Detection of Mismatch Repair Deficiency and Microsatellite Instability in Colorectal Adenocarcinoma by Targeted Next-Generation Sequencing

Detection of Mismatch Repair Deficiency and Microsatellite Instability in Colorectal Adenocarcinoma by Targeted Next-Generation Sequencing

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 ...

825KB Sizes 0 Downloads 77 Views

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 61 62

The Journal of Molecular Diagnostics, Vol. -, No. -, - 2016

jmd.amjpathol.org

Detection of Mismatch Repair Deficiency and Microsatellite Instability in Colorectal Adenocarcinoma by Targeted Next Generation Sequencing Q5

Jonathan A. Nowak,* Matthew B. Yurgelun,yz Jacqueline L. Bruce,* Vanesa Rojas-Rudilla,* Dimity L. Hall,* Priyanka Shivdasani,* Elizabeth P. Garcia,* Agoston T. Agoston,* Amitabh Srivastava,* Shuji Ogino,*zx Frank C. Kuo,* Neal I. Lindeman,* and Fei Dong* From the Department of Pathology,* Brigham and Women’s Hospital, Harvard Medical School, Boston; the Center for Medical Genetics and Preventiony and the Department of Medical Oncology,z Dana Farber Cancer Institute, Harvard Medical School, Boston; and the Department of Epidemiology,x Harvard T.H. Chan School of Public Health, Boston, Massachusetts Accepted for publication July 28, 2016. Address correspondence to Fei Dong, M.D., Department of Pathology, Brigham and Women’s Hospital, 75 Francis St., Amory 3, Boston, MA 02114. E-mail: fdong1@ partners.org.

Q3

Mismatch repair protein deficiency (MMR-D) and high microsatellite instability (MSI-H) are features of Lynch syndromeeassociated colorectal carcinomas and have implications in clinical management. We evaluate the ability of a targeted next-generation sequencing panel to detect MMR-D and MSI-H based on mutational phenotype. Using a criterion of >40 total mutations per megabase or >5 single-base insertion or deletion mutations in repeats per megabase, sequencing achieves 92% sensitivity and 100% specificity for MMR-D by immunohistochemistry in a training cohort of 149 colorectal carcinomas and 91% sensitivity and 98% specificity for MMR-D in a validation cohort of 94 additional colorectal carcinomas. False-negative samples are attributable to tumor heterogeneity, and next-generation sequencing results are concordant with analysis of microsatellite loci by PCR. In a subset of 95 carcinomas with microsatellite analysis, sequencing achieves 100% sensitivity and 99% specificity for MSI-H in the combined training and validation set. False-positive results for MMR-D and MSI-H are attributable to ultramutated cancers with POLE mutations, which are confirmed by direct sequencing of the POLE gene and are detected by mutational signature analysis. These findings provide a framework for a targeted tumor sequencing panel to accurately detect MMR-D and MSI-H in colorectal carcinomas. (J Mol Diagn 2016, -: 1e8; http://dx.doi.org/10.1016/j.jmoldx.2016.07.010)

Mismatch repair protein deficiency (MMR-D) and high microsatellite instability (MSI-H) reflect the mechanism of tumorigenesis in approximately 15% of colorectal carcinomas, which may be sporadic or inherited as part of Lynch syndrome. In MMR-D and MSI-H colorectal carcinomas, defective DNA mismatch repair machinery leads to hypermutation and instability of DNA repeat regions.1 Multiple laboratory tests are available to evaluate the status of the mismatch repair pathway. These include immunohistochemistry (IHC) staining for mismatch repair proteins, PCR to evaluate microsatellite instability, MLH1 promoter methylation analysis, and BRAF sequencing. These tests are performed directly on tumor samples and have prognostic and therapeutic implications for nearly all patients with colorectal carcinoma.2,3 In addition, the results

can be used to guide germline testing if the results indicate an increased risk of Lynch syndrome.4 As next-generation sequencing technology becomes increasingly available for clinical use, we hypothesize that much of the Lynch syndromeerelated testing in the anatomical and molecular pathology laboratory may be accomplished by a single assay. Sequencing of tumor specimens can test for mutational patterns characteristic of MMR-D and MSI-H while simultaneously detecting pathogenic alterations and facilitating downstream germline testing. Supported by the Department of Pathology, Brigham and Women’s Hospital and NIH grant R35 CA197735 (S.O.). Q1 Disclosures: None declared. Q2

Copyright ª 2016 American Society for Investigative Pathology and the Association for Molecular Pathology. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.jmoldx.2016.07.010

FLA 5.4.0 DTD  JMDI546_proof  14 November 2016  10:21 pm  EO: JMD16_0133

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 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124

125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186

Nowak et al Herein, we use a cancer gene panel to demonstrate an approach for MMR-D and MSI-H detection by targeted next-generation sequencing, and we correlate the sequencing results with DNA mismatch repair protein IHC and PCR-based microsatellite instability testing in a series of colorectal carcinomas.

