You are on page 1of 7

REVIEW ARTICLE

Molecular Pathology of Colorectal Cancer


Shuko Harada, MD and Diana Morlote, MD

MOLECULAR MECHANISMS OF COLORECTAL


Abstract: Colorectal cancer (CRC) is the third most commonly CARCINOGENESIS
diagnosed cancer. This review gives an overview of the current
knowledge of molecular mechanisms of colorectal carcinogenesis and Chromosomal Instability
the role of molecular testing in the management of CRC. The
majority of CRCs arise from precursor lesions such as adenoma,
The CIN pathway, which accounts for ~85% of CRCs, is
transforming to adenocarcinoma. Three molecular carcinogenesis characterized by alteration of the number and structure of
pathways have been identified; (1) chromosomal instability, (2) chromosomes, such as loss of chromosome 17p and 18q,
microsatellite instability (MSI), and (3) CpG island methylator phe- leading to aneuploidy (an abnormal chromosome number),
notype, each account for ~85%, 15%, and 17%, respectively. Evalu- subkaryotypic amplification, chromosomal rearrangement,
ation of MSI status, extended RAS mutation analysis, and BRAF and loss of heterozygosity at tumor suppressor gene loci.4,8 In
mutation analysis are recommended by the guideline published by addition, CIN tumors accumulate mutations in oncogenes and
joint effort from professional societies. MSI testing is important for tumor suppressor genes including APC, TP53, KRAS, and
identification of Lynch syndrome patients and prognostic and pre- BRAF.4 According to the well-accepted Vogelstein’s model,
dictive markers. Extended RAS testing is an important predictive
marker for antiepidermal growth factor receptor therapy. BRAF p.
inactivation of APC occurs first in the hyperproliferative epi-
V600 mutation status can be used as prognostic marker, but not thelium, followed by oncogenic mutations, most commonly
predictive marker for antiepidermal growth factor receptor therapies. KRAS, and eventually chromosomal alteration and inactiva-
Emerging technologies utilizing high throughput sequencing have tion of the tumor suppressor gene TP53.9 Typically, these
introduced novel biomarkers and testing strategies. Tumor mutation changes lead to the classic adenoma-carcinoma sequence. The
burden predicts immunotherapy response in addition to MSI status. Cancer Genome Atlas Network (TCGA) analysis found gains
Liquid biopsy can be utilized when adequate tissue sample is not of 1q, 7p, 7q, 8p, 8q, 12q, 13q, 19q, 20p, and 20q and loss of
available or for monitoring therapy response. However, assay stand- 1p, 4q, 5q, 8q, 14q, 20p, and 22q in CIN tumors.10 The study
ardization and guidelines and recommendations for utilization of also found 24 genes significantly mutated; in addition to the
these assay will be needed. The advancement in CRC research and
genes expected APC, TP53, SMAD4, PIK3CA and KRAS,
technologies will allow better prognostication and therapy strat-
ification for the management of patients with CRCs. frequent mutations were found in ARID1A, SOX9, and
FAM123B.10
Key Words: colorectal carcinoma, microsatellite instability (MSI), The most clinically relevant pathways involved in CIN
extended RAS, BRAF mutation, Lynch syndrome tumors are the Wnt and MAPK (mitogen-activating protein
kinase) pathways.11 The Wnt pathway is activated when the
(Adv Anat Pathol 2020;27:20–26)
Wnt protein binds to the Frizzled family receptor, which
removes Axin from the “destruction complex.” This leads to
β-catenin to accumulate in the nucleus and activates tran-

