL, Klein AP, Diaz LA, Jr., Allen PJ, Schmidt CM, Kinzler KW, Papadopoulos N, Hruban RH, Vogelstein B. Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Science translational medicine 2011; 3, 92ra66. 84. Wu J, Jiao Y, Dal Molin M, Maitra A, de Wilde RF, Wood LD, Eshleman JR, Goggins MG, Wolfgang CL, Canto MI, Schulick RD, Edil BH, Choti MA, Adsay V, Klimstra DS, Offerhaus GJ, Klein AP, Kopelovich L, Carter H, Karchin R, Allen PJ, Schmidt CM, Naito Y, Diaz LA, Jr., Kinzler KW, Papadopoulos N, Hruban RH, Vogelstein B. Whole-exome sequencing of neoplastic cysts of the pancreas reveals recurrent mutations in components of ubiquitin-dependent pathways. Proc Natl Acad Sci U S A 2011; 108, 21188-21193. 85. Singhi AD, McGrath K, Brand RE, Khalid A, Zeh HJ, Chennat JS, Fasanella KE, Papachristou GI, Slivka A, Bartlett DL, Dasyam AK, Hogg M, Lee KK, Marsh JW, Monaco SE, Ohori NP, Pingpank JF, Tsung A, Zureikat AH, Wald AI, Nikiforova MN. Preoperative next-generation sequencing of pancreatic cyst fluid is highly accurate in cyst classification and detection of advanced neoplasia. Gut 2018; 67, 21312141. 86. Kuboki Y, Fischer CG, Beleva Guthrie V, Huang W, Yu J, Chianchiano P, Hosoda W, Zhang H, Zheng L, Shao X, Thompson ED, Waters K, Poling J, He J, Weiss MJ, Wolfgang CL, Goggins MG, Hruban RH, Roberts NJ, Karchin R, Wood LD. Single-cell sequencing defines genetic heterogeneity in pancreatic cancer precursor lesions. The Journal of pathology 2019; 247, 347-356. 87. Pea A, Yu J, Rezaee N, Luchini C, He J, Dal Molin M, Griffin JF, Fedor H, Fesharakizadeh S, Salvia R, Weiss MJ, Bassi C, Cameron JL, Zheng L, Scarpa A, Hruban RH, Lennon AM, Goggins M, Wolfgang CL, Wood LD. Targeted DNA Sequencing Reveals Patterns of Local Progression in the Pancreatic Remnant Following Resection of Intraductal Papillary Mucinous Neoplasm (IPMN) of the Pancreas. Ann Surg 2017; 266, 133-141. 88. Basturk O, Berger MF, Yamaguchi H, Adsay V, Askan G, Bhanot UK, Zehir A, Carneiro F, Hong SM, Zamboni G, Dikoglu E, Jobanputra V, Wrzeszczynski KO, Balci S, Allen P, Ikari N, Takeuchi S, Akagawa H, Kanno A, Shimosegawa T, Morikawa T, Motoi F, Unno M, Higuchi R, Yamamoto M, Shimizu K, Furukawa T, Klimstra DS. Pancreatic intraductal tubulopapillary neoplasm is genetically distinct from intraductal papillary mucinous neoplasm and ductal adenocarcinoma. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 2017; 30, 1760-1772. 89. Berner O. Subkutane fettgewebe nekrose. Virchows Arch [A] Pathol Anat 1908; 27, 1029-1034. 29

