Background:

No consensus guideline has been established for microsatellite instability testing in upper gastrointestinal tract cancers. This study aims to determine whether targeted cancer next-generation sequencing can accurately detect microsatellite instability in upper gastrointestinal tract cancers and screen for patients with Lynch syndrome.

Methods:

In a cohort of 645 upper gastrointestinal tract cancers, targeted next-generation sequencing assessed microsatellite instability by identifying characteristic insertion and deletion mutations. Sequencing classification was compared with mismatch repair protein IHC. Cancers with microsatellite instability by sequencing were analyzed using a testing protocol to identify patients with Lynch syndrome.

Results:

Sequencing identified microsatellite instability in 3.6% (23/645) of upper gastrointestinal tract cancers, including 28% (8/29) of small intestinal and 9% (9/97) of gastric carcinomas. In 20 cancers classified as having microsatellite instability, 19 demonstrated loss of expression of MLH1, PMS2, MSH2, or MSH6, and one cancer was indeterminate by IHC. In contrast, 52 control cancers demonstrated retained expression of all mismatch repair proteins. Using targeted sequencing as the initial screening test, 1.1% (7/645) of patients were identified to have pathogenic germline variants confirming a diagnosis of Lynch syndrome.

Conclusions:

Targeted cancer next-generation sequencing is an accurate first-line test to detect microsatellite instability in upper gastrointestinal tract cancers.

Impact:

This study provides a proof of concept for the use of targeted next-generation sequencing to detect microsatellite instability and screen for Lynch syndrome.

Microsatellite instability defines a characteristic genomic phenotype of insertion and deletion mutations in DNA repeat regions (1). Microsatellite instability is caused by the genetic or epigenetic inactivation of DNA mismatch repair genes MLH1, MSH2, MSH6, or PMS2 and is a feature of some sporadic cancers and cancers associated with Lynch syndrome (2).

Microsatellite instability is most commonly observed in colorectal and endometrial cancers. Although the overall incidence of upper gastrointestinal tract cancers is lower than that of colorectal and endometrial cancers, recent studies evaluating large cancer datasets have identified significant rates of microsatellite instability in cancers of the stomach and small intestine (3–5). Clinical guidelines recommend universal screening for Lynch syndrome in colorectal cancers (6), but no consensus guidelines currently exist for microsatellite instability testing in upper gastrointestinal tract cancers. The accurate identification of microsatellite instability in upper gastrointestinal tract cancers may directly benefit patients in two ways. Microsatellite instability serves as a biomarker to predict response to immune checkpoint inhibitor therapy in solid tumors, including upper gastrointestinal tract cancers (5, 7). Patients who have cancers with microsatellite instability may be at increased risk for Lynch syndrome, and patients and affected family members may benefit from genetic counseling and germline testing (8).

As next-generation sequencing technology becomes clinically available for cancer genotyping to identify actionable driver mutations, we hypothesize that targeted cancer sequencing panels can also detect microsatellite instability and be used as a primary screening tool for Lynch syndrome in upper gastrointestinal tract cancers.

Patients were prospectively enrolled in an institutional cohort study for cancer genotyping. All participants provided written informed consent for tumor sequencing. This study was approved by the Institutional Review Board of the Dana Farber Cancer Institute and the Partners Human Research Committee.

Targeted next-generation sequencing on tumor tissue was performed as described previously (9). DNA was isolated from formalin-fixed, paraffin-embedded (FFPE) tissue with at least 20% tumor nuclei. Indexed sequencing libraries were enriched for exons of 275 genes (102 specimens) or 298 genes (543 specimens), encompassing 757,787 base pairs or 831,033 base pairs of targeted genome, respectively, using solution-based hybrid capture (Agilent SureSelect; Agilent Technologies). Massively parallel sequencing was performed using Illumina HiSeq2500 (Illumina, Inc.). Data analysis was performed using a custom pipeline, including Indelocator (GATK; Broad Institute) for calling insertion and deletion variants.

