You are on page 1of 15

IJC

International Journal of Cancer

Variant classification in precision oncology


Jonas Leichsenring1*, Peter Horak2,3*, Simon Kreutzfeldt2,3, Christoph Heining4,5, Petros Christopoulos6,7,
Anna-Lena Volckmar1, Olaf Neumann1, Martina Kirchner1, Carolin Ploeger1, Jan Budczies 1, Christoph E. Heilig2,
Barbara Hutter8, Martina Fröhlich8, Sebastian Uhrig8,9, Daniel Kazdal 1, Michael Allgäuer1, Alexander Harms1,
Eugen Rempel1, Ulrich Lehmann10, Michael Thomas 6,7, Nicole Pfarr11, Ninel Azoitei12, Irina Bonzheim13, Ralf Marienfeld14,
Peter Möller14, Martin Werner15, Falko Fend13, Melanie Boerries3,8,16,17, Nikolas von Bubnoff 3,8,18,19, Silke Lassmann15,
Thomas Longerich1,20, Michael Bitzer21, Thomas Seufferlein12, Nisar Malek21, Wilko Weichert11, Peter Schirmacher1,3,
Roland Penzel1, Volker Endris 1, Benedikt Brors2,3,8, Frederick Klauschen22, Hanno Glimm4, Stefan Fröhling 2,3,23† and
Albrecht Stenzinger 1,23†
1
Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
2
National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
3
German Cancer Consortium (DKTK), Heidelberg, Germany
4
National Center for Tumor Diseases (NCT), Dresden, Germany
5
Cancer Genetics and Epigenetics

University Hospital Carl Gustav Carus, Dresden, Germany


6
Thoraxklinik, Heidelberg, Germany
7
Translational Lung Cancer Research Heidelberg (TLCR-H), German Center for Lung Research (DZL), Giessen, Germany
8
German Cancer Research Center (DKFZ), Heidelberg, Germany
9
Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
10
Institute of Pathology, Hannover Medical School, Hanover, Germany
11
Institute of Pathology, Technische Universität München, Munich, Germany
12
Clinic of Internal Medicine I, University Hospital Ulm, Ulm, Germany

Key words: molecular pathology, variant classification, molecular tumor board, next-generation sequencing
Abbreviations: ACMG: American College of Medical Genetics and Genomics; AMP: Association for Molecular Pathology; ASCO: American
Society of Clinical Oncology; CAP: College of American Pathologists; CNV: copy number variation; CRC: colorectal cancer; DKFZ: German
Cancer Research Center; DKTK: German Cancer Consortium; ESCAT: ESMO Scale for Clinical Actionability of molecular Targets; ESMO:
European Society for Medical Oncology; IASLC: International Association for the Study of Lung Cancer; Indel: insertion/deletion; JCR: joint
consensus recommendation; MTB: molecular tumor board; NCT: National Center for Tumor Diseases; NGS: next-generation sequencing;
OSoverall survival; PARP: poly (ADP-ribose) polymerase; PFS: progression-free survival; RR: response rate; SNV: single nucleotide variants;
TKI: tyrosine kinase inhibitor; TMB: tumor mutational burden; TRK: tropomyosin receptor kinase; VUS: variant of unknown significance;
WES: whole-exome sequencing; WGS: whole-genome sequencing
Additional Supporting Information may be found in the online version of this article.
Grant sponsor: DKFZ-HIPO; Grant number: H021
Conflict of interests: AS: advisory board honoraria from Bayer, Novartis, BMS, AstraZeneca, ThermoFisher, Illumina; speaker’s honoraria
from Takeda, Roche, BMS, Illumina, AstraZeneca, Novartis, ThermoFisher, Bayer, MSD and research funding from Chugai and BMS. VE:
advisory board honoraria from Thermo Fischer. Speaker’s honoraria from AstraZeneca. ALV: speaker’s honoraria from AstraZeneca. PS:
advisory board honoraria, research funding and speaker’s honoraria from Novartis and AstraZeneca. WW: Advisory board and speaker’s
bureau from Roche, MSD, BMS, AstraZeneca, Pfizer, Merck, Lilly, Boehringer, Novartis, Takeda. Research funding from Roche, MSD, BMS,
Bruker. PC: Speakers honoraria from Roche, Novartis, Chugai. NP: Speaker’s honoraria and travel costs from Novartis, Roche, BMS. SL:
Research funding from Agilent, Illumina, Qiagen, BMS. Advisory boards: AstraZeneca, Novartis, Roche. Travel grants from Illumina, Agilent.
MW: Advisory boards: Roche, Novartis. Research funding from Agilent, Novartis, Roche. Honoraria and travel grants from Diakovere. MT:
advisory board honoraria from Novartis, Lilly, BMS, MSD, Roche, Celgene, Takeda, AbbVie, Boehringer, speaker’s honoraria from Lilly, MSD,
Takeda, research funding from AstraZeneca, BMS, Celgene, Roche, and travel grants from BMS, MSD, Novartis, Boehringer. NvB: Honoraria
from AstraZeneca, Amgen, Novartis, BMS. Research funding from Novartis. All other authors declared no conflicts of interest.
*J.L. and P.H. shared first authorship

S.F. and A.S. shared last authorship
[Correction added on September 21, 2019 after first online publication: affiliation updated.]
DOI: 10.1002/ijc.32358
History: Received 31 Jan 2019; Accepted 8 Apr 2019; Online 22 Apr 2019
Correspondence to: Stefan Fröhling, National Center for Tumor Diseases (NCT) Heidelberg, Im Neuenheimer Feld 460, 69120 Germany,
Tel.: +49-6221-566990, Fax: +49-6221-566967, E-mail: stefan.froehling@nct-heidelberg.de; or Albrecht Stenzinger, Institute of Pathology,
University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany, Tel.: +49-6221-5634380, Fax: +49-6221-565251,
E-mail: albrecht.stenzinger@med.uni-heidelberg.de

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Leichsenring et al. 2997

13
Institute of Pathology, University Hospital Tübingen, Tübingen, Germany
14
Institute of Pathology, University Hospital Ulm, Ulm, Germany
15
Institute of Pathology, Medical Center, University of Freiburg, Breisgau, Germany
16
Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
17
MIRACUM Consortium of the Medical Informatics Initiative, Freiburg, Germany
18
Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
19
Department of Hematology and Oncology, Medical Center, University of Schleswig-Holstein, Lübeck, Germany
20
Heidelberg-Göttingen-Hannover Medizininformatik (HiGHmed) Konsortium, Heidelberg, Germany
21
Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
22
Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
23
DKFZ-Heidelberg Center for Personalized Oncology (HIPO), Heidelberg, Germany

Next-generation sequencing has become a cornerstone of therapy guidance in cancer precision medicine and an indispensable
research tool in translational oncology. Its rapidly increasing use during the last decade has expanded the options for targeted
tumor therapies, and molecular tumor boards have grown accordingly. However, with increasing detection of genetic alterations,

Cancer Genetics and Epigenetics


their interpretation has become more complex and error-prone, potentially introducing biases and reducing benefits in clinical
practice. To facilitate interdisciplinary discussions of genetic alterations for treatment stratification between pathologists,
oncologists, bioinformaticians, genetic counselors and medical scientists in specialized molecular tumor boards, several systems
for the classification of variants detected by large-scale sequencing have been proposed. We review three recent and commonly
applied classifications and discuss their individual strengths and weaknesses. Comparison of the classifications underlines the
need for a clinically useful and universally applicable variant reporting system, which will be instrumental for efficient decision
making based on sequencing analysis in oncology. Integrating these data, we propose a generalizable classification concept
featuring a conservative and a more progressive scheme, which can be readily applied in a clinical setting.

What’s new?
With increasingly comprehensive molecular profiling of cancers, the clinical interpretation of genetic alterations is becoming
more and more challenging. Here the authors review several classifications for gene variant interpretation that were recently
introduced to guide clinical management. They highlight shared features and differences and point out major influencing
factors and unresolved issues that still need to be addressed. Based on this analysis, they propose a unified classification
concept that may become broadly implemented when remaining issues are solved.

