Open access peer-reviewed chapter

Challenges and Perspectives of the Risk Assessment of the Genetic Susceptibility to Cancer in the Next-Generation Sequencing Era

Written By

Israel Gomy

Submitted: 24 July 2017 Reviewed: 10 November 2017 Published: 23 December 2017

DOI: 10.5772/intechopen.72379

From the Edited Volume

Risk Assessment

Edited by Valentina Svalova

Chapter metrics overview

1,156 Chapter Downloads

View Full Metrics

Abstract

The risk assessment of the genetic susceptibility to cancer is the process of addressing and communicating the genetic risks to individuals and families with cancer. The recent breakthroughs of the next-generation sequencing era are adding new challenges to the precision clinical care.

Keywords

  • susceptibility
  • next-generation sequencing
  • cancer genetics

1. Introduction

New molecular biology technologies, such as whole-exome and whole-genome sequencing have been shedding new light on the understanding of inherited cancer susceptibility. At the same time, translational oncology researches on somatic and germline mutations in actionable genes have been opening new dilemmas of the next-generation sequencing era. A critical issue of the so-called precision medicine is the genetic counseling of individuals with cancer susceptibility.

Susceptibility to cancer depends on the penetrance of germline variants or inherited alleles, which may be classified into three groups such as highly penetrant, moderately penetrant and lowly penetrant alleles.

Alleles with high penetrance have the highest lifetime risk of cancer, frequently more than 10 times the relative risk, dramatically affecting the quality of life and decreasing its expectancy. More than 50 rare Mendelian cancer syndromes are caused by germline mutations affecting either tumor suppressor genes, DNA repair genes or proto-oncogenes, mostly with autosomal dominant inheritance (Table 1).

SyndromeGeneMutation statusPenetranceTumors
Hereditary breast and/or ovarian cancerBRCA1HeterozygousHighBreast cancer
BRCA2Ovarian cancer
RAD51 (B,C,D)ModeratePancreatic cancer
ATMModerateProstate cancer
CHEK2ModerateColorectal cancer
Lynch syndromeMLH1HeterozygousHighColorectal cancer
MSH2Endometrial cancer
MSH6Ovarian cancer
PMS2Gastric cancer
EPCAMLeukemia, lymphoma
MMR cancer syndromeMMR genesHomozygousHighRhabdomyosarcoma
Familial adenomatous polyposisAPCHeterozygousHighGastrointestinal adenomas
Colorectal cancer
Duodenal cancer
MYH-associated polyposisMUTYHHomozygousHighColorectal cancer
Polymerase proofreading-associated polyposisPOLEHeterozygoushighColorectal cancer
POLD1Endometrial cancer
Bloom syndromeBLM1HomozygousHighLeukemia
Colorectal cancer
Wilms tumor
Nijmegen syndromeNBS1HomozygousHighLymphoma
Medulloblastoma
Rhabdomyosarcoma
Fanconi anemiaFANC genes (includes BRCA2, PALB2, BRIP1)HomozygousHighLeukemia
Medulloblastoma
Wilms tumor
Li-Fraumeni syndromeTP53HeterozygousHighBreast cancer
Li-Fraumeni-like syndromeCHEK2ModerateSarcoma
Adrenocortical cancer
Brain tumor
Cowden syndromePTENHeterozygousHighHamartomatous polyps
Skin tumors
Breast cancer
Thyroid cancer
Endometrial cancer
Hereditary diffuse gastric cancerCDH1HeterozygousHighGastric cancer (diffuse)
Breast cancer (lobular)
Peutz-Jeghers syndromeSTK11HeterozygousHighHamartomatous polyps
Colorectal
Small bowel
Breast cancer
Pancreatic cancer
Juvenile polyposisSMAD4HeterozygousHighHamartomatous polyps
BMPR1AColorectal cancer
Pancreatic cancer
Melanoma syndromesCDKN2AHeterozygousHighMalignant melanoma
CDK4Pancreatic cancer
NeurofibromatosisNF1HeterozygousHighVestibular schwannoma
NF2Meningioma
Neurofibroma
Optic glioma
Tuberous sclerosisTSC1HeterozygousHighRenal angiomyolipoma
TSC2Subependymoma
Giant cell astrocytoma
Von Hippel-Lindau syndromeVHLHeterozygousHighHemangioblastomas
Renal cell cancer
Pheochromocytoma
Chuvash polycythemiaHomozygousHighVertebral angiomas
Birt-Hogg-Dubè syndromeFLCNHeterozygousHighRenal cell cancer
Skin tumors
Papillary renal cancer syndromesFHHeterozygousHighRenal cell cancer
MET
RetinoblastomaRB1HeterozygousHighRetinoblastoma
Hereditary ParagangliomaSDH (A, B, C, D)HeterozygousHighParaganglioma
Pheochromocytoma
Multiple endocrine neoplasia 1MEN1HeterozygousHighPituitary adenoma
Multiple endocrine neoplasia 2RETParathyroid adenoma
Medullar thyroid cancer
Pheochromocytoma

Table 1.