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

90. Webb JN. Acinar cell neoplasms of the exocrine pancreas. J Clin Pathol 1977; 30, 103-112. 91. Burns WA, Matthews MJ, Hamosh M, Weide GV, Blum R, Johnson FB. Lipase-secreting acinar cell carcinoma of the pancreas with polyarthropathy. A light and electron microscopic, histochemical, and biochemical study. Cancer 1974; 33, 1002-1009. 92. Morohoshi T, Kanda M, Horie A, Chott A, Dreyer T, Klöppel G, Heitz PU. Immunocytochemical markers of uncommon pancreatic tumors. Acinar cell carcinoma, pancreatoblastoma, and solid cystic (papillary-cystic) tumor. Cancer 1987; 59, 739-747. 93. Klimstra DS, Heffess CS, Oertel JE, Rosai J. Acinar cell carcinoma of the pancreas. A clinicopathologic study of 28 cases. Am J Surg Pathol 1992; 16, 815-837. 94. Hoorens A, Lemoine NR, McLellan E, Morohoshi T, Kamisawa T, Heitz PU, Stamm B, Ruschoff J, Wiedenmann B, Klöppel G. Pancreatic acinar cell carcinoma. An analysis of cell lineage markers, p53 expression, and Ki-ras mutation. American Journal of Pathology 1993; 143, 685-698. 95. Bergmann F, Aulmann S, Sipos B, Kloor M, von Heydebreck A, Schweipert J, Harjung A, Mayer P, Hartwig W, Moldenhauer G, Capper D, Dyckhoff G, Freier K, Herpel E, Schleider A, Schirmacher P, Mechtersheimer G, Klöppel G, Blaker H. Acinar cell carcinomas of the pancreas: a molecular analysis in a series of 57 cases. Virchows Arch 2014; 465, 661-672. 96. La Rosa S, Bernasconi B, Frattini M, Tibiletti MG, Molinari F, Furlan D, Sahnane N, Vanoli A, Albarello L, Zhang L, Notohara K, Casnedi S, Chenard MP, Adsay V, Asioli S, Capella C, Sessa F. TP53 alterations in pancreatic acinar cell carcinoma: new insights into the molecular pathology of this rare cancer. Virchows Arch 2016; 468, 289-296. 97. La Rosa S, Bernasconi B, Vanoli A, Sciarra A, Notohara K, Albarello L, Casnedi S, Billo P, Zhang L, Tibiletti MG, Sessa F. c-MYC amplification and c-myc protein expression in pancreatic acinar cell carcinomas. New insights into the molecular signature of these rare cancers. Virchows Arch 2018; 473, 435-441. 98. Jakel C, Bergmann F, Toth R, Assenov Y, van der Duin D, Strobel O, Hank T, Klöppel G, Dorrell C, Grompe M, Moss J, Dor Y, Schirmacher P, Plass C, Popanda O, Schmezer P. Genome-wide genetic and epigenetic analyses of pancreatic acinar cell carcinomas reveal aberrations in genome stability. Nature communications 2017; 8, 1323. 99. Jiao Y, Yonescu R, Offerhaus GJ, Klimstra DS, Maitra A, Eshleman JR, Herman JG, Poh W, Pelosof L, Wolfgang CL, Vogelstein B, Kinzler KW, Hruban RH, Papadopoulos N, Wood LD. Whole-exome sequencing of pancreatic neoplasms with acinar differentiation. The Journal of pathology 2014; 232, 428-435. 100. Abraham SC, Wu TT, Hruban RH, Lee JH, Yeo CJ, Conlon K, Brennan M, Cameron JL, Klimstra DS. Genetic and immunohistochemical analysis of pancreatic acinar cell carcinoma: frequent allelic loss on chromosome 11p and alterations in the APC/beta-catenin pathway. The American journal of pathology 2002; 160, 953-962. 101. Chmielecki J, Hutchinson KE, Frampton GM, Chalmers ZR, Johnson A, Shi C, Elvin J, Ali SM, Ross JS, Basturk O, Balasubramanian S, Lipson D, Yelensky R, Pao W, Miller VA, Klimstra DS, Stephens PJ. Comprehensive genomic profiling of pancreatic acinar cell carcinomas identifies recurrent RAF fusions and frequent inactivation of DNA repair genes. Cancer Discov 2014; 4, 1398-1405. 102. Becker WF. Pancreatoduodenectomy for carcinoma of the pancreas in an infant; report of a case. Annals of Surgery 1957; 145 864-870. 103. Horie A, Yano Y, Kotoo Y, Miwa A. Morphogenesis of pancreatoblastoma, infantile carcinoma of the pancreas: report of two cases. Cancer 1977; 39, 247-254. 104. Abraham SC, Wu TT, Klimstra DS, Finn L, Hruban RH. Distinctive molecular genetic alterations in sporadic and familial adenomatous polyposis-associated pancreatoblastomas: frequent alterations in the APC/beta-catenin pathway and chromosome 11p. American Journal of Pathology 2001; 159, 16191627. 30