Materials and Methods Patient Selection and Training and Validation Cohorts Patients were prospectively enrolled via Profile, an institutional cohort study for cancer genotyping.5 All participants provided written informed consent. A total of 243 colorectal adenocarcinomas were tested by targeted next-generation sequencing in the study. Of these, 149 were assigned to the training cohort, and 94 were assigned to the validation cohort based on period of enrollment and hybrid capture method (Library Preparation and Next-Generation Sequencing). All cases were evaluated for mismatch repair protein expression by IHC. Sixty-two of 149 specimens in the training cohort and 33 of 94 specimens in the validation cohort were also tested by PCR of microsatellite loci. PCR testing was performed clinically as requested by the treating physician. In addition, all cases with abnormal IHC results, except one in the training cohort without available material, were tested by PCR for the purposes of this study. Germline testing results were obtained by review of the medical record. This study was approved by the institutional review board of the Dana Farber Cancer Institute and the Partners Human Research Committee.

Library Preparation and Next-Generation Sequencing

genes implicated in Lynch syndrome, including MLH1, PMS2, MSH2, and MSH6, as well as genes frequently mutated in colorectal cancer, including KRAS, BRAF, PIK3CA, APC, and TP53 (complete gene list available in Supplemental Tables S1 and S2). EPCAM was not included in either panel. Massively parallel sequencing was performed using an Illumina HiSeq2500 (Illumina) in fast mode with 100  100 paired-end reads. Pooled sample reads were deconvoluted (demultiplexed) and sorted using Picard version 1.92 and later (Broad Institute, Cambridge, MA). Reads were aligned to the reference sequence b37 edition from the Human Genome Reference Consortium using BWA version 0.5.9 (Broad Institute). Duplicate reads were identified and removed using Picard. The median mean target coverage per sample after removal of duplicate reads was 169. The alignments were further refined using the Genome Analysis Toolkit version 1.6 and later (Broad Institute) for localized realignment around indel sites. Recalibration of the quality scores was performed using the Genome Analysis Toolkit. Mutation analysis for single-nucleotide variants was performed using MuTect version 1 0.27200 (Broad Institute). Insertions and deletions were called using Indelocator (Broad Institute). Integrative Genomics Viewer version 2.0.16 or later (Broad Institute) was used for visualization and interpretation. Variants were filtered to exclude synonymous variants, known germline variants in the Single Nucleotide Polymorphism database, and variants that occur at a population frequency of >0.1% in the Exome Sequencing Project database. Copy number detection was performed by analysis of fractional coverage of a defined genomic interval compared with pooled normal samples. Structural variant analysis was performed using BreaKmer to detect larger insertions and deletions.6

DNA was isolated from formalin-fixed, paraffin-embedded tumor tissue. Paired normal samples were not tested. Corresponding hematoxylin and eosinestained slides were reviewed and selected for areas with at least 20% tumor nuclei. Tumor-enriched areas were macrodissected from ten 4-mm sections. DNA was isolated using standard extraction methods (Qiagen, Valencia, CA) and quantified using PicoGreen-based dsDNA detection (Life Technologies, Carlsbad, CA). Indexed sequencing libraries were prepared from 50-ng sonically sheared DNA samples using Illumina TruSeq LT reagents (Illumina Inc, San Diego, CA). Using a custom RNA bait set created by Agilent SureSelect (Agilent Technologies, Santa Clara, CA), libraries were enriched for exons of genes implicated in cancer biology using solution-based hybrid capture. The RNA bait set for the training cohort was from an earlier enrollment period and included a basic panel that covered the coding region of 275 genes, which encompasses 757,787 bp of the genome. The RNA bait set for the validation cohort included an extended panel of 298 genes covering 831,033 bp. Both panels included coverage of DNA mismatch repair