Microsatellite instability as detected by next-generation sequencing was defined as greater than 3 microsatellite indel events per megabase (Mb) pair in the targeted genome, with events defined as single nucleotide insertion or deletion variants in homopolymeric DNA repeats of four or more nucleotides. Cancers with total microsatellite indel events below this threshold were classified as microsatellite stable. This criterion achieved 96% sensitivity and 99% specificity compared with mismatch repair protein IHC in colorectal adenocarcinomas (10).

IHC for MLH1, PMS2, MSH2, and MSH6 protein expression was performed as a case–control study. We tested all samples classified to have microsatellite instability with available pathology material. For each case sample predicted to have microsatellite instability, at least two control samples classified as microsatellite stable, matched by diagnosis, were tested. In addition, IHC was performed on all samples with greater than 2 but fewer than 3 microsatellite indel events per megabase if tissue was available. IHC and MLH1 promoter methylation analyses were performed per standard laboratory protocol as described previously (11).

Subsequent germline sequencing was performed using the same targeted next-generation sequencing assay on DNA isolated from nonneoplastic, FFPE tissue. The assay included coverage of coding regions of mismatch repair genes MLH1, PMS2, MSH2, and MSH6.

Patient characteristics

A total of 645 upper gastrointestinal tract carcinomas were sequenced, including 230 from esophagus or gastroesophageal junction, 199 from pancreas, 97 from stomach, 60 from bile duct, 29 from small intestine, 19 from gallbladder, and 11 from ampulla of Vater. A total of 426 patients (66%) were male, and 219 (34%) were female. The median age at the time of testing was 65.3 years (mean 64.2 years, range 19.0–93.6 years).

Detection of microsatellite instability in upper gastrointestinal tract cancers by targeted sequencing

Microsatellite instability was identified by next-generation sequencing in 23 of 645 (3.6%) upper gastrointestinal tract cancers. Cancers classified to have microsatellite instability had a median of 9.2 microsatellite indel events per megabase (mean 10.9, range 3.4–22.4). A total of 555 of 622 (89.2%) cancers classified as microsatellite stable had 0 microsatellite indel events. Fifty-five of 622 (8.8%) cancers classified as microsatellite stable had between 0 and 2 microsatellite indel events, and 12 of 622 (1.9%) had between 2 and 3 microsatellite indel events (Fig. 1A). Microsatellite instability was seen in 8 of 29 small intestinal (28%), 9 of 97 gastric (9%), 1 of 60 biliary tract (2%), 3 of 230 esophageal or gastroesophageal junction (1.3%), 2 of 199 pancreatic (1.0%), 0 of 19 gallbladder, and 0 of 11 ampullary cancers (Fig. 1B).

Figure 1.

Detection of microsatellite instability by next-generation sequencing. A, Number of microsatellite insertion or deletion events per megabase in upper gastrointestinal tract cancers. Cancers with greater than three events per megabase were classified as having microsatellite instability. B, Frequency of microsatellite instability in upper gastrointestinal tract cancers, by site. C, Validation of microsatellite instability classification by sequencing compared with IHC.

Figure 1.

Detection of microsatellite instability by next-generation sequencing. A, Number of microsatellite insertion or deletion events per megabase in upper gastrointestinal tract cancers. Cancers with greater than three events per megabase were classified as having microsatellite instability. B, Frequency of microsatellite instability in upper gastrointestinal tract cancers, by site. C, Validation of microsatellite instability classification by sequencing compared with IHC.

Close modal

Validation of results with mismatch repair protein IHC

Twenty of 23 cases classified to have microsatellite instability had available tumor tissue for IHC. Nineteen of 20 (95%) showed loss of expression of at least one mismatch repair protein including 14 with loss of MLH1 and PMS2, 2 with loss of PMS2, 2 with loss of MSH2 and MSH6, and 1 with loss of MSH6. The final case showed an unusual pattern, with nuclear staining of 5% of tumor nuclei for all four mismatch repair proteins, and was interpreted as indeterminate.