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

105. Frantz VK. Atlas of tumor pathology: Tumors of the pancreas. Washington, D.C.: Armed Forces Institute of Pathology, 1959. 106. Abraham SC, Klimstra DS, Wilentz RE, Wu TT, Cameron JL, Yeo CJ, Hruban RH. Solidpseudopapillary tumors of the pancreas are genetically distinct from pancreatic ductal adenocarcinomas and almost always harbor beta-catenin mutations. American Journal of Pathology 2002; 160, 1361-1369. 107. Klimstra DS, Wenig BM, Heffess CS. Solid-pseudopapillary tumor of the pancreas: a typically cystic carcinoma of low malignant potential. SeminDiagnPathol 2000; 17, 66-80. 108. Jensen RT, Bodei L, Capdevila J, Couvelard A, Falconi M, Glasberg S, Klöppel G, Lamberts S, Peeters M, Rindi G, Rinke A, Rothmund M, Sundin A, Welin S, Fazio N, The EMABP, Participants EMAB. Unmet Needs in Functional and Nonfunctional Pancreatic Neuroendocrine Neoplasms. Neuroendocrinology 2019; 108, 26-36. 109. Rindi G, Klersy C, Albarello L, Baudin E, Bianchi A, Buchler MW, Caplin M, Couvelard A, Cros J, de Herder WW, Delle Fave G, Doglioni C, Federspiel B, Fischer L, Fusai G, Gavazzi F, Hansen CP, Inzani F, Jann H, Komminoth P, Knigge UP, Landoni L, La Rosa S, Lawlor RT, Luong TV, Marinoni I, Panzuto F, Pape UF, Partelli S, Perren A, Rinzivillo M, Rubini C, Ruszniewski P, Scarpa A, Schmitt A, Schinzari G, Scoazec JY, Sessa F, Solcia E, Spaggiari P, Toumpanakis C, Vanoli A, Wiedenmann B, Zamboni G, Zandee WT, Zerbi A, Falconi M. Competitive Testing of the WHO 2010 versus the WHO 2017 Grading of Pancreatic Neuroendocrine Neoplasms: Data from a Large International Cohort Study. Neuroendocrinology 2018; 107, 375-386. 110. Partelli S, Bartsch DK, Capdevila J, Chen J, Knigge U, Niederle B, Nieveen van Dijkum EJM, Pape UF, Pascher A, Ramage J, Reed N, Ruszniewski P, Scoazec JY, Toumpanakis C, Kianmanesh R, Falconi M, Antibes Consensus Conference p. ENETS Consensus Guidelines for Standard of Care in Neuroendocrine Tumours: Surgery for Small Intestinal and Pancreatic Neuroendocrine Tumours. Neuroendocrinology 2017; 105, 255-265. 111. Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, Schulick RD, Tang LH, Wolfgang CL, Choti MA, Velculescu VE, Diaz LA, Jr., Vogelstein B, Kinzler KW, Hruban RH, Papadopoulos N. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 2011; 331, 1199-1203. 112. Heaphy CM, de Wilde RF, Jiao Y, Klein AP, Edil BH, Shi C, Bettegowda C, Rodriguez FJ, Eberhart CG, Hebbar S, Offerhaus GJ, McLendon R, Rasheed BA, He Y, Yan H, Bigner DD, Oba-Shinjo SM, Marie SK, Riggins GJ, Kinzler KW, Vogelstein B, Hruban RH, Maitra A, Papadopoulos N, Meeker AK. Altered telomeres in tumors with ATRX and DAXX mutations. Science 2011; 333, 425. 113. Yachida S, Vakiani E, White CM, Zhong Y, Saunders T, Morgan R, de Wilde RF, Maitra A, Hicks J, Demarzo AM, Shi C, Sharma R, Laheru D, Edil BH, Wolfgang CL, Schulick RD, Hruban RH, Tang LH, Klimstra DS, Iacobuzio-Donahue CA. Small Cell and Large Cell Neuroendocrine Carcinomas of the Pancreas are Genetically Similar and Distinct From Well-differentiated Pancreatic Neuroendocrine Tumors. Am J Surg Pathol 2012; 36, 173-184. 114. Tang LH, Basturk O, Sue JJ, Klimstra DS. A Practical Approach to the Classification of WHO Grade 3 (G3) Well-differentiated Neuroendocrine Tumor (WD-NET) and Poorly Differentiated Neuroendocrine Carcinoma (PD-NEC) of the Pancreas. Am J Surg Pathol 2016; 40, 1192-1202. 115. Iacovazzo D, Flanagan SE, Walker E, Quezado R, de Sousa Barros FA, Caswell R, Johnson MB, Wakeling M, Brandle M, Guo M, Dang MN, Gabrovska P, Niederle B, Christ E, Jenni S, Sipos B, Nieser M, Frilling A, Dhatariya K, Chanson P, de Herder WW, Konukiewitz B, Klöppel G, Stein R, Korbonits M, Ellard S. MAFA missense mutation causes familial insulinomatosis and diabetes mellitus. Proc Natl Acad Sci U S A 2018; 115, 1027-1032. 116. Zhou C, Dhall D, Nissen NN, Chen CR, Yu R. Homozygous P86S mutation of the human glucagon receptor is associated with hyperglucagonemia, alpha cell hyperplasia, and islet cell tumor. Pancreas 2009; 38, 941-946. 31