Introduction alterations beyond drug-approved entity-specific targets and


During the last few years, the use of diagnostic genetic analyses combinations thereof are identified in a broad range of tumor
of tumor samples has markedly expanded, facilitating the entities, offering potential access to off-label treatments and
growth of precision oncology through the identification of a molecularly stratified clinical trials. With growing complexity of
continuously increasing number of therapeutic targets and genetic data available for individual patients, pathological alter-
molecularly defined stratification of patients.1–7 Panel-based ations have to be interpreted according to predefined standards
next-generation sequencing (NGS) for the detection of tumor- and eventually reviewed in interdisciplinary as well as disease-
specific somatic mutations is already widely available in routine specific molecular tumor boards (MTB) engaging pathologists,
clinical use8 and a number of bioinformatics tools have been oncologists, genetic counselors, bioinformaticians and medical
developed for their assessment, including data quality, which is scientists. To facilitate and standardize communication and dis-
paramount for all subsequent steps (for summaries of these cussion of molecular findings and their clinical relevance, sev-
tools see Refs. 9,10). While most current targeted NGS eral classification systems have been proposed recently,4,9–17 but
approaches interrogate a few kilobases of the tumor exome to no definitive universal standard has been agreed upon.
identify specific targets for which approved drugs exist, more Supporting Information Appendix S1 provides an overview of
comprehensive sequencing efforts ranging from larger gene all major classifications that have been published recently.
panels detecting aberrations on both the DNA and RNA level A recent study18 compared the assessment of four fictional
to whole-exome and-genome sequencing (WES/WGS) as well patients by five MTBs from four countries. MTBs were pro-
as transcriptome sequencing are entering routine diagnostics.4 vided with curated mutational profiles without information on
As a direct consequence, an increasing number of genetic primary sequencing data. For all patients, different treatment

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2998 Variant classification in precision oncology

recommendations were made by each MTB and only for one National Center for Tumor Diseases (NCT) in Heidelberg4
case three MTBs agreed on the same treatment recommenda- and implemented by other institutions nationwide.6
tions. Importantly, heterogeneity of such recommendations The proposed classifications are supposed to facilitate
increased with the complexity of genomic information avail- interdisciplinary communication and comparability of results,
able. Differences were attributed to insufficient evidence for based on a wide spectrum of tumor genetic alterations ranging
specific alterations as well as to uncertainty regarding the from established molecular markers with direct clinical impli-
impact of cooccurring genetic alterations. It is noteworthy that cation (e.g., EGFR L858R in nonsmall cell lung cancer
in a recent survey by the Association for Molecular Pathology [NSCLC]) to alterations with currently unclear/unknown or
(AMP), only 12% and 5% of participating members reported no clinical significance.
experience with WES and WGS, respectively.10 This highlights
the importance of careful interpretation and standardized Joint consensus recommendation of AMP, ACMG, ASCO
reporting in light of ever more complex diagnostic data. and CAP
We analyze three recent and commonly applied approaches The classification jointly proposed by major clinical and patho-
to variant classifications with emphasis on their clinical implica- logical associations in the U.S.10 employs a four-tiered system
tions. Moreover, we devise an integrated classification approach (Table 1), which considers single nucleotide variants (SNV),
Cancer Genetics and Epigenetics

based on the comparative data, which can be readily applied in insertions/deletions (indels), gene fusions and copy number vari-
a clinical scenarios and will support harmonization of genetic ations (CNV). Genetic alterations are classified according to their
variant annotation, which is paramount for full exploitation clinical impact based on the level of clinical and experimental
of the precision oncology concept. evidence. Joint consensus recommendation (JCR) Tier 1 (strong
clinical significance) categorizes alterations with direct clinical
implication in a specific tumor type in terms of FDA-approved
Overview on Current Classifications therapies, inclusion in professional guidelines with expert con-
We analyzed the three most prominent classifications that sensus as well as results from well-powered clinical studies. JCR
have been developed over the last years: (i) A joint consensus Tier 2 (potential clinical significance) includes alterations with
recommendation (JCR) for the interpretation and reporting of FDA-approved therapies in a tumor type other than the tumor
sequence variants in cancer by the Association for Molecular type analyzed, investigational therapies, meta-analyses with some
Pathology (AMP), American College of Medical Genetics and consensus and preclinical studies as well as case reports. JCR
Genomics (ACMG), American Society of Clinical Oncology Tiers 3 and 4 (unknown clinical significance and benign variants,
(ASCO) and College of American Pathologists (CAP)10; (ii) A respectively) include alterations without convincing evidence or
framework for ranking molecular targets for cancer precision lacking evidence of cancer association (so far). The JCR also
medicine by the European Society for Medical Oncology acknowledges the importance of alterations associated with resis-
(ESMO)9; (iii) A classification developed for the MTBs within tance to targeted therapies. Uniquely, this classification also
the German Cancer Consortium (DKTK) applied at the includes diagnostic and prognostic biomarkers.

Table 1. Joint consensus recommendation of AMP, ACMG, ASCO and CAP for reporting genetic variants in cancer (From Li et al., J Mol Diagn,
2017, 19, 4–23 Elsevier, adapted by permission10)
Tier LoE Explanation
Tier 1 A.1 Biomarkers that predict response or resistance to FDA-approved therapies for a specific Variants of strong
type of tumor clinical
significance
A.2 Biomarkers included in professional guidelines that predict response or resistance to
therapies for a specific type of tumor
B Biomarkers that predict response or resistance to therapies for a specific type of tumor
based on well-powered studies with consensus from experts in the field
Tier 2 C.1 Biomarkers that predict response or resistance to therapies approved by the FDA or Variants of
professional societies for a different type of tumor potential clinical
significance
C.2 Biomarkers that serve as inclusion criteria for clinical trials
D Biomarkers that show plausible therapeutic significance based on preclinical studies
Tier 3 Not observed at a significant allele frequency in the general or specific subpopulation Variants of
databases, or pan-cancer or tumor-specific variant databases. No convincing unknown clinical
published evidence of cancer association significance
Tier 4 Observed at significant allele frequency in the general or specific subpopulation Benign or likely
databases. No existing published evidence of cancer association benign variants

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Leichsenring et al. 2999

ESMO scale for clinical actionability of molecular targets includes alterations for which objective responses have been
(ESCAT) shown in the absence of improved outcome, which might
The classification developed by the ESMO9 focuses on clinical show potential benefit in combination with other therapies.
relevance (Table 2). No specific molecular platform or assay Finally, ESCAT X categorizes alterations lacking actionability.
was considered during the development. Similar to Tier 1 of
the JCR, ESCAT I includes genetic alterations with matching
“standard of care” therapies, including alterations with a German National Center for Tumor Diseases classification
defined clinical benefit based on ESMO Magnitude of Clinical At the National Center for Tumor Diseases (NCT) in Heidelberg,
Benefit Scale (MCBS) 1.1,19 requiring the evaluation of single- a classification to assess molecular biomarkers beyond approved
arm clinical studies by the MTB for ESCAT I-B. Within the “standard of care” targets was established in 2015,4 which
MCBS, progression-free survival (PFS), overall survival (OS), has been modified and consented and is used within DKTK.
quality of life as well as grade 3 and 4 toxicities are taken into The MTB at NCT was originally launched to determine
consideration. Additionally, within ESCAT I, alteration-drug therapeutic choices for individual patients enrolled in the Molec-
matches in basket trials with clinical benefit across tumor ularly Aided Stratification for Tumor Eradication Research
types are included. ESCAT II includes alterations with (MASTER) program, a multicenter registry trial for prospec-
unknown magnitude of benefit. This category also considers tive, biology-driven stratification of younger adults with

Cancer Genetics and Epigenetics


results from retrospective analyses showing clinically mean- advanced-stage cancer across all histologies and patients with
ingful benefit (i.e., investigational therapeutic options). ESCAT rare tumors. Individual molecular alterations detected by
III includes alteration-drug matches within different tumor WES/WGS and transcriptome sequencing are validated at the
types as well as (downstream) alterations within the same sig- Center for Molecular Pathology, Institute of Pathology, Univer-
naling pathway as ESCAT I and II biomarkers. ESCAT IV sity Hospital Heidelberg,4,7 including SNVs, CNVs, gene fusions
groups alterations with preclinical evidence of utility as well as and gene expression changes. Actionable alterations are catego-
those with predicted actionability in silico, while ESCAT V rized according to their level of clinical (NCT evidence levels

Table 2. ESMO scale for clinical actionability of molecular targets (ESCAT; From Mateo et al., Ann Oncol, 2018, 29, 1895–1902 Oxford
Academic, adapted by permission9)
ESCAT LoE Explanation
I I-A Prospective, randomized clinical trial(s) show the alteration-drug match in a specific Drug match is associated
tumor type results in a clinically meaningful improvement of a survival endpoint with improved outcome
I-B Prospective, nonrandomized clinical trial(s) show that the alteration-drug match in a in clinical trials
specific tumor type results in clinically meaningful benefit as defined by ESMO
MCBS 1.1
I-C Clinical trials across tumor types or basket clinical trials show clinical benefit
associated with the alteration-drug match, with similar benefit observed across
tumor types
II II-A Retrospective studies show that patients with the specific alteration in a specific tumor Drug match is associated
type experience clinically meaningful benefit with matched drug compared to with antitumor activity,
alteration-negative patients but magnitude of
benefit is unknown
II-B Prospective clinical trial(s) show that the alteration-drug match in a specific tumor type
results in increased responsiveness when treated with a matched drug, however, no
data currently available on survival endpoints
III III-A Clinical benefit demonstrated in patients with the specific alteration (as ESCAT I and II Drug match suspected to
above) but in a different tumor type. Limited/ absence of clinical evidence available improve outcome based
for the patient-specific cancer type or broadly across cancer types on clinical trial data in
III-B An alteration that has a similar predicted functional impact as an already studied other tumor type(s) or
ESCAT I abnormality in the same gene or pathway, but does not have associated with similar molecular
supportive clinical data alteration
IV IV-A Evidence that the alteration or a functionally similar alteration influences drug Preclinical evidence of
sensitivity in preclinical in vitro or in vivo models actionability
IV-B Actionability predicted in silico
V Prospective studies show that targeted therapy is associated with objective responses, Drug match is associated
but this does not lead to improved outcome with objective response,
but without clinically
meaningful benefit
X No evidence that the genomic alteration is therapeutically actionable Lack of evidence for
actionability