In contrast, all 45 controls classified as microsatellite stable by sequencing showed intact staining for MLH1, PMS2, MSH2, and MSH6 (Fig. 1C). In addition, all 7 cases with between 2 and 3 microsatellite indel events per megabase (classified as microsatellite stable but near the threshold) and with available tissue showed intact staining for MLH1, PMS2, MSH2, and MSH6.

Identification of Lynch syndrome in patients with upper gastrointestinal tract cancers

For cases with microsatellite instability and available matched normal tissue, we followed a testing algorithm commonly applied to colorectal cancers to identify patients with Lynch syndrome (Fig. 2). For cancers with loss of MLH1 and PMS2 expression, MLH1 promoter methylation analysis was performed. Of 10 neoplasms with MLH1 and PMS2 loss and successful promoter methylation analysis, MLH1 promoter methylation was identified in 7.

Figure 2.

Study protocol for the evaluation of upper gastrointestinal tract cancers to identify patients with Lynch syndrome. Targeted next-generation sequencing was performed on 645 upper gastrointestinal tract cancers. Confirmatory IHC was performed on cancers classified as having microsatellite instability, and MLH1 promoter methylation analysis was tested for cancers with loss of MLH1 and PMS2 expression. Germline testing was performed on cases suspicious for Lynch syndrome–associated cancers, where nonneoplastic tissue was available.

Figure 2.

Study protocol for the evaluation of upper gastrointestinal tract cancers to identify patients with Lynch syndrome. Targeted next-generation sequencing was performed on 645 upper gastrointestinal tract cancers. Confirmatory IHC was performed on cancers classified as having microsatellite instability, and MLH1 promoter methylation analysis was tested for cancers with loss of MLH1 and PMS2 expression. Germline testing was performed on cases suspicious for Lynch syndrome–associated cancers, where nonneoplastic tissue was available.

Close modal

Nonneoplastic tissue for germline testing was available for 12 of 16 specimens with microsatellite instability and without MLH1 promoter methylation. Pathogenic Lynch syndrome variants were identified in 7 patients, representing 1.1% of upper gastrointestinal tract cancers in our cohort, including 4 small intestinal, 1 esophageal, 1 stomach, and 1 pancreatic cancers (Table 1; Supplementary Data). Patients with Lynch syndrome had a median age of 69.6 years (range 49.8–74.6 years). Upper gastrointestinal tract cancer was the first clinical manifestation of Lynch syndrome in 6 of 7 patients. Colonic adenocarcinoma was the first Lynch syndrome–associated cancer in the other patient.

Table 1.

A total of 23 upper gastrointestinal tract carcinomas classified as mismatch repair deficient by targeted panel sequencing, validation with IHC, and identification of pathogenic germline variants to diagnose Lynch syndrome