Hereditary cancer syndromes.

Alleles with moderate or intermediate penetrance increase the relative risk of about two to five times. Although they are rare in most populations, they may be frequently found in populations with consanguineous families due to founder effects. Affected relatives can be often identified, but the reduced penetrance of the alleles may skip generations and jeopardizes the family history.

Lowly penetrant alleles were discovered by genome-wide association studies (GWAS) and may put individuals to risk of cancer at slightly higher rates than those of the general population. This is due to a polygenic model, in which several alleles, mainly single nucleotide polymorphisms (SNPs), each one carrying a low risk, combine additively or multiplicatively to confer a range of risks in the population. In this model, individuals with few alleles would be at a reduced risk, whereas those with many alleles might suffer a lifetime risk as high as 50% [1]. It is estimated that more than 100 common variants with low risk may contribute to cancer susceptibility. Actually, they explain part of the excess familial risk, and the so-called “missing heritability” remains largely unknown [2]. Thus, it is very important to identify lowly penetrant alleles responsible for cancer genetic susceptibility. Most of these alleles are intergenic—lie between genes—and many neighbor tumor suppressor genes and proto-oncogenes, possibly affecting their expression. Nowadays, with the advance of next-generation sequencing and genotyping assays, more variants have been identified, shedding new light on the genomic architecture of the inherited susceptibility of cancer.

Advertisement

2. Risk assessment of the genetic susceptibility to cancer

The risk assessment of the genetic susceptibility to cancer (RAGSC) is a process to evaluate a personal risk of carrying a germline variant that is associated to the cancer development. RAGSC may be performed through statistical models that incorporate factors such as personal and familial history of tumors, ethnic background, and so on [3]. The advent of new sequencing technologies and bioinformatics has led to improvements of estimating more precisely risks of germline variants in many genes and assessing empiric risks of cancer.

Being part of this dynamic process [4], genetic counseling involves the analysis of pedigrees and risk assessment models to determine whether a family history is suggestive of sporadic, familial or hereditary cancer [5]. The main goal of genetic counseling is to inform susceptible individuals about their chances of developing cancer, helping them to make decisions about genetic testing, screening, prevention and treatments. Pretest and posttest genetic counseling are essential for the efficacy of implementing evidence-based protocols, in terms of reducing mortality rates [6].

Table 2 summarizes the RAGSC process. Three main risk categories can be derived on the basis of patient and family genetic information. In the low-risk category (near-population risk), management is based on population screening, and genetic tests are generally not cost-effective; in the moderate-risk group, genetic counseling, genetic testing and management are individual-based; in the high-risk group, genetic counseling, testing and management are evidence-based and improve survival [7].

Average riskHighModerate/intermediateLow/populational
Personal/family historyMendelian syndromesFamilial aggregationSporadic
Genetic testingSingle gene sequencing/NGS panels/WGS/WESNGS panels/WGS/WESDTC&/WGS/SNP genotyping
Genetic counselingMandatoryAdvisableAvailable
ManagementEvidence-basedIndividual-based1Not validated

Table 2.

Overview of the risk assessment of the genetic susceptibility to cancer.

Some evidence-based screening recommendations exist for breast and colorectal cancers.


Restricted by the US Food and Drug Administration.


DTC: direct-to-consumer tests; WGS: whole-genome sequencing; WES: whole-exome sequencing.

Advertisement

3. Referrals for RAGSC

Besides sex and age, familial history is the main unmodifiable risk factor of developing cancer.

Assessing the risk factors of cancer in an individual or family is complex and raises psychological, social and ethical issues. It requires the understanding of areas of medical genetics and oncology, besides the ability of communication, and it demands more time than just a regular consultation. The American Society of Clinical Oncology (ASCO), the National Society of Genetic Counselors (NSGC) and the Oncology Nursing Society (ONS) have published guidelines for the practice of genetic counseling, risk assessment and genetic testing [6, 8]. Moreover, it includes management of at-risk individuals so that they can make informed choices about cancer screening, prevention and targeted therapies [9]. In Table 3, there are some indications of referral for RAGSC.