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
3000 Variant classification in precision oncology

m1 and m2) as well as preclinical (NCT evidence levels m3 and Comparison of the Classification Systems
m4) evidence. As responses to individual drugs based on predic- Overview on similarities and differences of the
tive biomarkers can be highly dependent on tissue context,20 classifications
further stratification is made based on tumor type. NCT evi- Despite the differences between these classifications, significant
dence level m1 encompasses all recommendations made for the similarities exist (Fig. 1). The first and most obvious feature that
same tumor type, while evidence level m2 includes evidence for groups a set of genetic alterations, is evidence from clinical trials
the respective biomarker obtained in other tumor entities. showing improved outcome for a specific drug in a defined
Within the two clinical evidence levels, three subgroups are tumor entity (i.e., JCR Tier 1/ESCAT I/NCT level m1).4,9,10 The
defined: biomarkers for which clinical efficacy of the second group comprises genetic variants with potential clinical
corresponding drug was demonstrated in a molecularly stratified benefit, derived from clinical trials and case studies in other
cohort of a prospective study or a meta-analysis (m1A/m2A), tumor entities (JCR Tier 2 C.1/ESCAT III-A/NCT level m2).
biomarkers for which clinical effectiveness was shown in a However, emerging genetic alterations and their corresponding
molecularly stratified retrospective cohort or a case–control treatments are ranked differently within each classification based
study (m1B/m2B), and case studies or single unusual responders on the defined level of evidence including preclinical analyses,
that suggest clinical effectiveness of the drug (m1C/m2C). NCT case reports and retrospective analyses. A third group includes
Cancer Genetics and Epigenetics

level m3 encompasses preclinical data (e.g., cell culture and ani- genetic alterations, which might have a clinical benefit as
mal models). NCT level m4 incorporates alterations for which a informed by preclinical models and functional rationale (JCR Tier
theoretical biological rationale (including in silico prediction) 2 D/ESCAT III-B, -IV/NCT level m3, m4). The last group com-
exists but clinical or preclinical data are lacking. Additionally, prises genetic alterations with lacking or unknown association
the NCT classification includes suffixes for resistance and with cancer including variants of unknown (functional/clinical)
approval by the FDA and EMA, respectively (Table 3). While significance (VUS) as well as likely benign variants (JCR Tier
originally developed for comprehensive sequencing approaches, 3, 4/ESCAT X). This category does not exist within the NCT clas-
the use of this classification is currently being expanded to the sification, as such alterations are routinely found in broad geno-
interpretation of panel sequencing results. mic analyses and are not actionable. The NCT classification does

Table 3. Classification scheme developed for the MTBs of the National Center for Tumor Diseases (NCT) and within the German Cancer
Consortium (DKTK)
LoE Explanation
m1A Same entity Predictive value of the biomarker or clinical effectiveness of the corresponding drug in
a molecularly stratified cohort was demonstrated in a prospective study or a
meta-analysis in the same tumor type.
m1B Predictive value of the biomarker or clinical effectiveness of the drug in a molecularly
stratified cohort was demonstrated in a retrospective cohort or case–control study
in the same tumor type.
m1C Case study or single unusual responder indicates the biomarker is associated with
response to the drug in the same tumor type.
m2A Different entity Predictive value of the biomarker or clinical effectiveness of the corresponding drug in
a molecularly stratified cohort was demonstrated in a prospective study or a
meta-analysis in a different tumor type.
m2B Predictive value of the biomarker or clinical effectiveness of the drug in a molecularly
stratified cohort was demonstrated in a retrospective cohort or case–control study
in a different tumor type.
m2C Case study or single unusual responder indicates the biomarker is associated with
response to the drug in a different tumor type.
m3 Preclinical Preclinical data (in vitro, in vivo models or functional genomics studies) demonstrate
that the biomarker predicts response to a specific drug treatment, supported by
scientific rationale.
m4 Biological rationale Biological rationale that associates the biomarker with altered signaling pathway
activity or drug sensitivity without direct clinical or preclinical evidence for response
to the drug.
Additional modifiers is—several in situ data and studies on patient material (e.g., IHC, FISH) support the biomarker and the level of evidence.
iv—in vitro data
Z—Drug is approved for use with the specific biomarker [Z = EMA approval, Z(FDA) = FDA approval]
R—Biomarker predicts resistance to the drug.

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Leichsenring et al. 3001

Cancer Genetics and Epigenetics


Figure 1. Comparison of the JCR, ESCAT and NCT variant classifications systems. The individual evidence levels have been color-coded from
those with direct clinical implication (dark blue) to experimental and hypothetical biomarkers (light blue) and those with no clinical
implication (gray). [Color figure can be viewed at wileyonlinelibrary.com]

not explicitly include germline variant evaluation, which is JCR Tier 1 A.1 comprises drugs approved by the FDA,
reported separately and based on the ACMG/AMP consensus while ESCAT I-A includes alteration-drug matches in specific
guidelines21 as discussed below, but reports actionable targets tumor types with clinically meaningful improvement of a sur-
(e.g., pathogenic BRCA1/2 mutations) like ESCAT. Within the vival endpoint in a prospective, randomized clinical trial and
ESCAT and NCT systems, in silico predictions are classified as NCT level m1 includes predictive biomarkers with clinical
ESCAT IV-B and NCT level m4. No category for alterations lac- efficacy in biomarker-stratified prospective cohorts or meta-
king clinical or preclinical data has been considered within the analyses and is additionally labeled when the drug has been
JCR classification. Tables 4 and 5 exemplify the clinical applica- approved [NCT level m1 (Z)]. However, ESCAT differentiates
tion for data generated by panel-based NGS as well as whole- between randomized (ESCAT I-A) and nonrandomized
genome/exome sequencing. (ESCAT I-B) clinical trials. Taking NSCLC as an example,
tyrosine-kinase inhibitor (TKI)-responsive alterations in
EGFR+ and ALK+ tumors would be classified as ESCAT I-A,
Comparison of core themes across classification systems while TKI-responsive alterations in ROS1+ tumors (high
The following section addresses specific categorical topics response rate [RR] in single-arm study)41 would be classified
across classifications and highlights their impact on clinical as ESCAT I-B but JCR Tier 1 A.1 and NCT level m1A. Ana-
variant interpretation. lyses of EGFR, ALK and ROS1 are mandatory according to
the updated molecular testing guidelines for NSCLC by the
Impact of study design on the classification of genetic CAP, International Association for the Study of Lung Cancer
alterations with approved targeted therapies. The basis of (IASLC) and the AMP42 as well as by the ESMO,43 reflecting
oncological management is the use of approved (targeted) the high-level classification.
therapies that have shown clinical utility in prospective trials As another example, an activating BRAF V600E muta-
most often in conjunction with associated and defined bio- tion in NSCLC would be classified the same way, due to
markers (companion diagnostics). Hence, genetic variants FDA/EMA approval of treatment with dabrafenib/trametinib
proven to be biomarkers for approved targeted therapies are (Tier 1 A.1/NCT level m1A(Z)) but ESCAT I-B due to non-
found within the highest ranked category, albeit with differ- randomization during clinical trials.44 Notably, the use of clin-
ences between the JCR, ESCAT and NCT systems. ical trial randomization for the evaluation of predictive

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
3002 Variant classification in precision oncology

Table 4. Clinical application of somatic variant annotation based on data generated from targeted NGS of a cancer of unknown primary in a
74-year-old male patient
SNVs Allele frequency Function Treatment JCR ESCAT NCT
22,23
ERBB2 62.7% Likely activating Afatinib, two cases Tier 2 D – Level m2C
p.Gly660Asp
ERBB2 61.8% Likely activating Trastuzumab, in vitro24,25 Tier 2 D ESCAT IV-A Level m3
p.Ser310Phe
ATM 21.3% Deleterious Olaparib, one case,26 in vitro27 Tier 2 D – Level m2C
p.Ser2259fs*13
TP53 28% Deleterious – – – –
p.Arg248Trp
FBXW7 22.7% Likely deleterious Temsirolimus, one case28 Tier 2 D ESCAT IV-A Level m2C
p.Arg385His Everolimus, one case29 Tier 2 D – Level m2C-R
Anti-EGFR antibody, – – Level m2B-R
retrospective analysis30

