Genetics of Familial and Sporadic Pancreatic Cancer

Genetics of Familial and Sporadic Pancreatic Cancer

Gastroenterology 2019;-:1–15 Q1 Q2 Q3 Q33 Genetics of Familial and Sporadic Pancreatic Cancer Laura D. Wood1,2 Matthew B. Yurgelun3,4 PANCREATIC...

1019KB Sizes 1 Downloads 272 Views

Gastroenterology 2019;-:1–15

Q1 Q2 Q3

Q33

Genetics of Familial and Sporadic Pancreatic Cancer

Laura D. Wood1,2

Matthew B. Yurgelun3,4

PANCREATIC CANCER

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Michael G. Goggins1,2,5

1

Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; 2Department of Oncology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; 3Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; 4Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts; and 5Department of Medicine, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland In the previous decade, comprehensive genomic analyses have yielded important insights about the genetic alterations that underlie pancreatic tumorigenesis. Wholeexome and whole-genome sequencing of pancreatic ductal adenocarcinomas have confirmed the critical driver genes altered in the majority of pancreatic cancers, as well as identified numerous less frequently altered driver genes, and have delineated cancer subgroups with unique biological and clinical features. It is now appreciated that pancreatic susceptibility gene alterations are often identified in patients with pancreatic cancer without family histories suggestive of a familial cancer syndrome, prompting recent efforts to expand gene testing to all patients with pancreatic cancer. Studies of pancreatic cancer precursor lesions have begun to elucidate the evolutionary history of pancreatic tumorigenesis and to help us understand the utility of biomarkers for early detection and targets to develop new therapeutic strategies. In this review, we discuss the results of comprehensive genomic characterization of pancreatic ductal adenocarcinoma and its precursor lesions, and we highlight translational applications in early detection and therapy.

Keywords: Pancreatic Cancer; Mutation; Gene Testing; Progression Model.

P

ancreatic cancer is a deadly disease with a dismal prognosis that is predicted to be the second-leading cause of cancer death in the United States by 2030.1 The morbidity and mortality of pancreatic cancer are fueled by 2 clinical gaps—the lack of effective early detection approaches and the lack of efficacious therapies. Recent comprehensive genomic characterization of pancreatic cancers and their precursor lesions have provided critical insights that can help close these gaps. Identification of the somatic and germline genetic alterations that underlie pancreatic tumorigenesis can provide biomarkers for use in

early detection strategies and can form a rational foundation for the design of screening and surveillance programs. Moreover, knowledge of the genomic landscape of pancreatic cancer has informed clinical trials to develop new targeted therapeutic strategies. In this review, we discuss the results of comprehensive genomic characterization of pancreatic neoplasms, along with the translational application to early detection and therapy.

The Molecular Landscape of Sporadic Pancreatic Cancer The most frequently altered driver genes in pancreatic cancer were identified through traditional molecular genetic approaches before the broad application of genomesequencing technology (Table 1).2 Activating mutations in the oncogenic hotspots in exons 2 and 3 of KRAS occurs in the vast majority (>90%) of pancreatic ductal adenocarcinomas (PDACs), leading to activation of the downstream mitogen-activated protein kinase pathway.3–5 Mutations in 3 tumor suppressor genes also occur commonly in PDAC. Function of the p16 protein, a key regulator of the cell cycle encoded by the CDKN2A gene, is frequently lost in PDAC. The percentage of PDACs with mutational inactivation of CDKN2A underestimates its role as a tumor suppressor gene, because many PDACs inactivate CDKN2A by homozygous deletions that often goes undetected, and gene silencing by promoter methylation also occurs.6,7 Overall, approximately 90% of PDACs inactivate CDKN2A. Mutations Abbreviations used in this paper: ctCNA, circulating tumor DNA; ICGC, International Cancer Genome Consortium; IPMN, intraductal papillary mucinous neoplasm; PARP, poly(adenosine diphosphate) ribose polymerase; PanIN, pancreatic intraepithelial neoplasia; PDAC, pancreatic ductal adenocarcinoma; PDO, patient derived organoid; TGF, transforming growth factor. © 2019 by the AGA Institute 0016-5085/$36.00 https://doi.org/10.1053/j.gastro.2018.12.039

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120

2

PANCREATIC CANCER

121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180

Wood et al

Gastroenterology Vol.

in TP53, which encodes a protein critical for the DNA damage response, are also common (occurring in 70%–85% of PDACs), and most frequently involve missense mutation followed by loss of the second allele.8–10 Finally, loss of Smad4 function leading to altered transforming growth factor-b (TGFb) signaling, most commonly by inactivating somatic mutation or homozygous deletion of SMAD4, occurs in approximately 50% of PDACs.11 Overall, the cellular and molecular effects of these critical driver genes are complex and have been reviewed in detail elsewhere.12–15 The altered cellular signaling induced by these somatic mutations leads to phenotypes previously described as “hallmarks of cancer,” such as sustained proliferative signaling, resistance to cell death, and invasion.16 However, the precise mechanisms by which specific somatic mutations contribute to malignant cell phenotypes are still being elucidated. A decade ago, Jones et al17 published the first wholeexome sequencing analysis of pancreatic ductal adenocarcinoma, confirming these 4 mountains that comprise the frequently altered driver genes in the pancreatic cancer genome landscape: the oncogene KRAS and the tumor suppressor genes CDKN2A, TP53, and SMAD4.17 Since then, hundreds of PDACs have been comprehensively analyzed, including whole-exome and whole-genome sequencing approaches. These sequencing efforts, which include collaborative efforts by the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas, as well as individual research groups, have provided a wealth of information about the genetic alterations that underlie pancreatic tumorigenesis, allowing insights with clinical implications. These larger comprehensive PDAC sequencing studies have highlighted several important hills in the PDAC landscape, infrequently altered genes that are likely to be drivers of pancreatic tumorigenesis (Table 1). For example, in the first report of data from the ICGC effort, Biankin et al18 identified recurrent mutations in axon guidance pathway genes, including SLIT2 and ROBO2. Several studies have confirmed the importance of chromatin remodeling genes in pancreatic tumorigenesis, including MLL3, ARID1A, ARID2, and KDM6A18–20; importantly, an association has been reported between mutations in MLL genes and improved prognosis.20 Such comprehensive genetic analyses also highlight the importance of considering genes as components of integrated cellular pathways. For example, although KRAS is by far the most common method for activation of the mitogen-activated protein kinase pathway in PDAC, oncogenic hotspot mutations in BRAF have also been reported in KRAS wild-type tumors,21 and a small subset of PDACs harbor ERBB2 amplification.22 Other KRAS wild-type PDACs have been reported to have oncogenic mutations in CTNNB1, highlighting the importance of WNT signaling in at least a subset of PDACs.23 Similarly, although the TGFb pathway is commonly altered by inactivating SMAD4 mutation, mutations in TGFBR2, ACVRB1, and other related genes also occur.21,24 Other genes that are mutated in a small percentage of PDACs include FBXW7, which results in overexpression of cyclin E, and the spliceosome gene SF3B1,

-,

No.

-

which contributes to aberrant splicing. More recent studies have integrated genomic, transcriptomic, proteomic, and methylomic analyses.23 Widespread epigenetic changes occur during pancreatic tumorigenesis,25,26 and in a minority of pancreatic cancers these events can lead to gene silencing of tumor suppressor genes such as CDKN2A and CDH1.27,28 In addition to identifying mutated genes that drive pancreatic tumorigenesis, patterns of somatic mutations Q11 identified by whole-exome or whole-genome sequencing can also be used to suggest underlying mutagenic processes.29 Application of these patterns (so-called mutational signatures) to pancreatic cancer showed 7 different signatures in sequenced PDAC samples.30 Four major subtypes of PDAC were identified based on mutational signature: an age-related group, a group with defects in DNA doublestrand break repair, a group with defects in DNA mismatch repair, and a group characterized by a signature of unknown etiology (controversially associated with smoking). The group with the signature suggestive of defects in double-strand break repair had many structural rearrangements, and the group with the signature suggestive of mismatch repair defects had germline or somatic mutations in mismatch repair with resultant loss of protein expression, showing the expected result of these mutational processes. Although point mutations are critical drivers of pancreatic tumorigenesis, copy number alteration and chromosomal rearrangements are also likely to play an important role. Focal amplification is a known mechanism for oncogene activation and occurs at the MYC locus on chromosome 8q24 in a subset of PDACs with a worse prognosis, and homozygous deletion is a frequent mechanism for inactivation of well-characterized tumor suppressor genes such as CDKN2A and SMAD4.21 The role of chromosomal rearrangements has also been increasingly appreciated. Waddell et al19 reported whole-genome sequencing of 100 PDACs as part of the ICGC effort, identifying many chromosomal rearrangements but without detecting any recurrent gene fusions. The pattern of chromosomal rearrangements can be used to classify PDACs into 4 subtypes: stable, locally rearranged, scattered, and unstable. The unstable subtype, with >200 structural variation events, was associated with deleterious mutations in BRCA1, BRCA2, and PALB2, as well as with a mutation signature indicative of loss of function in BRCA pathway genes. Importantly, only half of the tumors with an unstable genome had identified mutations in BRCA pathway genes, suggesting alternative mechanisms for genomic instability in these tumors. These findings have important clinical implications, because patients with genomic instability due to defective DNA repair are especially susceptible to specific types of chemotherapy (see the “Translational Oncology” section). The importance of chromosomal rearrangements in pancreatic tumorigenesis was also highlighted by Notta et al31 in their whole-genome analysis of more than 100 PDACs. This study identified polyploidization (with either tetraploidy or hexaploidy) in almost half of PDACs; computational analyses of tumor sequencing data suggested

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240

-

Genetics of Familial and Sporadic Pancreatic Cancer

3

Table 1.Driver Genes With Frequent Somatic Mutations in Pancreatic Neoplasms Gene