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

117. Sipos B, Sperveslage J, Anlauf M, Hoffmeister M, Henopp T, Buch S, Hampe J, Weber A, Hammel P, Couvelard A, Hobling W, Lieb W, Boehm BO, Klöppel G. Glucagon cell hyperplasia and neoplasia with and without glucagon receptor mutations. J Clin Endocrinol Metab 2015; 100, E783-788. 118. Anlauf M, Bauersfeld J, Raffel A, Koch CA, Henopp T, Alkatout I, Schmitt A, Weber A, Kruse ML, Braunstein S, Kaserer K, Brauckhoff M, Dralle H, Moch H, Heitz PU, Komminoth P, Knoefel WT, Perren A, Klöppel G. Insulinomatosis: a multicentric insulinoma disease that frequently causes early recurrent hyperinsulinemic hypoglycemia. Am J Surg Pathol 2009; 33, 339-346. 119. Henopp T, Anlauf M, Schmitt A, Schlenger R, Zalatnai A, Couvelard A, Ruszniewski P, Schaps KP, Jonkers YM, Speel EJ, Pellegata NS, Heitz PU, Komminoth P, Perren A, Klöppel G. Glucagon cell adenomatosis: a newly recognized disease of the endocrine pancreas. J Clin Endocrinol Metab 2009; 94, 213-217. 120. Gepts W, De Mey J. Islet cell survival determined by morphology. An immunocytochemical study of the islets of Langerhans in juvenile diabetes mellitus. Diabetes 1978; 27 Suppl 1, 251-261. 121. Campbell-Thompson ML, Atkinson MA, Butler AE, Chapman NM, Frisk G, Gianani R, Giepmans BN, von Herrath MG, Hyoty H, Kay TW, Korsgren O, Morgan NG, Powers AC, Pugliese A, Richardson SJ, Rowe PA, Tracy S, In't Veld PA. The diagnosis of insulitis in human type 1 diabetes. Diabetologia 2013; 56, 2541-2543. 122. Campbell-Thompson M, Fu A, Kaddis JS, Wasserfall C, Schatz DA, Pugliese A, Atkinson MA. Insulitis and beta-Cell Mass in the Natural History of Type 1 Diabetes. Diabetes 2016; 65, 719-731. 123. Heitz PU, Klöppel G, Hacki WH, Polak JM, Pearse AG. Nesidioblastosis: the pathologic basis of persistent hyperinsulinemic hypoglycemia in infants. Morphologic and quantitative analysis of seven cases based on specific immunostaining and electron microscopy. Diabetes 1977; 26, 632-642. 124. Goossens A, Gepts W, Saudubray JM, Bonnefont JP, Nihoul F, Heitz PU, Klöppel G. Diffuse and focal nesidioblastosis. A clinicopathological study of 24 patients with persistent neonatal hyperinsulinemic hypoglycemia. Am J Surg Pathol 1989; 13, 766-775. 125. Thomas PM, Cote GJ, Wohllk N, Haddad B, Mathew PM, Rabl W, Aguilar-Bryan L, Gagel RF, Bryan J. Mutations in the sulfonylurea receptor gene in familial persistent hyperinsulinemic hypoglycemia of infancy. Science 1995; 268, 426-429. 126. Thomas PM, Wohllk N, Huang E, Kuhnle U, Rabl W, Gagel RF, Cote GJ. Inactivation of the first nucleotide-binding fold of the sulfonylurea receptor, and familial persistent hyperinsulinemic hypoglycemia of infancy. Am J Hum Genet 1996; 59, 510-518. 127. Glaser B, Kesavan P, Heyman M, Davis E, Cuesta A, Buchs A, Stanley CA, Thornton PS, Permutt MA, Matschinsky FM, Herold KC. Familial hyperinsulinism caused by an activating glucokinase mutation. The New England journal of medicine 1998; 338, 226-230. 128. Stanley CA. Perspective on the Genetics and Diagnosis of Congenital Hyperinsulinism Disorders. J Clin Endocrinol Metab 2016; 101, 815-826. 129. de Lonlay P, Fournet JC, Rahier J, Gross-Morand MS, Poggi-Travert F, Foussier V, Bonnefont JP, Brusset MC, Brunelle F, Robert JJ, Nihoul-Fekete C, Saudubray JM, Junien C. Somatic deletion of the imprinted 11p15 region in sporadic persistent hyperinsulinemic hypoglycemia of infancy is specific of focal adenomatous hyperplasia and endorses partial pancreatectomy. The Journal of clinical investigation 1997; 100, 802-807. 130. Sempoux C, Capito C, Bellanne-Chantelot C, Verkarre V, de Lonlay P, Aigrain Y, Fekete C, Guiot Y, Rahier J. Morphological mosaicism of the pancreatic islets: a novel anatomopathological form of persistent hyperinsulinemic hypoglycemia of infancy. J Clin Endocrinol Metab 2011; 96, 3785-3793. 131. Huopio H, Reimann F, Ashfield R, Komulainen J, Lenko HL, Rahier J, Vauhkonen I, Kere J, Laakso M, Ashcroft F, Otonkoski T. Dominantly inherited hyperinsulinism caused by a mutation in the sulfonylurea receptor type 1. The Journal of clinical investigation 2000; 106, 897-906.