C olorectal cancer (CRC) is the third most commonly


diagnosed cancer and the third leading cause among all
cancer deaths in the United States, although the incidence has
scription. When the signal is not activated, β-catenin is
degraded by APC-dependent destruction complex in the
cytoplasm. The Wnt signaling pathway was altered in 93%
been decreasing.1 Globally, CRC is the second and third of all tumors including biallelic inactivation of APC or
leading cause of cancer death in men and women, respectively.2 activation of CTNNB1 (β-catenin).10 These changes lead to
The vast majority of CRC develop sporadically, whereas <10% uncontrolled nuclear accumulation of β-catenin and cell
of cases result from a hereditary cancer syndrome.3 The proliferation. MAPK pathway is activated by a receptor
majority of CRCs arise from precursor lesions such as ade- tyrosine kinase. Activating mutation in one of the signaling
noma, transforming to adenocarcinoma. Three molecular car- molecules involved in this pathway, such as RAS, RAF, or
cinogenesis pathways have been identified; (1) chromosomal ERK, leads to the signaling molecule becoming con-
instability (CIN),4 (2) microsatellite instability (MSI),5,6 and (3) stitutively active, results in uncontrolled cell proliferation.
CpG island methylator phenotype (CIMP).7 Prognosis is
determined by multiple factors including TNM stage, location Microsatellite Instability
of the tumor, and histologic subtypes. In addition, there are MSI, which accounts for about 15% of CRCs, is
molecular biomarkers that predict prognosis and therapy characterized by generalized instability of short tandemly-
response. This review gives an overview of the current knowl- repeated DNA sequences known as microsatellites.12 MSI
edge of molecular mechanisms of colorectal carcinogenesis and may result from either mutation of one of mismatch repair
the role of molecular testing in the management of CRC. (MMR) genes, MLH1, MSH2, MSH6, or PMS2, or
silencing of the MLH1 promoter by hypermethylation.
Normally, when 2 strands of DNA replicate and nucleotide
From The University of Alabama at Birmingham, Birmingham, AL. mismatch occur, these errors are corrected by a MMR
The authors have no funding or conflicts of interest to disclose. enzyme. However, defects in this function result in a high
Reprints: Shuko Harada, MD, Department of Pathology, The University
of Alabama at Birmingham, 1802 6th Avenue South, WP P210E,
frequency of replication errors because of the slippage of the
Birmingham, AL 35249 (e-mail: sharada@uabmc.edu). DNA polymerase.3,8 Lynch syndrome is the most common
Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved. hereditary colon cancer syndrome, which is characterized by

20 | www.anatomicpathology.com Adv Anat Pathol  Volume 27, Number 1, January 2020


Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Adv Anat Pathol  Volume 27, Number 1, January 2020 Molecular Pathology of Colorectal Cancer

MSI.5 Lynch syndrome is associated with germline muta- MLH1 defect (Fig. 1). Only a small percentage of MLH1 loss
tions in one of the MMR genes and increases lifetime risk of tumors are because of Lynch syndrome, that is, because of
CRC to about 80% with early onset.5 Sporadic MSI tumors germline MLH1 mutation, whereas the majority of them result
can occur because of methylation of CpG-rich promoter from silencing of MLH1 expression because of promoter
sequence of MLH1. These cancers tend to arise in proximal hypermethylation.19 Hypermethylation of the MLH1 promoter
colon, tend to exhibit poor differentiation, mucinous cell is often a characteristic of CIMP tumors. These tumors often
type, and prominent lymphocytic infiltration.3 MSI tumors harbor a BRAF p.V600E mutation. BRAF p.V600 mutational
with hypermethylation account for three-quarters of analysis can be performed ahead of MLH1 promoter methyl-
hypermutated CRCs, whereas a quarter had somatic MMR ation analysis in tumors with MLH1 loss. The presence of a
gene mutation and polymerase ε mutations.10 BRAF mutation strongly favors sporadic pathogenesis.17 In
addition, BRAF mutation is associated with poor prognosis,