CNVs Chromosome
Cancer Genetics and Epigenetics

CDKN2B Whole-gene deletion 9p21.3


CDKN2A Whole-gene deletion 9p21.3
FGFR1 Low-level amplification 8p11.23
ERBB2 Low-level amplification 17q12
CDK12 Low-level amplification 17q12

biomarkers is a cause of discrepancies between the classifica- to assign clinical evidence to molecular alteration-drug combi-
tions and does not necessarily reflect quality of evidence, nations. Many molecular alterations with potential clinical
because in some cases with existing strong evidence for the benefit might not reach higher evidence levels due to their low
superiority of a compound, randomization against an alterna- incidence in a specific tumor type and would consequently
tive in a prospective clinical trial may not be considered always be inferior to any clinical evidence with low clinical
ethical. Moreover, future regulatory approvals of targeted benefit in an unselected cohort.
therapies may not be driven primarily by randomized studies,
leading to lower evidence level assessment of biomarker–drug Targetable alterations lacking approval. BRAF V600-mutant
combinations not always adequately reflecting their clinical NSCLC treated with a BRAF inhibitor, such as vemurafenib or
value or benefit. dabrafenib, or with dabrafenib in combination with trametinib,
Besides prospective randomized/nonrandomized single- has also shown increased PFS in prospective nonrandomized
entity trials, basket trials recently led to FDA approvals, as in trials,44,46,47 therefore these mutations would be classified as
the case of larotrectinib, a tropomyosin receptor kinase (TRK) JCR Tier 1 B/ESCAT I-B/NCT level m1A. Furthermore, retro-
inhibitor approved for solid tumors based on a prospective, spective studies (molecular single entity basket trial)48 suggest
nonrandomized basket trial including 55 patients with NTRK that some patients with non-V600E BRAF mutations may ben-
fusions and a high RR as well as improved PFS in a diverse efit from BRAF inhibition. Despite the retrospective nature of
range of tumors types including salivary-gland tumors, soft the study, non-V600E BRAF mutations would be classified as
tissue sarcomas, thyroid tumors, colon tumors and others, JCR Tier 2/ESCAT II-A/NCT level m1B. In melanoma, all
some of which (colon cancer, lung cancer, melanoma, GIST, BRAF V600-mutations are approved for vemurafenib therapy
cholangiocarcinoma, appendix carcinoma, breast cancer and in Europe (despite lacking trial evidence for non-V600E BRAF
pancreas cancer) were represented by single cases only.45 mutations), while in the U.S., only BRAF V600E mutations
Based on the classifications described above, the underlying are approved for treatment with vemurafenib by the FDA.
evidence would be classified as high as JCR Tier 1 A.1, ESCAT However, for BRAF V600 (including K/E) mutant melanoma,
I-B/I-C or NCT level m1A(Z) or as low as JCR Tier 2 C.1, combinatorial BRAF/MEK inhibitor regimens (encorafenib/
ESCAT I-C or NCT level m1C/m2C depending on the binimetinib,49 vemurafenib/cobimetinib50 and dabrafenib/
tumor type. trametinib51) have been approved by both regulatory agencies,
Overall, it has to be considered that evidence levels used in FDA and EMA. These examples highlight that country-specific
evidence-based medical practice were primarily designed to difference in the approval statuses of different biomarker–drug
rank the strength of a clinical trial or a research study in combinations can lead to the assignment of different classifica-
molecularly unstratified cohorts in order to guide rational tion tiers and should be considered when reading and inter-
clinical decisions. Such an evaluation might not be suited preting diagnostic reports and MTB decisions.

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Leichsenring et al. 3003

Table 5. Clinical application of somatic variant annotation based on data generated from whole-genome/exome sequencing of a cancer of
unknown primary in a male 59-year-old patient
SNVs Allele frequency Function Treatment JCR ESCAT NCT
ATR 34% Likely deleterious – – – –
p.Asp2027Tyr
BRCA2 54% Likely deleterious Olaparib31,32 Tier 3 ESCAT IV A Level m2A
p.Asp1807Phe
STK11 61% Likely deleterious Immune checkpoint-inhibitor, Tier 3 ESCAT IV A Level m4R
p.Glu14Gln retrospective analysis33,34 Tier 3 ESCAT IV A level m4
Metformin, in vitro35

CNVs Chromosome
IGF1R Low-level amplification 15q26.3
Complex biomarkers
Tumor mutational burden High Immune checkpoint-inhibitor, Tier 2 C.1 ESCAT III-A Level m2A
prospective clinical trial36–39

Cancer Genetics and Epigenetics


Mutational signatures AC4, AC24, AC2, AC13 – – – –
40
Homologous repair deficiency High Niraparib, prospective clinical trial – – Level m4

Targetable alterations with clinical evidence in other tumor classifications but are not explicitly mentioned in ESCAT. A
entities. In all three classifications, cross-entity basket trials well-known example includes oncogenic RAS mutations in
and translation of experiences from different entities are con- colorectal cancer, which indicate resistance to anti-EGFR anti-
sidered (JCR Tier 2 C.1/ESCAT I-C, III-A/NCT level m2). body therapy.54 Another example is RAS mutations or amplifi-
ESCAT specifically lists a cross-entity evidence class ESCAT cations of BRAF in melanoma, which lead to vemurafenib
I-C for clinical basket trials that have demonstrated clinically therapy resistance.55 Besides the JCR and NCT levels, several
meaningful benefit for the target–drug pair across different other classifications include biomarkers of resistance in their
tumor entities. TRK fusions as targets of specific TKI and mis- categorization (OncoKB, CIViC).14,56 An increasing number of
match repair deficiency (dMMR) for PD-1 blockade are mechanisms of primary and secondary resistance, which may
referred to as examples for this category. It remains unclear, also be tumor-type specific, will lead to an increasing complex-
which individual biomarkers fall into this category, as particu- ity of variant annotation, which might in turn complicate test-
larly for targets with very low prevalence, full evaluation of ing for targeted therapies in molecularly distinct subsets of
entity-associated responses can be limited and hard to inter- tumors. Additionally, detection of specific sensitizing concur-
pret. In scenarios with more prevalent genetic aberrations, rent mutations can also influence interpretation of initial vari-
recent data collectively show that clinical benefit across differ- ant classification. In case of EGFR L858R-mutated NSCLC,
ent entities can vary.20 As exemplified by BRAF V600E- simultaneous occurrence of an EGFR T790M mutation indi-
mutated NSCLC (JCR Tier 1 A.1/ESCAT I-B/NCT level m1A) cates resistance to first- and second-generation EGFR-TKI, but
and melanoma (JCR Tier 1 A.1/ESCAT I-A/NCT level m1A), not to osimertinib,57,58 where this constellation would indicate
BRAF inhibition would be indicated with response rates of drug sensitivity and is already regularly assessed in routine
4220 and 48%,52 respectively. At the same time, however, molecular diagnostics. Additionally, secondary mutations
response rates in colorectal cancer (CRC) are less than within the RAS signaling pathway result in resistance to EGFR-
5%.20,53 Therefore, histopathological evaluation of the tumor TKI55 and have to be considered in the overall interpretation of
tissue and contextual interpretation of molecular results is results. These fundamental examples of a complex decision-
paramount and genetic data can only be viewed in the histo- making process also serve as a cautionary note in light of the
pathological and clinical context of the disease. In order to rising number of automated decision-support tools for MTBs.
evaluate specific entity–biomarker–drug combinations in full,
we should consider clinical experiences in other entities for Targetable alterations based on preclinical data or biological
treatment in structured clinical trials or well-documented rationale. Categories JCR Tier 2 C.2, D/ESCAT III-B,
observational studies until sufficient data of individual -IV/NCT level m3–4 group alterations suggest benefit from
responses in the entity of question is available. targeted therapies based on functional or in silico data. In
addition, ESCAT IV differentiates between IV-A (evidence
Alterations associated with resistance and concurrent based on in vitro or in vivo models) and IV-B (in silico evi-
alterations. Alterations associated with resistance to specific dence). The JCR recommends against the use of prediction
therapies are classified in the JCR and by the NCT algorithms as sole evidence for variant classification or clinical

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


Cancer Genetics and Epigenetics

3004

Figure 2. Unifying classification concept. This concept may serve as a blueprint for harmonization of variant classifications and as a (pocket) guide for clinical decision making. Based
on implementation in patient treatment, evidence levels are color-coded: established predictive biomarkers/drug targets are highlighted in green. Biomarkers with potential clinical
implication are highlighted in different shades of orange based on the strengths of evidence, while variants without any major current clinical implication are highlighted in red. [Color
figure can be viewed at wileyonlinelibrary.com]