Mutation prevalencea

Gene type

Mutated neoplasms

Alteration type

KRAS CDKN2A

Common Common

Oncogene TSG

PanIN, IPMN, PDAC PanIN, IPMN, PDAC

Missense mutation Inactivating mutation/LOH, homozygous deletion Missense mutation/LOH

TP53

Common

TSG

PanIN, IPMN, PDAC

SMAD4

Common

TSG

PDAC

GNAS RNF43 ARID1A

Common Common Uncommon

Oncogene TSG TSG

IPMN, PDAC IPMN, PDAC PDAC

Inactivating mutation/LOH, homozygous deletion Missense mutation Inactivating mutation/LOH Inactivating mutation/LOH

SLIT2 BRCA2 MYC

Uncommon Uncommon Uncommon

TSG TSG Oncogene

PDAC PDAC PDAC

Mutation/LOH Inactivating mutation/LOHb Amplification

ERBB2c

Uncommon

Oncogene

PDAC

Amplification

Affected pathways MAPK signaling Cell cycle control DNA damage response TGFb signaling G-protein signaling Ubiquitin signaling Chromatin remodeling Axon guidance DNA repair Transcriptional regulation EGF signaling

Related altered genes BRAF FBXW7, RB1 — TGFBR2, ACVRB1 — — MLL3, ARID2, KDM6A ROBO2 ATM, PALB2 — —

LOH, loss of heterozygosity; MAPK, mitogen-activated protein kinase; TSG, tumor suppressor gene. a Common: >20% prevalence; uncommon: <10% prevalence. b Germline mutations also frequently occur. c Amplifications or fusions involving RET, BRAF, ALK, ROS1, and NTRK are rare (<1%) in PDAC but are therapeutically targetable.

that point mutations typically precede polyploidization, whereas copy number alterations occur afterward. In addition, they found that >60% of PDACs harbored at least 1 chromothripsis event, some of which altered known driver genes such as SMAD4. These results suggest the rapid (termed catastrophic) accumulation of copy number and chromosomal alterations in PDAC, which lies in contrast to the gradual accumulation of point mutations in key driver genes documented by analysis of pancreatic cancer precursor lesions (see the “Genetic Alterations in Pancreatic Cancer Precursor Lesions” section). Still, several genetic events are required for malignant transformation in the pancreas, including oncogene mutation and loss of both alleles of multiple tumor suppressor genes. Thus, multistep tumorigenesis occurs even if a copy number or chromothripsis event decreases the number of steps by targeting more than 1 driver gene at the same time. For example, a theoretical pancreatic cancer with an activating KRAS mutation and inactivation of CDKN2A and TP53 would require 5 gene alterations (KRAS mutation and inactivation of both copies of the 2 tumor suppressor genes). A chromothripsis event that caused simultaneous loss of 2 tumor suppressor loci would reduce the number of genetic events from 5 to 4, but it would not eliminate the need for sequential accumulation of genetic events. Comprehensive genetic analysis of primary and metastatic tumors from the same patients provides a unique opportunity to study the genetic basis and timing of metastatic progression. Analysis of such tumors harvested at autopsy from PDAC patients has shown that the clonal populations that give rise to the distant metastasis are present in the primary tumor, suggesting that there is not a recurrent genetic alteration that drives metastasis.32

Mathematical modeling from these data suggest a latent time of more than 15 years between the initiating mutation and the acquisition of metastatic ability, pointing to a broad time window for detection of a pancreatic neoplasm before metastasis.32 Moreover, mutations in well-characterized driver genes are present homogeneously throughout the primary tumor and metastasis, whereas the majority of alterations that are discordant between different metastases are likely passengers.33 Similarly, polyploidy, copy number alterations, and chromothripsis have been reported to be shared among the primary tumor and all metastases, suggesting that these types of alterations also precede the development of metastasis.31 In addition to the use of comprehensive DNA sequencing to identify somatic mutations, the gene expression of many PDACs has also been analyzed, either with microarrays or, more recently, RNA sequencing. Several studies have reported subtyping of pancreatic cancers based on transcriptomic profiling.34–36 Although the specific groups differ in each study, a shared theme is the distinction between a group with a classical/progenitor signature and a group with mesenchymal/basal features. A recent comprehensive study by The Cancer Genome Atlas confirmed the equivalence of these 2 expression groups across studies, and other reported subtypes are likely to reflect contamination of nonneoplastic exocrine or immune cells.23 These gene expression subtypes have clinical implications, with the mesenchymal/basal subtype showing worse prognosis in multiple studies.34–36 Deconvolution of gene expression data has also identified stroma-specific signatures, with activated stroma conferring a decreased survival.34 However, these epithelial and stromal transcriptomic subtypes have yet to be broadly applied for prognostication in clinical practice.

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

PANCREATIC CANCER

241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300

2019

301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360

4

Gastroenterology Vol.

Genetic Underpinnings of Familial Pancreatic Cancer

PANCREATIC CANCER

361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420

Wood et al

The major known familial pancreatic cancer susceptibility genes include the BRCA2, ATM, PALB2, CDKN2A, PRSS1, MLH1 and MSH2, STK11, and TP53 genes (Table 2).37–43 All of these genes (with the exception of PRSS1 mutations, which cause pancreatic cancer by predisposing patients to hereditary pancreatitis), are cancer susceptibility genes that predispose to other cancers. Two additional genes that have been recently implicated as pancreatic cancer susceptibility genes encode for pancreatic enzymes, CPA1 and CPB1, with rare deleterious variants identified in approximately 1% of patients with pancreatic cancer.44 Germline mutations in CPA1 are known to cause recurrent acute pancreatitis, but mutation carriers may develop pancreatic cancer without prior attacks of pancreatitis.44 Recent studies have investigated pancreatic cancer susceptibility by using gene panels of other cancer predisposition genes; these studies have also identified rare germline mutations in other genes in patients with pancreatic cancer, including RAD51C and others43,45–47; deleterious variants in these genes are so uncommon in the population that it is difficult even with large studies to establish whether these genes contribute to pancreatic cancer susceptibility.46 Mutations in other genes such as CHEK2 that are not rare in the population are associated with a moderately increased risk of breast cancer but have not been conclusively associated with pancreatic cancer, although they have been identified in some pancreatic cancer probands, possibly as incidental findings.46 The most commonly mutated germline susceptibility genes in patients with pancreatic cancer in most populations involve BRCA2 or ATM45,46; deleterious germline mutations involving other genes are much less common The mutation prevalence is higher in populations with common founder mutations, such as those with many individuals of Ashkenazi Jewish heritage because of the common BRCA2 founder mutation in that population48–53 or the Dutch founder mutation involving CDK2NA.54 The average lifetime risk of developing pancreatic cancer is highest among patients who carry germline mutations in PRSS1 (w30%),40 STK11 (w30%),55 and CDKN2A (w17%).56 Germline mutations in BRCA2 are thought to increase the risk of pancreatic cancer by approximately 6-fold, to a cumulative lifetime risk of approximately 7%.57–59 The penetrance of PALB2 mutations to cause pancreatic cancer is not yet known, but it is thought to be similar to those with BRCA2 mutations60; this is the case for breast cancer among PALB2 mutation carriers.61 Germline BRCA1 mutations increase the average overall risk of developing pancreatic cancer by approximately 2fold.41,43,62,63 The penetrance of ATM mutations to cause pancreatic and other cancers is not yet known. The cancer risk estimates for these susceptibility gene mutations are approximate and vary depending on the germline variant and other inherited and environmental factors. The role of other genes in pancreatic cancer susceptibility is still being evaluated. Inherited mutations in known pancreatic cancer susceptibility genes explain only a small

-,

No.

-

portion (<20%) of the familial clustering of pancreatic cancer, suggesting that other pancreatic cancer susceptibility genes remain to be identified. Some of the familial clustering of pancreatic cancer may be due to the combined effects of numerous low-penetrance variants (such as ABO, TERT, PDX1, and others) that have been identified in genome-wide association studies.64–70 One of the implications of the incomplete penetrance of cancer susceptibility gene variants is that evaluation of a careful cancer family history, although a very important cancer risk assessment tool,71 is not a comprehensive tool for identifying families with cancer susceptibility gene mutations. Therefore, it is not surprising that many individuals with germline mutations in cancer susceptibility genes have apparently sporadic forms of cancer. Recent studies have found that approximately 5% (and higher in certain populations47) of patients with apparently sporadic forms of pancreatic cancer (ie, without a family history of pancreatic cancer) have germline mutations in a pancreatic cancer susceptibility gene45,46; the prevalence among those with a family history of pancreatic cancer is somewhat higher,41,53,72–74 but overall, most germline mutations are found in individuals who do not have a family history of pancreatic cancer or a family history suggestive of an inherited cancer syndrome.45 Because of the potential value of identifying a germline susceptibility gene for patients and their families (more options for personalized medicine75 and the cancer screening and cancer prevention strategies family members can undertake54,76–79), routine gene testing for inherited susceptibility is now being advocated for patients with newly diagnosed pancreatic cancer. Recent National Comprehensive Q12 Cancer Network guidelines now include the recommendation to consider offering germline gene testing after appropriate counseling to all patients with a personal history of pancreatic cancer, regardless of family history or age at diagnosis. The clinical value of such routine germline gene testing of newly diagnosed pancreatic cancer is still being determined. Gene testing can be readily performed with panel testing that includes dozens of cancer susceptibility genes. The advantage of testing large panels of genes for variants is counterbalanced by the drawback that not infrequently, panel gene testing identifies 1 or more so-called deleterious variants that are only weakly associated with cancer risk, equivalent to the approximately 1.4-fold elevated risk of having pancreatic cancer for carriers of A or B blood groups, relative to blood group O.65,80 Interpreting such results can be challenging for practitioners not well grounded in genetic medicine and can create anxiety for patients. For gene mutation carriers, knowledge of their cancer susceptibility does hold the potential benefit of managing their risk with cancer screening. The utility of pancreatic screening and surveillance is still being evaluated, although recent evidence points to improvements in pancreatic cancer outcome for individuals who meet criteria for surveillance and who undertake regular surveillance. Initial studies from the Johns Hopkins Cancer of the Pancreas (ie, CAPS) program and from a European cohort that included individuals with the Dutch founder CDKN2A mutation found that regular surveillance with pancreatic imaging (mainly

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480

481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 -

2019

Table 2.Pancreatic Cancer Susceptibility Genes

Elevated relative risk of developing PDACa

BRCA2

w2

w6

Homologous repair

Breast, ovarian, prostate

ATM

w2

w6

DNA repair

Breast, prostate

BRCA1

0.5

w2.5-

Homologous repair

Breast, ovarian, prostate, colorectal

CDKN2A PALB2

0.4 0.4

w12–20 w6

Cell cycle Homologous repair

Melanoma Breast

MLH1

0.2

w7

Mismatch repair

MSH2

<0.1

w7

Mismatch repair

MSH6

0.4

w?