Personal history
Early onset of cancer diagnosis (e.g., breast cancer <45 years, colorectal cancer <50 years)
Multiple associated primary cancers: breast/ovary, colorectal/endometrium
Male breast cancer
Ovarian, fallopian tube, primary peritoneal cancer
Breast cancer and thyroid, sarcoma, adrenocortical carcinoma
Multiple colon polyps (>10 cumulative)
Colorectal or endometrial cancer with microsatellite instability and/or lack of expression of mismatch repair protein(s) by immunohistochemistry
Family history
Three close relatives (same side of family) with cancer of the same or syndromically related type (breast/ovary, colorectal/endometrium)
Two close relatives (same side of family) with cancer of the same or related type with at least one affected under 50 years
One first-degree relative with early onset cancer (breast <45 years, colorectal <50 years)
One fist-degree relative with multiple primary cancers
Two or more relatives with uncommon cancers (sarcoma, glioma, hemangioblastoma, etc.)
Relatives of patients with known BRCA, APC, MYH, Lynch syndrome mutations
Many relatives with cancer but no criteria for testing

Table 3.

Referrals for hereditary cancer risk assessment.

Advertisement

4. Next-generation sequencing

In 2013, at first, Roberts and Klein reported the use of next-generation sequencing (NGS) to identify a hereditary cancer syndrome. They found pathogenic germline variants in the ATM gene of six pancreatic cancer relatives from two different kindreds [10]. Jaeger et al. used whole-genome sequencing for the description of hereditary mixed polyposis syndrome [11].

More recently, multigene NGS panels have been used to analyze many highly and moderately penetrant variants. Although they use the same NGS technology, there is less information on predefined genes. In comparison with single-gene sequencing, panels are more time- and cost-efficient in many cases such as (1) when there is genetic or locus heterogeneity, (2) when there are actionable mutations in several genes and (3) when phenotype or family history is too unspecific or noninformative (e.g., adoption) [12].

One advantage of NGS is the possibility of including multiple genes in panels tailored to a certain familial aggregation of tumors such as breast or colon cancer. However, because of its economic viability, NGS has shifted the phenotype-driven hypothesis approach that is based on the characteristics of the syndrome. Slavin et al. found some interesting results about multigene panels. When they included only high-risk genes, the results were seldom positive, and there were more variants of unknown significance (VUS), probably because of the inclusion of more genes in the so-called “off-phenotype” pan-cancer panels [13]. Recently, evidence-based guidelines have included the utilization of multigene testing for hereditary breast and ovarian cancer risk assessment [14].

An important disadvantage of NGS is the probability of disclosing inconclusive or undetermined results. The interpretation of a VUS based on phenotype and genotype data is a difficult task and often jeopardizes the genetic counseling process. Choosing a panel with limited genes of high clinical utility specifically driven to the phenotype instead of pan-cancer panels with many low-risk genes can diminish the chances of finding variants with stressful interpretation [13]. Moreover, databases of variants with high and moderate risks are often not population-specific and may lead to misinterpretation of results.

Some ethical challenges are critical for implementing NGS in the clinics.

In March 2013, the American College of Medical Genetics and Genomics (ACMG) published recommendations on the reporting of incidental or secondary findings from NGS. The ACMG suggested the identification of 56 genes whose variants result in a high risk of developing a severe disease. Germline mutations of 16 of these genes cause hereditary cancer syndromes (Table 4) [15].

SyndromeGene
Li-FraumeniTP53
Peutz-JeghersSTK11
Familial adenomatous polyposisAPC
Von-Hippel LindauVHL
Multiple endocrine neoplasiaMEN1 (type 1); RET (type 2)
HamartomatosisPTEN
RetinoblastomaRB
Paraganglioma-pheochromocytomaSDHAF2, SDHB, SDHC, SDHD
Tuberous sclerosis complexTSC1, TSC2
Neurofibromatosis type 2NF2
WT1-related Wilms tumorWT1

Table 4.

ACMG list of hereditary cancer syndromes.

In 2015, the ACMG reviewed it based on the consensus that patients could opt out of the analysis of secondary findings. This decision must be made during the process of informed consent, before testing. As some of these cancer syndromes may have the onset during childhood, these guidelines may also be applied to children, whose parents should make the decision whether or not to opt out [16].

A recent review showed that following the recommendations of international human genetic societies, parents and their children must be previously informed by a written consent about which findings should be reported. The ordering clinician must discuss with the children´s parents all the possibilities of results, including the reporting of incidental findings, the “right not to know,” the risks and the benefits, as well is responsible to obtain the informed consent and to provide pre- and posttest genetic counseling [17].

Advertisement

5. Conclusions

Inevitably, more challenges will arise with the application of NGS in RAGSC.

First, pretest counseling and informed consent models need to be redesigned to address the multiplex testing. Novel approaches must be developed to ensure that individuals understand the risks and benefits of choices regarding these tests. Second, the clinical management of carriers of moderately penetrant variants is still poorly defined, although some evidence-based guidelines may include them [14]. Third, finding VUS is always a potential risk, and such identification complicates data interpretation and often requires further investigation and variant reclassification. In addition, management of patients with VUS is unclear. Finally, many hereditary cancer syndromes have locus heterogeneity, incomplete penetrance and may represent phenocopies, adding difficulty in RAGSC.

