CGC / ClinGen / VICC Classification Calculator + Emory Modification


Oncogenicity


Very Strong Evidence

OVS1: Null variant (nonsense, frameshift, canonical ±1 or 2 splice sites, initiation codon, single-exon or multiexon deletion) in a bona fide tumor suppressor gene.
  • Considering ClinGen guidance on the interpretation of loss-of-function variants.
  • Use caution interpreting pLOF variants at the extreme 3' end of a gene after the nonsense mediated decay site.
  • Use caution with splice variants that are predicted to lead to exon skipping but leave the remainder of the protein intact (in frame events).
  • MutSpliceDB maybe helpful to catalog RNA-sequencing based evidence for splice site variants effects on splicing.
  • Use caution if splice variant leads to expression of a well-known alternative iso-form that preserves tumor suppressor functionality.
  • Use caution in the presence of multiple transcripts.

Strong Evidence

OS1: Same amino acid change as a previously established oncogenic variant (using this standard) regardless of nucleotide change. Example: Val→Leu caused by either G>C or G>T in the same codon.
  • Beware of changes that impact splicing rather than the changes at the amino acid/protein level.
OS2: Well-established in vitro or in vivo functional studies, supportive of an oncogenic effect of the variant.
  • Functional studies that have been shown to be reproducible and robust are considered the most well established.
  • If OS1 is applicable, this rule can be used only if functional studies are based on the particular nucleotide change of the variant.
OS3: Located in one of the hotspots in cancerhotspots.org with at least 50 samples with a somatic variant at the same amino acid position, and the same amino acid change count in cancerhotspots.org in at least 10 samples.
  • Use caution with hotspots driven by truncating somatic variants.
  • Cancerhotspot has good coverage for many solid tumors; however, it has limited coverage for hematological, rare, and pediatric malignancies. If the somatic variant is in a tumor type not well covered by cancerhotspots.org, resources such as COSMIC or a tumor type-specific study could be used. Note that data in COSMIC is not adjusted by nucleotide mutability and varying gene-specific SNV rates.
  • For pediatric malignancies resources, PeCanPIE may be considered.
  • This rule cannot be used if OS1 is applicable, unless it is possible to observe hotspots on the basis of the particular nucleotide change.

Moderate Evidence

OM1: Located in a critical and well-established part of a functional domain (eg, active site of an enzyme).
  • This rule cannot be used if OS1 or OS3 is applicable.
OM2: Protein length changes as a result of in-frame deletions/insertions in a known oncogene or tumor suppressor gene or stop-loss variants in a known tumor suppressor gene.
  • This rule cannot be used if OVS1 is applicable
OM3: Missense variant at an amino acid residue where a different missense variant determined to be oncogenic (using this standard) has been documented. Amino acid difference from reference amino acid should be greater or at least approximately the same as for missense change determined to be oncogenic.
  • Example: p.Arg156His is oncogenic; now you observe p.Arg156Cys. This rule cannot be used if OS1 or OS3 or OM1 is applicable.
  • Beware of changes that impact splicing rather than the changes at the amino acid/protein level.
OM4: Located in one of the hotspots in cancerhotspots.org with <50 samples with a somatic variant at the same amino acid position, and the same amino acid change count in cancerhotspots.org is at least 10.
  • The difference of variant AA from reference AA should be greater or at least approximately the same as for the missense change that has already been determined to be oncogenic. See Grantham's Distance or Miyata’s distance
  • This rule cannot be used if OM1 or OM3 is applicable.
  • Use caution with hotspots driven by truncating somatic variants.
  • If the somatic variant is in a tumor type that is not covered well by cancerhotspots.org, resources such as COSMIC or a tumor type-specific study could be used.

Supporting Evidence

OP1: All used lines of computational evidence support an oncogenic effect of a variant (conservation/ evolutionary, splicing effect, etc.).
  • Because many in silico algorithms use the same or very similar input for their predictions, each algorithm should not be counted as an independent criterion.
  • Can be used only once in any evaluation of a variant.