CpG Island Methylator Phenotype especially in CIMP-low tumors.13 The absence of a BRAF
The CIMP pathway is characterized by widespread mutation does not exclude sporadic etiology and requires pro-
hypermethylation of numerous promoter CpG island loci and moter methylation analysis to exclude a diagnosis of Lynch
consequent inactivation of tumor suppressor genes.7 CIMP syndrome. Loss of other proteins (MSH2, MSH6, or PMS2)
tumors were initially identified in 30% to 35% of CRCs, but increases a probability for Lynch syndrome and genetic coun-
DNA methylation analysis of CIMP-specific promoters seling and germline gene sequencing is recommended (Table 1).
revealed that the CIMP pathway accounts for 17% of CRC.13 Germline mutations are typically nonsense or frameshift muta-
Although CIN and MSI pathways are usually exclusive,10 the tions, which result in loss of function. A hit to the second allele
CIMP pathway overlaps substantially with the MSI pathway. (second hit), as a result of either additional loss-of-function
In fact, sporadic MSI CRCs are almost exclusively associated mutation or loss of heterozygosity, silences the function of the
with CIMP-associated methylation of the MLH1 promoter MMR gene and results in tumor formation.
region.14 CIMP-positive tumors are shown to represent a Microsatellite Instability Testing by Polymerized
distinct subset with high BRAF mutation.13,14 CIMP has a
strong association with the serrated neoplasia.15 CIMP Chain Reaction
tumors tend to arise in the proximal colon, at an older age, MSI can be determined by a PCR-based assay. MSI is
and are more common in female individuals.16 defined as alterations in the lengths of microsatellites, also
referred as short tandem repeats, because of deletion or inser-
tion of repeating units to produce novel-length alleles in tumor
MICROSATELLITE INSTABILITY TESTING DNA when compared with the normal/germline DNA from
the same individual.20 There are > 1 million microsatellite loci
MMR Deficiency by Immunohistochemistry (IHC) throughout the genome.21 To standardize MSI analysis, the
Testing for MMR deficiency (dMMR) can be performed reference panel, referred to as the Bethesda panel, for the
by IHC for the 4 major MMR proteins (MLH1, MSH2, PMS2, detection of MSI was proposed by a 1997 National Cancer
and MSH6) or by MSI polymerase chain reaction (PCR)-based Institute (NCI) workshop and consists of 5 microsatellite
assay.17,18 Initial screening can be performed by either method- markers, 2 mononucleotide loci [big adenine tract (BAT)-25
ology depending on the availability. In general, IHC does not and BAT-26) and 3 dinucleotide loci (D2S123, D5S346, and
require a special laboratory to perform and can be done at low D17S250).22 Commercial assay kits have become available
cost. In addition, the assay can help screen for a possible since then, including the one from Promega Corp. (Madison,
defective gene. Therefore, MMR IHC is generally preferred. WI), which uses 5 nearly monomorphic mononucleotide
However, limitations include the fact that not all pathogenic microsatellite loci (BAT-25, BAT-26, NR-21, NR-24, and
mutations result in loss of expression and interpretation is MONO-27).20 The performance of these assays is similar, but
somewhat subjective.15 For the IHC assay, if all proteins are mononucleotide loci are shown to be more sensitive and specific
expressed in the nucleus, the tumor is considered microsatellite than dinucleotide loci.23 An MSI-H (high) tumor is defined as
stable (MSS). Loss of 1 or 2 protein expression indicates MMR the one that shows shift of the size in ≥ 2 out of 5 microsatellite
deficiency, which highly correlates with MSI. As MLH1/PMS2 loci, whereas a shift only at 1 locus is considered MSI-L (low).21
and MSH2/MSH6 form functional pairs and MLH1 and MSH2
are needed to stabilize the complex, when MLH1 or MSH2 are Emerging Technologies
lost, PMS2 or MSH6 are also lost (Table 1). The most common Recently, cartridge-based automated PCR assay was
MMR-deficient pattern is the loss of MLH1/PMS2 because of developed by Biocartis (Mechelen, BE) (Idylla MSI). The

