1Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
2Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
3Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
4Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
5Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
6Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
7Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
8Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
9Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
10Division of Oncology and Hematology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
11Division of Medical Oncology, Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
12Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
13Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
14Division of Medical Oncology, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
15Division of Hematology/Oncology, Department of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
16Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
17Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
18Department of Pathology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Korea
19Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
20Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
21Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
© The Korean Society of Pathologists/The Korean Society for Cytopathology
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Ethics Statement
Not applicable.
Availability of Data and Material
The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.
Code Availability
Not applicable.
Author Contributions
Conceptualization: JHK, WSK, MK. Data curation: all authors. Writing—original draft: all authors. Writing—review & editing: all authors. Approval of final manuscript: all authors.
Conflicts of Interest
S.Y.P., the editor-in-chief and H.S.L. and S.O.Y., contributing editors of the Journal of Pathology and Translational Medicine, were not involved in the editorial evaluation or decision to publish this article. All remaining authors have declared no conflicts of interest.
Funding Statement
This study was supported by the National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea (HA22C0052).
Level | Clinical implication | Required level of evidence |
---|---|---|
1 | Treatment should be considered standard of care | MFDS, FDA, EMA or equivalent-approved drug OR Prospective, randomized, phase III trials showing the benefit of survival endpoints |
2 | Treatment would be considered | Prospective phase I/II trials show clinical benefita |
3 | Clinical trials to be discussed with patients | A: Retrospective study or case series show potential clinical benefit in a specific tumor type |
B: Clinical studies show potential clinical benefit in other indications | ||
4 | Preclinical data only, lack of clinical data | Preclinical evidence suggests the potential benefit |
G | Suspicious germline variant on tumor tissue NGS | Suggestive actionable germline variant on tumor tissue testing |
R | Predictive biomarker of resistance | FDA-recognized predictive biomarker of resistance |
KPMNG, Korean Precision Medicine Networking Group; K-CAT, KPMNG scale of Clinical Actionability of molecular Targets; MFDS, Ministry of Food and Drug Safety; FDA, U.S. Food and Drug Administration; EMA, European Medicines Agency; NGS, next-generation sequencing.
aProspective phase I/II trials supporting level 2 targets include clinical trials across tumor types such as basket trials. In this case, the clinical benefit needs to be judged by expert consensus.
Gene/Alteration | Matched treatment | K-CAT | Reference |
---|---|---|---|
NTRK fusion | Entrectinib | 1 | [19,20] |
Larotrectinib | |||
BRAF V600E | Dabrafenib+trametinib (except colorectal cancer) | 1 | [11-17] |
RET fusion | Selpercatinib | 1 | [18] |
Microsatellite instability–high/Mismatch repair deficiency | Pembrolizumab | 1 | [9,40] |
High tumor mutation burden | Pembrolizumab | 1 | [10] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
EGFR | Exon 19 in-frame deletions, L858R, G719X, L861Q, S761I | 30–46 | 1 | [41-45] |
T790M | 50 of treated EGFR mutant NSCLC | 1, R | [29,46,47] | |
Exon 20 in-frame insertion | 3 | 1 | [48,49] | |
BRAF | V600E | 2–4 | 1 | [12,13,50] |
ALK | Rearrangement/Fusions | 3–5 | 1 | [36,37,51,52] |
KRAS | G12C | 13 | 1 | [53,54] |
MET | Exon 14 in-frame deletions, Exon 14 splice mutations | 3–4 | 1 | [55,56] |
Amplification | 3–5 | 2 | [56] | |
RET | Rearrangement/Fusions | 1.7 | 1 | [57,58] |
ROS1 | Rearrangement/Fusions | 2.6 | 1 | [59,60] |
ERBB2 | Exon 20 in-frame insertion | 2.3 | 1 | [61-64] |
Amplification | 2.4–38 | 2 | [65,66] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
ERBB2 | Amplifications | 15–20 | 1 | [67-71] |
Oncogenic mutations | 4 | 2 | [72,73] | |
PIK3CAa | Oncogenic mutations | 30–40 | 1 | [74,75] |
BRCA1/2 | Germline oncogenic mutations | 4 | 1 | [76,77] |
BRCA1/2b | Somatic oncogenic mutationsc | 3 | 2 | [78-80] |
PTEN | Oncogenic mutations | 7 | 2 | [81,82] |
ESR1 | Oncogenic mutations (mechanism of resistance) | 10 | R | [83] |
AKT1 | E17K | 5 | 2 | [82,84] |
PALB2d | Germline oncogenic mutations | 0.5–1 | 2 | [79,85] |
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; PARP, poly(adenosine diphosphate [ADP]–ribose) polymerase; HRD, homologous recombination deficiency.