IHC
SiteMicrosatellite events per MbMLH1PMS2MSH2MSH6MLH1 PromoterGermline variantGermline VAF or log2 ratioCancer VAF or log2 ratioSomatic variantCancer VAF or log2 ratio
Stomach 12.6 Loss Loss Intact Intact Methylated Not indicated     
Stomach 9.2 Loss Loss Intact Intact Methylated Not indicated     
Stomach 6.9 Loss Loss Intact Intact Methylated Not indicated     
Stomach 3.4 Loss Loss Intact Intact Methylated Not indicated     
Stomach 10.3 Loss Loss Intact Intact Unmethylated Wild-type   MLH1 deletion −0.7 
Stomach 4.6 Loss Loss Intact Intact Unmethylated MLH1 exon 13 del −0.7 −1.6   
Stomach 19.5 Loss Loss Intact Intact Failed Wild-type     
Stomach 9.2 Indeterminate Indeterminate Indeterminate Indeterminate Unmethylated Wild-type     
Stomach 13.7 Not available Not available Not available Not available Not available Not available     
Small intestine 18.3 Loss Loss Intact Intact Methylated Not indicated     
Small intestine 16.0 Loss Loss Intact Intact Methylated Not indicated     
Small intestine 5.7 Loss Loss Intact Intact Unmethylated MLH1 c.2194_2197dup (p.H733Qfs*14) 45% 43%   
Small intestine 11.5 Intact Loss Intact Intact Not indicated PMS2 c.137G>T (p.S46I) 51% 45% PMS2 c.23+1G>T 22% 
Small intestine 8.0 Intact Loss Intact Intact Not indicated PMS2 c.1500dup (p.V501Rfs*4) 58% 45%   
Small intestine 22.4 Intact Intact Loss Loss Not indicated Wild-type   MSH2 c.2581C>T (p.Q861*) 77% 
Small intestine 7.9 Intact Intact Loss Loss Not indicated MSH2 c.1786_1788del (p.N596del) 48% 43%   
Small intestine 6.9 Loss Loss Intact Intact Not available Not available     
Pancreas 5.7 Intact Intact Intact Loss Not indicated MSH2 c.1906G>C (p.A636P) 48% 54% MSH2 c.622G>T (p.G208*) 8% 
Pancreas 9.2 Loss Loss Intact Intact Not available Not available     
Esophagus 16.0 Loss Loss Intact Intact Methylated Not indicated     
Esophagus 14.9 Loss Loss Intact Intact Failed Wild-type     
Esophagus 14.9 Not available Not available Not available Not available Not available MLH1 c.2059C>T (p.R687W) 47% 51% MLH1 c.298C>T (p.R100*) 40% 
Bile duct 4.6 Not available Not available Not available Not available Not available Not available     
IHC
SiteMicrosatellite events per MbMLH1PMS2MSH2MSH6MLH1 PromoterGermline variantGermline VAF or log2 ratioCancer VAF or log2 ratioSomatic variantCancer VAF or log2 ratio
Stomach 12.6 Loss Loss Intact Intact Methylated Not indicated     
Stomach 9.2 Loss Loss Intact Intact Methylated Not indicated     
Stomach 6.9 Loss Loss Intact Intact Methylated Not indicated     
Stomach 3.4 Loss Loss Intact Intact Methylated Not indicated     
Stomach 10.3 Loss Loss Intact Intact Unmethylated Wild-type   MLH1 deletion −0.7 
Stomach 4.6 Loss Loss Intact Intact Unmethylated MLH1 exon 13 del −0.7 −1.6   
Stomach 19.5 Loss Loss Intact Intact Failed Wild-type     
Stomach 9.2 Indeterminate Indeterminate Indeterminate Indeterminate Unmethylated Wild-type     
Stomach 13.7 Not available Not available Not available Not available Not available Not available     
Small intestine 18.3 Loss Loss Intact Intact Methylated Not indicated     
Small intestine 16.0 Loss Loss Intact Intact Methylated Not indicated     
Small intestine 5.7 Loss Loss Intact Intact Unmethylated MLH1 c.2194_2197dup (p.H733Qfs*14) 45% 43%   
Small intestine 11.5 Intact Loss Intact Intact Not indicated PMS2 c.137G>T (p.S46I) 51% 45% PMS2 c.23+1G>T 22% 
Small intestine 8.0 Intact Loss Intact Intact Not indicated PMS2 c.1500dup (p.V501Rfs*4) 58% 45%   
Small intestine 22.4 Intact Intact Loss Loss Not indicated Wild-type   MSH2 c.2581C>T (p.Q861*) 77% 
Small intestine 7.9 Intact Intact Loss Loss Not indicated MSH2 c.1786_1788del (p.N596del) 48% 43%   
Small intestine 6.9 Loss Loss Intact Intact Not available Not available     
Pancreas 5.7 Intact Intact Intact Loss Not indicated MSH2 c.1906G>C (p.A636P) 48% 54% MSH2 c.622G>T (p.G208*) 8% 
Pancreas 9.2 Loss Loss Intact Intact Not available Not available     
Esophagus 16.0 Loss Loss Intact Intact Methylated Not indicated     
Esophagus 14.9 Loss Loss Intact Intact Failed Wild-type     
Esophagus 14.9 Not available Not available Not available Not available Not available MLH1 c.2059C>T (p.R687W) 47% 51% MLH1 c.298C>T (p.R100*) 40% 
Bile duct 4.6 Not available Not available Not available Not available Not available Not available     

NOTE: Log2 ratio, relative copy number, where 0 represents copy number neutral and −1 represents one copy loss in a specimen where 100% of cells are affected.