32

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

132. Rustgi AK. A historical perspective on clinical advances in pancreatic diseases. Gastroenterology 2013; 144, 1249-1251. 133. Kleeff J, Whitcomb DC, Shimosegawa T, Esposito I, Lerch MM, Gress T, Mayerle J, Drewes AM, Rebours V, Akisik F, Munoz JED, Neoptolemos JP. Chronic pancreatitis. Nat Rev Dis Primers 2017; 3, 17060. 134. Klöppel G. Progression from acute to chronic pancreatitis. A pathologist's view. The Surgical clinics of North America 1999; 79, 801-814. 135. Klöppel G, Adsay NV, Klimstra DS. Pancreatitis: an introduction to history, etiology and pathogenesis. Semin Diagn Pathol 2004; 21, 219-220. 136. Klöppel G. Chronic pancreatitis of alcoholic and nonalcoholic origin. Semin Diagn Pathol 2004; 21, 227-236. 137. Sarles H, Sarles JC, Muratore R, Guien C. Chronic inflammatory sclerosis of the pancreas--an autonomous pancreatic disease? Am J Dig Dis 1961; 6, 688-698. 138. Yoshida K, Toki F, Takeuchi T, Watanabe S, Shiratori K, Hayashi N. Chronic pancreatitis caused by an autoimmune abnormality. Proposal of the concept of autoimmune pancreatitis. Dig Dis Sci 1995; 40, 1561-1568. 139. Ito T, Nakano I, Koyanagi S, Miyahara T, Migita Y, Ogoshi K, Sakai H, Matsunaga S, Yasuda O, Sumii T, Nawata H. Autoimmune pancreatitis as a new clinical entity. Three cases of autoimmune pancreatitis with effective steroid therapy. Dig Dis Sci 1997; 42, 1458-1468. 140. Montefusco PP, Geiss AC, Bronzo RL, Randall S, Kahn E, McKinley MJ. Sclerosing cholangitis, chronic pancreatitis, and Sjogren's syndrome: a syndrome complex. Am J Surg 1984; 147, 822-826. 141. Kawaguchi K, Koike M, Tsuruta K, Okamoto A, Tabata I, Fujita N. Lymphoplasmacytic sclerosing pancreatitis with cholangitis: a variant of primary sclerosing cholangitis extensively involving pancreas. Hum Pathol 1991; 22, 387-395. 142. Scully KA, Li SC, Hebert JC, Trainer TD. The characteristic appearance of non-alcoholic duct destructive chronic pancreatitis: a report of 2 cases. Archives of pathology & laboratory medicine 2000; 124, 1535-1538. 143. Chen RY, Adams DB. IgG4 levels in non-Japanese patients with autoimmune sclerosing pancreatitis. The New England journal of medicine 2002; 346, 1919. 144. Hamano H, Kawa S, Horiuchi A, Unno H, Furuya N, Akamatsu T, Fukushima M, Nikaido T, Nakayama K, Usuda N, Kiyosawa K. High serum IgG4 concentrations in patients with sclerosing pancreatitis. The New England journal of medicine 2001; 344, 732-738. 145. Yoshida K, Toki F, Takeuchi T, Watanabe S, Shiratori K, Hayashi N. Chronic pancreatitis caused by an autoimmune abnormality. Proposal of the concept of autoimmune pancreatitis. Dig DisSci 1995; 40, 1561-1568. 146. Ito T, Nakano I, Koyanagi S, Miyahara T, Migita Y, Ogoshi K, Sakai H, Matsunaga S, Yasuda O, Sumii T, Nawata H. Autoimmune pancreatitis as a new clinical entity. Three cases of autoimmune pancreatitis with effective steroid therapy. DigDisSci 1997; 42, 1458-1468. 147. Hamano H, Kawa S, Horiuchi A, Unno H, Furuya N, Akamatsu T, Fukushima M, Nikaido T, Nakayama K, Usuda N, Kiyosawa K. High serum IgG4 concentrations in patients with sclerosing pancreatitis. New England Journal of Medicine 2001; 344, 732-738. 148. Shimosegawa T, Chari ST, Frulloni L, Kamisawa T, Kawa S, Mino-Kenudson M, Kim MH, Klöppel G, Lerch MM, Lohr M, Notohara K, Okazaki K, Schneider A, Zhang L, International Association of P. International consensus diagnostic criteria for autoimmune pancreatitis: guidelines of the International Association of Pancreatology. Pancreas 2011; 40, 352-358. 149. Detlefsen S, Mohr Drewes A, Vyberg M, Klöppel G. Diagnosis of autoimmune pancreatitis by core needle biopsy: application of six microscopic criteria. Virchows Arch 2009; 454, 531-539.