aThis applies only to breast cancer that is hormone receptor-positive/HER2-negative and has mutations including E542K, E545A, H1047R, H1047Y, Q546E, H1047L, Q546R, E545G, E545D, E545K, C420R. Other oncogenic mutations not included in this category, caution is needed, since it is unknown whether other mutations are associated with response to phosphoinositide 3-kinase inhibitor therapy;
bPhase III trials of PARP inhibitors have been conducted in patients with germline BRCA mutations, and their therapeutic effects have been confirmed. In some studies, the effects of PARP inhibitors have also been reported in patients with somatic BRCA mutations, and somatic tumor sequencing can identify many germline BRCA mutations;
cIn addition to BRCA 1/2, there are several other genes associated with homologous recombination deficiency, including ATRX, BLM, BRIP1, CHEK2, FANCA/C/D2/E/F/G/L, MRE11A, NBN, PALB2, and RAD50. Although the discovery frequency of each gene is very low, they are collectively found in approximately 8% of all breast cancers;
dThere are multiple germline mutations associated with HRD in breast cancer patients, but this table only includes the two most frequent ones.z
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
ERBB2 | Amplification | 3.9–10 | 2 | [86] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
ERBB2 | Amplification | 15 | 1 | [87-89] |
FGFR2a | Amplification | 5 | 2 | [90] |
MET | Amplification | 2–5 | 2 | [91] |
EGFR | Amplification | 5–10 | 3A | [92] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
KRAS | Oncogenic mutations | 40 | R | [93,94] |
NRAS | Oncogenic mutations | 3–5 | R | [95,96] |
BRAF | V600E | 5–10 | 1 | [96-98] |
Mismatch repair deficiency | MSI-H/MMR-D | 4–5 | 1 | [99,100] |
ERBB2 | Amplification | 4–5 | 1 | [101] |
KRAS | G12C | 3 | 2 | [102,103] |
POLE | Exonuclease domain mutations | 1–3 | 2 | [104-106] |
Gene | Alteration | Prevalence (%)a | K-CAT | Reference |
---|---|---|---|---|
NOTCH1, 2, 3 | Oncogenic mutations | 10–12 | 2 | [107,108] |
ERRB2 | Amplification | 30–40 | 2 | [109-111] |
FGFR1, 3 | Amplification/Oncogenic mutations | 1–7 | 2 | [112-114] |
MET | Amplification | 1 | 3A | [115,116] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
BRCA 1/2 | Germline oncogenic mutations | 1–4 | 1 | [117,118] |
PALB2 | Oncogenic mutations | 0.6 | 2 | [118] |
KRAS | G12C | 2–3 | 2 | [119,120] |
PIK3CA | Oncogenic mutations | 3 | 3A | [121] |
ERBB2 | Amplifications/Oncogenic mutations | 1–2 | 3A | [72,122] |
ALK | Rearrangement/Fusions | < 1 | 3A | [123] |
NRG1 | Rearrangement/Fusions | 1 | 3A | [124] |
ROS1 | Rearrangement/Fusions | < 1 | 3A | [125] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
IDH1 | Oncogenic mutations | 10–23 | 1 | [126,127] |
FGFR2 | Rearrangement/Fusions | 8–14 | 1 | [128-130] |
BRAF | V600E | 5 | 1 | [14,15] |
ERBB2 | Amplification | 10 | 2 | [131-133] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
ERBB2 | Amplification | 30 of uterine serous carcinoma | 2 | [134] |
AKT1 | E17K | 2 | 2 | [84] |
POLEa | Oncogenic mutations | 5–15 | NA | [135,136] |
TP53ab | Oncogenic mutations | 5–15 | NA | [135] |
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; NGS, next-generation sequencing; IHC, immunohistochemistry; TCGA, The Cancer Genome Atlas; MMR, mismatch repair.