Abbreviation: VAF, variant allele fraction. NOTE: Pathogenic variants identified by germline or somatic sequencing are highlighted in bold.

Somatic and second hit mutations in upper gastrointestinal tract cancers with microsatellite instability

We evaluated the tumor sequencing data to identify somatic second hit mutations in mismatch repair genes in patients with Lynch syndrome. In 3 of 7 patients with Lynch syndrome, a second loss-of-function mutation was identified in the tumor specimen. These mutations included a splice site variant and two nonsense variants. In 1 patient with an isolated MLH1 exon 13 germline deletion, the tumor specimen showed exon 13 deletion involving both copies of MLH1, and the second hit in this cancer was most likely due to loss of heterozygosity of the MLH1 gene locus. In the other 3 patients with Lynch syndrome, a second somatic mutation was not identified, and the observed variant allele fractions in tumor specimens did not support LOH. This result might be due to limitations of the study, including possibly limited ability to detect loss of heterozygosity in relatively low tumor purity conditions or the presence of sequence or structural alterations in noncoding regions of the gene, which were not assessed by this assay. In two upper gastrointestinal tract cancers with microsatellite instability, the full testing algorithm was completed, and germline testing did not identify a pathogenic Lynch syndrome variant. Tumor testing showed somatic inactivation of mismatch repair genes by mutation or focal gene deletion (Table 1).

Next-generation sequencing has emerged as an effective diagnostic tool in cancer care and is being rapidly adopted into clinical practice (12). In addition to the identification of oncogenic driver mutations, sequencing can identify patterns of passenger mutations associated with microsatellite instability. Multiple algorithms have been developed to identify microsatellite instability from sequencing assays (13–15), including from targeted panel sequencing of tumor only specimens (10, 16). Recently, tumor sequencing has been suggested as a replacement for traditional Lynch syndrome screening methods in colorectal cancer (17).

Compared with colorectal cancer, Lynch syndrome screening in the upper gastrointestinal tract faces practical and diagnostic challenges. Many upper gastrointestinal tract cancers are diagnosed from fine needle aspiration cytologic preparations or small biopsies, and patients are frequently treated with neoadjuvant therapy before surgical resection. These diagnostic and treatment patterns limit the availability of diagnostic tumor tissue and make accurate interpretation by IHC methods more difficult. In this setting, targeted cancer sequencing is an appealing alternative for microsatellite instability evaluation. The next-generation sequencing assay used in this study has been validated to be performed on as little as 50 ng of tumor-enriched input DNA.

Our findings demonstrate accurate microsatellite instability assessment in upper gastrointestinal tract cancers by sequencing, and our protocol identifies microsatellite instability in 3.6% and pathogenic germline Lynch syndrome variants in 1.1% of upper gastrointestinal tract cancers in this cohort. Although the frequency of microsatellite instability is lower than that of colorectal cancers, the overall rates of microsatellite instability and Lynch syndrome in upper gastrointestinal tract cancers are clinically significant. Microsatellite instability is a biomarker for response of solid tumors to immune checkpoint inhibitor therapy (5, 18), and the identification of microsatellite instability provides a treatment option for patients who have failed other systemic therapies. In addition, the ability to screen for microsatellite instability has implications for a subset of patients with upper gastrointestinal tract cancers and Lynch syndrome. Our study has identified 7 patients with Lynch syndrome in an unselected cohort. These patients are of similar age compared with patients with sporadic upper gastrointestinal tract cancers. Notably, three of seven pathogenic Lynch syndrome variants are missense variants, and two variants involve PMS2. Genetic alterations involving PMS2 have been associated with a moderately increased risk of colorectal and endometrial cancers, and the significance of PMS2-associated Lynch syndrome in cancer risk at other sites is controversial (19).