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


Variant classification in precision oncology

10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Leichsenring et al. 3005

decision making.10 In our opinion, information obtained by therapies are ranked first within both approaches, while data
preclinical evidence is relevant for patients with advanced, from current clinical trials and convincing evidence from dif-
treatment-refractory disease eligible for clinical trials based on ferent entities as well as case reports might be more applicable
molecular alterations or in well-documented observational in a progressive setting at experienced centers. Finally, exper-
studies. These alterations may not have an established benefit imental approaches based on clinical evidence within the
in clinical studies, though they may lead to valid therapeutic same or a different tumor entity as well as preclinical
reasoning considering the individual landscape of possible (in vitro/in silico) data should only be considered in the con-
driver mutations in the molecular context of the disease. text of clinical programs at specialized centers independent
of the therapeutic approach. Together with potential off-label
Assessment of case reports. Molecularly characterized case therapies, these options have to be discussed in depth in
reports and collections thereof (e.g., found in GENIE and interdisciplinary MTBs.
CIViC)56 have become increasingly valuable for the evaluation
of clinical benefit in a precision oncology setting. Such case Evolving Topics in Clinical Variant Classification
reports have been an integral part of the sequencing efforts With the continuous and accelerated development of new
within the MASTER program at NCT Heidelberg and are drugs and increasingly comprehensive molecular profiles,
taken into consideration through NCT levels m1C and m2C. MTBs will be facing a complex task of analyzing and inter-

Cancer Genetics and Epigenetics


Within the JCR, case reports are represented in Tier 2 if mul- preting not only a single molecular alteration but whole sets
tiple cases support the same conclusion but no consensus was of biomarkers, including continuous variables such as tumor
found, while ESCAT levels do not specifically address the pos- mutational burden (TMB), in the context of individual
sibility to obtain valid evidence from individual cases. patients’ tumor biology, histology and medical history. There-
Although this evidence may initiate further use of targeted fore, a number of additional factors, which are briefly
therapies in specific entity–biomarker–drug combinations and highlighted below, will play an increasingly important role in
may aid the development of prospective basket trials,59 obser- the near future.
vation of clinical impact in single patients represent an excep-
tional event associated with additional uncontrolled factors Biological themes recurring across tumor types
that can potentially interfere with response and outcome Some targetable genetic alterations have been identified in a
parameters. Hence, evidence from case studies (i.e. n = 1), diverse number of tumor types. However, many of these alter-
requires careful individual assessment and should not be gen- ations are too rare for prospective, randomized trials and thus
eralized prematurely. histology-agnostic basket trials were initiated to assess their
clinical impact. Larotrectinib, for example, demonstrated a RR
Alterations without clinical actionability. Tiers 3 and 4 of the of 75% in solid tumors with TRK fusions, resulting in FDA
JCR represent a collection of mutations without current clini- approval. For cases with n = 1, the evidence level could be
cal implication (mutations of unknown clinical significance considered equivalent to case reports, and the clinical rele-
and mutations without cancer association), while ESCAT X vance of different fusion partners with regard to clinical effi-
groups mutations lacking clinical actionability, independent of cacy60 cannot be evaluated based on individual patients.
their association with cancer. Listing benign or currently clini- Furthermore, within these molecular basket trials, control
cally irrelevant alterations is beyond the scope of a routine groups are absent, which influences outcome interpretation.
reporting of genetic profiles and certainly neither feasible nor Fortunately, additional data is generated in ongoing trials61
meaningful in a scenario where comprehensive genomic and the resulting data will have to be reevaluated.
assessment by WES/WGS is applied. Another example is the response to immunotherapy with
nivolumab in different mismatch repair deficient tumor
A Unifying Concept for Clinical Variant Classification entities.62,63 Trials including germline as well as sporadic
Based on the analysis shown above, we conceptualized our MSI-H colorectal cancer with nivolumab resulted in objec-
findings to devise an integrated, unifying classification tive RR of 31.1% (n = 74; 36% of patients had Lynch-
approach that can serve as a blueprint for harmonization of Syndrome),62 40% (n = 10; 80% had Lynch-Syndrome)64
the classification schemes that were proposed until now and 52% (n = 40; 50% had Lynch-Syndrome),65 suggesting
(Fig. 2). This conceptual approach also integrates the previous differences between germline and somatic mismatch repair
classification systems summarized in Supporting Information deficiency.
Appendix S1 and can be used as a (pocket) guide to somatic
variant classification for daily clinical decision making. This Multifactorial biomarkers: complexity is increasing
concept acknowledges that each classification system has a So far, most variant classifications are primarily designed to
subjective component that is either supporting a more conser- interpret and classify SNVs, indels, gene fusions and CNVs.
vative or a more progressive therapeutic approach. Of course, However, currently, a multitude of other relevant parameters,
variants and biomarkers associated with “standard of care” such as PD-1/PD-L1 expression, homologous recombination

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
3006 Variant classification in precision oncology

deficiency (HRD) and TMB66,67 have to be evaluated in predictive value of FGFR1 amplifications seen in BGJ398 tri-
MTBs. Not to mention global genetic (e.g., BRCAness),68,69 als78 to other pan-FGFR inhibitors or even to other multi-
gene expression and methylome profiles,70 which are currently kinase inhibitors with FGFR1-affinity.
not captured by most precision oncology approaches but will ESCAT groups alterations with similar predicted functional
play an increasing role in the future. impact in class ESCAT III-B but do not specify them further,
ESCAT refers to “molecular targets” instead of genetic leaving space for interpretation. The NCT classification does
alterations and therefore the PD-1/PD-L1 expression would not make assumptions about biological equivalency and
be classified as ESCAT I-A (at least in NSCLC).71 The JCR assigns these unknown variants to NCT level m4, leaving the
focuses on somatic variants only, including SNVs, indels, gene discussions of the biological implications to the MTB in
fusions and CNVs. NCT levels focus in a more general appropriate cases. For the JCR, such alterations might be
approach on biomarker-based recommendations, thus includ- summed up under JCR Tier 3. As of today, the issue on how
ing all measurable molecular properties, including immuno- to handle diagnostic and therapeutic discrepancies with regard
histochemistry, in situ hybridization, but also other possible to their biological equivalency in assigning evidence levels is
technologies, such as methylation studies or proteomic assays. not ultimately solved. However, it stresses the necessity to dis-
TMB, a continuous variable of probabilistic nature, would cuss patients with extensive genomic characterization in inter-
Cancer Genetics and Epigenetics

currently classify at best as JCR Tier 1 B/ESCAT I-C/NCT disciplinary MTBs to appropriately address these individual
level m1B.72 In addition, response to immune checkpoint constellations.
inhibitors has been reported to be influenced by a plethora of
molecular alterations, including STK11 or BRCA1/2 muta- Druggable targets associated with defects in the DNA repair
tions in association with other markers in a number of tumor machinery
entities.73,74 Identification and classification of BRCA1/2 variants and
Inclusion of the above alterations and biomarkers drasti- others are essential for risk assessment with respect to inher-
cally increases the complexity of an all-encompassing classi- itable disease. Such classification is performed according to
fication and requires ranking of each therapeutic option, the 2015 classification consensus by the ACMG-AMP21,79
taking into consideration the patient’s health status and taking into consideration population data, computational
preference, previous lines of therapy as well as therapy evidence, functional data, segregation data, de novo data,
availability. allelic data and “other data”. This classification, which is
based on a multiparametric probability model and originally
Biological equivalence of alterations and exchangeability of meant to classify a relative risk of developing cancer for
therapeutic regimens healthy carriers with a certain genetic make-up in the
Assessing therapies such as osimertinib in NSCLC patients germline, has been repurposed for the evaluation of drug
with somatic EGFR T790M mutations is relatively straight response associated with deleterious mutations in DNA
forward,57,58 but with the forthcoming implementation of repair genes. For example, likely pathogenic and pathogenic
large sequencing panels, WGS/WES and transcriptome variants (Plon 4/5 variants)80 in BRCA1/2 in breast81 and
sequencing, MTBs will frequently be challenged to assess rare ovarian cancer82,83 lead to homologous repair deficiency
alterations, which are not strictly identical to the published (HRD) sensitizing the tumor to Poly (ADP-ribose) polymer-
variants, or to consider therapeutic regimens which differ ase (PARP) inhibition, a concept termed synthetic lethality.84
from the ones used in a clinical trial. For example, the sensi- While the majority of these variants are germline events, the
tizing effect of BRAF V600 hotspot SNVs to selective BRAF classification system is also used to predict therapy response
inhibitors is observed in several entities, and similar effects for somatically acquired mutations.85 Such loss-of-function
have been assigned preclinically and clinically to certain alter- aberrations are classified as JCR Tier 1 A/ESCAT I-A/NCT
ations close by.75,76 However, it would be incorrect to apply level m1A in ovarian and breast cancer based on FDA
these findings to other unknown V600 or K601 SNVs, as clin- approval and a prospective, molecularly stratified, random-
ical evidence is lacking. Furthermore, care has to be taken ized trial.81 It should be noted, however, that the clinical rel-
before generalizing the effect of RAF/MEK inhibition in indi- evance and predictive values of deleterious variants in the
vidual responders with BRAF fusions.77 majority of the more than 100 core genes encoding DNA
Until now, many (potential) biomarkers remain incom- repair pathways are currently only poorly understood.86,87 As
pletely studied, hindering the assignment of evidence levels, a proof-of-concept, emerging data show that for example in
such as the interpretation of unknown mutations in kinase prostate cancer loss-of-function alterations beyond BRCA2
domains, mRNA expression corresponding to predictive pro- can be clinically relevant.26,88 As DNA repair operates in a
tein expressions, assessment of gene amplifications and their tumor agnostic manner, it is reasonable to assume that
cutoffs that define “amplified” = druggable, transferability of genetic lesions beyond BRCA1/2—whether germline or
predictive markers for PD1 inhibitors to PD-L1 inhibitors or somatic—can in principle be exploited for synthetic lethality
CTLA4 inhibitors as well as the generalization of the concepts across many cancer types.89