aAdjuvant treatment of endometrial cancer based on molecular classification;
bConsidering the coverage limitations of NGS for detecting p53 loss, a combined IHC approach is recommended. The TCGA approach results in the molecular stratification of endometrial cancer (EC) into four distinct molecular groups [137]; (1) ultramutated (> 100 mut/Mb) with pathogenic variations in the exonuclease domain of DNA polymerase epsilon (POLE)-ultramutated (POLEmut), (2) hypermutated (10–100 mut/Mb), microsatelliteunstable, (3) somatic copy number-high with frequent pathogenic variants in TP53, and (4) an MMR-proficient, low somatic copy number aberration subgroup with a low mutational burden. Extensive research on these surrogate markers has revealed a strong correlation with clinical outcome, thus proving their prognostic value [138-140]. POLEmut EC had generally has an excellent clinical outcome, while p53-abn EC has the worst, regardless of risk category, type of adjuvant treatment, tumor type, or grade. Adjuvant chemotherapy is beneficial in for patients with p53mut EC, while treatment de-escalation is being explored in patients with POLEmut EC [139], which exhibits a favorable outcome [141]. Consequently, all EC pathology specimens should undergo molecular classification, independent of histological type, using well-established IHC staining for p53 and MMR proteins (MLH1, PMS2, MSH2, MSH6), in conjunction with targeted tumor sequencing (POLE hotspot analysis). While POLE hotspot analysis is currently unavailable in Korea, and most NGS panels include the POLE gene, it has been incorporated into the recommendations. Moreover, since IHC plays a wellestablished role in identifying p53 mutations and NGS target sequencing of TP53 is insufficient to identify all loss of P53 function, IHC confirmation of p53 is recommended over NGS testing as a priority.
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
BRCA 1/2 | Oncogenic mutations | 5–15 | 1 | [142-149] |
HRD score | GIS, LOH | 50 | 1 | [142-144,146,148] |
AKT1 | E17K | 2 | 2 | [84] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
FGFR3 | Oncogenic mutations Rearrangement/Fusions | 13–15 | 1 | [150] |
FGFR2 | Rearrangement/Fusions | Unknown | 1 | [150] |
ERCC2 | Oncogenic mutations | 9–12 | 3A | [151,152] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
BRCA2 | Germline and/or somatic oncogenic mutations | 3–13 | 1 | [153,154] |
BRCA1 | Germline and/or somatic oncogenic mutations | 1 | 1 | [153,154] |
ATM | Oncogenic mutations | 6–7 | 1 | [153,154] |
BRIP1, BARD1, CDK12, CHEK1, CHEK2, FANCL, PALB2, RAD51B, RAD51C, RAD51D, RAD54L | Oncogenic mutations | < 1–5 | 1 | [153,154] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
VHL | Germline oncogenic mutations | 0.2 | 1 | [155] |
FH | Germline oncogenic mutations | 0.5 | 3A | [156,157] |
ALK | Rearrangement/Fusions | 0.3–0.5 | 3A | [158] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
BRAF | V600E/K | 35–50 | 1 | [11,159-162] |
V600 (excluding V600E/K) | ~5 | 1 | [163] | |
KIT | D579del and 12 other oncogenic mutations | 1–7 | 2 | [164,165] |
NRAS | Oncogenic mutations | ~20 | 2 | [166,167] |
BRAF | Rearrangement/Fusions | 3–7 | 3A | [168,169] |
K601, L597 | < 1 | 3A | [170-173] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
KIT | Oncogenic mutations | ~75–80 in GIST | 1 | [174,175] |
PDGFRA | Oncogenic mutations | ~8–10 in GIST | 1 | [175-177] |
PDGFB | Rearrangement/Fusions mostly COL1A1::PDGFB | ~90 in DFSP | 1 | [178,179] |
ALK | Rearrangement/Fusions | ~50 in IMT | 1 | [180-182] |
SMARCB1 | Deletion | ~83 in ES | 2 | [183] |
IDH1 | Oncogenic mutations | ~65 in chondrosarcoma | 2 | [184] |
TSC2 | Oncogenic mutations | ~30 in PEComa | 2 | [185,186] |
MDM2 | Amplification | ~90 in WDLPS/DDLPS; frequent in IS, low grade OSA | 2 | [187,188] |
CDK4 | Amplification | ~90 in WDLPS/DDLPS; frequent in IS, low grade OSA | 2 | [187,189] |
MET | Oncogenic mutations, Rearrangement/Fusions, Amplification | < 1% | 2 | [190] |
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; GIST, gastrointestinal stromal tumor; DFSP, dermatofibrosarcoma protuberans; ES, epithelioid sarcoma; IMT, inflammatory myofibroblastic tumor; WDLPS/DDLPS, well-differentiated/de-differentiated liposarcoma; IS, intimal sarcoma; OSA, osteosarcoma.