DNA Mismatch Repair IHC

2

jmd.amjpathol.org

IHC was conducted after pressure cooker heateinduced epitope retrieval (0.01 mol/L citrate buffer, pH 6.0) on 4-mm thick formalin-fixed, paraffin-embedded tissue sections using mouse anti-MLH1 monoclonal antibody (1:100 dilution; clone ES05; Novocastra, Buffalo Grove, IL), mouse anti-MSH2 monoclonal antibody (1:150 dilution; clone FE11; Calbiochem, San Diego, CA), mouse anti-PMS2 monoclonal antibody (1:50 dilution; clone MRQ-28; Cell Marque, Rocklin, CA), and mouse anti-MSH6 monoclonal antibody (1:50 dilution; clone PU29; Novocastra, Buffalo Grove, IL) using the Envision Plus Detection System (Dako, Carpinteria, CA).

Microsatellite Loci PCR Microsatellite instability was evaluated by amplification across five different microsatellite loci [four mononucleotide repeats (BAT25, BAT26, BAT40, BAT34c) and one dinucleotide repeat (D18S55)] with fluorescently labeled primers

-

The Journal of Molecular Diagnostics

FLA 5.4.0 DTD  JMDI546_proof  14 November 2016  10:21 pm  EO: JMD16_0133

187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248

POLE Sanger Sequencing

A

Statistical Analysis

Training 25 20 15 10 5 0

The Fisher exact test was used to compare differences in categorical variables. The Mann-Whitney test was used to compare distributions of continuous variables. All tests were two-sided with statistical significance set at P < 0.05.

10

100

Total Mutations (per Mb)

Validation

B 25

Results Molecular Phenotype of MMR-D in the Training Cohort Of 149 carcinomas in the training cohort, 13 (8.7%) had loss of nuclear expression of at least one mismatch repair protein and were classified as MMR-D. The remaining 136 carcinomas (91.3%) were classified as mismatch repair protein intact (MMR-I), meaning they had intact expression of all four mismatch repair proteins. Two variables from next-generation sequencing data were selected for analysis: the total mutational burden (substitutions and small insertion and deletion mutations) and the burden of single-nucleotide insertion or deletion mutations (indels) in mononucleotide repeat regions. A repeat region was defined in this study as two or more consecutive nucleotides. These variables were selected to reflect the expected molecular phenotype as a result of DNA repair defects associated with microsatellite instability.

The Journal of Molecular Diagnostics

-

20 15 10 5 print & web 4C=FPO

Q4

POLE was not part of the targeted next-generation sequencing panel, and the presence of POLE mutations in exons 9 and 13 was evaluated by Sanger sequencing in select cases. PCR amplification and sequencing primers were designed as follows: POLE exon 9 (forward: 50 -TGCTTATTTTGTCCCCACAG-30 , reverse: 50 -TACTTCCCAGAAGCCACCTG-30 ) and POLE exon 13 (forward: 50 -TCTGTTCTCATTCTCCTTCC-30 , reverse: 50 -CGGGATGTGGCTTACGTG-30 ). Bidirectional cycle sequencing was performed with the BigDye Terminator version 3.1 Cycle Sequencing Kit (Thermo Fisher, Cambridge, MA). Sequencing products were analyzed by capillary gel electrophoresis (3130xl Genetic Analyzer, Applied Biosystems), and results were visualized with Mutation Surveyor (Softgenetics, State College, PA) and Chromas Lite (Technelysium, South Brisbane, Australia).

311 The median total mutational burden in MMR-D 312 carcinomas was 62.0 per megabase (Mb) (range, 13.2 to 313 277.1 per Mb) compared with 15.8 per Mb (range, 5.3 to 314 39.6 per Mb) in MMR-I carcinomas (Mann-Whitney 315 P < 0.0001). 316 The median mutational burden of single-nucleotide indels 317 in repeat regions in MMR-D carcinomas was 11.9 per Mb 318 (range, 0 to 21.1 per Mb) compared with 0 per Mb (mean, 319 0.5 per Mb; range, 0 to 4.0 per Mb) in MMR-I carcinomas 320 321 (Mann-Whitney P < 0.0001). 322 On the basis of these observations, a criterion of total 323 mutations >40 per Mb or indels in repeat regions >5 per 324 Mb was selected for the detection of MMR-D. This criterion 325 achieved 92% sensitivity (n Z 12/13) and 100% specificity 326 (n Z 136/136) in the training cohort (Figure 1A). ½F1 327 In the single false-negative sample, the patient had two 328 primary colorectal carcinomas and liver metastasis. One of 329