Mismatch repair

TP53 STK11

0.1 <0.1

w7 w30

DNA repair AMPK signaling

PRSS1

<0.1

w10

CPA1

0.5

?

CPB1

0.5

?

Trypsin activation causing pancreatitis Acinar-cell ER stress causing pancreatitis Acinar-cell ER stress causing pancreatitis

Colorectal, gynecologic, urothelial, brain, stomach, intestinal, others Colorectal, gynecologic, urothelial, brain, stomach, intestinal, others Colorectal, gynecologic, urothelial, brain, stomach, intestinal, others Numerous Breast, GI tract, gynecologic, lung, others None

Gene

Predominant mechanism

Other cancers

Implications for PDAC patients

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

Platinum/PARP inhibitor–based therapyb Possible radiosensitivityc Platinum/PARP inhibitor based therapyb ? Platinum/PARP inhibitor based therapyb Checkpoint inhibitor immunotherapy

Comments

Q27

Common founder mutation in w1% of Jewish ethnicity Biallelic germline ATM mutations cause ataxiatelangiectasia syndrome Common founder mutation in w1% of Jewish ethnicity FAMMM syndrome —

Lynch syndrome

Checkpoint inhibitor immunotherapy

Lynch syndrome

Checkpoint inhibitor immunotherapy

Lynch syndrome

? —

Li-Fraumeni syndrome Peutz–Jeghers syndrome

?

Hereditary pancreatitis

None

?

None

?

Hereditary pancreatitis Subclinical pancreatitis Subclinical pancreatitis

AMPK, adenosine monophosphate–activated protein kinase; ER, endoplasmic reticulum; FAMMM, familial atypical multiple mole melanoma; GI, gastrointestinal. a From Hu et al.37 Chemosensitivity to PARP inhibitors/platinums if wild-type allele is inactivated in the cancer. b The prevalence of deleterious variants in other known cancer susceptibility genes is too low, making it difficult to determine their role in pancreatic cancer susceptibility. c Radiosensitivity shown in PDAC cells in vitro.

Q28

Q29 Q30

Genetics of Familial and Sporadic Pancreatic Cancer

Prevalence of deleterious germline mutations in patients with PDAC, %

5

PANCREATIC CANCER

541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600

6 Q13

PANCREATIC CANCER

601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660

Wood et al

Gastroenterology Vol.

magnetic resonance imaging/magnetic resonance cholangiopancreatography and endoscopic ultrasonography) leads to down-staging of pancreatic cancer and better 5year survival after a pancreatic cancer diagnosis.54,81 Pancreatic surveillance is offered to individuals of the appropriate age who are either 1) carriers of a germline mutation in a pancreatic cancer susceptibility gene and 2) individuals with multiple blood relatives with pancreatic cancer (generally at least 1 first-degree and 1 second-degree relative with pancreatic cancer).82 Pancreatic imaging with endoscopic ultrasonography and magnetic resonance imaging/magnetic resonance cholangiopancreatography has been used as the method of surveillance because such imaging can identify worrisome precancerous changes as well as small cancers, but surveillance could be enhanced with accurate blood tests, and many patients are found to have small pancreatic cysts of low malignant potential78 that can contribute additional worry for patients already concerned about their pancreatic cancer family history.83 The role of pancreatic surveillance is still undergoing evaluation and is best undertaken at expert centers as part of long-term research studies.

Genetic Alterations in Pancreatic Cancer Precursor Lesions PDAC arises from noninvasive precursor lesions that are curable if detected early—knowledge of the molecular alterations in these precursor lesions can provide a rational foundation for early-detection approaches. The most common PDAC precursor lesion is pancreatic intraepithelial neoplasia (PanIN), a microscopic lesion graded based on the degree of architectural and cytologic atypia. Low-grade PanINs (PanIN-1, PanIN-2) are common and have a low risk of malignant progression, whereas high-grade PanINs (PanIN-3) are much less frequent (<1% in autopsy series) and are considered to be carcinoma in situ.84 Several studies have documented the sequential accumulation of somatic mutations in PDAC driver genes in PanINs. KRAS mutation is the earliest known genetic alteration, present in >90% of all PanINs regardless of grade.85–87 Mutant KRAS DNA concentrations have been reported to increase with increasing grade of dysplasia in PanINs, raising the intriguing possibility that low-grade PanINs contain a mixture of KRAS mutant and wild-type cells.85 In contrast, inactivation of CDKN2A is uncommon in low-grade PanINs but has been reported to occur in >70% of high-grade PanINs.88 Mutations in TP53 and SMAD4 occur even later in PanIN progression, found almost exclusively in high-grade PanIN and invasive PDAC. Early studies showed aberrant p53 expression and loss of Smad4 expression at the protein level (indicative of somatic mutation/inactivation) in >30% of high-grade PanINs, whereas almost all low-grade PanINs retained normal expression.89,90 However, these studies analyzed PanIN in the setting of co-occurring invasive PDAC, which can confound such studies through intraductal spread of invasive carcinoma, a lesion that is histologically indistinguishable from high-grade PanIN.91 When analyzing only

-,

No.

-

high-grade PanINs in the absence of invasive PDAC, nextgeneration sequencing and immunohistochemistry showed that TP53 alterations were far less common (occurring in only 17% of high-grade PanINs) and that SMAD4 alterations were completely absent.92 In addition to somatic point mutations, other types of genetic alterations have been described in PanINs. These include telomere shortening, thought to be an early sign Q14 even in pancreatic tumorigenesis and frequently present in low-grade PanIN.93 Copy number alterations have been reported in PanIN; such alterations are uncommon in lowgrade PanIN but occur more frequently in high-grade PanIN, sometimes affecting known driver genes.94,95 Biallelic inactivation of SMAD4 was not identified in PanINs in these studies, supporting the concept that it is a driver of invasive PDAC. In addition, chromothripsis-like regions were identified in both low-grade and high-grade PanINs, showing that this phenomenon can precede the development of invasive PDAC.95 In addition to microscopic PanIN, pancreatic cancer can also arise from macroscopic cystic precursor lesions, most commonly intraductal papillary mucinous neoplasm (IPMN). IPMNs involve the pancreatic duct system and are by definition >1 cm in size, although many grow to be much larger. Like PanINs, IPMNs are graded (as low grade or high grade) based on the architectural and cytologic atypia of their lining epithelium, but they are also subtyped based on the direction of differentiation of this lining, including gastric, intestinal, and pancreatobiliary.84,96 IPMNs are driven by the 4 mountains of PanIN-based pancreatic tumorigenesis, including early alterations in KRAS and later mutations in CDKN2A, TP53, and SMAD4, with SMAD4 mutation typically limited to IPMN-associated invasive carcinoma.97–99 However, there are also 2 frequently altered driver genes that are specific to the IPMN pathway. Mutations in the oncogenic hotspot of GNAS occur early in IPMN tumorigenesis and are most common in IPMNs with intestinal differentiation.97,100,101 Inactivating mutations (often followed by loss of heterozygosity) in RNF43, which encodes a ubiquitin ligase involved in WNT signaling, are also common in IPMNs.102 Mutations in RNF43 have also been reported less commonly in PDACs not associated with IPMNs, as well as in other cystic precursor lesions (as will be discussed).19,102 The precise timing of RNF43 mutations in IPMN tumorigenesis has not yet been systematically investigated. Although IPMNs are the most common cystic precursor to PDAC, several other types of such precursor have also been studied with molecular techniques. Intraductal oncocytic papillary neoplasm, originally classified as a subtype of IPMN, has recently been shown to be driven by a distinct spectrum of genetic alterations, lacking mutations in KRAS and GNAS.103 Similarly, intraductal tubulopapillary neoplasms also lack KRAS mutations but do have uncommon mutations in the oncogene PIK3CA, which have also been reported in IPMNs.104,105 Mucinous cystic neoplasms (MCNs), which do not involve the pancreatic duct system and have a distinct pathognomonic ovarian-type stroma, have some genetic overlap with IPMNs; KRAS mutations are a common initiator,