Patients with Lynch syndrome and affected family members may benefit from enhanced surveillance for the prevention for additional primary cancers, including increased frequency of colonoscopy screening, consideration of prophylactic hysterectomy for women older than 40 years, and other emerging screening strategies (20). These potential benefits are in addition to the detection of driver oncogenic alterations, which may provide druggable targets in upper gastrointestinal tract cancers or determine eligibility in clinical trials (21, 22).

Our findings support the use of targeted cancer sequencing as a first-line screening test in upper gastrointestinal tract cancers to identify microsatellite instability and patients with Lynch syndrome. In laboratories already performing panel sequencing to identify driver mutations, the adoption of a similar protocol may benefit patients with upper gastrointestinal tract cancers.

M.B. Yurgelun reports receiving commercial research grant from Myriad Genetic Laboratories Inc. L.M. Sholl is a consultant/advisory board member for AstraZeneca, LOXO Oncology, and Foghorn Therapeutics. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A.G. Christakis, N.I. Lindeman, L.E. MacConaill, F. Dong

Development of methodology: A.G. Christakis, N.I. Lindeman, L.E. MacConaill, F. Dong

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.G. Christakis, D.J. Papke, N.I. Lindeman, L.E. MacConaill, F. Dong

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.G. Christakis, D.J. Papke, M.B. Yurgelun, A.T. Agoston, N.I. Lindeman, L.E. MacConaill, L.M. Sholl, F. Dong

Writing, review, and/or revision of the manuscript: A.G. Christakis, D.J. Papke, J.A. Nowak, M.B. Yurgelun, N.I. Lindeman, L.E. MacConaill, L.M. Sholl, F. Dong

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.G. Christakis, N.I. Lindeman, F. Dong

Study supervision: F. Dong

This work was supported by the Brigham and Women's Hospital Department of Pathology and the Dana Farber Cancer Institute.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Thibodeau
SN
,
Bren
G
,
Schaid
D
. 
Microsatellite instability in cancer of the proximal colon
.
Science
1993
;
260
:
816
9
.
2.
Lynch
HT
,
Snyder
CL
,
Shaw
TG
,
Heinen
CD
,
Hitchins
MP
. 
Milestones of Lynch syndrome: 1895–2015
.
Nat Rev Cancer
2015
;
15
:
181
94
.
3.
Hause
RJ
,
Pritchard
CC
,
Shendure
J
,
Salipante
SJ
. 
Classification and characterization of microsatellite instability across 18 cancer types
.
Nat Med
2016
;
22
:
1342
50
.
4.
Bonneville
R
,
Krook
MA
,
Kautto
EA
,
Miya
J
,
Wing
MR
,
Chen
HZ
, et al
Landscape of microsatellite instability across 39 cancer types
.
JCO Precis Oncol
2017
;
2017
:
1
15
.
5.
Le
DT
,
Durham
JN
,
Smith
KN
,
Wang
H
,
Bartlett
BR
,
Aulakh
LK
, et al
Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade
.
Science
2017
;
357
:
409
13
.
6.
Giardiello
FM
,
Allen
JI
,
Axilbund
JE
,
Boland
CR
,
Burke
CA
,
Burt
RW
, et al
Guidelines on genetic evaluation and management of Lynch syndrome: a consensus statement by the U.S. Multi-Society Task Force on colorectal cancer
.
Gastroenterology
2014
;
147
:
502
26
.
7.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
8.
Engel
C
,
Loeffler
M
,
Steinke
V
,
Rahner
N
,
Holinski-Feder
E
,
Dietmaier
W
, et al
Risks of less common cancers in proven mutation carriers with Lynch syndrome
.
J Clin Oncol
2012
;
30
:
4409
15
.
9.
Garcia
EP
,
Minkovsky
A
,
Jia
Y
,
Ducar
MD
,
Shivdasani
P
,
Gong
X
, et al
Validation of OncoPanel: a targeted next-generation sequencing assay for the detection of somatic variants in cancer
.
Arch Pathol Lab Med
2017
;
141
:
751
8
.
10.
Papke
DJ
,
Nowak
JA
,
Yurgelun
MB
,
Frieden
A
,
Srivastava
A
,
Lindeman
NI
, et al
Validation of a targeted next-generation sequencing approach to detect mismatch repair deficiency in colorectal adenocarcinoma
.
Mod Pathol
2018
;
31
:
1882
90
.
11.
Nowak
JA
,
Yurgelun
MB
,
Bruce
JL
,
Rojas-Rudilla
V
,
Hall
DL
,
Shivdasani
P
, et al
Detection of mismatch repair deficiency and microsatellite instability in colorectal adenocarcinoma by targeted next-generation sequencing
.
J Mol Diagn
2017
;
19
:
84
91
.
12.
Nagarajan
R
,
Bartley
AN
,
Bridge
JA
,
Jennings
LJ
,
Kamel-Reid
S
,
Kim
A
, et al
A window into clinical next-generation sequencing-based oncology testing practices
.
Arch Pathol Lab Med
2017
;
141
:
1679
85
.
13.
Niu
B
,
Ye
K
,
Zhang
Q
,
Lu
C
,
Xie
M
,
McLellan
MD
, et al
MSIsensor: microsatellite instability detection using paired tumor-normal sequence data
.
Bioinformatics
2014
;
30
:
1015
6
.
14.
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
.
15.
Kautto
EA
,
Bonneville
R
,
Miya
J
,
Yu
L
,
Krook
MA
,
Reeser
JW
, et al
Performance evaluation for rapid detection of pan-cancer microsatellite instability with MANTIS
.
Oncotarget
2017
;
8
:
7452
63
.
16.
Salipante
SJ
,
Scroggins
SM
,
Hampel
HL
,
Turner
EH
,
Pritchard
CC
. 
Microsatellite instability detection by next generation sequencing
.
Clin Chem
2014
;
60
:
1192
9
.
17.
Hampel
H
,
Pearlman
R
,
Beightol
M
,
Zhao
W
,
Jones
D
,
Frankel
WL
, et al
Assessment of tumor sequencing as a replacement for Lynch syndrome screening and current molecular tests for patients with colorectal cancer
.
JAMA Oncol
2018
;
4
:
806
13
.
18.
Lemery
S
,
Keegan
P
,
Pazdur
R
. 
First FDA approval agnostic of cancer site - when a biomarker defines the indication
.
N Engl J Med
2017
;
377
:
1409
12
.
19.
Ten Broeke
SW
,
van der Klift
HM
,
Tops
CMJ
,
Aretz
S
,
Bernstein
I
,
Buchanan
DD
, et al
Cancer risks for PMS2-associated Lynch syndrome
.
J Clin Oncol
2018
;
36
:
2961
8
.
20.
Vasen
HFA
,
Blanco
I
,
Aktan-Collan
K
,
Gopie
JP
,
Alonso
A
,
Aretz
S
, et al
Revised guidelines for the clinical management of Lynch syndrome (HNPCC): recommendations by a group of European experts
.
Gut
2013
;
62
:
812
23
.
21.
Bang
YJ
,
Van Cutsem
E
,
Feyereislova
A
,
Chung
HC
,
Shen
L
,
Sawaki
A
, et al
Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial
.
Lancet
2010
;
376
:
687
97
.
22.
Schwaederle
M
,
Zhao
M
,
Lee
JJ
,
Lazar
V
,
Leyland-Jones
B
,
Schilsky
RL
, et al
Association of biomarker-based treatment strategies with response rates and progression-free survival in refractory malignant neoplasms: a meta-analysis
.
JAMA Oncol
2016
;
2
:
1452
9
.

Supplementary data