Next Article in Journal
Prenatal Diagnosis of Mucopolysaccharidosis-Plus Syndrome (MPSPS)
Next Article in Special Issue
The Human Microbiome and Its Role in Musculoskeletal Disorders
Previous Article in Journal
Multiple Sclerosis Heritability Estimation on Sardinian Ascertained Extended Families Using Bayesian Liability Threshold Model
Previous Article in Special Issue
Rare Coding Variants in Patients with Non-Syndromic Vestibular Dysfunction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Co-Occurrence of Germline Genomic Variants and Copy Number Variations in Hereditary Breast and Colorectal Cancer Patients

by
Luiza Côrtes
1,2,†,
Tatiane Ramos Basso
1,†,
Rolando André Rios Villacis
3,
Jeferson dos Santos Souza
4,
Mads Malik Aagaard Jørgensen
1,
Maria Isabel Achatz
5 and
Silvia Regina Rogatto
1,6,7,*
1
Department of Clinical Genetics, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark
2
Tocogynecoly Graduation Program, Botucatu Medical School, University of São Paulo State—UNESP, Botucatu 18618-687, SP, Brazil
3
Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília—UnB, Brasília 70910-900, DF, Brazil
4
Health Technology Institute, SENAI CIMATEC, Salvador 41650-010, BA, Brazil
5
Cancer Genetics Unit, Oncology Branch, Hospital Sirio-Libanês, São Paulo 01308-050, SP, Brazil
6
Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, 5000 Odense, Denmark
7
Danish Colorectal Cancer Center South, 7100 Vejle, Denmark
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Genes 2023, 14(8), 1580; https://doi.org/10.3390/genes14081580
Submission received: 11 July 2023 / Revised: 24 July 2023 / Accepted: 25 July 2023 / Published: 3 August 2023
(This article belongs to the Special Issue Feature Papers in Human Genomics and Genetic Diseases 2023)

Abstract

:
Hereditary Breast and Ovarian Cancer (HBOC) syndrome is an autosomal dominant disease associated with a high risk of developing breast, ovarian, and other malignancies. Lynch syndrome is caused by mutations in mismatch repair genes predisposing to colorectal and endometrial cancers, among others. A rare phenotype overlapping hereditary colorectal and breast cancer syndromes is poorly characterized. Three breast and colorectal cancer unrelated patients fulfilling clinical criteria for HBOC were tested by whole exome sequencing. A family history of colorectal cancer was reported in two patients (cases 2 and 3). Several variants and copy number variations were identified, which potentially contribute to the cancer risk or prognosis. All patients presented copy number imbalances encompassing PMS2 (two deletions and one duplication), a known gene involved in the DNA mismatch repair pathway. Two patients showed gains covering the POLE2 (cases 1 and 3), which is associated with DNA replication. Germline potentially damaging variants were found in PTCH1 (patient 3), MAT1A, and WRN (patient 2). Overall, concurrent genomic alterations were described that may increase the risk of cancer appearance in HBOC patients with breast and colorectal cancers.