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720

721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780

2019

Genetics of Familial and Sporadic Pancreatic Cancer

but GNAS mutations have not been reported in mucinous cystic neoplasms.102,106 Mutations in other tumor suppressor genes, including RNF43, occur later.102,106–108 Recent genetic analyses of pancreatic cancer precursor lesions have shown genetic heterogeneity with respect to driver gene mutations, suggesting a more complex evolutionary trajectory than was previously appreciated. One study in PanINs that used ultrasensitive mutation techniques showed multiple KRAS mutations in almost 25% of lesions.85 Similarly, analysis of cyst fluid showed that 13% of IPMN samples contained multiple KRAS mutations and that 4% contained multiple GNAS mutations.100 Dissection and subsequent next-generation sequencing of 2 different IPMN regions showed that 23% contained multiple KRAS or GNAS mutations, and up to 4 different KRAS mutations were detected within a single IPMN.109 Recent single-cell DNA sequencing analyses of IPMNs have shown that these heterogeneous KRAS mutations occur in different cells, raising the possibility of polyclonal origin of at least a subset of IPMNs.110 In addition to this heterogeneity within a single precursor lesion, multiple studies have provided evidence for multifocal neoplasia in the pancreas. Targeted nextgeneration sequencing showed that the vast majority of co-occurring low-grade and high-grade PanINs did not share any genetic alterations and, thus, likely arose independently.92 Similarly, a sizable proportion (18%) of cooccurring IPMNs and PDACs were genetically independent despite their close proximity.109 In addition, more than half of the pancreatic neoplasms that occurred in the remnant pancreas after IPMN resection were likely independent due to lack of shared genetic alterations.111 Taken together, these studies highlight the frequency at which multiple seemingly independent neoplasms occur in the same patient’s pancreas. It is currently not clear whether this risk of multifocal neoplasia represents the result of normal aging or varies based on as yet undefined molecular factors. Although some interpret these data as suggesting a field defect that puts the entire pancreas at risk for neoplasia, the mechanistic basis of such a field defect is unknown. If such a field defect does exist, its mechanism does not appear to be a somatic mutation in a well-characterized driver gene. In contrast to the occurrence of multiple independent neoplasms in close proximity, recent work also highlights the ability of a single neoplasm to move throughout the pancreatic ductal system. Whole-exome sequencing of cooccurring yet anatomically distant PanINs and invasive carcinomas suggests that neoplastic clones can spread throughout the pancreatic ductal system.112

Translational Application of Molecular Insights DNA harboring acquired mutations in KRAS and other genes is shed from pancreatic cancers into the blood, pancreatic fluids, and stool and has potential value in earlydetection tests. Circulating tumor DNA (ctDNA) has been extensively studied as a tool for the detection and surveillance of multiple cancer types.113,114 However, even with assays designed to detect mutations in low abundance, the

7

amount of ctDNA detected in most patients with pancreatic cancer is very low (often in concentrations in the 0.1% range), and the percentage of patients with stage I/II disease who have any ctDNA detectable with existing technologies is low (w30%–40%).20,114,115 Patients with pancreatic cancer and a positive ctDNA test result have a poorer prognosis vs those without.115 Somatic mutations are also shed into the pancreatic ductal system, and the detection of these mutations in pancreatic cyst fluid and pancreatic juice has been studied to determine how the detection of these mutations can improve the evaluation of patients undergoing pancreatic surveillance. For example, the mutation profile of pancreatic cyst fluid can be used to reliably classify cysts into mucinous and nonmucinous cysts,116,117 and the neoplastic grade of pancreatic cysts can be determined with very good accuracy by using a test panel that combines the detection of mutations with chromosomal copy number alterations.117 Similarly, pancreatic juice collected from the duodenum after secretin infusion contains mutations that reflect neoplastic changes within the pancreas.118,119 Pancreatic juice is thought to contain DNA shed from the pancreatic ductal system and may be helpful in identifying occult neoplasia not visible by pancreatic imaging tests.120,121 The mutational profiles in pancreatic cyst fluid and pancreatic juice samples mirror the timing of mutations and other genetic alterations that arise during pancreatic neoplastic development, with KRAS and GNAS mutations detected with the presence of low-grade dysplasia in PanINs and IPMNs, as well as later stages of neoplastic progression and invasive cancer, whereas mutations that arise during later stages of neoplastic development (TP53 and SMAD4) are usually detected only in the presence of high-grade dysplasia or invasive cancer.118,119 Protein markers,122 aberrantly methylated DNA,123 telomerase activity,124 and other markers are also being evaluated to understand the most cost effective and accurate diagnostic marker panel for pancreatic cyst fluids.

Translational Oncologic Therapeutics in Pancreatic Cancer At first glance, the field of pancreatic adenocarcinoma therapeutics might seem like a failure with regard to targeted and/or immune-based therapeutics. More than 90% of pancreatic adenocarcinomas harbor activating mutations in KRAS,23 for which targeted therapies have been notoriously ineffective across all cancer types. Furthermore, aside from erlotinib125 (the use of which has largely become obsolete with the improvements from multiagent cytotoxic chemotherapy with folinic acid, fluorouracil, irinotecan, Q15 and oxaliplatin [FOLFIRINOX]126 and gemcitabine/nabpaclitaxel127), no molecularly targeted agents have been approved by the US Food and Drug Administration (FDA) for treating pancreatic adenocarcinoma, despite numerous randomized trials128 examining therapies directed at PI3K,129 IGF1R,130 EGFR,131 KIT,132 the VEGF pathway,133–135 and other theoretically druggable targets.136 Additionally, immune checkpoint inhibitors have failed to show any

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

PANCREATIC CANCER

-

781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840

PANCREATIC CANCER

841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900

Wood et al

Gastroenterology Vol.

-,

No.

-

print & web 4C=FPO

8

Figure 1. The PanIN progression model. More than 90% of low-grade PanIN-1 lesions have KRAS codon 12 mutations and have telomere shortening. The percentage of PanIN DNA with KRAS mutations increases with PanIN grade. Progression of low-grade PanIN-1 to low-grade PanIN-2 and high-grade PanINs (PanIN-3) is usually associated with inactivation of both copies of CDKN2A. TP53 mutation is rarely detected in low-grade PanIN but is often found in high-grade PanIN and invasive cancer. Inactivation of both copies of SMAD4 is found in invasive cancer. Loss of chromosomal arms contributes to biallelic inactivation and are increasingly found as PanINs progress to low-grade PanIN-2 to high-grade PanIN and invasive cancer. In addition to the loss of a chromosomal arm, shattering and rearrangement of one or more chromosomal regions (chromothripsis) may occur. Bi-allelic inactivation of BRCA2 inactivation of BRCA2 is thought to occur in the late stages of PanIN progression. The timing of mutation of other low-frequency driver genes during PanIN progression (eg, ATM and ARID1A) has not been extensively studied.

meaningful therapeutic benefit in treating pancreatic adenocarcinoma137–139 (with the notable exception140 of the 1% of tumors141–143 with high-level microsatellite instability/mismatch repair deficiency). Despite the current dearth of FDA-approved noncytotoxic treatment options for pancreatic cancer, recent years have brought about an abundance of novel genomic, molecular, and immunologic insights into the diverse and complex biology of this notoriously “undruggable” malignancy, providing hope that rational translational oncologic therapeutics can soon become critical, effective, feasible, and routine components of pancreatic cancer care. KRAS wild-type pancreatic cancers appear to be a particularly appealing subgroup for targeted therapies, with various investigators having found such cases to be enriched for targetable alterations, amplifications, or fusions in oncogenes including BRAF, ROS1, NRG1, RET, ALK, and ERBB2, with real-world cases of such patients showing substantial responses to matched targeted therapies.143–145 The unique Know Your Tumor initiative143 led by the Pancreatic Cancer Action Network (ie, PanCAN) advocacy group recently showed the potential feasibility and benefits of using multi-omic tumor profiling to guide targeted therapy use among 640 pancreatic cancer patients from 44 different states. This effort identified potentially actionable alterations in 165 (26%) of all individuals (nearly all of which were associated with therapies already FDA approved for other malignancies), and an overall 50% rate of potentially actionable findings. Although the most common such alterations were in DNA damage repair/homologous recombination (HR) genes (as will be discussed), other

tumors were noted to have potentially targetable variants, amplifications, or fusions in ERBB2, RET, BRAF, ALK, ROS1, and NTRK.143 Furthermore, individuals with pancreatic cancers found to have such alterations in this study had superior progression-free survival when their treatment was guided by these genomic data vs those receiving nontargeted therapies, although with a small magnitude of difference (4.1 months vs 1.9 months).143 A recent singleinstitution feasibility study145 incorporating real-time whole-exome and RNA sequencing into the clinical treatment workflows of individuals with pancreatic cancer similarly showed 48% of all individuals to have potentially actionable findings (again, with the most common such findings being sequence alterations and/or mutational signatures of HR deficiency), with 30% of individuals undergoing a therapeutic change in management based on their tumor profiling. Other investigators have similarly reported the feasibility of real-time whole-genome and RNA sequencing to guide the management of pancreatic cancer patients with median turnaround times of slightly more than a month.146 Despite such promising preliminary data, however, the oncologic literature is littered with numerous examples of theoretically “druggable” somatic alterations that have proven to be markedly more difficult to target in real-world treatment than initially assumed, especially when extrapolating across different tumor types (eg, the anti-ERBB2 agents lapatinib and pertuzumab, each of which has failed to show benefit in randomized phase III studies of ERBB2amplified gastroesophageal cancers, in spite of their significant activity in ERRB2-amplified breast cancers).147,148 The