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Leichsenring et al. 3007

Diagnostic biomarkers, pharmacogenomics and risk For the remaining, newly discovered, rare or individual
assessment genetic alterations, each classification has its advantages and
Beyond predictive genetic lesions, only the JCR10 explicitly drawbacks and not all alterations are classified equally
acknowledges diagnostic (e.g., the PML-RARA fusion in acute throughout the different schemes. Differences depend on the
promyelocytic leukemia) and prognostic (e.g., NPM1 muta- specific evidence levels defining the categories within each
tions in acute myeloid leukemia) biomarkers and includes scheme, which are influenced by clinical and biological ratio-
them into the classification approach. However, the use of sin- nale, but also by different socioeconomic circumstances
gle genetic events as clear-cut diagnostic biomarkers should be including various regulatory approvals of targeted compounds
viewed with caution. Even gene fusions involving, for exam- in different health care systems.
ple, ALK or EWSR1 can occur in tumor types other than clas- While the JCR defines a range of evidence levels including
sic index cancer types NSCLC and Ewing sarcoma. Recently markers of resistance, variants of unknown significance and
published examples are EWSR1 translocations in pancreatic benign variants, it is tailored to the U.S. health care system,
neuroendocrine tumors90 or ALK fusions in colorectal can- focusing on FDA approval status, which is not necessarily
cer.91 While the overall prevalence might be lower compared applicable outside of the U.S.
to the classic tumor types where the initial discoveries were ESCAT, like the JCR, covers the range from standard of
made,92,93 one should be aware that the concept of a single care to alterations with no evidence of therapeutic applicabil-

Cancer Genetics and Epigenetics


pathognomonic genetic event has limitations, particularly ity but does not take into consideration markers of resistance.
when it comes to the interpretation of individual molecular Furthermore, it relies on its own standards for determination
data. Importantly, the role of pharmacogenomics, that is, of a clinical benefit, which appears to be less subjective than a
determining drug response based on sequencing results, is broad definition of “well-powered studies” or “expert opin-
becoming more important with accumulating evidence ion.” However, this adds another layer of complexity and
(e.g., in colorectal cancer)94 and may be more broadly incor- might sometimes be the source of difficulties or discrepancies
porated in oncological decision making in the near future. A in diagnostic assessment. Consideration of the randomization
third emerging topic is specific germline aberrations that status of clinical trials can also be a cause of inconsistency
require genetic counseling. With more comprehensive molec- with the other systems and the in silico predictors mentioned
ular profiling entering routine diagnostics, this field will in this proposal are rarely useful.
strongly expand in the next couple of years: our current The NCT MTB classification addresses the complexity of
knowledge of cancer risk-associated variants is confined to a evaluating predictive molecular biomarkers by focusing on two
relatively small set of genes compared to, for instance, the main criteria, tumor entity and clinical relevance. By avoiding
number of genes that are implicated in the various DNA- parameters beside primary scientific data (such as the design of
repair pathways.95,96 prospective clinical trials, the secondary evaluation of clinical
benefit and the drug approval status) it is less complex than the
other classification schemes, thus easy to apply and communi-
Predictive molecular biomarkers for conventional therapies
cate. In our experience, the classification provides graded and
As exemplified for BRCA1/2 variants in triple-negative breast
evaluable therapeutic recommendations for advanced cancer
cancer, genetic aberrations can predict efficacy of conventional
patients and is well suited for cross-institutional use. Further-
therapeutic approaches such as platinum-based chemother-
more, similar to the JCR, it specifically allows incorporation of
apy.97 The authors assume that the rapid progress in our
case studies, which, in rare entities, might be and remain the
understanding of tumor biology will lead to the discovery of
only level of evidence available.
many molecular profiles that can predict response or resis-
A unifying integrative classification concept, as presented
tance to specific chemotherapy and radiotherapy regimens,
by us, can support daily clinical decision making and con-
which will need to be considered in future adaptations of the
siders a more conservative and more progressive treatment
variant classification systems.
approach.

Summary and Outlook Conclusion


The growing amount of molecular data correlates with This head-to-head comparison of three different classifica-
increasing diagnostic and therapeutic complexity.98 All variant tions is meant to support the clinical interpretation of com-
classifications analyzed herein provide a framework for the mon and rare druggable cancer biomarkers. As we have
clinical assessment of the detected genetic alterations and shown, MTB reports must be supported by rational,
facilitate prioritization of the corresponding targeted therapies, evidence-based decisions, which are reproducible and readily
while not absolutely ranking them in terms of overall patient transferrable between institutions. This is a prerequisite for
management. All three classifications are relatively easily the implementation of interinstitutional precision oncology
applicable for most well-characterized alterations, which are programs and any future standardization in the field. Such a
already considered standard of care. common standard, for which we are providing a conceptual

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
3008 Variant classification in precision oncology

approach, needs to address the complexity and uniqueness of predictive value of biomarkers and the clinical efficacy of
molecular alterations within individual tumors while being associated drugs.
flexible enough to allow application in a routine clinical
setting—even for rare tumors and rare genetic events. By Acknowledgements
All members of the Center for Molecular Pathology at the University Hos-
focusing on implementation of molecularly targeted thera-
pital Heidelberg, DKFZ-HIPO and the members of the Molecular Tumor
pies in a comprehensively characterized patient cohort in Board at NCT Heidelberg for generating the genetic data and continuous
well documented clinical trials or observational studies, we discussions and interpretation. This work was supported by grant H021
might be able to “bootstrap” our knowledge about the from DKFZ-HIPO to C.H., H.G., A.S. and S.F.

References
1. Roychowdhury S, Iyer MK, Robinson DR, et al. 13. Meric-Bernstam F, Johnson A, Holla V, PARP inhibitor olaparib. Transl Oncol 2017;10:
Personalized oncology through integrative high- et al. A decision support framework 190–6.
throughput sequencing: a pilot study. Sci Transl for Genomically informed investigational 28. Villaruz LC, Socinski MA. Temsirolimus
Med 2011;3:111ra21. cancer therapy. J Natl Cancer Inst 2015;107: therapy in a patient with lung adenocarcinoma
2. Burkard ME, Deming DA, Parsons BM, et al. djv098. harboring an FBXW7 mutation. Lung Cancer
Implementation and clinical utility of an inte- 14. Chakravarty D, Gao J, Phillips S, et al. OncoKB: a 2014;83:300–1.
Cancer Genetics and Epigenetics