OP2: Somatic variant in a gene in a malignancy with a single genetic etiology. Example: retinoblastoma is caused by bi-allelic RB1 inactivation.
  • A small fraction of cases may be caused by an alternative mechanism; histological similarities may cause misdiagnosis.
OP3: Located in one of the hotspots in cancerhotspots. org and the particular amino acid change count in cancerhotspots.org is below 10.
  • Use caution with hotspots driven by truncating somatic variants.
  • If somatic variant is in a tumor type that is not covered well by cancerhotspots.org, resources such as COSMIC or a tumor type–specific study could be used.
OP4: Absent from controls (or at an extremely low frequency) in gnomAD.
  • Population data for insertions/deletions may be poorly called by next-generation sequencing. Population data may contain somatic variants associated with clonal hematopoiesis.
Emory OP5: In a gene frequently seen in disease in a LOW TMB case.
  • In cases with low TMB, any mutation should be carefully evaluated because it is unlikely to be a random variant or VUS.
Emory OP6: Bi-allelic in Autosomal Recessive/Predisposition/Tumor suppressor gene.
  • Compound heterozygous mutations in tumor suppressor genes would more likely result in loss of function.
Emory OP7: Clearly not a germline variant by % in a LOW TMB case.
  • Similar to OP5, somatic mutations in cases with low TMB are more likely oncogenic and should be evaluated carefully.


Benign Effect


Very Strong Evidence

SBVS1: Minor allele frequency is >5% in gnomAD in any 5 general continental populations: African, East Asian, European (non-Finnish), Latino, and South Asian.
  • If the somatic variant is in a gene known to cause predisposition to hereditary cancer, ACMG/AMP ClinGen germline expert panel gene-specific guidelines (if they exist) must be consulted to establish a cutoff that takes disease prevalence into account.

Strong Evidence

SBS1: Minor allele frequency is >1% in gnomAD in any 5 general continental populations: African, East Asian, European (non-Finnish), Latino, and South Asian.
  • Indels may be underrepresented in gnomAD in part due to technical challenges associated with their detection.
  • Some variants in gnomAD (and similar databases) may actually be somatic and not germline, owing to clonal hematopoiesis (CHIP).
  • If the somatic variant is in a gene known to cause predisposition to hereditary cancer, ACMG/AMP ClinGen germline expert panel gene-specific guidelines (if they exist) must be consulted to establish a cutoff that takes disease prevalence into account.
SBS2: Well-established in vitro or in vivo functional studies show no oncogenic effects.

Supporting Evidence

SBP1: All used lines of computational evidence suggest no effect of a variant (conservation/evolutionary, splicing effect, etc.).
  • Because many in silico algorithms use the same or very similar input for their predictions, each algorithm cannot be counted as an independent criterion. Can be used only once in any evaluation of a variant.
SBP2: A synonymous (silent) variant for which splicing prediction algorithms predict no effect on the splice consensus sequence nor the creation of a new splice site and the nucleotide is not highly conserved.



Final Calculation


CGC Classification: VUS

The total score is: 0

Emory v0.1 Classification: VUS

The total score is: 0

Calculation Explanation

CGC

Benign
<= -7
Likely Benign
-1 to -6
VOUS
0 to 5
Likely Oncogenic
6 to 9
Oncogenic
>=10

Emory v0.1

Benign
<= -7
Likely Benign
-1 to -6
VOUS
0 to 4
Potential Oncogenic
5 to 6
Likely Oncogenic
7 to 9
Oncogenic
>=10

Calculation based on the following publication: Horak P, et al. Standards for the classification of pathogenicity of somatic variants in cancer (oncogenicity): Joint recommendations of ClinGen, CGC, and VICC. Genetics in Medicine 2022;24(5):986-998. (PMID: 35101336)


JavaScript-based webpage written by Thomas Schneider, MD; contents updated by Linsheng Zhang, MD, PhD, Department of Pathology and Laboratory Medicine, Emory University School of Medicine. October 2022