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

Q26

901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960

961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020

2019

Genetics of Familial and Sporadic Pancreatic Cancer

aforementioned studies145,146,149 illustrate the relative rarity of each individual potentially targetable alteration, amplification, and rearrangement in pancreatic cancer, strongly suggesting that pancreatic cancer-specific clinical trials to evaluate the efficacy of given drugs for each individual molecular target are highly likely to be unfeasible. Although they may not be able to account for the possibility of heterogeneous responses across different tumor types, basket trials have proven to be a promising approach for evaluating the efficacy of targeted therapies for rare and uncommon molecular targets, and they are likely to be a critical approach to advancing the therapeutic landscape for the fraction of pancreatic cancers with such molecular targets. Indeed, recent basket trials examining larotrectinib in tumors with NTRK fusions150 and pembrolizumab in tumors with mismatch repair deficiency140 both identified significant responses in the few enrolled pancreatic cancer patients, illustrating the potential for such study designs to help make inroads in learning how to therapeutically leverage such molecular targets. Further expanding on the notion of using tumor biopsy specimens to guide personalized therapy, emerging data on pancreatic adenocarcinoma patient-derived organoids (PDOs) hold additional promise in helping to expand and refine treatment options for individuals with pancreatic cancer. One recent study suggested that in vitro PDO sensitivity (or lack thereof) to cytotoxic agents appeared highly concordant with the corresponding patients’ realworld responses (or lack thereof) to systemic chemotherapy.151 Perhaps more important, however, this work also demonstrated the ability to use such PDOs for multiomic tumor profiling and also to assay for sensitivity to various molecularly targeted agents, particularly among specimens that appear to be inherently resistant to cytotoxic chemotherapy.151 The most common subtype of potentially therapeutically targetable alterations identified in pancreatic adenocarcinomas are those with somatic and/or germline defects in DNA damage repair pathways, particularly those involved in homologous recombination (HR).143,145 Recent data suggest that anywhere from 3.2-8.1% of all pancreatic adenocarcinoma cases arise in the setting of germline variants in HR genes, most commonly BRCA2, ATM, BRCA1, PALB2, and CHEK2.45,46,152,153 In parallel, an additional 2.5%–9.9% of pancreatic adenocarcinomas harbor somatic alterations in these same genes, and 7.0%–7.6% have mutational signatures of HR deficiency without identifiable germline or somatic HR gene alterations.30,154 A number of small studies have suggested that such germline HR gene variants and/or HR deficiency mutational signatures likely predict response to cisplatin- or oxaliplatin-based chemotherapy in pancreatic adenocarcinoma.152,155–158 Such findings have naturally led to the hypothesis that poly(adenosine diphosphate) ribose polymerase (PARP) inhibitors, known to have significant benefit in breast and ovarian cancers with HR deficiency, may have therapeutic benefit in this important subset of pancreatic adenocarcinomas. A phase I study159 of olaparib combined with cisplatin, irinotecan, and mitomycin-C in 18 patients with

9

advanced pancreatic cancer showed a 23% objective response rate, though only 2 participants were known to carry germline BRCA2 variants (the others had unknown BRCA1/2 status), 1 of whom experienced more than 4 years of disease control. Unfortunately, 2 participants (including the BRCA2 carrier with prolonged disease control) developed therapy-related myelodysplastic syndrome, and thus the regimen was believed to be unacceptably toxic for further study.159 Another multiagent phase I study160 examined veliparib in combination with cisplatin and gemcitabine as first-line therapy in 17 patients with advanced pancreatic cancer (7 of whom were known to carry germline BRCA1 or BRCA2 variants). Overall, this combination showed a 41.2% overall objective response rate, although all of the responses occurred in the BRCA1/2 carriers (who overall had a 77.8% objective response rate).160 The BRCA1/2 carriers in this study also had significantly superior median overall survival compared with noncarriers (23.3 months vs 11 months).160 As single agents, on the other hand, PARP inhibitors have shown modest activity (0%–22% objective response rates) in small studies of pancreatic adenocarcinoma patients with germline BRCA1 or BRCA2 gene variants,161–164 although these rates appear to be lower than what has been observed in HR-deficient breast and ovarian cancers.165–167 One potential explanation for this lower response rate may stem from the observation152 that roughly half of pancreatic adenocarcinomas arising in the setting of germline HR gene variants lack an identifiable somatic “second hit” within the tumor, thereby suggesting that a substantial fraction of such tumors may not be truly HR deficient.151 Such data call into question the paradigm used in most PARP inhibitor trials for pancreatic cancer to date, in which the presence of a pathogenic germline HR gene variant alone (rather than paired germline and somatic HR gene defects) is used to determine eligibility. Ongoing randomized clinical trials are evaluating PARP inhibitors in combination with platinum-based chemotherapy (NCT10585805) and as maintenance therapy after an initial response to multiagent platinum-based chemotherapy (NCT02184195) in individuals with pathogenic germline HR gene variants, although studies examining PARP inhibitors in pancreatic adenocarcinoma patients with somatic HR gene alterations and/or with mutational signatures of HR deficiency would be of significant interest, so as to attempt to expand the therapeutic reach of these agents. The abundance of pancreatic cancers with defects in HR machinery is particularly intriguing with regard to expanding the use of immunotherapy in pancreatic cancer. Various in vitro and in vivo data have strongly suggested the potential for synergy between PARP inhibitors and immune checkpoint blockade in breast and ovarian cancers,168–170 possibly via PARP inhibitor-induced DNA damage leading to increased neoantigen production and immunogenicity. Furthermore, mutational signature data from pancreatic cancer specimens suggest a strong correlation between tumors with DNA damage repair gene alterations/signatures and markers of immunogenicity, including neoantigen load, PD-L1 expression, CTLA-4 expression, and CD8þ T-cell activation.30

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

PANCREATIC CANCER

-

1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080

10

PANCREATIC CANCER

1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140

Wood et al

Gastroenterology Vol.

Conclusion Pancreatic cancer represents one of the best characterized cancer types at the genetic level, with comprehensive genetic analysis (whole-exome or whole-genome sequencing) to identify somatic alterations reported on more than 500 individual cancers. In addition, extensive investigation into germline alterations that underlie inherited pancreatic cancer predisposition has been performed, and premalignant lesions have been characterized at the molecular level. These comprehensive molecular analyses have led to several clinically important insights, informing both early detection approaches and targeted therapeutics. Future work will further refine these clinical translations so that the molecular basis of pancreatic cancer can be better leveraged to improve the lives of patients with this disease.

References 1. Rahib L, Smith BD, Aizenberg R, et al. 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. 2. Rozenblum E, Schutte M, Goggins M, et al. Tumorsuppressive pathways in pancreatic carcinoma. Cancer Res 1997;57:1731–1734. 3. Almoguera C, Shibata D, Forrester K, et al. Most human carcinomas of the exocrine pancreas contain mutant cK-ras genes. Cell 1988;53:549–554. 4. Hruban RH, van Mansfeld AD, Offerhaus GJ, et al. K-ras oncogene activation in adenocarcinoma of the human pancreas. A study of 82 carcinomas using a combination of mutant-enriched polymerase chain reaction analysis and allele-specific oligonucleotide hybridization. Am J Pathol 1993;143:545–554. 5. Smit VT, Boot AJ, Smits AM, et al. KRAS codon 12 mutations occur very frequently in pancreatic adenocarcinomas. Nucleic Acids Res 1988;16:7773–7782. 6. Caldas C, Hahn SA, da Costa LT, et al. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nat Genet 1994; 8:27–32. 7. Fukushima N, Sato N, Ueki T, et al. Aberrant methylation of preproenkephalin and p16 genes in pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma. Am J Pathol 2002;160:1573–1581. 8. Scarpa A, Capelli P, Mukai K, et al. Pancreatic adenocarcinomas frequently show p53 gene mutations. Am J Pathol 1993;142:1534–1543. 9. Pellegata NS, Sessa F, Renault B, et al. K-ras and p53 gene mutations in pancreatic cancer: ductal and nonductal tumors progress through different genetic lesions. Cancer Res 1994;54:1556–1560. 10. Redston MS, Caldas C, Seymour AB, et al. p53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Res 1994;54:3025–3033. 11. Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 1996;271:350–353.

-,

No.

-

12. Nussinov R, Tsai CJ, Jang H. Oncogenic Ras isoforms signaling specificity at the membrane. Cancer Res 2018; 78:593–602. 13. LaPak KM, Burd CE. The molecular balancing act of p16(INK4a) in cancer and aging. Mol Cancer Res 2014; 12:167–183. 14. Kastenhuber ER, Lowe SW. Putting p53 in context. Cell 2017;170:1062–1078. 15. Colak S, Ten Dijke P. Targeting TGF-beta signaling in cancer. Trends Cancer 2017;3:56–71. 16. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–674. 17. 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. 18. Biankin AV, Waddell N, Kassahn KS, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012;491:399–405. 19. Waddell N, Pajic M, Patch AM, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015;518:495–501. 20. Sausen M, Phallen J, Adleff V, et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat Commun 2015;6:7686. 21. Witkiewicz AK, McMillan EA, Balaji U, et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun 2015;6:6744. 22. Safran H, Steinhoff M, Mangray S, et al. Overexpression of the HER-2/neu oncogene in pancreatic adenocarcinoma. Am J Clin Oncol 2001;24:496–499. 23. Raphael BJ, Hruban RH, Aguirre AJ, et al. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 2017;32:185–203. 24. Goggins M, Shekher M, Turnacioglu K, et al. Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Res 1998;58:5329–5332. 25. Nones K, Waddell N, Song S, et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int J Cancer 2014;135:1110–1118. 26. Vincent A, Omura N, Hong SM, et al. Genome-wide analysis of promoter methylation associated with gene expression profile in pancreatic adenocarcinoma. Clin Cancer Res 2011;17:4341–4354. 27. Sato N, Fukushima N, Matsubayashi H, et al. Aberrant methylation of Reprimo correlates with genetic instability and predicts poor prognosis in pancreatic ductal adenocarcinoma. Cancer 2006;107:251–257. 28. Winter JM, Ting AH, Vilardell F, et al. Absence of E-cadherin expression distinguishes noncohesive from cohesive pancreatic cancer. Clin Cancer Res 2008;14:412–418. 29. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013;500:415–421. 30. Connor AA, Denroche RE, Jang GH, et al. Association of distinct mutational signatures with correlates of increased immune activity in pancreatic ductal adenocarcinoma. JAMA Oncol 2017;3:774–783.