TABLE 1. Interpretation of Mismatch Repair Protein Immunohistochemistry Test Results and Further Action
Nuclear Expression of Protein

MLH1 PMS2 MSH2 MSH6 Interpretation Action


+ + + + MSS No further action
− − + + MSI, MLH1 loss MLH1 promoter heypermethylation analysis,
if no methylation, MLH1 mutation analysis,
and genetic counseling
+ − + + MSI, PMS2 loss PMS2 mutation analysis and genetic counseling
+ + − − MSI, MSH2 loss MSH2 mutation analysis and genetic counseling
+ + + − MSI, MSH6 loss MSH6 mutation analysis and genetic counseling
MSI indicates microsatellite instability; MSS, microsatellite stable.

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved. www.anatomicpathology.com | 21
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Harada and Morlote Adv Anat Pathol  Volume 27, Number 1, January 2020

FIGURE 1. Mismatch repair immunohistochemistry showing loss of MLH1 (B). PMS2 is also lost when MLH1 is lost (E). MSH2 and MSH6
are intact (C and D, respectively). Hematoxylin and eosin staining image is shown in (A). All 200× magnification. Note the presence of
internal control staining in lymphocytes and stromal cells (B and E).

performance has been shown to be comparable with the NGS achieved 98% sensitivity and 100% specificity for the
conventional PCR-based assay with 93.6% agreement and detection of MSI status as compared with the PCR assay.
lower failure rate (4% vs. 11.9%).24 Although further vali- Waalkes et al26 also demonstrated high sensitivity and specificity
dation by large number of the cases is warranted, these for CRC (both 100%), whereas endometrial and prostate cancer
automated tools may give small laboratories the ability to had false-negative cases. An advantage of using NGS is the
perform MSI assays on cases with equivocal results by IHC. ability to analyze over 100 loci for instability, whereas the assay
Next-generation sequencing (NGS) can also be used for requires special instrumentation and a bioinformatics pipeline.
determination of MSI status.25,26 Zhu et al25 showed MSI by Comparison of different techniques is summarized in Table 2.