Level | Clinical implication | Required level of evidence |
---|---|---|
1 | Treatment should be considered standard of care | MFDS, FDA, EMA or equivalent-approved drug OR Prospective, randomized, phase III trials showing the benefit of survival endpoints |
2 | Treatment would be considered | Prospective phase I/II trials show clinical benefit |
3 | Clinical trials to be discussed with patients | A: Retrospective study or case series show potential clinical benefit in a specific tumor type |
B: Clinical studies show potential clinical benefit in other indications | ||
4 | Preclinical data only, lack of clinical data | Preclinical evidence suggests the potential benefit |
G | Suspicious germline variant on tumor tissue NGS | Suggestive actionable germline variant on tumor tissue testing |
R | Predictive biomarker of resistance | FDA-recognized predictive biomarker of resistance |
Gene/Alteration | Matched treatment | K-CAT | Reference |
---|---|---|---|
NTRK fusion | Entrectinib | 1 | [19,20] |
Larotrectinib | |||
BRAF V600E | Dabrafenib+trametinib (except colorectal cancer) | 1 | [11-17] |
RET fusion | Selpercatinib | 1 | [18] |
Microsatellite instability–high/Mismatch repair deficiency | Pembrolizumab | 1 | [9,40] |
High tumor mutation burden | Pembrolizumab | 1 | [10] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
EGFR | Exon 19 in-frame deletions, L858R, G719X, L861Q, S761I | 30–46 | 1 | [41-45] |
T790M | 50 of treated EGFR mutant NSCLC | 1, R | [29,46,47] | |
Exon 20 in-frame insertion | 3 | 1 | [48,49] | |
BRAF | V600E | 2–4 | 1 | [12,13,50] |
ALK | Rearrangement/Fusions | 3–5 | 1 | [36,37,51,52] |
KRAS | G12C | 13 | 1 | [53,54] |
MET | Exon 14 in-frame deletions, Exon 14 splice mutations | 3–4 | 1 | [55,56] |
Amplification | 3–5 | 2 | [56] | |
RET | Rearrangement/Fusions | 1.7 | 1 | [57,58] |
ROS1 | Rearrangement/Fusions | 2.6 | 1 | [59,60] |
ERBB2 | Exon 20 in-frame insertion | 2.3 | 1 | [61-64] |
Amplification | 2.4–38 | 2 | [65,66] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
ERBB2 | Amplifications | 15–20 | 1 | [67-71] |
Oncogenic mutations | 4 | 2 | [72,73] | |
PIK3CA |
Oncogenic mutations | 30–40 | 1 | [74,75] |
BRCA1/2 | Germline oncogenic mutations | 4 | 1 | [76,77] |
BRCA1/2 |
Somatic oncogenic mutations |
3 | 2 | [78-80] |
PTEN | Oncogenic mutations | 7 | 2 | [81,82] |
ESR1 | Oncogenic mutations (mechanism of resistance) | 10 | R | [83] |
AKT1 | E17K | 5 | 2 | [82,84] |
PALB2 |
Germline oncogenic mutations | 0.5–1 | 2 | [79,85] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
ERBB2 | Amplification | 3.9–10 | 2 | [86] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
ERBB2 | Amplification | 15 | 1 | [87-89] |
FGFR2 |
Amplification | 5 | 2 | [90] |
MET | Amplification | 2–5 | 2 | [91] |
EGFR | Amplification | 5–10 | 3A | [92] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
KRAS | Oncogenic mutations | 40 | R | [93,94] |
NRAS | Oncogenic mutations | 3–5 | R | [95,96] |
BRAF | V600E | 5–10 | 1 | [96-98] |
Mismatch repair deficiency | MSI-H/MMR-D | 4–5 | 1 | [99,100] |
ERBB2 | Amplification | 4–5 | 1 | [101] |
KRAS | G12C | 3 | 2 | [102,103] |