grated academic-community regional molecular precision oncology Knowledge Base. JCO Precis 29. Jardim DL, Wheler JJ, Hess K, et al. FBXW7
tumor board. JCO Precis Oncol 2017;1:1–10. Oncol 2017;1:1–16. mutations in patients with advanced cancers: clin-
3. Dalton WB, Forde PM, Kang H, et al. Personalized 15. Starlinger J, Pallarz S, Seva J, et al. Variant infor- ical and molecular characteristics and outcomes
medicine in the oncology clinic: implementation mation systems for precision oncology. BMC Med with mTOR inhibitors. PLoS One 2014;9:e89388.
and outcomes of the Johns Hopkins molecular Inform Decis Mak 2018;18:107. 30. Lupini L, Bassi C, Mlcochova J, et al. Prediction
tumor board. JCO Precis Oncol 2017;1:1–19. 16. Damodaran S, Miya J, Kautto E, et al. Cancer of response to anti-EGFR antibody-based thera-
4. Horak P, Klink B, Heining C, et al. Precision driver log (CanDL): catalog of potentially action- pies by multigene sequencing in colorectal cancer
oncology based on omics data: the NCT Heidel- able cancer mutations. J Mol Diagn 2015;17: patients. BMC Cancer 2015;15:808.
berg experience. Int J Cancer 2017;141:877–86. 554–9. 31. Ledermann J, Harter P, Gourley C, et al. Olaparib
5. Zehir A, Benayed R, Shah RH, et al. Mutational 17. Ritter DI, Roychowdhury S, Roy A, et al. Somatic maintenance therapy in patients with platinum-
landscape of metastatic cancer revealed from pro- cancer variant curation and harmonization sensitive relapsed serous ovarian cancer: a
spective clinical sequencing of 10,000 patients. through consensus minimum variant level data. preplanned retrospective analysis of outcomes by
Nat Med 2017;23:703–13. Genome Med 2016;8:117. BRCA status in a randomised phase 2 trial. Lancet
6. Hoefflin R, Geißler A-L, Fritsch R, et al. Personal- 18. Rieke DT, Lamping M, Schuh M, et al. Compari- Oncol 2014;15:852–61.
ized clinical decision making through implemen- son of treatment recommendations by molecular 32. Pujade-Lauraine E, Ledermann JA, Selle F, et al.
tation of a molecular tumor board: a German tumor boards worldwide. JCO Precis Oncol Olaparib tablets as maintenance therapy in
single-center experience. JCO Precis Oncol 2018; 2018;1–14. patients with platinum-sensitive, relapsed ovarian
2:1–16. 19. Cherny NI, Dafni U, Bogaerts J, et al. ESMO- cancer and a BRCA1/2 mutation
7. Lier A, Penzel R, Heining C, et al. Validating magnitude of clinical benefit scale version 1.1. (SOLO2/ENGOT-Ov21): a double-blind,
comprehensive next-generation sequencing results Ann Oncol 2017;28:2340–66. randomised, placebo-controlled, phase 3 trial.
for precision oncology: the NCT/DKTK molecu- 20. Hyman DM, Puzanov I, Subbiah V, et al. Lancet Oncol 2017;18:1274–84.
larly aided stratification for tumor eradication Vemurafenib in multiple nonmelanoma cancers 33. Rizvi H, Sanchez-Vega F, La K, et al. Molecular
research experience. JCO Precis Oncol with BRAF V600 mutations. N Engl J Med 2015; determinants of response to anti-programmed cell
2018;2:1–13. 373:726–36. death (PD)-1 and anti-programmed death-ligand
8. Volckmar AL, Leichsenring J, Kirchner M, et al. 21. Richards S, Aziz N, Bale S, et al. Standards and 1 (PD-L1) blockade in patients with non-small-cell
Combined targeted DNA and RNA sequencing of guidelines for the interpretation of sequence vari- lung cancer profiled with targeted next-generation
advanced NSCLC in routine molecular diagnos- ants: a joint consensus recommendation of the sequencing. J Clin Oncol 2018;36:633–41.
tics: analysis of the first 3,000 Heidelberg cases. American College of Medical Genetics and Geno- 34. Skoulidis F, Goldberg ME, Greenawalt DM, et al.
Int J Cancer 2019; https://doi.org/10.1002/ mics and the Association for Molecular Pathology. STK11/LKB1 mutations and PD-1 inhibitor resis-
ijc.32133. [Epub ahead of print] Genet Med 2015;17:405–24. tance in KRAS-mutant lung adenocarcinoma.
9. Mateo J, Chakravarty D, Dienstmann R, et al. A 22. Yamamoto H, Toyooka S, Ninomiya T, et al. Cancer Discov 2018;8:822–35.
framework to rank genomic alterations as Therapeutic potential of Afatinib for cancers with 35. Shackelford DB, Abt E, Gerken L, et al. LKB1
targets for cancer precision medicine: the ERBB2 (HER2) transmembrane domain mutations inactivation dictates therapeutic response of non-
ESMO scale for clinical actionability of G660D and V659E. Oncologist 2018;23:150–4. small cell lung cancer to the metabolism drug
molecular targets (ESCAT). Ann Oncol 2018;29: 23. Ou S-HI, Schrock AB, Bocharov EV, et al. HER2 phenformin. Cancer Cell 2013;23:143–58.
1895–902. transmembrane domain (TMD) mutations 36. Hellmann MD, Callahan MK, Awad MM, et al.
10. Li MM, Datto M, Duncavage EJ, et al. Standards (V659/G660) that stabilize homo- and Tumor mutational burden and efficacy of
and guidelines for the interpretation and reporting heterodimerization are rare oncogenic drivers in Nivolumab monotherapy and in combination
of sequence variants in cancer: a joint consensus lung adenocarcinoma that respond to afatinib. with Ipilimumab in small-cell lung cancer. Cancer
recommendation of the Association for Molecular J Thorac Oncol 2017;12:446–57. Cell 2018;33:853–61 e4.
Pathology, American Society of Clinical Oncology, 24. Herter-Sprie GS, Greulich H, Wong K-K. Activat- 37. Hellmann MD, Ciuleanu TE, Pluzanski A, et al.
and College of American Pathologists. J Mol ing mutations in ERBB2 and their impact on Nivolumab plus ipilimumab in lung cancer with a
Diagn 2017;19:4–23. diagnostics and treatment. Front Oncol 2013;3:86. high tumor mutational burden. N Engl J Med
11. Andre F, Mardis E, Salm M, et al. Prioritizing tar- 25. Kavuri SM, Jain N, Galimi F, et al. HER2 activat- 2018;378:2093–104.
gets for precision cancer medicine. Ann Oncol ing mutations are targets for colorectal cancer 38. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer
2014;25:2295–303. treatment. Cancer Discov 2015;5:832–41. immunology. Mutational landscape determines
12. Van Allen EM, Wagle N, Stojanov P, et al. 26. Mateo J, Carreira S, Sandhu S, et al. DNA-repair sensitivity to PD-1 blockade in non-small cell
Whole-exome sequencing and clinical interpreta- defects and Olaparib in metastatic prostate cancer. lung cancer. Science 2015;348:124–8.
tion of formalin-fixed, paraffin-embedded tumor N Engl J Med 2015;373:1697–708. 39. Van Allen EM, Miao D, Schilling B, et al. Geno-
samples to guide precision cancer medicine. Nat 27. Wang C, Jette N, Moussienko D, et al. ATM- mic correlates of response to CTLA-4 blockade in
Med 2014;20:682. deficient colorectal cancer cells are sensitive to the metastatic melanoma. Science 2015;350:207–11.

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Leichsenring et al. 3009

40. Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib MEK1. Proc Natl Acad Sci U S A 2012;109: following immunotherapy among patients with
maintenance therapy in platinum-sensitive, recur- E2127–33. TP53 and KRAS mutated stage IV NSCLC.
rent ovarian cancer. N Engl J Med 2016;375: 56. Griffith M, Spies NC, Krysiak K, et al. CIViC is a J Thorac Oncol 2018;13:S486.
2154–64. community knowledgebase for expert 74. Strickland KC, Howitt BE, Shukla SA, et al. Asso-
41. Shaw AT, S-HI O, Bang Y-J, et al. Crizotinib in crowdsourcing the clinical interpretation of vari- ciation and prognostic significance of BRCA1/2--
ROS1-rearranged non–small-cell lung cancer. N ants in cancer. Nat Genet 2017;49:170–4. mutation status with neoantigen load, number of
Engl J Med 2014;371:1963–71. 57. Mok TS, Wu Y-L, Ahn M-J, et al. Osimertinib or tumor-infiltrating lymphocytes and expression of
42. Lindeman NI, Cagle PT, Aisner DL, et al. platinum–pemetrexed in EGFR T790M–positive PD-1/PD-L1 in high grade serous ovarian cancer.
Updated molecular testing guideline for the selec- lung cancer. N Engl J Med 2017;376:629–40. Oncotarget 2016;7:13587–98.
tion of lung cancer patients for treatment with 58. Yang JC-H, Ahn M-J, Kim D-W, et al. 75. Richtig G, Hoeller C, Kashofer K, et al. Beyond the
targeted tyrosine kinase inhibitors. J Mol Diagn Osimertinib in pretreated T790M-positive BRAFV600E hotspot: biology and clinical implica-
2018;20:129–59. advanced non–small-cell lung cancer: AURA tions of rare BRAF gene mutations in melanoma
43. Planchard D, Popat S, Kerr K, et al. Metastatic study phase II extension component. J Clin Oncol patients. Br J Dermatol 2017;177:936–44.
non-small cell lung cancer: ESMO clinical practice 2017;35:1288–96. 76. Ponti G, Pellacani G, Tomasi A, et al. The somatic
guidelines for diagnosis, treatment and follow-up. 59. Heining C, Horak P, Uhrig S, et al. NRG1 fusions affairs of BRAF: tailored therapies for advanced
Ann Oncol 2018;29:iv192–237. in KRAS wild-type pancreatic cancer. Cancer malignant melanoma and orphan non-V600E
44. Planchard D, Smit EF, Groen HJM, et al. Discov 2018;8:1087–95. (V600R-M) mutations. J Clin Pathol 2013;66:
Dabrafenib plus trametinib in patients with previ- 60. Childress MA, Himmelberg SM, Chen H, et al. 441–5.
ously untreated BRAFV600E-mutant metastatic ALK fusion partners impact response to ALK 77. Ross JS, Wang K, Chmielecki J, et al. The distribu-
non-small-cell lung cancer: an open-label, phase inhibition: differential effects on sensitivity, cellu- tion of BRAF gene fusions in solid tumors and
2 trial. Lancet Oncol 2017;18:1307–16. lar phenotypes, and biochemical properties. Mol response to targeted therapy. Int J Cancer 2016;