Indels in Homopolymer Region (per Mb)

in paired tumor-normal samples. PCR products were analyzed by capillary gel electrophoresis (3130xl Genetic Analyzer, Applied Biosystems, Foster City, CA). Microsatellite instability was defined by alteration in the length of the PCR product in the tumor sample compared with normal. Samples with instability in greater than or equal to two of five loci are classified as MSI-H. Samples with instability in one of five loci are classified as low microsatellite instability. Samples without instability in any of the five loci are classified as microsatellite stable (MSS).

Indels in Homopolymer Region (per Mb)

249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310

Microsatellite Instability by Sequencing

0 10

100

Total Mutations (per Mb) MMR-D

MMR-I

Figure 1

Mismatch repair protein deficiency status compared with mutational burden and singleebase pair insertion and deletion mutations in mononucleotide repeat (homopolymer) regions detected by next-generation sequencing in the training (A) and validation (B) data sets. MMR-D, mismatch repair protein deficient; MMR-I, mismatch repair protein intact.

jmd.amjpathol.org

FLA 5.4.0 DTD  JMDI546_proof  14 November 2016  10:21 pm  EO: JMD16_0133

3

330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372

373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434

Nowak et al the two primary colorectal carcinomas had loss of MSH2 and MSH6 staining, whereas the other primary carcinoma had intact IHC staining for all four mismatch repair proteins. IHC was not available for the liver metastasis. Sequencing was performed on the metastatic carcinoma in the liver, which revealed a KRAS mutation, a TP53 mutation, two APC frameshift mutations, and copy number changes consistent with carcinogenesis via the chromosomal instability pathway. Microsatellite PCR performed on the liver sample was MSS, consistent with next-generation sequencing results.

Application in the Validation Cohort Of 94 carcinomas in the validation cohort, 11 (11.7%) were MMR-D, and 83 (88.3%) were MMR-I. Application of the training cohort criterion achieved 91% sensitivity (n Z 10/11) and 98% specificity (n Z 81/83) in the validation cohort (Figure 1B). In the single false-negative sample in validation, IHC revealed heterogeneous loss of MSH2 and MSH6 within the carcinoma. Next-generation sequencing results were concordant with MSS status as determined by microsatellite PCR testing. Sequencing identified a TP53 mutation, two APC nonsense mutations, and copy number changes consistent with carcinogenesis via the chromosomal instability pathway. The results were explained by tumor heterogeneity and limited sampling for molecular analysis. The only two false-positive carcinomas with retained MMR protein expression were determined to be POLEassociated ultramutated colorectal carcinomas (Phenotype of POLE-Associated Ultramutated Colorectal Carcinoma). We confirmed that using both total mutational rate and the rate of insertion and deletion mutations resulted in better sensitivity than using either variable alone. Using only >40 total mutations per Mb, the assay achieved 73% sensitivity (n Z 8/11) and 98% specificity (n Z 81/83) in the validation cohort. Using only >5 single-nucleotide indels in repeat regions per Mb, the assay achieved 82%sensitivity (n Z 9/11) and 100% specificity (n Z 83/83). Other than the carcinoma with heterogenous IHC staining, the only MMR-D carcinoma not detected using single-nucleotide indels in repeat regions had isolated MSH6 deficiency.

99% (n Z 72/73), respectively, with the only discordant result attributable to a POLE-mutated colorectal carcinoma.

Phenotype of POLE-Associated Ultramutated Colorectal Carcinoma Two carcinomas in the validation cohort were predicted by next-generation sequencing to have MMR-D but had retained mismatch repair protein expression by IHC. One carcinoma was MSS by PCR and the other was MSI-H, with instability observed in two of five microsatellite loci. Both carcinomas had a very high mutation burden (300.8 and 290.0 per Mb) and a low burden of indels in mononucleotide repeat regions (2.4 and 0 per Mb). Sanger sequencing of POLE exons 9 and 13 had a POLE c.857C>G (p.Pro286Arg) mutation in each sample. As a control, POLE Sanger sequencing was performed for the two MSI-H carcinomas from this study with the highest mutational burden. Both MSI-H carcinomas tested negative for POLE exon 9 or exon 13 mutations (Figure 2). ½F2 Furthermore, POLE-associated ultramutated colorectal carcinomas had distinctive mutational signatures compared with colorectal carcinomas with MSI-H. Both cases were highly enriched for cytosine to adenine transitions at TpCpT sites. In contrast, carcinomas with MSI-H had a predominance of cytosine to thymine transversions at CpG sites (Figure 3). These findings were consistent with previously ½F3 described POLE-associated and MSI-Heassociated mutational signatures.7