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200

1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245Q16 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260

2019

Genetics of Familial and Sporadic Pancreatic Cancer

31. Notta F, Chan-Seng-Yue M, Lemire M, et al. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature 2016;538:378–382. 32. Yachida S, Jones S, Bozic I, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 2010;467:1114–1117. 33. Makohon-Moore AP, Zhang M, Reiter JG, et al. Limited heterogeneity of known driver gene mutations among the metastases of individual patients with pancreatic cancer. Nat Genet 2017;49:358–366. 34. Moffitt RA, Marayati R, Flate EL, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nature Genet 2015;47:1168–1178. 35. Collisson EA, Sadanandam A, Olson P, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med 2011;17:500–503. 36. Bailey P, Chang DK, Nones K, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016;531:47–52. 37. Kastrinos F, Mukherjee B, Tayob N, et al. Risk of pancreatic cancer in families with Lynch syndrome. JAMA 2009;302:1790–1795. 38. Roberts NJ, Jiao Y, Yu J, et al. ATM mutations in patients with hereditary pancreatic cancer. Cancer Discov 2012;2:41–46. 39. Klein AP. Genetic susceptibility to pancreatic cancer. Mol Carcinog 2012;51:14–24. 40. Lowenfels AB, Maisonneuve P, DiMagno EP, et al. Hereditary pancreatitis and the risk of pancreatic cancer. International Hereditary Pancreatitis Study Group. J Natl Cancer Inst 1997;89:442–446. 41. Zhen DB, Rabe KG, Gallinger S, et al. BRCA1, BRCA2, PALB2, and CDKN2A mutations in familial pancreatic cancer: a PACGENE study. Genet Med 2015;17:569–577. 42. Jones S, Hruban RH, Kamiyama M, et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science 2009;324:217. 43. Roberts NJ, Norris AL, Petersen GM, et al. Whole genome sequencing defines the genetic heterogeneity of familial pancreatic cancer. Cancer Discov 2016; 6:166–175. 44. Tamura K, Yu J, Hata T, et al. Mutations in the pancreatic secretory enzymes CPA1 and CPB1 are associated with pancreatic cancer. Proc Natl Acad Sci U S A 2018; 115:201720588. 45. Shindo K, Yu J, Suenaga M, et al. Deleterious germline mutations in patients with apparently sporadic pancreatic adenocarcinoma. J Clin Oncol 2017;35:3382–3390. 46. Hu C, Hart SN, Polley EC, et al. Association between inherited germline mutations in cancer predisposition genes and risk of pancreatic cancer. JAMA 2018; 319:2401–2409. 47. Lowery MA, Wong W, Jordan EJ, et al. Prospective evaluation of germline alterations in patients with exocrine pancreatic neoplasms. J Natl Cancer Inst 2018; 110:1067–1074. 48. Couch FJ, Farid LM, DeShano ML, et al. BRCA2 germline mutations in male breast cancer cases and breast cancer families. Nat Genet 1996;13:123–125.

11

49. Neuhausen S, Gilewski T, Norton L, et al. Recurrent BRCA2 6174delT mutations in Ashkenazi Jewish women affected by breast cancer. Nat Genet 1996;13:126–128. 50. Oddoux C, Struewing JP, Clayton CM, et al. The carrier frequency of the BRCA2 6174delT mutation among Ashkenazi Jewish individuals is approximately 1%. Nat Genet 1996;14:188–190. 51. Figer A, Irmin L, Geva R, et al. The rate of the 6174delT founder Jewish mutation in BRCA2 in patients with noncolonic gastrointestinal tract tumours in Israel. Br J Cancer 2001;84:478–481. 52. Ozcelik H, Schmocker B, Di Nicola N, et al. Germline BRCA2 6174delT mutations in Ashkenazi Jewish pancreatic cancer patients. Nat Genet 1997;16:17–18. 53. Salo-Mullen EE, O’Reilly EM, Kelsen DP, et al. Identification of germline genetic mutations in patients with pancreatic cancer. Cancer 2015;121:4382–4388. 54. Vasen H, Ibrahim I, Ponce CG, et al. Benefit of surveillance for pancreatic cancer in high-risk individuals: outcome of long-term prospective follow-up studies from three European expert centers. J Clin Oncol 2016; 34:2010–2019. 55. Giardiello FM, Brensinger JD, Tersmette AC, et al. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology 2000;119:1447–1453. 56. Vasen HF, Gruis NA, Frants RR, et al. Risk of developing pancreatic cancer in families with familial atypical multiple mole melanoma associated with a specific 19 deletion of p16 (p16-Leiden). Int J Cancer 2000; 87:809–811. 57. van Asperen CJ, Brohet RM, Meijers-Heijboer EJ, et al. Cancer risks in BRCA2 families: estimates for sites other than breast and ovary. J Med Genet 2005;42:711–719. 58. Struewing JP, Abeliovich D, Peretz T, et al. The carrier frequency of the BRCA1 185delAG mutation is approximately 1 percent in Ashkenazi Jewish individuals. Nat Genet 1995;11:198–200. 59. Ferrone CR, Levine DA, Tang LH, et al. BRCA germline mutations in Jewish patients with pancreatic adenocarcinoma. J Clin Oncol 2009;27:433–438. 60. Hu C, LaDuca H, Shimelis H, et al. Multigene hereditary cancer panels reveal high-risk pancreatic cancer susceptibility genes. JCO Precis Oncol 2018. 61. Antoniou AC, Casadei S, Heikkinen T, et al. Breastcancer risk in families with mutations in PALB2. N Engl J Med 2014;371:497–506. 62. Thompson D, Easton DF. Cancer incidence in BRCA1 mutation carriers. J Natl Cancer Inst 2002;94:1358– 1365. 63. Mocci E, Milne RL, Mendez-Villamil EY, et al. Risk of pancreatic cancer in breast cancer families from the breast cancer family registry. Cancer Epidemiol Biomarkers Prev 2013;22:803–811. 64. Wolpin BM, Chan AT, Hartge P, et al. ABO blood group and the risk of pancreatic cancer. J Natl Cancer Inst 2009;101:424–431. 65. Wolpin BM, Kraft P, Gross M, et al. Pancreatic cancer risk and ABO blood group alleles: results from the pancreatic cancer cohort consortium. Cancer Res 2010; 70:1015–1023.

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

PANCREATIC CANCER

-

Q17

1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320

12

PANCREATIC CANCER

1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380

Q18

Wood et al

Gastroenterology Vol.

66. Wolpin BM, Kraft P, Xu M, et al. Variant ABO blood group alleles, secretor status, and risk of pancreatic cancer: results from the pancreatic cancer cohort consortium. Cancer Epidemiol Biomarkers Prev 2010;19:3140–3149. 67. Amundadottir L, Kraft P, Stolzenberg-Solomon RZ, et al. Genome-wide association study identifies variants in the ABO locus associated with susceptibility to pancreatic cancer. Nat Genet 2009;41:986–990. 68. Petersen GM, Amundadottir L, Fuchs CS, et al. A genome-wide association study identifies pancreatic cancer susceptibility loci on chromosomes 13q22.1, 1q32.1 and 5p15.33. Nat Genet 2010;42:224–228. 69. Childs EJ, Chaffee KG, Gallinger S, et al. Association of common susceptibility variants of pancreatic cancer in higher risk patients: a PACGENE study. Cancer Epidemiol Biomarkers Prev 2016. 70. Wolpin BM, Rizzato C, Kraft P, et al. Genome-wide association study identifies multiple susceptibility loci for pancreatic cancer. Nat Genet 2014;46:994–1000. 71. Lu KH, Wood ME, Daniels M, et al. American Society of Clinical Oncology Expert Statement: collection and use of a cancer family history for oncology providers. J Clin Oncol 2014;32:833–840. 72. Holter S, Borgida A, Dodd A, et al. Germline BRCA mutations in a large clinic-based cohort of patients with pancreatic adenocarcinoma. J Clin Oncol 2015;33:3124– 3129. 73. Hu C, Hart SN, Bamlet WR, et al. Prevalence of pathogenic mutations in cancer predisposition genes among pancreatic cancer patients. Cancer Epidemiol Biomarkers Prev 2016;25:207–211. 74. Lucas AL, Frado LE, Hwang C, et al. BRCA1 and BRCA2 germline mutations are frequently demonstrated in both high-risk pancreatic cancer screening and pancreatic cancer cohorts. Cancer 2014;120:1960–1967. 75. Knudsen ES, O’Reilly EM, Brody JR, et al. Genetic diversity of pancreatic ductal adenocarcinoma and opportunities for precision medicine. Gastroenterology 2016;150:48–63. 76. Manchanda R, Loggenberg K, Sanderson S, et al. Population testing for cancer predisposing BRCA1/BRCA2 mutations in the Ashkenazi-Jewish community: a randomized controlled trial. J Natl Cancer Inst 2015; 107:379. 77. Metcalfe K, Lynch HT, Foulkes WD, et al. Effect of oophorectomy on survival after breast cancer in BRCA1 and BRCA2 mutation carriers. JAMA Oncol 2015;1:306–313. 78. Canto MI, Hruban RH, Fishman EK, et al. Frequent detection of pancreatic lesions in asymptomatic highrisk individuals. Gastroenterology 2012;142:796–804. 79. Stoffel EM, Mangu PB, Gruber SB, et al. Hereditary colorectal cancer syndromes: American Society of Clinical Oncology Clinical Practice Guideline endorsement of the familial risk-colorectal cancer: European Society for Medical Oncology Clinical Practice Guidelines. J Clin Oncol 2015;33:209–217. 80. Klein AP, Wolpin BM, Risch HA, et al. Genome-wide meta-analysis identifies five new susceptibility loci for pancreatic cancer. Nat Commun 2018;9:556.

-,

No.