POLE | Exonuclease domain mutations | 1–3 | 2 | [104-106] |
Gene | Alteration | Prevalence (%) |
K-CAT | Reference |
---|---|---|---|---|
NOTCH1, 2, 3 | Oncogenic mutations | 10–12 | 2 | [107,108] |
ERRB2 | Amplification | 30–40 | 2 | [109-111] |
FGFR1, 3 | Amplification/Oncogenic mutations | 1–7 | 2 | [112-114] |
MET | Amplification | 1 | 3A | [115,116] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
BRCA 1/2 | Germline oncogenic mutations | 1–4 | 1 | [117,118] |
PALB2 | Oncogenic mutations | 0.6 | 2 | [118] |
KRAS | G12C | 2–3 | 2 | [119,120] |
PIK3CA | Oncogenic mutations | 3 | 3A | [121] |
ERBB2 | Amplifications/Oncogenic mutations | 1–2 | 3A | [72,122] |
ALK | Rearrangement/Fusions | < 1 | 3A | [123] |
NRG1 | Rearrangement/Fusions | 1 | 3A | [124] |
ROS1 | Rearrangement/Fusions | < 1 | 3A | [125] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
IDH1 | Oncogenic mutations | 10–23 | 1 | [126,127] |
FGFR2 | Rearrangement/Fusions | 8–14 | 1 | [128-130] |
BRAF | V600E | 5 | 1 | [14,15] |
ERBB2 | Amplification | 10 | 2 | [131-133] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
ERBB2 | Amplification | 30 of uterine serous carcinoma | 2 | [134] |
AKT1 | E17K | 2 | 2 | [84] |
POLE |
Oncogenic mutations | 5–15 | NA | [135,136] |
TP53 |
Oncogenic mutations | 5–15 | NA | [135] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
BRCA 1/2 | Oncogenic mutations | 5–15 | 1 | [142-149] |
HRD score | GIS, LOH | 50 | 1 | [142-144,146,148] |
AKT1 | E17K | 2 | 2 | [84] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
FGFR3 | Oncogenic mutations Rearrangement/Fusions | 13–15 | 1 | [150] |
FGFR2 | Rearrangement/Fusions | Unknown | 1 | [150] |
ERCC2 | Oncogenic mutations | 9–12 | 3A | [151,152] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
BRCA2 | Germline and/or somatic oncogenic mutations | 3–13 | 1 | [153,154] |
BRCA1 | Germline and/or somatic oncogenic mutations | 1 | 1 | [153,154] |
ATM | Oncogenic mutations | 6–7 | 1 | [153,154] |
BRIP1, BARD1, CDK12, CHEK1, CHEK2, FANCL, PALB2, RAD51B, RAD51C, RAD51D, RAD54L | Oncogenic mutations | < 1–5 | 1 | [153,154] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
VHL | Germline oncogenic mutations | 0.2 | 1 | [155] |
FH | Germline oncogenic mutations | 0.5 | 3A | [156,157] |
ALK | Rearrangement/Fusions | 0.3–0.5 | 3A | [158] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
BRAF | V600E/K | 35–50 | 1 | [11,159-162] |
V600 (excluding V600E/K) | ~5 | 1 | [163] | |
KIT | D579del and 12 other oncogenic mutations | 1–7 | 2 | [164,165] |
NRAS | Oncogenic mutations | ~20 | 2 | [166,167] |
BRAF | Rearrangement/Fusions | 3–7 | 3A | [168,169] |
K601, L597 | < 1 | 3A | [170-173] |
Gene | Alteration | Prevalence (%) | K-CAT | Reference |
---|---|---|---|---|
KIT | Oncogenic mutations | ~75–80 in GIST | 1 | [174,175] |
PDGFRA | Oncogenic mutations | ~8–10 in GIST | 1 | [175-177] |
PDGFB | Rearrangement/Fusions mostly COL1A1::PDGFB | ~90 in DFSP | 1 | [178,179] |
ALK | Rearrangement/Fusions | ~50 in IMT | 1 | [180-182] |
SMARCB1 | Deletion | ~83 in ES | 2 | [183] |
IDH1 | Oncogenic mutations | ~65 in chondrosarcoma | 2 | [184] |