Gene Mutations in MMR-D Colorectal Carcinomas We also evaluated differences in mutational frequencies of specific genes between MMR-D and MMR-I colorectal carcinomas. This analysis was limited to frequently mutated oncogenes and tumor suppressor genes in colorectal cancer: KRAS, BRAF, PIK3CA, APC and TP53 (Figure 4). MMR-D ½F4 carcinomas were associated with a higher frequency of BRAF mutations (42% versus 12% in MMR-I carcinomas, P Z 0.0007) and a lower frequency of TP53 mutations (33% versus 74% in MMR-I tumors, P < 0.0001).

Concordance with Microsatellite PCR Using the criterion established for detecting MMR-D, namely, total mutation burden >40 per Mb or number of indels in repeat regions >5 per Mb, next-generation sequencing achieved 100% sensitivity (n Z 11/11) and 100% specificity (n Z 51/51) for MSI-H in the training cohort and 100% sensitivity (n Z 11/11) and 95% specificity (n Z 21/22) for MSI-H in the validation cohort. Sensitivity and specificity for next-generation sequencingebased detection of MSI-H in the combined training and validation cohorts are 100% (n Z 22/22) and

4

Detection of Pathogenic Mutations in Patients with Lynch Syndrome Of the 243 patients in the combined training and validation cohort, 10 had clinical germline testing for Lynch syndromeeassociated genes based on abnormal mismatch repair protein IHC or microsatellite PCR results. Seven patients had pathogenic variants in MLH1, PMS2, MSH2 or MSH6, including sequence alterations and copy number changes, identified on germline testing that confirmed a diagnosis of Lynch syndrome. All seven variants were also detected in

jmd.amjpathol.org

-

The Journal of Molecular Diagnostics

FLA 5.4.0 DTD  JMDI546_proof  14 November 2016  10:21 pm  EO: JMD16_0133

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

print & web 4C=FPO

A

B

DNA C AAG TTT CCT GAT GCT G Sequence G Amino Acid sequence Codon

C

D

C AAG TTT CCT GAT GCT G G

C AAG TTT CCT GAT GCT G

C AAG TTT CCT GAT GCT G

Lys Phe Pro Asp Ala Arg 284 285 286 287 288

Lys Phe Pro Asp Ala

Lys Phe Pro Asp Ala

284 285 286 287 288

284 285 286 287 288

Lys Phe Pro Asp Ala Arg 284 285 286 287 288

Figure 2 A and B: POLE c.857C>G (p.Pro286Arg) mutations were identified in two colorectal carcinomas with high mutational burdens but without increased indels in homopolymer regions (arrowheads indicate affected nucleotide). C and D: The two high microsatellite instability carcinomas with the highest mutational burden were wild type for POLE mutations.

½T1

the next-generation sequencing panel of tumor tissue (Table 1).

Discussion Lynch syndrome confers an increased lifetime risk of cancer development in affected individuals and is implicated in the pathogenesis of approximately 3% of colorectal cancers.

A

A diagnosis of Lynch syndrome has clinical implications for the patient and family members, and many laboratories have instituted universal screening for Lynch syndrome in newly diagnosed colorectal carcinomas using immunohistochemical staining of DNA mismatch repair genes or microsatellite instability PCR or both.8 Abnormal results may be evaluated by additional testing with MLH1 promoter methylation analysis or BRAF gene sequencing.9 This algorithmic approach requires a complex clinical decision

B

C>A

T>A

C>G

T>C

C

C>A

C>G

C>A

C>G

T>A

T>C

T>A

T>C

D 0.3 0.2

0.1

print & web 4C=FPO

0.0 C>A

C>G

C>T

T>A

T>C

T>G

TA GTC GTG GTT TTA TTC TTG TTT

497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558

Microsatellite Instability by Sequencing

C>T

T>G

Figure 3

Mutational signatures of POLE-associated ultramutated colorectal carcinomas (A and B) compared with colorectal carcinomas with high microsatellite instability (C and D).

The Journal of Molecular Diagnostics

-

jmd.amjpathol.org

FLA 5.4.0 DTD  JMDI546_proof  14 November 2016  10:21 pm  EO: JMD16_0133

5

559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620

1.0

Proportion of Carcinomas Mutated

621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682

Nowak et al MMR-D MMR-I

0.8 0.6 0.4 0.2 0.0

KRAS

BRAF PIK3CA

APC

TP53

Figure 4 Commonly mutated colorectal cancer genes in mismatch repair protein deficient (MMR-D) and mismatch repair protein intact (MMR-I) carcinomas. MMR-D carcinomas had a higher frequency of BRAF mutations (P Z 0.0007) and lower frequency of TP53 mutations (P < 0.0001).

tree and may require a single sample to be tested by multiple assays. We hypothesize that much of the testing performed for Lynch syndrome screening may be streamlined with a single next-generation sequencing assay. Prior work using next-generation sequencing to predict microsatellite status has focused on targeted sequencing of known microsatellite loci10 or analysis of microsatellite regions using new informatics tools.11e13 We found that the mutational phenotype from a standard informatics pipeline restricted to coding regions of genes is also able to detect microsatellite instability with high sensitivity and specificity. In the validation cohort, the only discordant results compared with microsatellite PCR testing are found to be due to carcinomas harboring POLE p.Pro286Arg mutations. POLE p.Pro286Arg is an exon 9 hotspot mutation in the exonuclease domain of POLE and has been associated with

an elevated rate of mutation in vitro14 and an ultramutated phenotype in human cancer.15 In our analysis, the POLEassociated ultramutated phenotype can be resolved by POLE gene sequencing and mutational signature analysis. Our results are comparable to an analysis by Stadler et al,16 who observed that mutational burden alone is highly sensitive and specific for MMR-D in a similar targeted nextgeneration sequencing panel. Compared with their assay, we perform sequencing on tumor-only samples without paired normal tissue and rely on the informatics pipeline to filter common germline events. As a result, the assay by Stadler et al16 is expected to be more specific for the detection of true somatic events. We found that tumor-only sequencing is equally effective in detecting MMR-D with the additional benefits of decreasing sequencing cost, simplifying the laboratory workflow and making testing available to patients in situations in which paired normal DNA may be difficult to obtain. Furthermore, we identify recurrent insertion and deletion mutations in repeat regions as a key phenotypic feature of MMR-D, which may be more specific for MSI-H compared with other mechanisms of hypermutation, such as POLE mutation. We also observe a low rate of discrepancy between MMR-D and MSI-H attributable to tumor heterogeneity, and we validate next-generation sequencing against microsatellite PCR, the current molecular gold standard. We describe the resolution of MSI-H from POLE-associated ultramutated carcinomas through analysis of mutational signatures. Finally, we found that our sequencing panel detects pathogenic mutations in DNA mismatch repair genes, which can guide downstream germline testing. Notably, our approach for MMR-D and MSI-H screening of using mutational patterns rather than individual gene mutations may be applied to sequencing panels with different gene combinations as long as the panels are of

Table 1 Mismatch Repair Gene Mutations Detected by Tumor and Germline Sequencing in Lynch SyndromeeAssociated Colorectal Carcinomas

Mismatch repair IHC

Singleebase pair indels in Total mutations repeat regions, per Mb Tumor sequencing MSI PCR per Mb

MSH2 and MSH6 loss MSI-H

97.7

10.6

PMS2 loss MSI-H MSH2 and MSH6 loss MSI-H MSH2 and MSH6 loss MSI-H

88.4 73.4 66.2

14.5 15.4 12.0

PMS2 loss MSH6 loss

MSI-H MSI-H

59.4 59.0

11.8 3.6

MLH1 and PMS2 loss

MSI-H

37.3

10.8

Allele fraction, % Germline sequencing

MSH2 c.1861C>T (p.R621*), MSH2 c.1165C>T (p.R389*), MSH6 c.845dupT (p.V282fs), MSH6 c.3103C>T (p.R1035*) PMS2 c.137G>T (p.S46I) MSH2 c.1684G>T (p.E562*) MSH2 c.2131C>T (p.R711*), MSH2 c.1251dupT (p.I418fs) PMS2 exon 1e5 deletion MSH6 c.3939_3957dup19 (p.Arg1318fs), PMS2 c.2347G>A (p.V783I) MLH1 exon 13 deletion

16 26 39 24 60 71 45 20 NA NA NA

MSH6 c.845dupT (p.V282fs)

PMS2 c.137G>T (p.S46I) MSH2 c.1684G>T (p.E562*) MSH2 c.2131C>T (p.R711*) PMS2 exon 1e5 deletion MSH6 c.3939_3957dup19 (p.Arg1318fs) MLH1 exon 13 deletion

IHC, immunohistochemistry; MSI, microsatellite instability; MSI-H, high microsatellite instability; NA, not applicable.

6

jmd.amjpathol.org

-

The Journal of Molecular Diagnostics

FLA 5.4.0 DTD  JMDI546_proof  14 November 2016  10:21 pm  EO: JMD16_0133

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

745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806

Microsatellite Instability by Sequencing sufficient scale to estimate mutational burden with statistical power. In many cases, laboratories already using nextgeneration sequencing for cancer may be able to evaluate mismatch repair pathway status using existing data at minimal additional cost. In addition to providing an accurate measure of mismatch repair pathway status, next-generation sequencing allows for quantification of the degree of mismatch repair deficiency or the overall mutational burden. Increased neoantigen load has been strongly linked to clinical response to immune checkpoint inhibitors in multiple cancer types, including MSI-H colorectal carcinomas,17 and MSI-H colorectal carcinomas harbor immune microenvironments favorable to checkpoint blockade.18 Beyond evaluation of the mismatch repair pathway, sequencing identifies clinically actionable gene mutations. Pathway-activating mutations in KRAS and BRAF predict lack of response to anti-EGFR antibody therapy in patients with advanced colorectal cancer.19,20 Given the increasing availability of pathway-specific targeted therapies, next-generation sequencing may guide enrollment into clinical trials. As these advances broaden the indication for the sequencing of cancer genomes, it is rational to use data derived from a single test to fulfill multiple clinical roles, including the evaluation of hereditary cancer risk. In summary, the mutational phenotype of colorectal carcinomas by targeted next-generation sequencing, as defined by the burden of total mutations and singlenucleotide indels in repeat regions, can accurately detect MMR-D and MSI-H compared with current methods. Nextgeneration sequencing may be a useful tool for colorectal carcinomas by providing a means of generating molecular results to guide therapeutic decisions while simultaneously enabling risk assessment for Lynch syndrome.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

Supplemental Data Supplemental material for this article can be found at http://dx.doi.org/10.1016/j.jmoldx.2016.07.010.

15.

References 1. Lynch HT, Snyder CL, Shaw TG, Heinen CD, Hitchins MP: Milestones of Lynch syndrome: 1895e2015. Nat Rev Cancer 2015, 15: 181e194 2. Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM, Hamilton SR, Laurent-Puig P, Gryfe R, Shepherd LE, Tu D, Redston M, Gallinger S: Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med 2003, 349:247e257 3. Lochhead P, Kuchiba A, Imamura Y, Liao X, Yamauchi M, Nishihara R, Qian ZR, Morikawa T, Shen J, Meyerhardt JA, Fuchs CS, Ogino S: Microsatellite instability and BRAF mutation testing in colorectal cancer prognostication. J Natl Cancer Inst 2013, 105: 1151e1156 4. Giardiello FM, Allen JI, Axilbund JE, Boland CR, Burke CA, Burt RW, Church JM, Dominitz JA, Johnson DA, Kaltenbach T, Levin TR, Lieberman DA, Robertson DJ, Syngal S, Rex DK:

The Journal of Molecular Diagnostics

-

16.

17.

Guidelines on genetic evaluation and management of lynch syndrome: a consensus statement by the US Multi-Society Task Force on Colorectal Cancer. Gastroenterology 2014, 147:502e526 MacConaill LE, Garcia E, Shivdasani P, Ducar M, Adusumilli R, Breneiser M, Byrne M, Chung L, Conneely J, Crosby L, Garraway L, Gong X, Hahn W, Hatton C, Kantoff P, Kluk M, Kuo F, Jia Y, Joshi R, Longtine J, Manning A, Palescandolo E, Sharaf N, Sholl L, van Hummelen P, Wade J, Wollinson BM, Zepf D, Rollins BJ, Lindeman NI: Prospective enterprise-level molecular genotyping of a cohort of cancer patients. J Mol Diagn 2014, 16:660e672 Abo RP, Ducar M, Garcia EP, Thorner AR, Rojas-Rudilla V, Lin L, Sholl LM, Hahn WC, Meyerson M, Lindeman NI, Van Hummelen P, MacConnaill LE: BreaKmer: detection of structural variation in targeted massively parallel sequencing data using Kmers. Nucleic Acids Res 2014, 43:e19 Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, et al: Signatures of mutational processes in human cancer. Nature 2013, 500:415e421 Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, Nakagawa H, Sotamaa K, Prior TW, Westman J, Panescu J, Fix D, Lockman J, Comeras I, de la Chapelle A: Screening for the Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med 2005, 352:1851e1860 Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, Kang GH, Widschwendter M, Weener D, Buchanan D, Koh H, Simms L, Barker M, Leggett B, Levine J, Kim M, French AJ, Thibodeau SN, Jass J, Haile R, Laird PW: CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 2006, 38:787e793 Gan C, Love C, Beshay V, Macrae F, Fox S, Waring P, Taylor G: Applicability of next generation sequencing technology in microsatellite instability testing. Genes (Basel) 2015, 6:46e59 Niu B, Ye K, Zhang Q, Lu C, Xie M, McLellan MD, Wendl MC, Ding L: MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics 2014, 30:1015e1016 Salipante SJ, Scroggins SM, Hampel HL, Turner EH, Pritchard CC: Microsatellite instability detection by next generation sequencing. Clin Chem 2014, 60:1192e1199 Huang MN, McPherson JR, Cutcutache I, Teh BT, Tan P, Rozen SG: MSIseq: software for assessing microsatellite instability from catalogs of somatic mutations. Sci Rep 2015, 5:13321 Kane DP, Shcherbakova PV: A common cancer-associated DNA polymerase E mutation causes an exceptionally strong mutator phenotype, indicating fidelity defects distinct from loss of proofreading. Cancer Res 2014, 74:1895e1901 Church DN, Briggs SEW, Palles C, Domingo E, Kearsey SJ, Grimes JM, Gorman M, Martin L, Howarth KM, Hodgson SV; NSECG Collaborators, Kaur K, Taylor J, Tomlinson IP: DNA polymerase E and D exonuclease domain mutations in endometrial cancer. Hum Mol Genet 2013, 22:2820e2828 Stadler ZK, Battaglin F, Middha S, Hechtman JF, Tran C, Cercek A, Yaeger R, Segal NH, Varghese AM, Reidy-Lagunes DL, Kemeny NE, Salo-Mullen EE, Ashraf A, Weiser MR, Garcia-Aguilar J, Robson ME, Offit K, Arcila ME, Berger MF, Shia J, Solit DB, Saltz LB: Reliable detection of mismatch repair deficiency in colorectal cancers using mutational load in next-generation sequencing panels. J Clin Oncol 2016, 34:2141e2147 Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Duffy SM, Goldberg RM, de la Chapelle A, Koshiji M, Bhaijee F, Huebner T, Hruban RH, Wood LD, Cuka N, Pardoll DM, Papadopoulos N, Kinzler KW, Zhou S, Cornish TC, Taube JM, Anders RA, Eshleman JR, Vogelstein B, Diaz LA: PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015, 372: 2509e2520

jmd.amjpathol.org

FLA 5.4.0 DTD  JMDI546_proof  14 November 2016  10:21 pm  EO: JMD16_0133

7

807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868

869 870 871 872 873 874 875 876

Nowak et al 18. Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, Blosser RL, Fan H, Wang H, Luber BS, Zhang M, Papadopoulos N, Kinzler KW, Vogelstein B, Sears CL, Anders RA, Pardoll DM, Housseau F: The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counterinhibitory checkpoints. Cancer Discov 2015, 5:43e51 19. Lièvre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, Côté JF, Tomasic G, Penna C, Ducreux M, Rougier P, Penault-Llorca F,

Laurent-Puig P: KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 2006, 66: 3992e3995 20. Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, De Dosso S, Mazzucchelli L, Frattini M, Siena S, Bardelli A: Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol 2008, 26: 5705e5712

8

jmd.amjpathol.org

-

The Journal of Molecular Diagnostics

FLA 5.4.0 DTD  JMDI546_proof  14 November 2016  10:21 pm  EO: JMD16_0133

877 878 879 880 881 882 883 884