-

81. Canto MI, Almario JA, Schulick RD, et al. Risk of neoplastic progression in high-risk individuals undergoing long-term surveillance for pancreatic cancer. Gastroenterology 2018;155:740–751. 82. Canto MI, Harinck F, Hruban RH, et al. International consensus recommendations on the management of patients with increased risk for familial pancreatic cancer (The Cancer of the Pancreas Screening (CAPS) Consortium Summit). Gut 2013;62:339–347. 83. Konings IC, Harinck F, Kuenen MA, et al. Factors associated with cancer worries in individuals participating in annual pancreatic cancer surveillance. Fam Cancer 2017;16:143–151. 84. Basturk O, Hong SM, Wood LD, et al. 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. 85. Kanda M, Matthaei H, Wu J, et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology 2012;142:730–733. 86. Lohr M, Kloppel G, Maisonneuve P, et al. Frequency of K-ras mutations in pancreatic intraductal neoplasias associated with pancreatic ductal adenocarcinoma and chronic pancreatitis: a meta-analysis. Neoplasia 2005; 7:17–23. 87. Murphy SJ, Hart SN, Lima JF, et al. Genetic alterations associated with progression from pancreatic intraepithelial neoplasia to invasive pancreatic tumor. Gastroenterology 2013;145:1098–1109. 88. Wilentz RE, Geradts J, Maynard R, et al. Inactivation of the p16 (INK4A) tumor-suppressor gene in pancreatic duct lesions: loss of intranuclear expression. Cancer Res 1998;58:4740–4744. 89. Wilentz RE, Iacobuzio-Donahue CA, Argani P, et al. Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Res 2000;60:2002– 2006. 90. Luttges J, Galehdari H, Brocker V, et al. Allelic loss is often the first hit in the biallelic inactivation of the p53 and DPC4 genes during pancreatic carcinogenesis. Am J Pathol 2001;158:1677–1683. 91. Hutchings D, Waters KM, Weiss MJ, et al. Cancerization of the pancreatic ducts: demonstration of a common and under-recognized process using immunolabeling of paired duct lesions and invasive pancreatic ductal adenocarcinoma for p53 and Smad4 expression. Am J Surg Pathol 2018;42:1556–1561. 92. Hosoda W, Chianchiano P, Griffin JF, et al. Genetic analyses of isolated high-grade pancreatic intraepithelial neoplasia (HG-PanIN) reveal paucity of alterations in TP53 and SMAD4. J Pathol 2017;242:16–23. 93. 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. 94. Hong SM, Vincent A, Kanda M, et al. Genome-wide somatic copy number alterations in low-grade PanINs and IPMNs from individuals with a family history of pancreatic cancer. Clin Cancer Res 2012;18:4303–4312.

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440

1441 1442 1443 Q19 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500

2019

Genetics of Familial and Sporadic Pancreatic Cancer

95. Hata T, Suenaga M, Marchionni L, et al. Genome-wide somatic copy number alterations and mutations in highgrade pancreatic intraepithelial neoplasia. Am J Pathol 2018;188:1723–1733. 96. Adsay NV, Merati K, Basturk O, et al. Pathologically and biologically distinct types of epithelium in intraductal papillary mucinous neoplasms: delineation of an “intestinal” pathway of carcinogenesis in the pancreas. Am J Surg Pathol 2004;28:839–848. 97. Amato E, Molin MD, Mafficini A, et al. Targeted nextgeneration sequencing of cancer genes dissects the molecular profiles of intraductal papillary neoplasms of the pancreas. J Pathol 2014;233:217–227. 98. 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. 99. Biankin AV, Biankin SA, Kench JG, et al. Aberrant p16(INK4A) and DPC4/Smad4 expression in intraductal papillary mucinous tumours of the pancreas is associated with invasive ductal adenocarcinoma. Gut 2002; 50:861–868. 100. Wu J, Matthaei H, Maitra A, et al. Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Sci Transl Med 2011;3:92ra66. 101. Furukawa T, Kuboki Y, Tanji E, et al. Whole-exome sequencing uncovers frequent GNAS mutations in intraductal papillary mucinous neoplasms of the pancreas. Sci Rep 2011;1:161. 102. Wu J, Jiao Y, Dal Molin M, et al. Whole-exome sequencing of neoplastic cysts of the pancreas reveals recurrent mutations in components of ubiquitindependent pathways. Proc Natl Acad Sci U S A 2011; 108:21188–21193. 103. Basturk O, Tan M, Bhanot U, et al. The oncocytic subtype is genetically distinct from other pancreatic intraductal papillary mucinous neoplasm subtypes. Mod Pathol 2016;29:1058–1069. 104. Yamaguchi H, Kuboki Y, Hatori T, et al. Somatic mutations in PIK3CA and activation of AKT in intraductal tubulopapillary neoplasms of the pancreas. Am J Surg Pathol 2011;35:1812–1817. 105. Schonleben F, Qiu W, Ciau NT, et al. PIK3CA mutations in intraductal papillary mucinous neoplasm/carcinoma of the pancreas. Clin Cancer Res 2006;12:3851–3855. 106. Jimenez RE, Warshaw AL, Z’Graggen K, et al. Sequential accumulation of K-ras mutations and p53 overexpression in the progression of pancreatic mucinous cystic neoplasms to malignancy. Ann Surg 1999;230:501–509. 107. Iacobuzio-Donahue CA, Wilentz RE, Argani P, et al. Dpc4 protein in mucinous cystic neoplasms of the pancreas: frequent loss of expression in invasive carcinomas suggests a role in genetic progression. Am J Surg Pathol 2000;24:1544–1548. 108. Conner JR, Marino-Enriquez A, Mino-Kenudson M, et al. Genomic characterization of low- and high-grade pancreatic mucinous cystic neoplasms reveals recurrent KRAS alterations in “high-risk” lesions. Pancreas 2017;46:665–671.

13

109. Felsenstein M, Noe M, Masica DL, et al. IPMNs with cooccurring invasive cancers: neighbours but not always relatives. Gut 2018;67:1652–1662. 110. Kuboki Y, Fischer CG, Beleva Guthrie V, et al. Single-cell sequencing defines genetic heterogeneity in pancreatic cancer precursor lesions. J Pathol 2018;247. 111. Pea A, Yu J, Rezaee N, et al. 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. 112. Makohon-Moore AP, Matsukuma K, Zhang M, et al. Precancerous neoplastic cells can move through the pancreatic ductal system. Nature 2018;561:201–205. 113. Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6:224ra24. 114. Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018;359(6378):926–930. 115. Cohen JD, Javed AA, Thoburn C, et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc Natl Acad Sci U S A 2017;114:10202–10207. 116. Singhi AD, McGrath K, Brand RE, et al. Preoperative next-generation sequencing of pancreatic cyst fluid is highly accurate in cyst classification and detection of advanced neoplasia. Gut 2018;67:2131–2141. 117. Springer S, Wang Y, Molin MD, et al. A combination of molecular markers and clinical features improve the classification of pancreatic cysts. Gastroenterology 2015;149:1501–1510. 118. Yu J, Sadakari Y, Shindo K, et al. Digital next-generation sequencing identifies low-abundance mutations in pancreatic juice samples collected from the duodenum of patients with pancreatic cancer and intraductal papillary mucinous neoplasms. Gut 2017;66:1677–1687. 119. Suenaga M, Yu J, Shindo K, et al. Pancreatic juice mutation concentrations can help predict the grade of dysplasia in patients undergoing pancreatic surveillance. Clin Cancer Res 2018. 120. Kanda M, Knight S, Topazian M, et al. Mutant GNAS detected in duodenal collections of secretin-stimulated pancreatic juice indicates the presence or emergence of pancreatic cysts. Gut 2013;62:1024–1033. 121. Eshleman JR, Norris AL, Sadakari Y, et al. KRAS and guanine nucleotide-binding protein mutations in pancreatic juice collected from the duodenum of patients at high risk for neoplasia undergoing endoscopic ultrasound. Clin Gastroenterol Hepatol 2015;13:963–969. 122. Jabbar KS, Arike L, Verbeke CS, et al. Highly accurate identification of cystic precursor lesions of pancreatic cancer through targeted mass spectrometry: a phase IIc diagnostic study. J Clin Oncol 2017:JCO.2017.73.7288. 123. Hata T, Dal Molin M, Hong SM, et al. Predicting the grade of dysplasia of pancreatic cystic neoplasms using cyst fluid DNA methylation markers. Clin Cancer Res 2017;23:3935–3944. 124. Hata T, Dal Molin M, Suenaga M, et al. Cyst fluid telomerase activity predicts the histologic grade of cystic

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

Q20

PANCREATIC CANCER

-

Q21

1501 1502 1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560

14

PANCREATIC CANCER

1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599 1600 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611 1612 1613 1614 1615 1616 1617 1618 1619 1620

Wood et al

Gastroenterology Vol.

neoplasms of the pancreas. Clin Cancer Res 2016; 22:5141–5151. 125. Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 2007;25:1960–1966. 126. Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011;364:1817–1825. 127. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013;369:1691–1703. 128. Ottaiano A, Capozzi M, De Divitiis C, et al. Gemcitabine mono-therapy versus gemcitabine plus targeted therapy in advanced pancreatic cancer: a meta-analysis of randomized phase III trials. Acta Oncol 2017;56:377–383. 129. O’Neil BH, Scott AJ, Ma WW, et al. A phase II/III randomized study to compare the efficacy and safety of rigosertib plus gemcitabine versus gemcitabine alone in patients with previously untreated metastatic pancreatic cancer. Ann Oncol 2015;26:1923–1929. 130. Fuchs CS, Azevedo S, Okusaka T, et al. A phase 3 randomized, double-blind, placebo-controlled trial of ganitumab or placebo in combination with gemcitabine as first-line therapy for metastatic adenocarcinoma of the pancreas: the GAMMA trial. Ann Oncol 2015; 26:921–927. 131. Philip PA, Benedetti J, Corless CL, et al. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. J Clin Oncol 2010;28:3605–3610. 132. Deplanque G, Demarchi M, Hebbar M, et al. A randomized, placebo-controlled phase III trial of masitinib plus gemcitabine in the treatment of advanced pancreatic cancer. Ann Oncol 2015;26:1194–1200. 133. Kindler HL, Ioka T, Richel DJ, et al. Axitinib plus gemcitabine versus placebo plus gemcitabine in patients with advanced pancreatic adenocarcinoma: a doubleblind randomised phase 3 study. Lancet Oncol 2011; 12:256–262. 134. Kindler HL, Niedzwiecki D, Hollis D, et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303). J Clin Oncol 2010;28:3617–3622. 135. Rougier P, Riess H, Manges R, et al. Randomised, placebo-controlled, double-blind, parallel-group phase III study evaluating aflibercept in patients receiving firstline treatment with gemcitabine for metastatic pancreatic cancer. Eur J Cancer 2013;49:2633–2642. 136. Sheahan AV, Biankin AV, Parish CR, et al. Targeted therapies in the management of locally advanced and metastatic pancreatic cancer: a systematic review. Oncotarget 2018;9:21613–21627. 137. Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366:2455–2465.