TSC2 | Oncogenic mutations | ~30 in PEComa | 2 | [185,186] |
MDM2 | Amplification | ~90 in WDLPS/DDLPS; frequent in IS, low grade OSA | 2 | [187,188] |
CDK4 | Amplification | ~90 in WDLPS/DDLPS; frequent in IS, low grade OSA | 2 | [187,189] |
MET | Oncogenic mutations, Rearrangement/Fusions, Amplification | < 1% | 2 | [190] |
KPMNG, Korean Precision Medicine Networking Group; K-CAT, KPMNG scale of Clinical Actionability of molecular Targets; MFDS, Ministry of Food and Drug Safety; FDA, U.S. Food and Drug Administration; EMA, European Medicines Agency; NGS, next-generation sequencing. Prospective phase I/II trials supporting level 2 targets include clinical trials across tumor types such as basket trials. In this case, the clinical benefit needs to be judged by expert consensus.
FDA, U.S. Food and Drug Administration; K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; NSCLC, non–small cell lung cancer.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; PARP, poly(adenosine diphosphate [ADP]–ribose) polymerase; HRD, homologous recombination deficiency. This applies only to breast cancer that is hormone receptor-positive/HER2-negative and has mutations including E542K, E545A, H1047R, H1047Y, Q546E, H1047L, Q546R, E545G, E545D, E545K, C420R. Other oncogenic mutations not included in this category, caution is needed, since it is unknown whether other mutations are associated with response to phosphoinositide 3-kinase inhibitor therapy; Phase III trials of PARP inhibitors have been conducted in patients with germline In addition to There are multiple germline mutations associated with HRD in breast cancer patients, but this table only includes the two most frequent ones.z
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; ctDNA, circulating tumor DNA. FGFR2b overexpression or
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; MSI-H, microsatellite instability–high; MMRD, mismatch repair deficiency.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets. The above prevalence is about the representative subtype among various subtypes of head and neck cancer.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; NGS, next-generation sequencing; IHC, immunohistochemistry; TCGA, The Cancer Genome Atlas; MMR, mismatch repair. Adjuvant treatment of endometrial cancer based on molecular classification; Considering the coverage limitations of NGS for detecting p53 loss, a combined IHC approach is recommended. The TCGA approach results in the molecular stratification of endometrial cancer (EC) into four distinct molecular groups [
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; HRD, homologous recombination deficiency; GIS, genomic instability scores; LOH, loss of heterozygosity.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets.
K-CAT, Korean Precision Medicine Networking Group scale of Clinical Actionability of molecular Targets; GIST, gastrointestinal stromal tumor; DFSP, dermatofibrosarcoma protuberans; ES, epithelioid sarcoma; IMT, inflammatory myofibroblastic tumor; WDLPS/DDLPS, well-differentiated/de-differentiated liposarcoma; IS, intimal sarcoma; OSA, osteosarcoma.