Cancer Genetics and Epigenetics


45. Drilon A, Laetsch TW, Kummar S, et al. Efficacy Cancer Res 2018;16:1724–36. 138:881–90.
of Larotrectinib in TRK fusion-positive cancers in 61. Lassen UN, Albert CM, Kummar S, et al. 78. Nogova L, Sequist LV, Perez Garcia JM, et al.
adults and children. N Engl J Med 2018;378: 409OLarotrectinib efficacy and safety in TRK Evaluation of BGJ398, a fibroblast growth factor
731–9. fusion cancer: an expanded clinical dataset show- receptor 1–3 kinase inhibitor, in patients with
46. Planchard D, Kim TM, Mazieres J, et al. ing consistency in an age and tumor agnostic advanced solid tumors harboring genetic alter-
Dabrafenib in patients with BRAFV600E-positive approach. Ann Oncol 2018;29:mdy279.397. ations in fibroblast growth factor receptors: results
advanced non-small-cell lung cancer: a single- 62. Overman MJ, McDermott R, Leach JL, et al. of a global phase I, dose-escalation and dose-
arm, multicentre, open-label, phase 2 trial. Lancet Nivolumab in patients with metastatic DNA expansion study. J Clin Oncol 2017;35:157–65.
Oncol 2016;17:642–50. mismatch repair-deficient or microsatellite 79. Nykamp K, Anderson M, Powers M, et al. She-
47. Subbiah V, Gervais R, Riely GJ, et al. Efficacy of instability-high colorectal cancer (CheckMate rloc: a comprehensive refinement of the ACMG-
vemurafenib in patients (pts) with non-small cell 142): an open-label, multicentre, phase 2 study. AMP variant classification criteria. Genet Med
lung cancer (NSCLC) with BRAFV600 mutation. Lancet Oncol 2017;18:1182–91. 2017;19:1105–17.
J Clin Oncol 2017;35:9074. 63. Mittica G, Ghisoni E, Giannone G, et al. Checkpoint 80. Plon SE, Eccles DM, Easton D, et al. Sequence
48. Gautschi O, Milia J, Cabarrou B, et al. Targeted inhibitors in endometrial cancer: preclinical rationale variant classification and reporting: recommenda-
therapy for patients with BRAF-mutant lung can- and clinical activity. Oncotarget 2017;8:90532–44. tions for improving the interpretation of cancer
cer results from the European EURAF cohort. 64. Le DT, Uram JN, Wang H, et al. PD-1 blockade susceptibility genetic test results. Hum Mutat
J Thorac Oncol 2015;10:1451–7. in tumors with mismatch-repair deficiency. N 2008;29:1282–91.
49. Dummer R, Ascierto PA, Gogas HJ, et al. Overall Engl J Med 2015;372:2509–20. 81. Robson M, Im SA, Senkus E, et al. Olaparib for
survival in patients with BRAF-mutant melanoma 65. Le DT, Durham JN, Smith KN, et al. Mismatch metastatic breast cancer in patients with a germline
receiving encorafenib plus binimetinib versus repair deficiency predicts response of solid tumors BRCA mutation. N Engl J Med 2017;377:523–33.
vemurafenib or encorafenib (COLUMBUS): a to PD-1 blockade. Science 2017;357:409–13. 82. Ledermann J, Harter P, Gourley C, et al. Olaparib
multicentre, open-label, randomised, phase 3 trial. 66. Allgäuer M, Budczies J, Christopoulos P, et al. maintenance therapy in platinum-sensitive
Lancet Oncol 2018;19:1315–27. Implementing tumor mutational burden (TMB) relapsed ovarian cancer. N Engl J Med 2012;366:
50. Ascierto PA, McArthur GA, Dréno B, et al. analysis in routine diagnostics—a primer for 1382–92.
Cobimetinib combined with vemurafenib in molecular pathologists and clinicians. Transl Lung 83. Moore K, Colombo N, Scambia G, et al. Mainte-
advanced BRAFV600-mutant melanoma (coB- Cancer Res 2018;7:703–15. nance Olaparib in patients with newly diagnosed
RIM): updated efficacy results from a randomised, 67. Chan TA, Yarchoan M, Jaffee E, et al. Develop- advanced ovarian cancer. N Engl J Med 2018;379:
double-blind, phase 3 trial. Lancet Oncol 2016;17: ment of tumor mutation burden as an immuno- 2495–505.
1248–60. therapy biomarker: utility for the oncology clinic. 84. Lord CJ, Ashworth A. PARP inhibitors: synthetic
51. Long GV, Eroglu Z, Infante J, et al. Long-term Ann Oncol 2018;30:44–56. lethality in the clinic. Science 2017;355:1152–8.
outcomes in patients with BRAF V600-mutant 68. Nik-Zainal S, Davies H, Staaf J, et al. Landscape of 85. Dougherty BA, Lai Z, Hodgson DR, et al. Biologi-
metastatic melanoma who received Dabrafenib somatic mutations in 560 breast cancer whole-genome cal and clinical evidence for somatic mutations in
combined with Trametinib. J Clin Oncol 2018;36: sequences. Nature 2016;534:47–54. BRCA1 and BRCA2 as predictive markers for
667–73. 69. Gröschel S, Hübschmann D, Raimondi F, et al. olaparib response in high-grade serous ovarian
52. Chapman PB, Hauschild A, Robert C, et al. Defective homologous recombination DNA repair cancers in the maintenance setting. Oncotarget
Improved survival with Vemurafenib in mela- as therapeutic target in advanced chordoma. Nat 2017;8:43653–61.
noma with BRAF V600E mutation. N Engl J Med Commun 2019;10:1635. 86. Wood RD, Mitchell M, Sgouros J, et al. Human
2011;364:2507–16. 70. Capper D, Jones DTW, Sill M, et al. DNA DNA Repair Genes. Science 2001;291:1284–9.
53. Kopetz S, Desai J, Chan E, et al. Phase II pilot methylation-based classification of central nervous 87. Heeke AL, Pishvaian MJ, Lynce F, et al. Preva-
study of Vemurafenib in patients with metastatic system tumours. Nature 2018;555:469–74. lence of homologous recombination-related gene
BRAF-mutated colorectal cancer. J Clin Oncol 71. Reck M, Rodríguez-Abreu D, Robinson AG, et al. mutations across multiple cancer types. JCO Precis
2015;33:4032–8. Pembrolizumab versus chemotherapy for PD- Oncol 2018;2:1–13.
54. Porru M, Pompili L, Caruso C, et al. Targeting L1-positive non-small-cell lung cancer. N Engl J 88. Hussain M, Daignault-Newton S,
KRAS in metastatic colorectal cancer: current Med 2016;375:1823–33. Twardowski PW, et al. Targeting androgen recep-
strategies and emerging opportunities. J Exp Clin 72. Goodman AM, Kato S, Bazhenova L, et al. Tumor tor and DNA repair in metastatic castration-
Cancer Res 2018;37:57. mutational burden as an independent predictor of resistant prostate cancer: results from NCI 9012.
55. Ohashi K, Sequist LV, Arcila ME, et al. response to immunotherapy in diverse cancers. J Clin Oncol 2018;36:991–9.
Lung cancers with acquired resistance to EGFR Mol Cancer Ther 2017;16:2598–608. 89. Pearl LH, Schierz AC, Ward SE, et al. Therapeutic
inhibitors occasionally harbor BRAF gene muta- 73. Marmarelis M, Bange E, Bagley S, et al. P1.01-64 opportunities within the DNA damage response.
tions but lack mutations in KRAS, NRAS, or impact of STK11 co-mutation on outcomes Nat Rev Cancer 2015;15:166–80.

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC


10970215, 2019, 11, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ijc.32358, Wiley Online Library on [26/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
3010 Variant classification in precision oncology

90. Scarpa A, Chang DK, Nones K, et al. Whole- 93. Cantile M, Marra L, Franco R, et al. Molecular 96. Hakem R. DNA-damage repair; the good, the bad,
genome landscape of pancreatic neuroendocrine detection and targeting of EWSR1 fusion and the ugly. EMBO J 2008;27:589–605.
tumours. Nature 2017;543:65–71. transcripts in soft tissue tumors. Med Oncol 2013; 97. Tutt A, Tovey H, Cheang MCU, et al.
91. Aisner DL, Nguyen TT, Paskulin DD, et al. 30:412. Carboplatin in BRCA1/2-mutated and
ROS1 and ALK fusions in colorectal cancer, 94. Patel JN, Fong MK, Jagosky M. Colorectal cancer triple-negative breast cancer BRCAness sub-
with evidence of intratumoral heterogeneity for biomarkers in the era of personalized medicine. groups: the TNT trial. Nat Med 2018;24:
molecular drivers. Mol Cancer Res 2014;12:111–8. J Pers Med 2019;9:E3. 628–37.
92. Soda M, Choi YL, Enomoto M, et al. Identification 95. Ciccia A, Elledge SJ. The DNA damage response: 98. Abernethy AP, Etheredge LM, Ganz PA, et al.
of the transforming EML4-ALK fusion gene in non- making it safe to play with knives. Mol Cell 2010; Rapid-learning system for cancer care. J Clin
small-cell lung cancer. Nature 2007;448:561–6. 40:179–204. Oncol 2010;28:4268–74.
Cancer Genetics and Epigenetics

Int. J. Cancer: 145, 2996–3010 (2019) © 2019 UICC

You might also like