33

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

150. Kawaguchi K, Koike M, Tsuruta K, Okamoto A, Tabata I, Fujita N. Lymphoplasmacytic sclerosing pancreatitis with cholangitis: a variant of primary sclerosing cholangitis extensively involving pancreas. Human Pathology 1991; 22, 387-395. 151. Ectors N, Maillet B, Aerts R, Geboes K, Donner A, Borchard F, Lankisch P, Stolte M, Lüttges, J, Kremer B, Klöppel G. Non-alcoholic duct destructive chronic pancreatitis. Gut 1997; 41, 263-268. 152. Zamboni G, Lüttges, J, Capelli P, Frulloni L, Cavallini G, Pederzoli P, Leins A, Longnecker D, Klöppel G. Histopathological features of diagnostic and clinical relevance in autoimmune pancreatitis: a study on 53 resection specimens and 9 biopsy specimens. Virchows Arch 2004; 445, 552-563. 153. Notohara K, Burgart LJ, Yadav D, Chari S, Smyrk TC. Idiopathic chronic pancreatitis with periductal lymphoplasmacytic infiltration: clinicopathologic features of 35 cases. Am J Surg Pathol 2003; 27, 1119-1127. 154. Deshpande V, Zen Y, Chan JK, Yi EE, Sato Y, Yoshino T, Klöppel G, Heathcote JG, Khosroshahi A, Ferry JA, Aalberse RC, Bloch DB, Brugge WR, Bateman AC, Carruthers MN, Chari ST, Cheuk W, Cornell LD, Fernandez-Del Castillo C, Forcione DG, Hamilos DL, Kamisawa T, Kasashima S, Kawa S, Kawano M, Lauwers GY, Masaki Y, Nakanuma Y, Notohara K, Okazaki K, Ryu JK, Saeki T, Sahani DV, Smyrk TC, Stone JR, Takahira M, Webster GJ, Yamamoto M, Zamboni G, Umehara H, Stone JH. Consensus statement on the pathology of IgG4-related disease. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 2012; 25, 1181-1192. 155. Sugumar A, Klöppel G, Chari ST. Autoimmune pancreatitis: pathologic subtypes and their implications for its diagnosis. The American journal of gastroenterology 2009; 104, 2308-2310; quiz 2311. 156. Adsay NV, Pierson C, Sarkar F, Abrams J, Weaver D, Conlon KC, Brennan MF, Klimstra DS. Colloid (mucinous noncystic) carcinoma of the pancreas. AmJ SurgPathol 2001; 25, 26-42. 157. Ikeura T, Detlefsen S, Zamboni G, Manfredi R, Negrelli R, Amodio A, Vitali F, Gabbrielli A, Benini L, Klöppel G, Okazaki K, Vantini I, Frulloni L. Retrospective comparison between preoperative diagnosis by International Consensus Diagnostic Criteria and histological diagnosis in patients with focal autoimmune pancreatitis who underwent surgery with suspicion of cancer. Pancreas 2014; 43, 698-703. 158. Shiokawa M, Kodama Y, Sekiguchi K, Kuwada T, Tomono T, Kuriyama K, Yamazaki H, Morita T, Marui S, Sogabe Y, Kakiuchi N, Matsumori T, Mima A, Nishikawa Y, Ueda T, Tsuda M, Yamauchi Y, Sakuma Y, Maruno T, Uza N, Tsuruyama T, Mimori T, Seno H, Chiba T. Laminin 511 is a target antigen in autoimmune pancreatitis. Science translational medicine 2018; 10. 159. Kato Y, Azuma K, Someda H, Shiokawa M, Chiba T. Case of IgG4-associated sclerosing cholangitis with normal serum IgG4 concentration, diagnosed by anti-laminin 511-E8 antibody: a novel autoantibody in patients with autoimmune pancreatitis. Gut 2019. 160. Detlefsen S, Brasen JH, Zamboni G, Capelli P, Klöppel G. Deposition of complement C3c, immunoglobulin (Ig)G4 and IgG at the basement membrane of pancreatic ducts and acini in autoimmune pancreatitis. Histopathology 2010; 57, 825-835. 161. Whitcomb DC, Gorry MC, Preston RA, Furey W, Sossenheimer MJ, Ulrich CD, Martin SP, Gates LK, Jr., Amann ST, Toskes PP, Liddle R, McGrath K, Uomo G, Post JC, Ehrlich GD. Hereditary pancreatitis is caused by a mutation in the cationic trypsinogen gene. Nature genetics 1996; 14, 141-145. 162. Jalaly NY, Moran RA, Fargahi F, Khashab MA, Kamal A, Lennon AM, Walsh C, Makary MA, Whitcomb DC, Yadav D, Cebotaru L, Singh VK. An Evaluation of Factors Associated With Pathogenic PRSS1, SPINK1, CTFR, and/or CTRC Genetic Variants in Patients With Idiopathic Pancreatitis. The American journal of gastroenterology 2017; 112, 1320-1329. 163. Mounzer R, Whitcomb DC. Genetics of acute and chronic pancreatitis. Current opinion in gastroenterology 2013; 29, 544-551.