Graphical Abstract

1. Background

Breast cancer (BC) and colorectal cancer (CRC) are frequent neoplasms, with more than 4 million people worldwide diagnosed yearly [1]. Individuals with hereditary cancer syndromes are at an increased risk of developing second primary tumors, such as BC and CRC [2,3].
Pathogenic variants in known cancer-predisposition genes are responsible for 5% to 10% of BC and CRC cases [4]. The most common pathogenic variants related to hereditary breast and ovarian cancer syndrome (HBOC) occur in BRCA1 and BRCA2 genes [5]. Lynch syndrome (LS) is a common condition of hereditary colorectal cancer syndromes. LS is predominantly associated with CRC and endometrial cancer and an increased lifetime risk of developing multiple primary cancer types (small bowel, urothelium, biliary tract, and stomach). Although under debate, several studies suggested that BC may be included as extracolonic cancer in LS [6,7]. LS is related to pathogenic germline variants in DNA mismatch repair (MMR) genes (MLH1, MSH2, MSH6, PMS2) or EPCAM and [8,9]. Of note, many patients that fulfill the criteria of hereditary cancer predisposition syndromes do not exhibit pathogenic variants in the main genes described as altered in these syndromes [4,10,11].
In addition to germline genetic variants, copy number variations (CNVs) can explain the heritability of several cancer syndromes, including HBOC and LS [12,13,14,15]. Previously, we reported a germline deletion mapped in the transcriptional regulatory region of ROBO1 in three patients fulfilling the criteria of hereditary breast and colorectal cancer [13]. We also described rare CNVs in 11 of 22 patients with multiple primary tumors, including the EPCAM/MSH2 deletion in one Lynch syndrome patient [14]. Rare germline CNVs have been reported in patients with a family history of BC, negative for BRCA1 and BRCA2 pathogenic variants [13,16,17].
Multiple primary cancers occur in a substantial proportion of cancer patients (10–25%) [18,19]. Patients with breast cancer are at higher risk of developing CRC, and patients with CRC show an increased incidence of BC [3,20,21]. Pathogenic variants of BRCA1, BRCA2, MLH1, MSH2, MSH6, PALB2, CHEK2, ATM, and TP53 were associated with an increased risk of breast and colorectal cancer [4,22,23]. Herein, the whole exome sequencing results were described in three unrelated patients with a personal and/or family history of BC and CRC. These patients have no germline pathogenic variants in the major predisposition genes related to their clinical phenotype.

2. Methods

2.1. Patients

Three BC patients with second primary CRC were selected from a cohort of 445 patients that fulfilled the testing criteria for inherited breast and ovarian cancer (National Comprehensive Cancer Network—NCCN—Guidelines Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic v2.2021). Patients prospectively consented to blood sample sequencing via whole exome sequencing. The study was conducted according to the ethical guidelines and regulations from the Declaration of Helsinki, and the Human Research Ethics Committee approved the study (CONEP Protocol # 2136/15). The patients were treated at A.C. Camargo Cancer Center, São Paulo, Brazil. All patients were tested for TP53, BRCA1, BRCA, MLH1, and MSH2 by Sanger sequencing, and no pathogenic variants were found.

2.2. DNA Extraction, Whole Exome Sequencing (WES), and Data Analysis

Genomic DNA from peripheral blood lymphocytes was extracted using the QIAamp DNA Blood Mini QIAcube Kit (Qiagen, Hilden, Germany), following the manufacturer’s instructions. DNA quality and quantification assessments were performed with the Genomic DNA ScreenTape assay (Agilent Technologies Inc., Santa Clara, CA, USA) and the Qubit dsDNA BR assay Kit (Thermo Fisher Scientific Inc., Waltham, MA, USA). Libraries were prepared using TruSeq Rapid (Illumina, San Diego, CA, USA), and the pair end (PE150) sequencing was performed on a NextSeq550 System (Illumina, San Diego, CA, USA), following the manufacturer’s recommendations. The sequence reads were aligned (Burrows-Wheeler Alignment, BWA-mem) to the human genome using the UCSC hg19, recalibrated, and realigned with the Genome Analysis Toolkit (GATK) [24]. The alignment statistics were performed with Sequence Alignment/Map (SAMtools), Picard, and Binary Alignment Map (BAMtools). Variant calling and merging were carried out with GATK HaplotypeCaller.
Germline variants annotation and selection were carried out using VarSeq™ version 2.2.2 (Golden Helix Inc., Bozeman, MT, USA). Single nucleotide variants (SNVs) and InDels were identified and classified using ACMG guidelines (revised in November 2022). Variants mapped in homopolymer regions or predicted as benign (B) or likely benign (LB) (ACMG or ClinVar classification), and those with frequency > 0.2 in the GnomAD v2.1 (The Genome Aggregation Database) and-/or in the ABRaOM (Online Archive of Brazilian Mutations) datasets were excluded [25,26]. The variants were manually curated using the Genome Browse software (Golden Helix Inc.). Only nonsynonymous, protein-damaging, and rare variants were included and manually curated. Pathogenic (P) and likely pathogenic (LP) variants were selected using ACMG and ClinVar classification. The variants of uncertain significance (VUS) potentially damaging were selected using deleterious effects filtering. Variants associated with loss of function or splice altering predictions (Ada scores > 0.6) or predicted as damaging in at least two independent functional prediction algorithms, including Sorting Intolerant From Tolerant (SIFT) [27], Polymorphism Phenotyping v2—Polyphen2 HVAR [28], MutationTaster [29], MutationAssessor [30], Functional Analysis through Hidden Markov Models—FATHMM, and FATHMM-MKL Coding [31], or Combined Annotation Dependent Depletion (CADD) score > 3 were selected. The list of variants was compared with the Catalogue Of Somatic Mutations In Cancer (COSMIC, https://cancer.sanger.ac.uk/cosmic, accessed on 30 May 2023).
Genomic CNVs were retrieved using CNV caller on target regions algorithm (Golden Helix VarSeq v.2.2.2). CNVs with a p-value < 0.001 were included, and those variants with a frequency ≥ 0.01 on the Database of Genomic Variant (DGV) were excluded. A circus plot was constructed to visualize the CNVs identified [32].

3. Results

The most relevant clinical and pathological features of each patient are summarized in Table 1. The patients met the NCCN v2.2021 criteria for HBOC and Hereditary Breast and Colorectal Cancer (HBCC), according to Meijers-Heijboer et al. [33] and Naseem et al. [22]. The representative pedigrees of these patients are depicted in Figure 1. Index patient 1 developed bilateral BC (64 years) and CRC (at age 71); her sister had BC diagnosed at age 68 (Figure 1A). Patient 2 was diagnosed with synchronous bilateral BC (55 years) and CRC (57 years). One sister had BC (40 years) and CRC (48 years), thyroid cancer was reported in another sister (45 years), and her maternal aunt presented osteosarcoma at age 52 (Figure 1B). Patient 3 was diagnosed with BC (48 years) and CRC (56 years), had four relatives with BC (mother, sister, and two paternal cousins), and a maternal cousin with CRC (Figure 1C).
Germline SNVs and CNVs were investigated in all three index patients (Figure 2, Table 2 and Table S1). Patient 1 presented the germline LP variant CYP1B1 c.1159G > A (Table 2), along with 49 VUS, including two missense variants NTRK1 c.16C > T and NFATC2 gene c.1952G > A reported as cancer-associated (COSMIC, https://cancer.sanger.ac.uk/cosmic (accessed on 30 May 2023)). This patient also presented 212 rare CNVs (p < 0.001), such as the heterozygous deletion of the PMS2 gene (start: 5764955, end: 6045662, spanning 280,707 base pairs, and affecting exons 2 to 15) (Figure 1D), a gain of POLE2 gene (start: 50154854, end: 50160008) (Figure 1E), and a heterozygous deletion of NOMO1 (start: 14946287, end: 14959012) (Table 2). Copy number variants of PMS2 were also found in all index patients, while gains of POLE2 were detected in patients 1 and 3.
The index patient 2 presented three LP variants (SLC3A1 c.1400T > C, MAT1A c.826dupG, and PTGIS c.824G > A) and 59 VUS, including the missense variant WRN c.95A > G, which is also in the COSMIC gene list. Seventeen rare CNVs were identified, including a gain of 9592 bp in the PMS2 gene (start: 6013030, end: 6022622, spanning exons 12 to 15) (Figure 1D and Table 2). This region has high homology with the PMS2CL pseudogene.
The index patient 3 presented 33 VUS, including the missense variants GNAQ c.286A > T and PTCH1 c.140G > T, previously described as cancer-associated genes (COSMIC gene list). Among the 150 rare CNVs found in this patient, a heterozygous deletion of PMS2 (start: 6048628, end: 6049129, 501 bp at exon 1) was identified (Figure 1D), a region with no homology with the PMS2CL pseudogene. Additionally, a heterozygous deletion of the ASH2L gene (start: 37914599, end: 37963256) and a gain in POLE2 (start: 50120873, end: 50131392) (Figure 1E) were found (Table 2).

4. Discussion

The accumulation of germline variants and CNVs in cancer-related genes may explain the development of multiple primary tumors in individual patients [34]. In this study, we investigated the germline whole exome sequencing of three breast and colorectal cancer individuals fulfilling the HBOC and HBCC criteria. Pathogenic and likely pathogenic variants were not detected in two index patients. All patients presented variants with uncertain significance; a set of these genes was described as cancer-related. Moreover, SNVs and CNVs that may contribute to the cancer risk were identified in all three index patients. Previously, we reported that early-onset rectal cancer (<40 years) patients exhibited a significantly higher number of germline variants, as well as multiple variants in oncogenes, tumor suppressors, and mismatch repair genes associated with hereditary colorectal cancer [35]. In early-onset breast cancer, concurrent germline variants of BRCA1, BRCA2, PMS2, and PALB2 were reported [36]. Analysis of hereditary cancer cases (732 BC patients, 189 CRC patients, and 490 cancer-free elderly controls) revealed that 1% presented concurrent germline pathogenic variants in BRCA1, BRCA2, MUTYH, ATM, CHECK2, NBN, and RAD50 [37]. The germline genomic profile of BC/CRC patients fulfilling the criteria of HBOC and HBCC is poorly explored in the literature.
Patient 1 presented the LP variant CYP1B1 c.1159G > A. The protein encoded by CYP1B1 plays a role in the metabolism of steroid hormones and the progression of certain cancers, such as breast and prostate cancer [38,39]. Previous single nucleotide polymorphism studies of CYP1B1 showed an association with an increased risk of developing BC and CRC [40,41]. In CRC samples, high CYP1B1 expression was significantly associated with lower overall and disease-free survival rates [42]. Among the 49 VUS identified in the index patient 1, two genes were cancer-related: NTRK1c.16C > T and NFATC2 c.1952G > A. Gene fusions involving NTRK1 can confer oncogenic potential in several tumors, including BC and CRC [43,44]. These genetic fusions in the tyrosine receptor kinases have emerged as targets for cancer therapy [45]. NFATC2 codifies a member of transcription factors activated by calcium signaling pathways that acts in the immune response control [46,47]. NFATC2 regulates the self-renewal capacity and tumorigenic initiation of tumor stem cells and is a promising therapeutic target for colorectal cancer treatment [48]. Overall, CYP1B1, NTRK1, and NFATC2 are not related to cancer risk but have potential roles as prognostic markers and actionable drug targets.
Index patient 1 presented 213 rare copy number variations (p < 0.0001), including those mapped in PMS2, POLE2, and NOMO1 genes. A heterozygous deletion of the PMS2 gene was also found in patient 3, while a significant gain was detected in patient 2. A heterozygous pathogenic deletion of PMS2 (exons 13–15) was reported in two unrelated BC patients with no clinical or familial criteria for LS phenotype [15]. Interestingly, this PMS2 pathogenic deletion was also identified in two patients with transverse colon cancer [49]. The PMS2 gene (PMS1 homolog 2, mismatch repair system component, mapped on 7p22.1) encodes a key protein involved in the DNA mismatch repair, and it is associated with LS [50]. The molecular diagnosis of PMS2 in hereditary cancer syndromes is challenging due to a family of pseudogenes highly homologous to PMS2 [51,52]. Fourteen pseudogenes showed high homology with the 5′end (exons 1 to 5) of PMS2, while the pseudogene PMS2CL shares high sequence similarity to exons 9 and exons 11–15 of PMS2 [51,52]. The large heterozygous deletion of PMS2 (280,707 bp; spanning exons 2 to 15) found in patient 1 has partial homology with the pseudogenes and, therefore, can impact the cancer risk of this patient and her family members. The PMS2 gain identified in patient 2 and the heterozygous loss detected in patient 3 are questionable as to their clinical impact. The biological significance of these alterations for cancer risk should be confirmed. However, protocols to ensure CNVs have challenges: they are expensive, time-consuming, and show gene-specific limitations, such as the availability of MLPA kits (MRC Holland), and do not always result in practical conclusions [53,54]. Although some alterations should be better investigated, reports presenting findings in rare diseases are critical to identifying their clinical and biological consequences [55]. A study focusing on CRC at a young age showed that women with PMS2 germline variants had a 3-fold increased risk for BC development [56]. It is reasonable to suggest that CNVs of PMS2 contribute to the cancer risk in HBOC and HBCC patients. Copy number gains of POLE2 were identified in two patients (1 and 3). The POLE2 protein (centromere-associated protein E) is a subunit of DNA polymerase epsilon (Pol ε), which is one of the main enzymes involved in DNA replication [57]. POLE2 mutations have been associated with colorectal cancer, but the POLE2 CNV related to hereditary cancer syndrome is a new finding [57,58,59]. Gains of POLE2 can alter the protein function and contribute to cancer risk. Heterozygous loss of NOMO1 (start: 14946287, end: 14959012) was also detected in index case 1. Colorectal cancer frequently exhibits NOMO1 deletion [60,61]. This biomarker is a potential therapeutic target for early-onset colorectal cancer [60].
Index patient 2 presented three likely pathogenic variants SLC3A1 c.1400T > C, MAT1A c.826dupG, and PTGIS c.824G > A. Breast cancer cells have elevated expression of SLC3A1, which accelerates cysteine uptake and accumulates reduced glutathione and decreases reactive oxygen species in tumor cells [62,63]. Dysregulation of MAT1A in tumor cells was associated with replication errors and genetic instability [64,65]. Furthermore, MAT1A dysregulation increases homocysteine levels, promoting cancer progression [66]. The PTGIS (Prostaglandin I2 Synthase) gene encodes a cytochrome P450 oxidase family member. In colorectal cancer, PTGIS dysregulation may contribute to chromosomal instability and tumor progression [67]. High PTGIS expression can promote infiltration of immune cells, such as tumor-associated macrophages and regulatory T cells, into the tumor microenvironment, which impairs the prognosis of patients with lung, ovarian, and gastric cancer [68,69]. The missense variant WRNc.95A > G was also identified among the 59 VUS of index case 2. The tumor suppressor WRN encodes an enzyme from the RECQ gene family, which is essential for maintaining genomic stability [70]. Germline variants of WRN were described in HBOC and early-onset familial colorectal cancer [71,72,73]. The heterozygous loss at exon 1 of PMS2 was identified among the 17 rare CNVs detected in this patient, as discussed above. We suggest that MAT1A and WRN variants and PMS2 loss could contribute to the hereditary cancer predisposition phenotype observed in this family.
Although index patient 3 had no P/LP variants, 33 VUS, and two potentially damaging missense variants, GNAQc.286A > T and PTCH1c.140G > T (COSMIC) were identified. Mutations in the GNAQ/GNA11 gene activate the MAP kinase signaling pathway, a key factor in the development of uveal melanoma [74]. The GNAQ c.286A > T variant detected in the index patient was reported in a small percentage of colorectal cancers assessed by gene panel sequencing [75]. Germline inactivating mutations in PTCH1 were associated with Gorlin Syndrome, a genetic predisposition to multiple basal cell carcinoma and medulloblastoma [76]. Recently, Yoshida et al. reported a PTCH1 germline variant in one patient with second malignant neoplasms (primary: medulloblastoma; second: osteosarcoma and acute myeloid leukemia) [77]. Germline pathogenic PTCH1 variant was also described in one colorectal cancer patient whose cancer showed microsatellite instability and wild-type MMR genes [78]. Germline variants in cancer predisposition genes not related to the patient phenotype need investigation to result in adequate clinical management.
Moreover, 150 rare CNVs were found in the index patient 3. Among them, a heterozygous deletion of 501 bp in PMS2 (exon 1), as described above, a heterozygous deletion of ASH2L, and a gain of POLE2 (start: 50120873, end: 50131392). ASH2L gene encodes a protein involved in the epigenetic regulation and transcriptional activation of various biological processes, including embryonic development, stem cell self-renewal, hematopoiesis, and tumorigenesis [79,80]. Elevated expression of ER alpha is frequently correlated with an increase in ASH2L expression in BC cell lines and primary BC [81]. In colorectal cancer, ASH2L acts positively by being recruited to the promoter region of genes associated with p53-mediated apoptosis, potentiating its function and inducing cell death [82]. These results provide promising clues for developing therapeutic targets in tumors related to ER alpha, such as breast cancer [80,83].

5. Conclusions

Concurrent germline SNVs and CNVs potentially associated with the risk of development of BC and CRC were identified in three HBOC/HBCC families. Prognostic markers and potential drug targets were also described. Early diagnosis of these patients and comprehensive genomic analysis are crucial to implementing more effective prevention or individualized treatment strategies that reduce the risk of a second primary tumor. Additional efforts are required to increase the number of BC/CRC families evaluated by the whole exome sequencing and clarify the impact of these alterations on cancer risk and prognosis.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14081580/s1. Table S1. List of germline variants (pathogenic, likely pathogenic and variants with uncertain significance) and copy number variations in three patients fulfilling HBOC criteria.

Author Contributions

Conceptualization, M.I.A. and S.R.R.; Data curation, S.R.R.; Formal analysis, L.C., T.R.B. and R.A.R.V.; Funding acquisition, S.R.R.; Investigation, R.A.R.V. and S.R.R.; Methodology, L.C., T.R.B. and R.A.R.V.; Project administration, S.R.R.; Resources, S.R.R.; Software, J.d.S.S. and M.M.A.J.; Supervision, M.I.A. and S.R.R.; Writing—original draft, L.C., T.R.B., R.A.R.V. and S.R.R.; Writing—review and editing, S.R.R. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by grants from the National Institute of Science and Technology in Oncogenomics (FAPESP 2008/57887–9 and CNPq 573589/08–9), BR and Region of Southern Denmark Research Fund, DK. L.C. was granted a scholarship from the Brazilian Federal Agency for Support and Evaluation of Graduate Education (CAPES).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of the A.C. Camargo Cancer Center (São Paulo, Brazil) (CONEP Protocol # 2136/15 and date of approval: 4 October 2016).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The whole exome sequencing data will not be made publicly available due to restraints imposed by the ethics committee; requests for data can be made to the corresponding author.

Acknowledgments

The authors would like to thank the patients for accepting to participate in the study and the Nucleic Acid Bank of A.C. Camargo Cancer Center (São Paulo, Brazil) for sample processing. We are grateful to Maria Nirvana da Cruz Formiga, and Luisa Matos do Canto Alvim for their assistance during the development of this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Dash, C.; Lu, J.; Parikh, V.; Wathen, S.; Shah, S.; Shah Chaudhari, R.; Adams-Campbell, L. Disparities in colorectal cancer screening among breast and prostate cancer survivors. Cancer Med. 2021, 10, 1448–1456. [Google Scholar] [CrossRef]
  3. Halamkova, J.; Kazda, T.; Pehalova, L.; Gonec, R.; Kozakova, S.; Bohovicova, L.; Krakorova, D.A.; Slaby, O.; Demlova, R.; Svoboda, M.; et al. Second primary malignancies in colorectal cancer patients. Sci. Rep. 2021, 11, 2759. [Google Scholar] [CrossRef]
  4. Akcay, I.M.; Celik, E.; Agaoglu, N.B.; Alkurt, G.; Kizilboga Akgun, T.; Yildiz, J.; Enc, F.; Kir, G.; Canbek, S.; Kilic, A.; et al. Germline pathogenic variant spectrum in 25 cancer susceptibility genes in Turkish breast and colorectal cancer patients and elderly controls. Int. J. Cancer 2021, 148, 285–295. [Google Scholar] [CrossRef] [PubMed]
  5. Yoshida, R. Hereditary breast and ovarian cancer (HBOC): Review of its molecular characteristics, screening, treatment, and prognosis. Breast Cancer 2021, 28, 1167–1180. [Google Scholar] [CrossRef]
  6. Sheehan, M.; Heald, B.; Yanda, C.; Kelly, E.D.; Grobmyer, S.; Eng, C.; Kalady, M.; Pederson, H. Investigating the Link between Lynch Syndrome and Breast Cancer. Eur. J. Breast Health 2020, 16, 106–109. [Google Scholar] [CrossRef]
  7. Schwartz, C.J.; da Silva, E.M.; Marra, A.; Gazzo, A.M.; Selenica, P.; Rai, V.K.; Mandelker, D.; Pareja, F.; Misyura, M.; D’Alfonso, T.M.; et al. Morphologic and Genomic Characteristics of Breast Cancers Occurring in Individuals with Lynch Syndrome. Clin. Cancer Res. 2022, 28, 404–413. [Google Scholar] [CrossRef] [PubMed]
  8. Win, A.K.; Jenkins, M.A.; Dowty, J.G.; Antoniou, A.C.; Lee, A.; Giles, G.G.; Buchanan, D.D.; Clendenning, M.; Rosty, C.; Ahnen, D.J.; et al. Prevalence and penetrance of major genes and polygenes for colorectal cancer. Cancer Epidemiol. Biomark. Prev. 2017, 26, 404–412. [Google Scholar] [CrossRef] [Green Version]
  9. Biller, L.H.; Creedon, S.A.; Klehm, M.; Yurgelun, M.B. Lynch Syndrome-Associated Cancers Beyond Colorectal Cancer. Gastrointest Endosc. Clin. 2022, 32, 75–93. [Google Scholar] [CrossRef] [PubMed]
  10. McBride, K.A.; Ballinger, M.L.; Killick, E.; Kirk, J.; Tattersall, M.H.N.; Eeles, R.A.; Thomas, D.M.; Mitchell, G. Li-Fraumeni syndrome: Cancer risk assessment and clinical management. Nat. Rev. Clin. Oncol. 2014, 11, 260–271. [Google Scholar] [CrossRef] [PubMed]
  11. Watanabe, T.; Soeda, S.; Endo, Y.; Okabe, C.; Sato, T.; Kamo, N.; Ueda, M.; Kojima, M.; Furukawa, S.; Nishigori, H.; et al. Rare Hereditary Gynecological Cancer Syndromes. Int. J. Mol. Sci. 2022, 23, 1563. [Google Scholar] [CrossRef] [PubMed]
  12. Silva, F.C.; Lisboa, B.C.; Figueiredo, M.C.; Torrezan, G.T.; Santos, E.M.; Krepischi, A.C.; Rossi, B.M.; Achatz, M.I.; Carraro, D.M. Hereditary breast and ovarian cancer: Assessment of point mutations and copy number variations in Brazilian patients. BMC Med. Genet. 2014, 15, 55. [Google Scholar] [CrossRef]
  13. Villacis, R.A.R.; Abreu, F.B.; Miranda, P.M.; Domingues, M.A.C.; Carraro, D.M.; Santos, E.M.M.; Andrade, V.P.; Rossi, B.M.; Achatz, M.I.; Rogatto, S.R. ROBO1 deletion as a novel germline alteration in breast and colorectal cancer patients. Tumour Biol. 2016, 37, 3145–3153. [Google Scholar] [CrossRef] [PubMed]
  14. Villacis, R.A.R.; Basso, T.R.; Canto, L.M.; Pinheiro, M.; Santiago, K.M.; Giacomazzi, J.; de Paula, C.A.A.; Carraro, D.M.; Ashton-Prolla, P.; Achatz, M.I.; et al. Rare germline alterations in cancer-related genes associated with the risk of multiple primary tumor development. J. Mol. Med. 2017, 95, 523–533. [Google Scholar] [CrossRef] [PubMed]
  15. Boujemaa, M.; Hamdi, Y.; Mejri, N.; Romdhane, L.; Ghedira, K.; Bouaziz, H.; El Benna, H.; Labidi, S.; Dallali, H.; Jaidane, O.; et al. Germline copy number variations in BRCA1/2 negative families: Role in the molecular etiology of hereditary breast cancer in Tunisia. PLoS ONE 2021, 16, e0245362. [Google Scholar] [CrossRef] [PubMed]
  16. Krepischi, A.C.; Achatz, M.I.; Santos, E.M.; Costa, S.S.; Lisboa, B.C.; Brentani, H.; Santos, T.M.; Gonçalves, A.; Nóbrega, A.F.; Pearson, P.L.; et al. Germline DNA copy number variation in familial and early-onset breast cancer. Breast Cancer Res. 2012, 14, R24. [Google Scholar] [CrossRef]
  17. Pylkäs, K.; Vuorela, M.; Otsukka, M.; Kallioniemi, A.; Jukkola-Vuorinen, A.; