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
Diagnostic Detection of Allelic Losses and Imbalances by Next-Generation Sequencing 1P/19Q Co-deletion Analysis of Gliomas Q33
Hendrikus J. Dubbink,* Peggy N. Atmodimedjo,* Ronald van Marion,* Peter H.J. Riegman,* Johan M. Kros,* Martin J. van den Bent,y and Winand N.M. Dinjens* From the Departments of Pathology* and Neuro-Oncology,y Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands Accepted for publication June 1, 2016.
Q3
Q6
Address correspondence to Hendrikus J. Dubbink, Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands. E-mail: h.dubbink@ erasmusmc.nl.
Cancer cells are genomic unstable and accumulate tumor typeespecific molecular aberrations, which may represent hallmarks for predicting prognosis and targets for therapy. Co-deletion of chromosomes 1p and 19q marks gliomas with an oligodendroglioma component and predicts a better prognosis and response to chemotherapy. In the current study, we present a novel method to detect chromosome 1p/19q co-deletion or loss of heterozygosity (LOH) in a diagnostic setting, based on single-nucleotide polymorphism (SNP) analysis and next-generation sequencing (NGS) on an Ion Torrent platform. We selected highly polymorphic SNPs distributed evenly over both chromosome arms. To experimentally determine the sensitivity and specificity of targeted SNP analysis, we used DNAs extracted from 49 routine formalin-fixed, paraffin-embedded glioma tissues and compared the outcome with diagnostic microsatellite-based LOH analysis and calculated estimates. We show that targeted SNP analysis by NGS allows reliable detection of 1p and/or 19q deletion in a background of 70% of normal cells according to calculated outcomes, is more sensitive than microsatellite-based LOH analysis, and requires much less DNA. This specific and sensitive SNP assay is broadly applicable for simultaneous allelic imbalance analysis of multiple genomic regions and can be incorporated easily into NGS mutation analyses. The combined mutation and chromosomal imbalance analysis in a single NGS assay is suited perfectly for routine glioma diagnostics and other diagnostic molecular pathology applications. (J Mol Diagn 2016, -: 1e12; http://dx.doi.org/10.1016/j.jmoldx.2016.06.002)
Cancer cells are genomic unstable and accumulate mutations (ie, point and small mutations) and large aberrations including amplification or deletion of whole chromosomes, chromosome arms, or intrachromosomal deletions or amplifications. Detection of these tumor-specific molecular aberrations is of increasing importance in routine pathology diagnostics.1 In this respect, well-known examples are EGFR mutations in nonesmall-cell lung cancer,2 ERBB2 (HER2) amplifications in breast cancer,3 and 1p/19q codeletions in gliomas.4 Molecular classification of malignancies from the central nervous system is important to establish the diagnosis and prognosis and to predict treatment outcome of histologically similar tumors. Oligodendrogliomas are a glioma subtype
Q4
Q5
characterized by concurrent loss of the entire chromosomal arms 1p and 19q, resulting from an unbalanced t(1;19) (q10;p10).4,5 Complete 1p/19q loss also has been observed in Q7 oligoastrocytomas, but at a lower frequency. In contrast to tumors with an oligodendroglial component, astrocytomas rarely present with complete 1p/19q loss, but often have smaller losses of genetic material from 1p and 19q.6 Codeletion of the entire chromosome 1p and 19q predicts a better prognosis for patients with oligodendrogliomas independent of treatment, and an improved outcome to treatment Supported by the Department of Pathology, Erasmus MC Cancer Q1 Institute, Rotterdam, the Netherlands. Q2 Disclosures: None declared.
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.2016.06.002
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
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
Dubbink et al with adjuvant chemotherapy resulting in an increased progression-free survival and overall survival as compared with patients with tumors without typical 1p/19q co-deletion or with smaller losses of 1p and/or 19q.6e8 Currently, most routine diagnostics for detection of 1p/19q co-deletion or loss of heterozygosity (LOH) is performed by fluorescent in situ hybridization,9 microsatellite analysis,10 or multiplex ligation-dependent probe amplification.11 Newer comprehensive molecular approaches for the evaluation of chromosomal gains and losses include whole-exome next-generation sequencing (NGS), comparative genomic hybridization arrays, and high-density singlenucleotide polymorphism (SNP) arrays.12 However, these techniques require specific equipment, experience with data analyses, and large amounts of DNA, which often is not present in biopsy specimens or in cases in which only (immune) stained sections or cytology preparations are available, and cannot always be combined with mutation analysis. In addition, the quality and quantity of DNA from formalin-fixed, paraffin-embedded (FFPE) tissues is suboptimal for reliable quantitative analysis of the data from these assays.13 Recent developments in NGS technology allow costeffective clinical implementation of targeted sequencing of only small amounts (10 ng) of DNA from archival FFPE material.1 However, because of the repetitive nature of microsatellites, these markers cannot (yet) be analyzed reliably by most NGS platforms. Several laboratories have shown the usefulness of amplicon-based NGS strategies to generate information on copy number alterations (CNAs).14e18 In the current study, we developed an application of NGS-based, low-density analysis of highly polymorphic SNPs for the routine detection of LOH, in particular 1p/19q co-deletion, with high sensitivity and specificity and a fast turnaround time. This novel assay paves the way toward simultaneous detection of both allelic imbalances and mutations in small amounts of DNA retrieved from FFPE tissues for glioma subtype diagnostics in particular, but also for routine implementation in molecular diagnostics of other tumor types in a pathology laboratory.
Selected tissue areas enriched for a high percentage of neoplastic cells were microdissected manually from hematoxylin-stained 4-mm sections of FFPE glioma tissues. The percentage of neoplastic cells was estimated by our local neuropathologist (J.M.K.). DNA was extracted by proteinase K digestion for 16 hours at 56 C in the presence of 5% Chelex 100 resin and used without further purification after Q9 inactivation of proteinase K and removal of cell debris and the Chelex resin, as previously described.21,22 DNA concentration was measured with the Qubit 2.0 fluorometer (Thermo Fisher Scientific), as described by the manufacturer. Q10 In most cases no (adjacent) normal tissue was available.
Microsatellite-Based LOH Analysis of Chromosomes 1p and 19q LOH analysis was performed within 10 polymorphic microsatellite markers scattered across the entire chromosomes 1p and 19q (Figure 1A). Microsatellite markers and their ½F1 chromosomal localization (human reference genome 19) are shown in Table 1. Microsatellite amplification was per- ½T1 formed after initial denaturation at 95 C for 3 minutes for 28 cycles of 15 seconds at 95 C, 15 seconds at 60 C, and 15 seconds at 72 C, followed by 7 minutes at 72 C. PCR products were analyzed on an ABI 3730 genetic analyzer (Applied Biosystems). Allelic losses were assessed based on Q11 the analysis of multiple informative markers, as described elsewhere.10 Typical oligodendroglial co-deletion of 1p/19q was defined as LOH of all informative markers on both chromosomal arms. If not all informative markers were lost, a chromosome was considered partially lost.
SNP-Based LOH Analysis of Chromosomes 1p and 19q
Forty-nine glioma tissues were collected between 1997 and 2003 during the European Organization of Research and Treatment of Cancer study 26951 on adjuvant procarbazine, lomustine, and vincristine chemotherapy of anaplastic oligodendrogliomas and anaplastic oligoastrocytomas.19 Normal autopsy brain FFPE tissue was obtained from the tissue bank of the Department of Pathology. All tissue samples were assessed anonymously according to the Proper Secondary Use of Human Tissue code established by the Dutch Federation of Medical Scientific Societies.20
A custom primer panel was designed that includes SNPs on chromosomes 1p and 19q using the Ion AmpliSeq Designer Q12 2.0.23 Highly polymorphic SNPs on both chromosomes were selected via the NCBI SNP database with a global Q13 minor allele frequency of at least 45% to obtain a high number of informative SNPs in each assay. The mean SNP density for chromosomes 1p and 19q was set arbitrarily to approximately 1 SNP per 3.5 Mb and 1 SNP per 2 Mb, respectively, yielding a total of 29 SNPs on chromosome 1p and 16 SNPs on chromosome 19q that covered the entire chromosomal arms (Figure 1A). Selected SNPs and their chromosomal localization (SNP database 138) are shown in Table 2. ½T2 Next-generation targeted sequencing was performed by semiconductor sequencing with the Ion Torrent Personal Genome Machine with the suppliers’ materials and protocols (Life Technologies, Carlsbad, CA) as previously described.24 Briefly, DNA input varied between 1 and 10 ng, dependent on the amount of tissue or DNA available. Library and template preparations were performed
2
jmd.amjpathol.org
Materials and Methods Glioma Tissues
Q8
DNA Extraction
-
The Journal of Molecular Diagnostics
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
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
Figure 1 Overview on marker location and single-nucleotide polymorphism (SNP)-based loss of heterozygosity (LOH) analysis by targeted nextgeneration sequencing. A: Location of SNP and microsatellite markers on chromosomes 1p and 19q (not at scale, size bars shown in Mbp). The relative positions and distribution of the microsatellite markers (green dots above) and SNPs (blue dots below) on chromosomes 1p and 19q used in this study are indicated. The SNP and microsatellite markers and their exact locations are shown in Tables 1 and 2. B: Explanatory figure showing the possible outcomes of SNP-based LOH analysis of chromosome 1p. The x axis shows the chromosomal position in million base pairs (Mbp), and the y axis shows the percentage variant (B-allele frequency).
print & web 4C=FPO
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
Detection of Chromosomal Loss by NGS
Q14
consecutively with the AmpliSeq Library Kit 2.0-384 LV and the Ion Personal Genome Machine Template OT2 200 kit. Templates were sequenced using the Ion Personal Genome Machine Sequencing 200 Kit v2 on an Ion 318v2 chip. Sequence information was analyzed with Variant Table 1
Q21
Q22
Caller v3.6 (Life Technologies), and variants were annotated in a local Galaxy pipeline using ANNOVAR.25e27 Because of the semiquantitative nature of NGS analysis, even more with inferior DNA quality, a SNP was considered to be imbalanced or relatively lost when the variant B-allele
Chromosomal Localization of the Microsatellite Markers Used in This Study
Microsatellite marker (GRCh37/human reference genome 19) Marker
Position
chromosome 1p D1S199 19956993-19957152 D1S513 31331730-31331987 D1S197 50750477-50750694 D1S2806 67868153-67868355 D1S495 102561337-102561767 chromosome 19q D19S875 29797108-29797470 D19S198 42153083-42153201 D19S412 47010944-47011334 D19S606 47973563-47973917 D19S572 54105351-54105693
Locus
Primer forward*
Primer reverse
1p36.13 1p35.2 1p32.3 1p31.3 1p21.1
50 -GATCATGACACTACACTTCAG-30 50 -ATCGCAAGACACAGGCACTG-30 50 -TCATGTCCCTCCTCCCAAAG-30 50 -CATTACATCACAGCCTGATTAGA-30 50 -ACCAAACCTTTGCAGAGGA-30
50 -ACCATGTGCTCCGTAAATAAG-30 50 -ACTAACACACACCATTGCAAG-30 50 -GAGCAAGCATCCAAAAACGA-30 50 -CCACCATGCCTGACCT-30 50 -AACCCTGGTATGCCATCA-30
19q12 19q13.2 19q13.32 19q13.32 19q13.42
50 -TGGTTCTGTGATGACTACTACATGC-30 50 -GAAAGTGTCCACAACGGTAGC-30 50 -CAGCCTGAGCGACAGAATG-30 50 -AGGGCTGGGACCTCAC-30 50 -TTTGGGTGTGCTGACACGTG-30
50 -AACTTGGTTTATGATGTCTCTTGC-30 50 -AAAGGGGTTAGAGGAGAAGGC-30 50 -TGTCTCCTCCTTGGTGCATG-30 50 -TACGAGGCTGTGCCTGTAG-30 50 -TGAAGATCTCGGGCCACATC-30 Q30
*FAM label.
The Journal of Molecular Diagnostics
-
jmd.amjpathol.org
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
3
311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372
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
Dubbink et al Table 2
Chromosomal Localization of the SNPs Used in This Study Q31
SNP (database 138) SNP
Position
chromosome 1p rs7663 16112795 rs169957 19683301 rs169885 21628545 rs742358 22459170 rs309481 23210600 rs189882 24868045 rs9259 25168124 rs7491 25895238 rs159525 26213991 rs7504 27238150 rs6564 28212975 rs157208 29245406 rs6425953 36168038 rs7686 38268918 rs7315 40306898 rs7903 45976472 rs504816 53307957 rs7374 55316322 rs87061 60594980 rs11811946 65952428 rs5680 71477315 rs191142 76990862 rs12754569 85462971 rs54396 88776278 rs106075 91604522 rs1132 95394352 rs8888 101338324 rs6604120 109289487 rs8128 115110683 chromosome 19q rs7283 30106659 rs2542297 31883906 rs33841 34011248 rs12852 35615179 rs1291 38229378 rs17628 39926509 rs166539 40931717 rs3817 44090195 rs10113 47112648 rs8355 48833800 rs6521 49519873 rs11573 51359497 rs193040 53073605 rs3814 53611187 rs10217 56030428 rs10448 59093239
Primer forward
Primer reverse
50 -CATTTTGTACGGCACGGACAA-30 50 -GGTGCCAGAGCCAGATTTGT-30 50 -CTCACTAGGCTCCCCTGATG-30 50 -GGAGCCACCTGCATCCC-30 50 -ATCTCTGCCTGTTGGAATCTTCAG-30 50 -GACCTGAGTTCAACCCCTGT-30 50 -TGCCTAAAAGCATTTATCCTTCATACCA-30 50 -CTGACACAGGCCAGCCTT-30 50 -GGGAATGGAGTACAAGGGCTAT-30 50 -GCAGCCTAAGCTTTCATTCTCATC-30 50 -ATGTGGTTAACATGGATTAATGTGGGA-30 50 -AGCATTTAGTTAGAGGAGAGGAGAGG-30 50 -TGGGCTGTTTCCTCCCTTCTA-30 50 -GACTGAACCCCTGCCAACTAA-30 50 -GCAGCTCGCAAATTTCAAAGTCT-30 50 -TTCCCATGCAGCCCTTTGAATA-30 50 -GCAGGTTATAAGGGTCTTCTCGAGT-30 50 -CTGAGAGGATTCTGGCACCTG-30 50 -TCAAGTTAGAATGACCACTTTCCGTATG-30 50 -GGGAGATGAGCTGAAAGTTCCA-30 50 -CAATTTGGGTAGTCACAAACTCCAT-30 50 -AAATGACTTTCTGGAAAATGAGGTCTATTCA-30 50 -GGGACTTATTCCACGCTTCAG-30 50 -TCTTGCCTTCATCACAGGTTGG-30 50 -TTCACCTGTCTCTTACCCCCATA-30 50 -ACTTGCTAATTTTGTCCAAAGGGAGTA-30 50 -GGTAAACAGAGGCCTAGTTAAGAATTCC-30 50 -GTCGTGTGGCAGAGTGAGT-30 50 -CAAATAGAATTACCACAGCAGCCTACA-30
50 -CTCTAGGCTCAGGGCAAGAC-30 50 -AGGAAGGGAGATGTTAGGATGACC-30 50 -CTAAGGTTGGTTTCTGACTAAGACCTAA-30 50 -GGTTGCTCAGCTCCTTCCT-30 50 -ACTCATGTCTCTCATTCATTCACACAA-30 50 -GCCACTTTCTTGTAAAGGTGTGTT-30 50 -AGGTGCCCATCAGTTTCTCTTC-30 50 -TTTCACAAATAAAGCACAGCAAGACTT-30 50 -CCCCATTTTCTTCCTTTTCTTCCATAC-30 50 -TGTATAGACAGCACTTGGCTCCT-30 50 -CTCAGGTCTCCATAAGGGTCTTCT-30 50 -CCAGTGTGCACTCCAGAGTA-30 50 -GCTTTCCTTCTTTCTGGAATTTCTGTT-30 50 -TGGATAAAGATTGAAGAGCCACAGG-30 50 -TGAGCCACATATTGGGAGTTCTAGAT-30 50 -CCAAGGTGTTACATTTTGTTTCACTACA-30 50 -GCCCCTGAAAATCTGGCAACA-30 50 -TCATTTGTGTGGAAAGTCAGAGGAA-30 50 -CTGAGGATAAAGAGGTCTCTTCAACTG-30 50 -CATATGGCCCACCTCATGTTCT-30 50 -ACAGCAAATTAGCTCCTAACCTAACAAA-30 50 -TGCAGGCCTATGGGAAATGTTC-30 50 -CTGCTTACTTCCTCGGCTCTT-30 50 -TGGTGTGAAAGTAGGATGAAAACCTT-30 50 -TCCTCTGTGTGGTTTGGTAAATTACAT-30 50 -CTCCTAGGTCCCAAAGAAATGTGG-30 50 -TGCATCAATTCATTCTTAAGGTTGCCTA-30 50 -GGAAAGCAGGAAGAAGTCCTCA-30 50 -CGGCAAGACTTCTGAAAAGACAATTTA-30
50 -TGCTCTATGATAAATCAGTTGCATCTGT-30 50 -GAGAGCCATTGTCCCAAATATGGA-30 50 -CACCAGCCTGCTCAGTCTAC-30 50 -CCCAATACCTAATAAGGCATGCGAAA-30 50 -CTCTCTCAATTGCTAAAGCCATACCTA-30 50 -GCTCCCATGTTACCCCCTAGAT-30 50 -CAGCACGTAGAGGTCCGT-30 50 -TTTATTCATTCCTCCAAAGAGCACCA-30 50 -CCAAGCTGCATGATTGCTCTTT-30 50 -CTACGGAAGCGGGAGTGA-30 50 -CCCTGAGGTGGCAGCAT-30 50 -CGTGTCTTTTCAAACCCACATCCTAA-30 50 -GGCCTGGCTTTAGTCCTGT-30 50 -TGAGACAACCAGTGACTTCAAACA-30 50 -GTGTGACCCTCTCCAGGATTT-30 50 -GGTCGAGGGTGATTCGCT-30
50 -GCCATCCAATGGACCTTTGGG-30 50 -TCAACTGGTCCTCTCCTACCC-30 50 -TGCTAGTCCTTCAGCAATGAGAC-30 50 -TGTTTGAGGGATAAAACGGCATGA-30 50 -TGGGAAAACCTTCAGATATGGTTCAG-30 50 -CGCGGTGAGGTTGTCTAGTC-30 50 -GGCTACCTCTTCGTTCTGATTGG-30 50 -GGCATGGAAAACTTGGGTGAGT-30 50 -CCATCCGGGCTGGAGGA-30 50 -GTTCAGGAGAGGTCAAGCGT-30 50 -ATCCAGGGAGCCGCTTC-30 50 -CCAGGGTTGGGAATGCTTCTC-30 50 -ACGTCCCAGGTACGGAAGT-30 50 -GTGAGGAATAATTCAGTCTAGTTGTGCT-30 50 -CAGTCTTGTGATTCTGTCTTTCTAGGATT-30 50 -AACACCAGCCAGAGCTTATTCAT-30
SNP, single-nucleotide polymorphism.
frequency of a heterozygous SNP was either higher than 55% or lower than 45%. All variant frequencies between 45% and 55% were considered not to be aberrant. Similarly, cut-off lines were indicated at 5% and 95%, if not otherwise stated. These values are not absolute but were helpful in interpretation of the outcome. Typical oligodendroglial co-deletion of
1p and 19q was defined as imbalance to a similar degree of all informative SNP on both chromosomes. Partial allelic Q15 imbalance or loss was considered only if at least two consecutive informative SNPs showed imbalance to minimize the chance that apparent losses or imbalances are the result of preferential amplification of one of the alleles.
4
jmd.amjpathol.org
-
The Journal of Molecular Diagnostics
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
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
Results Interpretation of Low-Density, SNP-Based LOH Analysis Targeted, amplicon-based NGS analysis of SNPs yields different outcomes per SNP (Figure 1B). SNPs may be noninformative homozygous for either a reference or variant SNP (B-allele), or heterozygous and informative. In case of tumor tissues, either the reference or variant heterozygous SNP may be entirely lost, duplicated, or amplified in the neoplastic cells. These genetic aberrations result in an increased or decreased frequency of the variant SNP relative to the reference SNP. Because a tumor is an admixture of neoplastic and normal cells, the degree of the increase or decrease depends on the percentage of neoplastic cells. The higher the percentage of neoplastic cells the larger the shift away from the normal heterozygous situation, and if the percentage is more than 95% all targeted SNPs on a chromosome may seem to be homozygous and noninformative.
Normal Variation of B-Allele Frequencies Using Normal Autopsy Brain FFPE Tissues Because of the frequent occurrence and diagnostic and clinical relevance of 1p and 19q deletions in brain tumors, we used glioma tissues to study the performance of SNPbased LOH analysis by NGS. For NGS detection of allelic imbalances, we selected 29 and 16 highly polymorphic SNPs on chromosomes 1p and 19q, respectively, scattered over both chromosomal arms, and compared the results with classic microsatellite-based LOH analysis using five markers on each chromosomal arm. To define suitable cut-off values for interpretation of the data, we analyzed 10 normal autopsy brain FFPE tissues. As expected, homozygous SNPs showed hardly any variation and
print & web 4C=FPO
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
Detection of Chromosomal Loss by NGS
The Journal of Molecular Diagnostics
-
559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 SNP versus Microsatellite-Based LOH Analysis of 580 581 Chromosomes 1p and 19q 582 Figure 3 shows representative results obtained by SNP-based ½F3 583 584 LOH analysis of DNA isolated from a high percentage of 585 neoplastic cells of FFPE astrocytoma and oligodendroglioma 586 tissue. The astrocytoma showed an equal distribution of 587 informative and noninformative SNPs over both chromosomal 588 arms without obvious LOH related to the percentage of 589 neoplastic cells (Figure 3C). Because of the relative low 590 quality of the DNA from FFPE tissue, informative SNPs 591 without relative loss often are not on a straight line at a B-allele 592 frequency of 50%, but mostly are scattered between B-allele 593 594 frequencies of 45% to 55%. Genomic imbalances therefore are called only if the B-allele frequencies of at least two SNPs Q17 595 596 clearly deviate from the 45% and 55% lines. The oligoden597 droglioma clearly showed 1p/19q co-deletion, leading to an 598 599 600 601 602 Figure 2 Distribution of B-allele frequencies of Q23 603 604 single-nucleotide polymorphisms (SNPs) in next605 generation sequencing (NGS) analysis of normal brain tissues. NGS analyses of 10 normal autopsy 606 brain formalin-fixed, paraffin-embedded tissues 607 were performed. The mean B-allele frequencies of 608 chromosome 1p SNPs, which are either homozygous 609 referent, homozygous variant, or heterozygous, are 610 shown. Corresponding standard deviations were 611 determined based on the number of times the SNPs 612 were homozygous referent, homozygous variant, or 613 heterozygous, which may vary per SNP. The x axis 614 shows the chromosomal position in million base 615 pairs (Mbp), and the y axis shows the percentage variant (B-allele frequency). 616 617 618 619 620
are located close to allele frequencies of 0% or 100%, Q16 respectively. In contrast, heterozygous SNPs are more scattered around the expected B-allele frequency of 50%, probably because of the inferior quality of FFPE DNA (Figure 2). ½F2 Therefore, we set cut-off values at 45% and 55% for heterozygous SNPs to discriminate normal from aberrant allele frequencies. One SNP (in red) is located systematically outside these cut-off values, likely owing to preferential amplification of the B-allele and therefore should be interpreted carefully or removed from the panel or bed file. In addition, cut-off values, for homozygous SNPs only, were set at 5% and 95% to help in data analysis. For the same reason and if not otherwise indicated, SNPs with B-allele frequencies between 5% and 45% and 55% and 95%, respectively, are indicated in red dots. SNPs showing other B-allele frequencies are shown in blue, even though at a very high percentage of neoplastic cells aberrant heterozygous SNPs might be located between frequencies of 0% and 5% or 95% and 100%.
jmd.amjpathol.org
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
5
print & web 4C=FPO
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
Dubbink et al
Figure 3
Representative examples of single-nucleotide polymorphism (SNP)-based loss of heterozygosity (LOH) analysis of glioma tissues. Hematoxylin and eosin stain of the corresponding astrocytoma with an estimated neoplastic cell content of 60% (A), and an oligodendroglioma with 90% neoplastic cell histology (B). C: Astrocytoma without obvious genomic alteration of chromosomes 1p (above) and 19q (below). The x axis shows the chromosomal position in million base pairs (Mbp), and the Y axis shows the percentage variant (B-allele frequency). D: Oligodenglioma with typical 1p/19q codeletion. Note the similar degree of LOH (ie, shifts relative to the location of nonaffected informative SNPs) of both chromosome 1p (above) and 19q (below). The x axis shows the chromosomal position in million base pairs (Mbp), and the y axis shows the percentage variant (B-allele frequency). Original magnification: 20 (B and C).
equal decrease or increase of the B-allele frequency of all informative SNPs on both chromosomes (Figure 3D). Microsatellite-based LOH analysis confirmed these observations (data not shown). Of note, the latter method required 10 times more DNA because for analysis of each of the 10 microsatellite markers the same amount of DNA as for NGSbased SNP analysis of both chromosomal arms is needed.
Q24 Q25 Q26
Next, we addressed the sensitivity of SNP-based versus microsatellite-based LOH analysis. DNA from glioma tissue
with known 1p/19q co-deletion and an estimated percentage of neoplastic cells of >90% was diluted with DNA isolated from adjacent near-100% normal tissue and the sensitivity of both LOH detection methods was compared using the same tumor/normal DNA dilutions (Figure 4A). Dilution experi- ½F4 ments were performed in duplicate and also with glioma tissue from another patient (data not shown). The results of one representative experiment of the microsatellite analysis are shown, the second experiment showed similar results. SNP-based LOH analysis was at least as accurate as and even more sensitive than the microsatellite-based method (Figure 4, B and C). Moreover, the data were much easier to interpret in the absence of normal tissue and, importantly, in
6
jmd.amjpathol.org
Highly Sensitive and Easily Interpretable SNP-Based LOH Analysis
-
The Journal of Molecular Diagnostics
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
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
Figure 4
Sensitivity of single-nucleotide polymorphism (SNP)-based compared with microsatellitebased loss of heterozygosity (LOH) analysis. A: Schematic view of serial dilution preparation from oligodendroglioma tissue (>90% neoplastic cells) with known 1p/19q deletion and adjacent normal tissue. SNP- and microsatellitebased LOH analyses were performed in parallel with the same amount of DNA from the same DNA preparations. B: Serial dilution analysis to test the sensitivity of SNP-based LOH detection. Only results of the chromosome 1p analysis are shown. Experiments were performed in duplicate. The results are indicated per experiment by blue dots and red crosses, respectively. The estimated percentages of neoplastic cells are indicated in the graphs. Calculated outcomes of SNP-based LOH analysis of indicated different admixtures of normal and neoplastic cells are indicated by the lines. The x axis shows the chromosomal position in million base pairs (Mbp), and the y axis shows the percentage variant (B-allele frequency). C: Serial dilution analysis to test the sensitivity of microsatellite-based LOH detection. Results with one (D1S513) of five other chromosome 1p markers are shown. Experiments were performed in duplicate, and one representative experiment is shown. Similar results were obtained with other markers (data not shown). Estimated percentages of neoplastic cells are indicated between the graphs. The x axis shows the DNA fragment size in base pairs (bp), and the y axis shows the relative fluorescence unit (RFU).
print & web 4C=FPO
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
Detection of Chromosomal Loss by NGS
The Journal of Molecular Diagnostics
-
jmd.amjpathol.org
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
7
Q27
Q28
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
Dubbink et al 869 contrast to microsatellite-based LOH analysis, there was no 870 need for comparison with normal DNA. For example, at 20% 871 neoplastic cells there was unambiguous LOH of chromosome 872 1p in the SNP assay, whereas LOH of the microsatellite 873 markers was disputable and certainly impossible to determine 874 in the absence of reference normal tissue. Furthermore, at 875 near-100% neoplastic cells the microsatellite marker appar876 ently was noninformative and interpretation required results 877 of matched normal DNA, whereas in the SNP assay the 878 879 absence of all highly polymorphic informative markers was a 880 strong argument for LOH, certainly in cases with an esti881 mated high percentage of neoplastic cells. The experimental 882 data are in accordance with the calculated B-allele fre883 quencies in relation to the neoplastic cell percentage 884 (Figure 4B). Apparently, the NGS method is very repro885 ducible, as can be deduced from the small differences be886 tween the B-allele frequencies of the two experiments shown 887 in Figure 4B. In addition, independent dilution experiments 888 with glioma tissue from another patient showed a high 889 concordance between the separate experiments and a similar 890 891 limit of detection (data not shown). 892 893 Performance and Validation of SNP-Based 1p/19q LOH 894 Analysis of FFPE Glioma Tissues 895 896 The performance of NGS-based LOH analysis was exam897 ined further by analysis of a series of 49 glioma samples 898 collected within a prospective trial between 1997 and 899 2003.19 The estimated percentage of neoplastic cells varied 900 901 ½T3 between 50% and >90% (Table 3). No normal reference adjacent tissue was available. 902 903 In all 49 cases, both microsatellite- and SNP-based LOH 904 analysis made a decision possible on the 1p and 19q status 905 (Table 3). In 39 cases, concordant results were obtained with 906 both methods for both chromosomal arms, and in 10 cases the 907 results were distinct for either 1p or 19q. In nine cases, SNP 908 analysis showed partial or complete 1p or 19q LOH, which 909 was not shown by or was not completely overlapping with 910 microsatellite analysis (Table 3). In one case, glioma 1, 911 unambiguous LOH of two of four informative microsatellite 912 markers on 19q was observed, whereas no partial intra913 chromosomal LOH was shown in the SNP analysis. 914 915 Most of these discordant results (seven cases) could be 916 explained by the observed superior lower limit of detection 917 ½F5 of the SNP analyses (Figure 5). Aberrations as shown in 918 Figure 5 may reflect subclonal allelic losses or aberrant copy 919 numbers of the specific chromosomal regions. Partial LOH 920 in glioma 20 might have been missed because probably only 921 one apparently noninformative microsatellite marker 922 (D19S572) was present in the LOH region. The other two 923 discrepancies (gliomas 1 and 26) involving partial losses are 924 more difficult to explain without extensive research, but 925 might reflect the distinct, nonoverlapping, genomic loca926 927 tions and differences in density of the SNP and microsat928 ellite markers and underlying complex chromosomal 929 rearrangements. 930
8
Discussion We describe here, to our knowledge, a thus far unrecognized diagnostic NGS application based on targeted low-density SNP profiling that allows sensitive detection of CNAs, including allelic imbalances and chromosomal losses or LOH. Our method was developed for the detection of molecular aberrations in neoplastic cells surrounded by distinct numbers of normal cells. A simple algorithm was used to calculate allele frequencies from the relative presence of SNPs in genomic regions of interest in tumor tissues. A ratio less than 45% or more than 55% of consecutive informative SNPs was supposed to be indicative for loss of genetic material or for the presence of aberrant numbers of the Q18 respective alleles (allelic imbalance), respectively. Our main purpose was to develop a diagnostic test for implementation in running NGS tests in molecular pathology laboratories. In that respect, NGS-based LOH analysis has several important advantages over alternative approaches, including microsatellite-based LOH detection and multiplex ligation-dependent probe amplification. Probably because of the quantitative nature of NGS, the method has a higher sensitivity than microsatellite-based LOH analysis and even may detect LOH in neoplastic cells in a background of up to 80% normal cells. The sensitivity of multiplex ligation-dependent probe amplification also is lower because it requires at least 50% neoplastic cells for reliable detection of genomic losses.11 The NGS approach is capable to detect LOH in very small amounts of DNA, ranging from as low as 1 to 10 ng, whereas microsatellite marker analysis requires at least 10 times more DNA input and multiplex ligation-dependent probe amplification greater than 250 ng.11 This makes this SNP-based method very suitable in a routine molecular pathology setting because often only limited amounts of tissue, small biopsy specimens, routine immune- or hematoxylin and eosinestained archival sections, or cytology preparations are available. Because of the small-amplicon design, the NGS method appeared very powerful for investigating relatively low-quality DNA from FFPE tissues. For validation we used multicenter archival FFPE glioma tissues obtained between 1997 and 2003, illustrating the robustness of NGS-based LOH detection.19 The limited amount and low quality of DNA of this material is most likely the reason why frequently no results were obtained with individual markers in the microsatellite analysis (Supplemental Table S1). In addition, detection of LOH by NGS is less dependent on normal reference tissue. This is important because in many diagnostic cases, including brain malignancies, tumor biopsy specimens, and cytology specimens, no adjacent normal tissue is available. For microsatellite analyses, simultaneous analysis of adjacent normal tissue is preferable, especially in cases with borderline percentages of neoplastic cells or if the percentage of neoplastic cells is too high (Figure 4C). Because of a high percentage of
jmd.amjpathol.org
-
The Journal of Molecular Diagnostics
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
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 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
Detection of Chromosomal Loss by NGS Table 3
Parallel Examination of 49 Gliomas by NGS-Based SNP Profiling and Microsatellite Analysis SNP-based LOH
Glioma
Percentage of neoplastic cells
Microsatellite-based LOH
1p
19q
1p
19q
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
80 90 80 80 70 80 85 80 70 70 50 90 60 90 90 70 75 100 90 90 60 90 80 75 90 85 80 80 80 60 90 70 80 70 70 90 80 85 75 90 50 90 75 75 90 50 60 90 80
No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH LOH LOH No LOH LOH LOH LOH No LOH No LOH LOH LOH LOH LOH No LOH LOH LOH LOH LOH LOH LOH LOH LOH No LOH No LOH Partial LOH LOH Partial LOH No LOH LOH No LOH Partial LOH No LOH No LOH Partial LOH LOH No LOH Partial LOH
No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH LOH LOH LOH LOH LOH LOH Partial LOH Partial LOH LOH LOH LOH LOH Partial LOH LOH LOH LOH LOH LOH LOH LOH LOH LOH Partial LOH Partial LOH No LOH LOH LOH LOH LOH LOH LOH Partial LOH LOH LOH LOH LOH
No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH LOH LOH No LOH LOH LOH LOH No LOH No LOH LOH LOH LOH LOH No LOH LOH LOH LOH LOH LOH LOH LOH LOH No LOH No LOH Partial LOH LOH Partial LOH No LOH Partial LOH No LOH Partial LOH No LOH No LOH No LOH LOH No LOH Partial LOH
Partial LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH No LOH LOH LOH LOH LOH LOH LOH No LOH Partial LOH LOH LOH LOH LOH No LOH LOH LOH LOH LOH LOH LOH LOH LOH No LOH No LOH No LOH No LOH Partial LOH LOH LOH LOH LOH LOH No LOH LOH LOH LOH LOH
LOH, loss of heterozygosity; SNP, single-nucleotide polymorphism.
neoplastic cells in many 1p/19q co-deleted cases, almost all microsatellite markers apparently were noninformative, with only one or two markers left per chromosome arm that seemed informative as a result of the presence of a very
The Journal of Molecular Diagnostics
-
weak signal of a putative second alternative allele. Decisions on the presence of LOH therefore often are made based on the assumption that it is unlikely that all microsatellite markers are noninformative when using
jmd.amjpathol.org
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
9
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 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
Figure 5 Examples of partial imbalance as observed in single-nucleotide polymorphism (SNP)-based loss of heterozygosity (LOH) analysis of glioma tissues. Unambiguous (partial) allelic imbalance was observed in case of SNP-based LOH analysis, but not with in-parallel microsatellite-based analysis (data not shown). A: Glioma 46 shows a large telomeric imbalance of chromosome 1p; with classic fluorescent in situ hybridization probes this tumor might have been considered 1p/19q co-deleted. Glioma 36 (B) and glioma 37 (C) show both telomeric imbalances of chromosome 19q. See additional information on these gliomas, including percentages of neoplastic cells, in Table 3 and Supplemental Table S1. The x axis shows the chromosomal position in million base pairs, and the y axis shows the percentage variant (B-allele frequency).
polymorphic microsatellite markers.10 However, for a solid conclusion, adjacent normal reference material, tumor tissue with a lower percentage of neoplastic cells, or blood should be tested in parallel if any of these sources is available. In fact, this also may be true for SNP analyses in exceptional cases with extremely high percentages (>90%) of neoplastic cells. However, the chance that all SNPs are noninformative is much lower because there are more putative informative markers that also are distributed better over the chromosomal arms in the SNP test. Another important feature of targeted NGS-based LOH analysis is that it produces quantitative information that, in contrast to microsatellite analysis, can be used to estimate whether LOH likely has resulted from loss of genetic material or from other CNA phenomena. For this purpose, it is essential to relate the degree of LOH (ie, shifts with respect to nonaffected informative SNPs; Figure 1B) to the estimated percentage of neoplastic cells and, if applicable, the observed frequencies of other oncogenic mutations if the assay is combined with simultaneous mutation detection (see later). This is particularly important for molecular classification of gliomas because it allows discrimination between typical oligodendroglial cases of combined 1p/19q deletion (Figure 3D) and an atypical combined 1p/19q LOH pattern caused by complex genetic changes not specific for oligodendroglioma (data not shown). We have shown that a mean number of 1 SNP per 2 to 4 Mb is sufficient to detect LOH of chromosome 1p and 19q. The test can be adapted easily for analyses of other chromosomal regions or increase the density of SNPs to be able to detect smaller losses in, for example, selected genes. Moreover, the method can be extended for simultaneous detection of mutations in any gene of interest without the need for more DNA input. As an example, we recently showed that molecular classification of gliomas better defines clinically relevant subgroups than histologic grading by combining CNA detection, including chromosomal losses and imbalances, of chromosomes 1p, 7, 10q, and 19q with mutation analysis of, among others, IDH1, IDH2, TP53, PTEN, ATRX, CIC, and FUBP1.22 This analysis currently is part of our routine molecular diagnostics of gliomas. In contrast to microsatellite markers, SNPs are compatible with LOH detection in microsatellite-unstable malignancies. Therefore, another obvious diagnostic application is combined mutation and LOH analysis of microsatellite-unstable tumors, for example, to molecularly classify microsatelliteunstable gliomas or to establish the sporadic nature of microsatellite unstable Lynch Syndromeelike tumors, for which no causal germline aberration has been found.24,28,29 Finally, SNP-based LOH analyses extend coverage analyses as described by other groups14e18 and further enhances the information that can be extracted from NGS data. In addition, it allows LOH detection by means of small NGS panels. Classification of cancers for optimal treatment is driven increasingly by the presence of specific molecular
10
jmd.amjpathol.org
print & web 4C=FPO
1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 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
Dubbink et al
Q29
-
The Journal of Molecular Diagnostics
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
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 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 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
Detection of Chromosomal Loss by NGS
Q19
aberrations, including actionable mutations for precision medicine. Assessment of personalized treatment options would benefit enormously from a comprehensive test that encompasses all essential molecular alterations whether or not for specific tumor types or for the purpose of testing distinct tumor types. NGS rapidly becomes part of standard care delivered by molecular pathology laboratories and allows implementation of single tests to study distinct molecular aberrations that previously could be analyzed only by the use of several methods. Our study adds another option to implement in novel NGS panels and underscores the usefulness of low-density SNP profiling that might be used in parallel with mutation detection and coverage analysis to better address all different types of CNA, including genomic losses, allelic imbalances, and gene amplifications.17 Currently, we are developing a web-based, open-source, diagnostic SNP analysis tool (manuscript in preparation). In summary, we have shown that NGS analysis, apart from detection of mutations and amplifications, is also very suitable and can be diagnostically implemented easily for simultaneous and adequate detection of LOH in archival FFPE tissue. Combined NGS-based LOH and mutation analysis therefore is suited perfectly to become standard practice for routine glioma diagnostics and other diagnostic molecular pathology applications.
4.
5.
6.
7.
8.
9.
Acknowledgments We thank Lotte Douglas-Berger, Gerard Gathier, and Cathleen van der Lee-Haarloo for technical assistance, Monique Oomen and Shazia Arshad for collecting the tissue bank samples, and Frank van der Panne for preparation of the figures.
10.
11.
Supplemental Data Supplemental material for this article can be found at http://dx.doi.org/10.1016/j.jmoldx.2016.06.002.
12.
References 13. 1. Dubbink HJ, Deans ZC, Tops BB, van Kemenade FJ, Koljenovic S, van Krieken HJ, Blokx WA, Dinjens WN, Groenen PJ: Next generation diagnostic molecular pathology: critical appraisal of quality assurance in Europe. Mol Oncol 2014, 8:830e839 2. Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic S, Giaccone G, Jenkins RB, Kwiatkowski DJ, Saldivar JS, Squire J, Thunnissen E, Ladanyi M: Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Mol Diagn 2013, 15:415e453 3. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, Cameron D, Dowsett M, Barrios CH, Steger G, Huang CS, Andersson M, Inbar M, Lichinitser M, Lang I, Nitz U, Iwata H,
The Journal of Molecular Diagnostics
-
14.
15.
16.