34

Journal Pre-proof

Jo

ur

na

lP

re

-p

ro

of

164. Lowenfels AB, Maisonneuve P, DiMagno EP, Elitsur Y, Gates LK, Jr., Perrault J, Whitcomb DC. Hereditary pancreatitis and the risk of pancreatic cancer. International Hereditary Pancreatitis Study Group. J Natl Cancer Inst 1997; 89, 442-446. 165. Williams JA. The nobel pancreas: a historical perspective. Gastroenterology 2013; 144, 11661169. 166. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 2014; 74, 2913-2921. 167. https://nyaspubs.onlinelibrary.wiley.com/doi/abs/10.1111/j.2164-0947.1980.tb02775.x. Accessed August 23, 2019.

35

Journal Pre-proof Table : Timeline of milestones in the pathology of epithelial neoplasms with ductal differentiation

1970- Cystic neoplasms lumped together. No driver genes known. 1978- Serous neoplasms separated from mucin-producing neoplasms 1982- Intraductal papillary mucinous neoplasms described 1988- KRAS gene mutations described in pancreatic cancer

of

1991- TP53 gene mutations described in pancreatic cancer 1994- p16/CDKN2A gene mutations described in pancreatic cancer

ro

1996- SMAD4 discovered in pancreatic cancer

-p

2004- Sendai Criteria for managing cystic neoplasms 2008- Whole-exome sequencing of pancreatic cancer

re

2011- Whole-exome sequencing of cystic neoplasms

lP

2014- Fukuoka criteria for managing cystic neoplasms

2015- Whole-genome sequencing of pancreatic cancer by the International Cancer Genome Consortium

na

2016- Whole-genome sequencing of the germline of familial pancreatic cancer

Jo

ur

2017- The Cancer Genome Atlas (TCGA) program reports its characterization of pancreatic cancer

36

Figure 1

Figure 2

Figure 3

Figure 4

Figure 5

Figure 6

Figure 7