-,

No.

-

138. Royal RE, Levy C, Turner K, et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother 2010;33:828–833. 139. Foley K, Kim V, Jaffee E, et al. Current progress in immunotherapy for pancreatic cancer. Cancer Lett 2016; 381:244–251. 140. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409–413. 141. Humphris JL, Patch AM, Nones K, et al. Hypermutation in pancreatic cancer. Gastroenterology 2017;152:68–74. 142. Hu ZI, Shia J, Stadler ZK, et al. Evaluating mismatch repair deficiency in pancreatic adenocarcinoma: challenges and recommendations. Clin Cancer Res 2018; 24:1326–1336. 143. Pishvaian MJ, Bender RJ, Halverson D, et al. Molecular profiling of pancreatic cancer patients: initial results from the know your tumor initiative. Clin Cancer Res 2018; 24:5018–5027. 144. Heining C, Horak P, Uhrig S, et al. NRG1 fusions in KRAS wild-type pancreatic cancer. Cancer Discov 2018; 8:CD-18-0036. 145. Aguirre AJ, Nowak JA, Camarda ND, et al. Real-time genomic characterization of advanced pancreatic cancer to enable precision medicine. Cancer Discov 2018; 8:1096–1111. 146. Aung KL, Fischer SE, Denroche RE, et al. Genomicsdriven precision medicine for advanced pancreatic cancer: early results from the COMPASS trial. Clin Cancer Res 2018;24:1344–1354. 147. Hecht JR, Bang YJ, Qin SK, et al. Lapatinib in combination with capecitabine plus oxaliplatin in human epidermal growth factor receptor 2-positive advanced or metastatic gastric, esophageal, or gastroesophageal adenocarcinoma: TRIO-013/LOGiC—a randomized phase III trial. J Clin Oncol 2016;34:443–451. 148. Tabernero J, Hoff PM, Shen L, et al. Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): final analysis of a double-blind, randomised, placebo-controlled phase 3 study. Lancet Oncol 2018; 19:1372–1384. 149. Pishvaian MJ, Bender RJ, Halverson D, et al. Molecular profiling of patients with pancreatic cancer: initial results from the know your tumor initiative. Clin Cancer Res 2018;24:5018–5027. 150. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med 2018;378:731–739. 151. Tiriac H, Belleau P, Engle DD, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov 2018;8:CD-180349. 152. Yurgelun MB, Chittenden AB, Morales-Oyarvide V, et al. Germline cancer susceptibility gene variants, somatic second hits, and survival outcomes in patients with resected pancreatic cancer. Genet Med 2018; 21:213–223.

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:26 pm  ce

Q31

1621 1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633 1634 1635 1636 1637 1638 1639 1640 1641 1642 1643 1644 1645 1646 1647 1648 1649 1650 1651 1652 1653 1654 1655 1656 1657 1658 1659 1660 1661 1662 1663 1664 1665 1666 1667 1668 1669 1670 1671 1672 1673 1674 1675 1676 1677 1678 1679 1680

1681 1682Q22 1683 1684 1685 1686 1687 1688 1689 1690 1691 1692 1693 1694 1695 1696 1697 1698Q23 1699 1700 1701 1702Q24 1703 1704 1705 1706 1707 1708 1709 1710 1711 1712 1713 1714 1715 1716 1717 1718 1719 1720 1721 1722 1723Q25 1724 1725 1726 1727 1728 1729 1730 1731 1732 1733 1734 1735 1736 1737 1738 1739 1740

2019

Genetics of Familial and Sporadic Pancreatic Cancer

153. Brand R, Borazanci E, Speare V, et al. Prospective study of germline genetic testing in incident cases of pancreatic adenocarcinoma. Cancer 2018;124(17). 154. Aguirre AJ, Nowak JA, Camarda ND, et al. Real-time genomic characterization of advanced pancreatic cancer to enable precision medicine. Cancer Discov 2018; 8:1096–1111. 155. Kondo T, Kanai M, Kou T, et al. Association between homologous recombination repair gene mutations and response to oxaliplatin in pancreatic cancer. Oncotarget 2018;9:19817–19825. 156. Golan T, Sella T, O’Reilly EM, et al. Overall survival and clinical characteristics of BRCA mutation carriers with stage I/II pancreatic cancer. Br J Cancer 2017; 116:697–702. 157. Shahda S, Timms KM, Ibrahim AA, et al. Homologous recombination deficiency in patients with pancreatic ductal adenocarcinoma and response to chemotherapy. JCO Precis Oncol 2018. 158. Reiss KA, Yu S, Judy R, et al. Retrospective survival analysis of patients with advanced pancreatic ductal adenocarcinoma and germline BRCA or PALB2 mutations. JCO Precis Oncol 2018. 159. Yarchoan M, Myzak MC, Johnson BA 3rd, et al. Olaparib in combination with irinotecan, cisplatin, and mitomycin C in patients with advanced pancreatic cancer. Oncotarget 2017;8:44073–44081. 160. O’Reilly EM, Lee JW, Lowery MA, et al. Phase 1 trial evaluating cisplatin, gemcitabine, and veliparib in 2 patient cohorts: germline BRCA mutation carriers and wildtype BRCA pancreatic ductal adenocarcinoma. Cancer 2018;124:1374–1382. 161. de Bono J, Ramanathan RK, Mina L, et al. Phase I, dose-escalation, two-part trial of the PARP inhibitor talazoparib in patients with advanced germline BRCA1/2 mutations and selected sporadic cancers. Cancer Discov 2017;7:620–629. 162. Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol 2015; 33:244–250. 163. Shroff RT, Hendifar A, McWilliams RR, et al. Rucaparib monotherapy in patients with pancreatic cancer and a known deleterious BRCA mutation. JCO Precis Oncol 2018. 164. Lowery MA, Kelsen DP, Capanu M, et al. Phase II trial of veliparib in patients with previously treated BRCAmutated pancreas ductal adenocarcinoma. Eur J Cancer 2018;89:19–26.

15

165. Litton JK, Rugo HS, Ettl J, et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med 2018;379:753–763. 166. Robson M, Im SA, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med 2017;377:523–533. 167. Kim G, Ison G, McKee AE, et al. FDA approval summary: olaparib monotherapy in patients with deleterious germline BRCA-mutated advanced ovarian cancer treated with three or more lines of chemotherapy. Clin Cancer Res 2015;21:4257–4261. 168. Higuchi T, Flies DB, Marjon NA, et al. CTLA-4 blockade synergizes therapeutically with PARP inhibition in BRCA1-deficient ovarian cancer. Cancer Immunol Res 2015;3:1257–1268. 169. Jiao S, Xia W, Yamaguchi H, et al. PARP inhibitor upregulates PD-L1 expression and enhances cancerassociated immunosuppression. Clin Cancer Res 2017; 23:3711–3720. 170. Lee JM, Cimino-Mathews A, Peer CJ, et al. Safety and clinical activity of the programmed death-ligand 1 inhibitor durvalumab in combination with poly (ADPRibose) polymerase inhibitor olaparib or vascular endothelial growth factor receptor 1-3 inhibitor cediranib in women’s cancers: a dose-escalation, phase I study. J Clin Oncol 2017;35:2193–2202.

1741 1742 1743 1744 1745 1746 1747 1748 1749 1750 1751 1752 1753 1754 1755 1756 1757 1758 1759 1760 1761 1762 1763 1764 1765 1766 1767 Received September 20, 2018. Accepted December 5, 2018. 1768 1769 Reprint requests 1770 Address requests for reprints to: Laura Wood, MD, PhD, Johns Hopkins Medical Institutions, CRB2 351, 1550 Orleans Street, Baltimore, Maryland 1771 Q4 Q5 21231. e-mail: [email protected]; fax: 410-614-0671; Matthew B. Yurgelun, 1772 MD, Johns Hopkins Medical Institutions, CRB2 351, 1550 Orleans Street, Baltimore, Maryland 21231. e-mail: [email protected]; fax: 1773 410-614-0671; Michael Goggins, MD, Johns Hopkins Medical Institutions, Q6 1774 CRB2 351, 1550 Orleans Street, Baltimore, Maryland 21231. e-mail: [email protected]; fax: 410-614-0671. 1775 1776 Q7 Conflicts of interest 1777 The authors disclose no conflicts. 1778 Funding 1779 Q8 This work was supported by National Institutes of Health grants U01210170, R01CA176828, K08 DK107781, and CA62924; Susan Wojcicki and Dennis 1780 Troper; the Pancreatic Cancer Action Network; the Lustgarten Foundation for Q32 1781 Pancreatic Cancer Research; Stand Up to Cancer; Buffone Family Gastrointestinal Cancer Research Fund; Kaya Tuncer Career Development 1782 Award in Gastrointestinal Cancer Prevention; American Gastroenterological Q9 1783 Association–Bernard Lee Schwartz Foundation Research Scholar Award in Pancreatic Cancer; Sidney Kimmel Foundation for Cancer Research Kimmel 1784 Scholar Award; American Association of Cancer Research–Incyte Q10 1785 Corporation Career Development Award for Pancreatic Cancer Research; 1786 Rolfe Pancreatic Cancer Foundation; Joseph C. Monastra Foundation; and The Gerald O. Mann Charitable Foundation. Michael G. Goggins is the Sol 1787 Goldman Professor of Pancreatic Cancer Research. 1788 1789 1790 1791 1792 1793 1794 1795 1796 1797 1798 1799 1800

FLA 5.5.0 DTD  YGAST62391_proof  1 April 2019  6:27 pm  ce

PANCREATIC CANCER

-