Thomssen C, Lohrisch C, Suter TM, Ruschoff J, Suto T, Greatorex V, Ward C, Straehle C, McFadden E, Dolci MS, Gelber RD; Herceptin Adjuvant Trial Study Team: Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 2005, 353:1659e1672 Reifenberger J, Reifenberger G, Liu L, James CD, Wechsler W, Collins VP: Molecular genetic analysis of oligodendroglial tumors shows preferential allelic deletions on 19q and 1p. Am J Pathol 1994, 145:1175e1190 Jenkins RB, Blair H, Ballman KV, Giannini C, Arusell RM, Law M, Flynn H, Passe S, Felten S, Brown PD, Shaw EG, Buckner JC: A t(1; 19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res 2006, 66:9852e9861 Vogazianou AP, Chan R, Backlund LM, Pearson DM, Liu L, Langford CF, Gregory SG, Collins VP, Ichimura K: Distinct patterns of 1p and 19q alterations identify subtypes of human gliomas that have different prognoses. Neuro Oncol 2010, 12:664e678 Intergroup Radiation Therapy Oncology Group Trial 9402, Cairncross G, Berkey B, Shaw E, Jenkins R, Scheithauer B, Brachman D, Buckner J, Fink K, Souhami L, Laperierre N, Mehta M, Curran W: Phase III trial of chemotherapy plus radiotherapy compared with radiotherapy alone for pure and mixed anaplastic oligodendroglioma: Intergroup Radiation Therapy Oncology Group Trial 9402. J Clin Oncol 2006, 24:2707e2714 van den Bent MJ, Brandes AA, Taphoorn MJ, Kros JM, Kouwenhoven MC, Delattre JY, Bernsen HJ, Frenay M, Tijssen CC, Grisold W, Sipos L, Enting RH, French PJ, Dinjens WN, Vecht CJ, Allgeier A, Lacombe D, Gorlia T, Hoang-Xuan K: Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951. J Clin Oncol 2013, 31:344e350 Woehrer A, Sander P, Haberler C, Kern S, Maier H, Preusser M, Hartmann C, Kros JM, Hainfellner JA; Research Committee of the European Confederation of Neuropathological Societies: FISH-based detection of 1p 19q codeletion in oligodendroglial tumors: procedures and protocols for neuropathological practice - a publication under the auspices of the Research Committee of the European Confederation of Neuropathological Societies (Euro-CNS). Clin Neuropathol 2011, 30: 47e55 Hatanpaa KJ, Burger PC, Eshleman JR, Murphy KM, Berg KD: Molecular diagnosis of oligodendroglioma in paraffin sections. Lab Invest 2003, 83:419e428 Jeuken J, Cornelissen S, Boots-Sprenger S, Gijsen S, Wesseling P: Multiplex ligation-dependent probe amplification: a diagnostic tool for simultaneous identification of different genetic markers in glial tumors. J Mol Diagn 2006, 8:433e443 Harada S, Henderson LB, Eshleman JR, Gocke CD, Burger P, Griffin CA, Batista DA: Genomic changes in gliomas detected using single nucleotide polymorphism array in formalin-fixed, paraffinembedded tissue: superior results compared with microsatellite analysis. J Mol Diagn 2011, 13:541e548 Nikiforova MN, Hamilton RL: Molecular diagnostics of gliomas. Arch Pathol Lab Med 2011, 135:558e568 Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, et al: Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol 2013, 31:1023e1031 Boeva V, Popova T, Lienard M, Toffoli S, Kamal M, Le Tourneau C, Gentien D, Servant N, Gestraud P, Rio Frio T, Hupe P, Barillot E, Laes JF: Multi-factor data normalization enables the detection of copy number aberrations in amplicon sequencing data. Bioinformatics 2014, 30:3443e3450 Pritchard CC, Salipante SJ, Koehler K, Smith C, Scroggins S, Wood B, Wu D, Lee MK, Dintzis S, Adey A, Liu Y, Eaton KD, Martins R, Stricker K, Margolin KA, Hoffman N, Churpek JE, Tait JF, King MC, Walsh T: Validation and implementation of targeted capture and sequencing for the detection of actionable mutation, copy number
jmd.amjpathol.org
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
11
1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 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
Dubbink et al 1365 1366 1367 17. 1368 1369 1370 1371 18. 1372 1373 1374 1375 1376 1377 1378 1379 1380 19. 1381 1382 1383 1384 1385 1386 1387 1388 20. 1389 1390 1391 Q20 Q32 1392 21. 1393 1394 1395 1396 22. 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
12
variation, and gene rearrangement in clinical cancer specimens. J Mol Diagn 2014, 16:56e67 Grasso C, Butler T, Rhodes K, Quist M, Neff TL, Moore S, Tomlins SA, Reinig E, Beadling C, Andersen M, Corless CL: Assessing copy number alterations in targeted, amplicon-based nextgeneration sequencing data. J Mol Diagn 2015, 17:53e63 Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, Chandramohan R, Liu ZY, Won HH, Scott SN, Brannon AR, O’Reilly C, Sadowska J, Casanova J, Yannes A, Hechtman JF, Yao J, Song W, Ross DS, Oultache A, Dogan S, Borsu L, Hameed M, Nafa K, Arcila ME, Ladanyi M, Berger MF: Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn 2015, 17:251e264 van den Bent MJ, Carpentier AF, Brandes AA, Sanson M, Taphoorn MJ, Bernsen HJ, Frenay M, Tijssen CC, Grisold W, Sipos L, Haaxma-Reiche H, Kros JM, van Kouwenhoven MC, Vecht CJ, Allgeier A, Lacombe D, Gorlia T: Adjuvant procarbazine, lomustine, and vincristine improves progression-free survival but not overall survival in newly diagnosed anaplastic oligodendrogliomas and oligoastrocytomas: a randomized European Organisation for Research and Treatment of Cancer phase III trial. J Clin Oncol 2006, 24:2715e2722 Federation of Medical Scientific Societies: Code for proper secondary use of human tissue in the Netherlands. Available at https://www.federa. org/sites/default/files/bijlagen/coreon/codepropersecondaryuseofhumant issue1_0.pdf van Lier MG, Wagner A, van Leerdam ME, Biermann K, Kuipers EJ, Steyerberg EW, Dubbink HJ, Dinjens WN: A review on the molecular diagnostics of Lynch syndrome: a central role for the pathology laboratory. J Cell Mol Med 2010, 14:181e197 van den Bent MJ, Dubbink HJ, Sanson M, van der Lee-Haarloo CR, Hegi M, Jeuken JW, Ibdaih A, Brandes AA, Taphoorn MJ, Frenay M, Lacombe D, Gorlia T, Dinjens WN, Kros JM: MGMT promoter
23.
24.
25.
26.
27.
28.
29.
methylation is prognostic but not predictive for outcome to adjuvant PCV chemotherapy in anaplastic oligodendroglial tumors: a report from EORTC Brain Tumor Group Study 26951. J Clin Oncol 2009, 27:5881e5886 Dubbink HJ, Atmodimedjo PN, Kros JM, French PJ, Sanson M, Idbaih A, Wesseling P, Enting R, Spliet W, Tijssen C, Dinjens WN, Gorlia T, van den Bent MJ: Molecular classification of anaplastic oligodendroglioma using next-generation sequencing: a report of the prospective randomized EORTC Brain Tumor Group 26951 phase III trial. Neuro Oncol 2016, 18:388e400 Geurts-Giele WR, Leenen CH, Dubbink HJ, Meijssen IC, Post E, Sleddens HF, Kuipers EJ, Goverde A, van den Ouweland AM, van Lier MG, Steyerberg EW, van Leerdam ME, Wagner A, Dinjens WN: Somatic aberrations of mismatch repair genes as a cause of microsatellite-unstable cancers. J Pathol 2014, 234:548e559 Giardine B, Riemer C, Hardison RC, Burhans R, Elnitski L, Shah P, Zhang Y, Blankenberg D, Albert I, Taylor J, Miller W, Kent WJ, Nekrutenko A: Galaxy: a platform for interactive large-scale genome analysis. Genome Res 2005, 15:1451e1455 Goecks J, Nekrutenko A, Taylor J, Galaxy T: Galaxy: a comprehensive approach for supporting accessible, reproducible, and transparent computational research in the life sciences. Genome Biol 2010, 11:R86 Wang K, Li M, Hakonarson H: ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res 2010, 38:e164 Mensenkamp AR, Vogelaar IP, van Zelst-Stams WA, Goossens M, Ouchene H, Hendriks-Cornelissen SJ, Kwint MP, Hoogerbrugge N, Nagtegaal ID, Ligtenberg MJ: Somatic mutations in MLH1 and MSH2 are a frequent cause of mismatch-repair deficiency in Lynch syndromelike tumors. Gastroenterology 2014, 146:643e646.e8 Haraldsdottir S, Hampel H, Tomsic J, Frankel WL, Pearlman R, de la Chapelle A, Pritchard CC: Colon and endometrial cancers with mismatch repair deficiency can arise from somatic, rather than germline, mutations. Gastroenterology 2014, 147:1308e1316.e1
jmd.amjpathol.org
-
The Journal of Molecular Diagnostics
FLA 5.4.0 DTD JMDI519_proof 23 July 2016 4:14 am EO: JMD15_0282
1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 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