In summary, the biggest challenge in counseling families with cancer is conferring precise information regarding genetic susceptibilities because it allows a better informed decision-making process about risk management, clinical surveillance, targeted therapies and preventive measures.

References

  1. 1. Pharoah PD, Antoniou A, Bobrow M, Zimmern RL, Easton DF, Ponder BA. Polygenic susceptibility to breast cancer and implications for prevention. Nature Genetics. 2002;31:33-36
  2. 2. Stadler ZK, Tom P, Robson ME, Weitzel JN, Kauff ND, Hurley KE, Devlin V, Gold B, Klein RJ, Offit K. Genome-wide association studies of cancer. Journal of Clinical Oncology. 2010;28:4255-4267
  3. 3. Riley BD, Culver JO, Skrzynia C, Senter LA, Peters JA, Costalas JW, Callif-Daley F, Grumet SC, Hunt KS, Nagy RS, et al. Essential elements of genetic cancer risk assessment, counseling, and testing: Updated recommendations of the National Society of Genetic Counselors. Journal of Genetic Counseling. 2012;21:151-161
  4. 4. Peters JA, Stopfer JE. Role of the genetic counselor in familial cancer. Oncology. 1996;10:159-166 discussion, 176-176, 178
  5. 5. Schneider K, Garber J. Counseling About Cancer: Strategies for Genetic Counselors. 2nd ed. New York: Wiley; 2001. p. 1904-1909
  6. 6. Trepanier A, Ahrens M, McKinnon W, Peters J, Stopfer J, Grumet SC, Manley S, Culver JO, Acton R, Larsen-Haidle J, et al. Genetic cancer risk assessment and counseling: Recommendations of the National Society of Genetic Counselors. Journal of Genetic Counseling. 2004;13:83-114
  7. 7. Schwartz GF, Hughes KS, Lynch HT, Fabian CJ, Fentiman IS, Robson ME, Domchek SM, Hartmann LC, Holland R, Winchester DJ, The Consensus Conference Committee. (2009) Proceedings of the international consensus conference on breast cancer risk, genetics & risk management. The Breast Journal. April, 2007;15:4-16
  8. 8. Robson ME, Storm CD, Weitzel J, Wollins DS, Offit K. American Society of Clinical Oncology policy statement update: Genetic and genomic testing for cancer susceptibility. Journal of Clinical Oncology. 2010;28:893-901
  9. 9. Weitzel JN, Blazer KR, Mac Donald DJ, Culver JO, Offit K. Genetics, genomics and risk assessment. State of the art and future directions in the era of personalized medicine. CA: A Cancer Journal for Clinicians. 2011;61:327-359
  10. 10. Roberts NJ, Klein AP. Genome-wide sequencing to identify the cause of hereditary cancer syndromes: With examples from familial pancreatic cancer. Cancer Letters. 2013;340:227-233
  11. 11. Jaeger E, Leedham S, Lewis A, Segditsas S, Becker M, Cuadrado PR, Davis H, Kaur K, Heinimann K, Howarth K, et al. Hereditary mixed polyposis syndrome is caused by a 40-kb upstream duplication that leads to increase and ectopic expression of the BMP antagonist GREM1. Nature Genetics. 2012;44:699-703
  12. 12. Domchek SM, Bradbury A, Garber JE, Offit K, Robson ME. Multiplex genetic testing for cancer susceptibility: Out on the high wire without a net? Journal of Clinical Oncology. 2013;31:1267-1270
  13. 13. Slavin TP, Niell-Swiller M, Solomon I, Nehoray B, Rybak C, Blazer KR, Weitzel JN. Clinical application of multigene panels: Challenges of next-generation counseling and cancer risk management. Frontiers in Oncology. 2015;5:208
  14. 14. NCCN. NCCN Clinical Practice Guidelines in Oncology V.1.2018: Genetic/Familial High-risk Assessment: Breast and Ovarian. NCCN Clinical Practice Guidelines [Internet]. 2017. Available from: http://www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf
  15. 15. Greem RC, Berg JS, Grody WW, Kalia SS, Br K, Martin CL, McGuire AL, Nussbaum RL, O’Daniel JM, Ormond KE, et al. ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing. Genetics in Medicine. 2013;15:565-574
  16. 16. ACMG Board of Directors. ACMG policy statement: Updated recommendations regarding analysis and reporting of secondary findings in clinical genome-scale sequencing. Genetics in Medicine. 2015;17:68-69
  17. 17. Kuhlen M, Borkhardt A. Cancer susceptibility syndromes in children in the area of broad clinical use of massive parallel sequencing. European Journal of Pediatrics. 2015;174:987-997

Written By

Israel Gomy

Submitted: 24 July 2017 Reviewed: 10 November 2017 Published: 23 December 2017