22 | www.anatomicpathology.com Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Adv Anat Pathol  Volume 27, Number 1, January 2020 Molecular Pathology of Colorectal Cancer

TABLE 2. Comparison of Assay Technologies to Determine Mismatch Repair Gene Status23–25


MMR IHC MSI PCR Automated MSI NGS
Description MLH1, MSH2, MSH6, 5 mononucleotide 7 microsatellite loci, PCR, Over 100 microsatellite
PMS2 protein expression microsatellite loci, PCR, analyzed by high-resolution loci, NGS analysis
determined by IHC and fragment analysis melting detection
Sensitivity 94% 83%-98% No data 98%
Specificity 100% 100% No data 100%
Pros No need of molecular Require small amount Short hands-on time; Ability to analyze many
laboratory; work on low of tumor; scalable; fast turnaround time more loci, reduce
tumor cellularity samples; objective interpretation equivocal results
identify a defective gene
Cons Some mutations do not Need molecular lab; need Further validation study Expensive; need a special
result in expression loss; normal tissue; labor is needed; difficult to instrument and
subjective interpretation intensive troubleshoot when failed bioinformatics
IHC indicates immunohistochemistry; MMR, mismatch repair; MSI, microsatellite instability; NGS, next-generation sequencing; PCR, polymerase chain
reaction.

Testing Algorithm mutational status, not detected (“wild type”) or abnormal


MSI/MMR testing was introduced initially to identify (mutated), indicated that patients with KRAS mutation in
Lynch syndrome patients. Bethesda guidelines were devel- codon 12 or 13 did not derive benefit from treatment with
oped by the NCI to identify individuals who should be cetuximab or panitumumab.35 Therefore, the American
tested for MSI.27,28 Those included age under 50 years, Society of Clinical Oncology (ASCO) published its clinical
strong family history, presence of synchronous or meta- opinion stating all patients with metastatic CRC who are
chronous colorectal or Lynch syndrome-associated tumors, candidates for anti-EGFR antibody therapy should have
and CRC with MSI-H histology.28 However, 10 of 23 pro- their tumor tested for KRAS mutations in a CLIA-certified
bands with Lynch syndrome were over 50 years of age and 5 laboratory. If KRAS mutation in codon 12 or 13 is detected,
did not meet the Amsterdam criteria or the Bethesda then patients should not receive anti-EGFR antibody ther-
guidelines.29 Therefore, universal screening of CRC for apy as part of their treatment.35 In addition to codons 12
Lynch syndrome has been recommended by many pro- and 13 of KRAS, mutations in other codons of KRAS and in
fessional organizations including the National Compre- NRAS are found to render tumors resistant to anti-EGFR
hensive Cancer Network (2014).30 antibody therapy.36,37
In addition to identifying Lynch syndrome patients, Therefore, a recent guideline from the American Society
MSI testing can be used as a prognostic and predictive for Clinical Pathology, the College of American Pathologists,
marker. MSI tumors tend to be earlier stage and have lower the Association for Molecular Pathology, and the ASCO
recurrence rates than MSS tumors.31 The evidence suggests recommends mutational testing for EGFR signaling pathway
that MSI tumors do not benefit from fluorouracil-based genes as they provide clinically actionable information as
adjuvant chemotherapy in stage II or stage III colon negative predictors of benefit to anti-EGFR monoclonal
cancer.32 Recently, MSI status was recognized as a bio- antibody therapies for targeted therapy of CRC.17 The
marker for immune checkpoint inhibitor therapy.33 This will guideline states that patients with colorectal carcinoma being
be discussed in the separate section.

PREDICTIVE MARKERS FOR TARGETED


THERAPIES
Extended RAS
Signaling pathway through the epidermal growth fac-
tor receptor (EGFR) plays an important role in CRC
(Fig. 2). Anti-EGFR monoclonal antibodies such as cetux-
imab and panitumumab have been shown to bind to the
extracellular domain of EGFR and block downstream sig-
naling, that is, RAS/MRAF/MEK/ERK pathway, and have
a significant clinical benefit in patients with metastatic
CRC.34 However, if a downstream signaling molecule is
mutated, the anti-EGFR antibody cannot block the path-
way, resulting in resistance. Activation mutations in the
KRAS gene are seen in 30% to 50% CRC. Five randomized
trials of cetuximab or panitumumab, when used as either
monotherapy or combination with chemotherapy, have
shown that patients with metastatic CRC may benefit from
these therapies. Both agents are approved by the US Food FIGURE 2. EGFR pathway. EGFR indicates epidermal growth
and Drug Administration (FDA) for treatment of metastatic factor receptor; mTOR, mammalian target of rapamycin; P,
CRC. Stratified analyses of data from these trials by KRAS phosphorylation; PI3K, phosphatidylinositol 3 kinase.

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved. www.anatomicpathology.com | 23
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Harada and Morlote Adv Anat Pathol  Volume 27, Number 1, January 2020

considered for anti-EGFR therapy must receive RAS muta- cancer looking for additional targeted therapy or clinical trial
tional testing. Mutational analysis should include KRAS and options. Overall, a molecular testing algorithm on the basis of
NRAS codons 12 and 13 of exon 2, 59, and 61 of exon 3, and current NCCN guidelines (version 2.2019; www.nccn.org/
117 and 146 of exon 4 (“expanded” or “extended” RAS). In professionals/physician_gls/pdf/colon.pdf) and biomarker
addition, BRAF p.V600 [BRAF c.1799 (p.V600)] mutational guidelines17 is illustrated in Figure 3.
analysis should be performed in CRC tissue in patients with
colorectal carcinoma for prognostic stratification. However, Immunotherapy
they state there is insufficient evidence to recommend BRAF Immune checkpoint inhibitors are an emerging field for
p.V600 mutational status as a predictive biomarker for oncology therapy. In 2015, patients with MSI tumors were
response to anti-EGFR inhibitors.17 Additional biomarkers shown to benefit from immune checkpoint blockade with
have been investigated for possible targeted therapies. How- pembrolizumab.33 Subsequently, the same group expanded
ever, currently there is insufficient evidence to recommend their study to various tumor types and showed the large pro-
PIK3CA or PTEN mutational analysis of colorectal carci- portion of mutant neoantigens in MMR-deficient cancers make
noma tissue for therapy selection outside of a clinical trial.17 them sensitive to immune checkpoint blockade, regardless of
the cancers’ tissue of origin.43 On the basis of this information,
Testing Algorithm the FDA-approved pembrolizumab for unresectable or meta-
Extended RAS and BRAF mutation analysis can be static MSI-H solid tumors in 2017. In the same year, the FDA
performed by any technologies that can detect sequence also granted accelerated approval to Nibolumab for dMMR
variations, that is, Sanger sequence, allele-specific real-time and MSI-H metastatic CRC (http://www.fda.gov/drugs/drug-
PCR, pyrosequencing, or NGS. When the guideline rec- approvals-and-databases).44 Another predictive marker for
ommended testing exons 2, 3, and 4 of KRAS and NRAS, immune checkpoint inhibitor therapy is tumor mutational
then single-gene assays became cumbersome and inefficient. burden (TMB). A recent study suggests that TMB seems to be
NGS-based targeted gene panels have become more efficient an important independent biomarker within MSI-H mCRC to
and cost-effective. In fact, there is a significant increase in stratify patients for likelihood of response to immune check-
laboratories using NGS for RAS testing from ∼10% in 2014 point inhibitors.45 However, currently, there are no guidelines
to 2015 to 23.1% in 2015 to 2016.38 In the same study, they for TMB estimation and reporting, which results in large var-
found that only a third to a quarter of participating labo- iability. Two organizations, Friends of Cancer Research
ratories met the guideline recommendations for extended (Friends) and the Quality Assurance Initiative Pathology
RAS testing, suggesting a delay in implementation at the (QuIP), initiated an effort to standardize the process and have
time of survey38 although much improvement is expected by proposed evidence-based recommendations for achieving con-
now. NGS-based assays, however, require a specific instru- sistent TMB estimation and reporting in clinical samples across
ment, a complex workflow, a bioinformatics pipeline, higher assays and centers.46
quality and quantity of DNA, and longer turnaround time. Recent preclinical and clinical studies suggest that the
Recently, a rapid and fully automated system has been checkpoint inhibitors can be expanded to microsatellite-
evaluated, and concordances between the Idylla NRAS- stable CRC in combinations with chemotherapy, molecular
BRAF or KRAS Mutation Test and reference test results targeted therapy, radiation, or novel immunomodulatory
were found in almost perfect agreement.39–41 A study by agents.47 Many clinical trials are currently in progress,
Franczak et al42 showed that the Idylla platform retrieved which may open a door for new treatment options for
49/67 (73.1%) samples that did not reach DNA quality microsatellite-stable tumors. Duan et al48 demonstrated
requirements for NGS. A caveat is an assay does not cover effective immunotherapy of CRC through systemic delivery
some rare variants (eg, KRAS p.G12F, p.G13C) compared of an immunostimulatory chemotherapeutic combination in
with sequence-based assays. Initial quick screening for nanoscale coordination polymer core-shell particles, syner-
extended RAS and BRAF status can be performed by an gizing with anti-PD-L1 antibody in murine model. As these
automated system and large NGS-targeted panel can be per- novel agents progress through the phases of clinical devel-
formed for patients with recurrent or resistant advanced-stage opment, novel predictive markers specific to these agents

FIGURE 3. Biomarker testing algorithm for colon cancer. IHC indicates immunohistochemistry; MSI, microsatellite instability; NGS, next-
generation sequencing; PCR, polymerase chain reaction.

24 | www.anatomicpathology.com Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Adv Anat Pathol  Volume 27, Number 1, January 2020 Molecular Pathology of Colorectal Cancer

may become important to optimally select patients most information could provide etiological insights into tailored
likely to benefit from them.47 Comprehensive analysis of strategies of disease prevention and treatment.53
multiple markers to identify sensitive patients to immune
checkpoint blockade therapy and its standardization will be REFERENCES
needed.
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA
Cancer J Clin. 2018;68:7–30.
Liquid Biopsy 2. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer
Most tissue used in CRC biomarker testing is derived statistics 2018: GLOBOCAN estimates of incidence and
from formalin-fixed paraffin-embedded (FFPE) tissue. How- mortality worldwide for 36 cancers in 185 countries. CA Cancer
ever, adequate tumor cells may not be available in tissue block J Clin. 2018;68:394–424.
for genomic profiling, especially in small biopsies in an 3. Soreide K, Janssen EA, Soiland H, et al. Microsatellite
advanced-stage setting. Small biopsy samples may not be instability in colorectal cancer. Br J Surg. 2006;93:395–406.
4. Lengauer C, Kinzler KW, Vogelstein B. Genetic instability in
representative when the tumor shows heterogeneity. In addi- colorectal cancers. Nature. 1997;386:623–627.
tion, repeat biopsies for recurrent or metastatic lesions or for 5. Lynch HT, de la Chapelle A. Hereditary colorectal cancer.
monitoring the response to the therapy are not practical. The N Engl J Med. 2003;348:919–932.
term “liquid biopsy” has been introduced to indicate the 6. Gryfe R, Kim H, Hsieh ET, et al. Tumor microsatellite
sampling from nonsolid biological tissue, most commonly of instability and clinical outcome in young patients with color-
blood.49 Plasma cell-free DNA (cfDNA) or circulating tumor ectal cancer. N Engl J Med. 2000;342:69–77.
DNA (ctDNA) has been widely accepted among oncologists 7. Toyota M, Ahuja N, Ohe-Toyota M, et al. CpG island
as a promising noninvasive biomarker for longitudinal mon- methylator phenotype in colorectal cancer. Proc Natl Acad Sci
itoring of a tumor. Recent prospective multicenter study of U S A. 1999;96:8681–8686.
8. Markowitz SD, Bertagnolli MM. Molecular origins of cancer:
patients with stage I to III CRC revealed that ctDNA-positive
molecular basis of colorectal cancer. N Engl J Med. 2009;361:
patients at postoperative day 30 or after adjuvant chemo- 2449–2460.
therapy were likely to relapse (7 times or 17 times, respec- 9. Fearon ER, Vogelstein B. A genetic model for colorectal
tively), and ctDNA status was independently associated with tumorigenesis. Cell. 1990;61:759–767.
relapse after adjusting for known clinicopathologic risk 10. The Cancer Genomic Atlas Network. Comprehensive molec-
factors.50 ctDNA analysis can potentially change the post- ular characterization of human colon and rectal cancer. Nature.
operative management of CRC by enabling risk stratification, 2012;487:330–337.
adjuvant chemotherapy monitoring, and early relapse 11. Guinney J, Dienstmann R, Wang X, et al. The consensus
detection.50 In a proper setting, ctDNA can be utilized as a molecular subtypes of colorectal cancer. Nat Med. 2015;21:
1350–1356.
noninvasive biomarker to guide management of the patient.
12. Aaltonen LA, Peltomaki P, Leach FS, et al. Clues to the
However, there are disadvantages including high false- pathogenesis of familial colorectal cancer. Science. 1993;260:
positive and false-negative rates. Therefore, cfDNA is cur- 812–816.
rently not generally recommended for initial disease diagnosis, 13. Ogino S, Nosho K, Kirkner GJ, et al. CpG island methylator
early detection, or screening. Detection of cfDNA mutations phenotype, microsatellite instability, BRAF mutation and
(eg, TP53, KRAS, and APC) has limited diagnostic sensitivity clinical outcome in colon cancer. Gut. 2009;58:90–96.
(40% to 60%).51 Testing for methylated DNA in gene such as 14. Weisenberger DJ, Siegmund KD, Campan M, et al. CpG island
SEPT9, SFRP1, SDC2 has increased sensitivity (up to 90%) methylator phenotype underlies sporadic microsatellite insta-
for CRC.51 Improvements in technologies such as NGS and bility and is tightly associated with BRAF mutation in
colorectal cancer. Nat Genet. 2006;38:787–793.
digital droplet PCR are rapidly overcoming some of the issues
15. Fischer J, Walker LC, Robinson BA, et al. Clinical implications
of analytical sensitivity, and it is likely that mutation and of the genetics of sporadic colorectal cancer. ANZ J Surg. 2019.
methylation analysis of ctDNA will improve specificity for the Epub ahead of print.
diagnosis of cancer in the future.52 Currently, there is no 16. Rhee YY, Kim KJ, Kang GH. CpG island methylator
standardization for the assay and preanalytical, analytical, phenotype-high colorectal cancers and their prognostic impli-
and postanalytical considerations were identified that need to cations and relationships with the serrated neoplasia pathway.
be addressed.52 Guidelines and recommendations for uti- Gut Liver. 2017;11:38–46.
lization of ctDNA in clinical practice are needed. 17. Sepulveda AR, Hamilton SR, Allegra CJ, et al. Molecular
biomarkers for the evaluation of colorectal cancer: Guideline
From the American Society for Clinical Pathology, College of
CONCLUSIONS American Pathologists, Association for Molecular Pathology,
and American Society of Clinical Oncology. J Mol Diagn.
This review discussed molecular mechanisms of color-
2017;19:187–225.
ectal carcinogenesis and current biomarker guidelines. 18. Funkhouser WK Jr, Lubin IM, Monzon FA, et al. Relevance,
Current standard molecular workup includes evaluation of pathogenesis, and testing algorithm for mismatch repair-
MSI status and extended RAS and BRAF status. We also defective colorectal carcinomas: a report of the association for
discussed emerging technologies utilizing high throughput molecular pathology. J Mol Diagn. 2012;14:91–103.
sequencing for TMB and ctDNA. New consensus and 19. Boland CR, Goel A. Microsatellite instability in colorectal
guidelines/recommendations are needed for these new tech- cancer. Gastroenterology. 2010;138:2073.e3–2087.e3.
nologies, and pathologists should be actively involved in the 20. Murphy KM, Zhang S, Geiger T, et al. Comparison of the
process. The advancement in CRC research and tech- microsatellite instability analysis system and the Bethesda panel
nologies will allow better prognostication and therapy for the determination of microsatellite instability in colorectal
cancers. J Mol Diagn. 2006;8:305–311.
stratification for the management of patients with CRCs. 21. Ellegren H. Microsatellites: simple sequences with complex
Although not discussed in this review, early detection, and evolution. Nat Rev Genet. 2004;5:435–445.
prevention of CRC is an ultimate goal for precision medi- 22. Boland CR, Thibodeau SN, Hamilton SR, et al. A National
cine and public health. Molecular pathologic epidemiology Cancer Institute Workshop on Microsatellite Instability for
(MEP) approach integrating genetic and environmental cancer detection and familial predisposition: development of

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved. www.anatomicpathology.com | 25
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Harada and Morlote Adv Anat Pathol  Volume 27, Number 1, January 2020

international criteria for the determination of microsatellite 38. Richman SD, Fairley J, Butler R, et al. How close are we to
instability in colorectal cancer. Cancer Res. 1998;58:5248–5257. standardised extended RAS gene mutation testing? The UK
23. Zhang L. Immunohistochemistry versus microsatellite insta- NEQAS evaluation. J Clin Pathol. 2017;70:58–62.
bility testing for screening colorectal cancer patients at risk for 39. Zekri J, Baghdadi MA, Alardati H, et al. Evaluation of the
hereditary nonpolyposis colorectal cancer syndrome: part II. Idylla KRAS and NRAS mutation test in colorectal cancer
The utility of microsatellite instability testing. J Mol Diag. tissue. Exp Mol Pathol. 2019;110:104270.
2008;10:301–307. 40. Prieto-Potin I, Montagut C, Bellosillo B, et al. Multicenter
24. Maertens G, De Craene B, Rondelez E, et al. Detection of evaluation of the Idylla NRAS-BRAF mutation test in
microsatellite instability (MSI) in colorectal cancer samples with metastatic colorectal cancer. J Mol Diagn. 2018;20:664–676.
the automated Idylla MSI test. Ann Oncol. 2017;28(suppl 5): 41. Solassol J, Vendrell J, Markl B, et al. Multi-center evaluation of
v22–v42. the fully automated PCR-based Idylla KRAS mutation assay
25. Zhu L, Huang Y, Fang X, et al. A novel and reliable method to for rapid kras mutation status determination on formalin-fixed
detect microsatellite instability in colorectal cancer by next- paraffin-embedded tissue of human colorectal cancer. PloS
generation sequencing. J Mol Diagn. 2018;20:225–231. One. 2016;11:e0163444.
26. Waalkes A, Smith N, Penewit K, et al. Accurate pan-cancer 42. Franczak C, Dubouis L, Gilson P, et al. Integrated routine
molecular diagnosis of microsatellite instability by single- workflow using next-generation sequencing and a fully-auto-
molecule molecular inversion probe capture and high-through- mated platform for the detection of KRAS, NRAS and BRAF
put sequencing. Clin Chem. 2018;64:950–958. mutations in formalin-fixed paraffin embedded samples with
27. Rodriguez-Bigas MA, Boland CR, Hamilton SR, et al. poor DNA quality in patients with colorectal carcinoma. PloS
A National Cancer Institute Workshop on Hereditary Non- One. 2019;14:e0212801.
polyposis Colorectal Cancer Syndrome: meeting highlights 43. Le DT, Durham JN, Smith KN, et al. Mismatch repair
and Bethesda guidelines. J Natl Cancer Inst. 1997;89: deficiency predicts response of solid tumors to PD-1 blockade.
1758–1762. Science. 2017;357:409–413.
28. Umar A, Boland CR, Terdiman JP, et al. Revised Bethesda 44. Zhao P, Li L, Jiang X, et al. Mismatch repair deficiency/
Guidelines for hereditary nonpolyposis colorectal cancer microsatellite instability-high as a predictor for anti-PD-1/PD-
(Lynch syndrome) and microsatellite instability. J Natl Cancer L1 immunotherapy efficacy. J Hematol Oncol. 2019;12:54.
Inst. 2004;96:261–268. 45. Schrock AB, Ouyang C, Sandhu J, et al. Tumor mutational
29. Hampel H, Frankel WL, Martin E, et al. Screening for the burden is predictive of response to immune checkpoint inhibitors
Lynch syndrome (hereditary nonpolyposis colorectal cancer). N in MSI-high metastatic colorectal cancer. Ann Oncol. 2019. Epub
Engl J Med. 2005;352:1851–1860. ahead of print.
30. Hampel H. NCCN increases the emphasis on genetic/familial 46. Stenzinger A, Allen JD, Maas J, et al. Tumor mutational
high-risk assessment in colorectal cancer. J Natl Compr Cancer burden standardization initiatives: recommendations for con-
Netw. 2014;12(suppl 5):829–831. sistent tumor mutational burden assessment in clinical samples
31. Malesci A, Laghi L, Bianchi P, et al. Reduced likelihood of to guide immunotherapy treatment decisions. Genes Chromo-
metastases in patients with microsatellite-unstable colorectal somes Cancer. 2019;58:578–588.
cancer. Clin Cancer Res. 2007;13:3831–3839. 47. Hermel DJ, Sigal D. The emerging role of checkpoint inhibition in
32. Ribic CM, Sargent DJ, Moore MJ, et al. Tumor microsatellite- microsatellite stable colorectal cancer. J Pers Med. 2019:9:pii:E5.
instability status as a predictor of benefit from fluorouracil- 48. Duan X, Chan C, Han W, et al. Immunostimulatory nano-
based adjuvant chemotherapy for colon cancer. N Engl J Med. medicines synergize with checkpoint blockade immunotherapy
2003;349:247–257. to eradicate colorectal tumors. Nat Commun. 2019;10:1899.
33. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors 49. Castro-Giner F, Gkountela S, Donato C, et al. Cancer
with mismatch-repair deficiency. N Engl J Med. 2015;372: diagnosis using a liquid biopsy: challenges and expectations.
2509–2520. Diagnostics (Basel). 2018:8:pii:E31.
34. Cunningham D, Humblet Y, Siena S, et al. Cetuximab 50. Reinert T, Henriksen TV, Christensen E, et al. Analysis of
monotherapy and cetuximab plus irinotecan in irinotecan- plasma cell-free DNA by ultradeep sequencing in patients with
refractory metastatic colorectal cancer. N Engl J Med. 2004;351: stages I to III colorectal cancer. JAMA Oncol. 2019. Epub ahead
337–345. of print.
35. Allegra CJ, Jessup JM, Somerfield MR, et al. American Society 51. Molnar B, Galamb O, Kalmar A, et al. Circulating cell-free
of Clinical Oncology provisional clinical opinion: testing for nucleic acids as biomarkers in colorectal cancer screening and
KRAS gene mutations in patients with metastatic colorectal diagnosis—an update. Expert Rev Mol Diagn. 2019;19:477–498.
carcinoma to predict response to anti-epidermal growth factor 52. Cree IA, Uttley L, Buckley Woods H, et al. The evidence base
receptor monoclonal antibody therapy. J Clin Oncol. 2009;27: for circulating tumour DNA blood-based biomarkers for the
2091–2096. early detection of cancer: a systematic mapping review. BMC
36. Douillard JY, Rong A, Sidhu R. RAS mutations in colorectal Cancer. 2017;17:697.
cancer. N Engl J Med. 2013;369:2159–2160. 53. Hamada T, Nowak JA, Milner DA Jr, et al. Integration of
37. Douillard JY, Oliner KS, Siena S, et al. Panitumumab- microbiology, molecular pathology, and epidemiology: a new
FOLFOX4 treatment and RAS mutations in colorectal cancer. paradigm to explore the pathogenesis of microbiome-driven
N Engl J Med. 2013;369:1023–1034. neoplasms. J Pathol. 2019;247:615–628.

26 | www.anatomicpathology.com Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.

You might also like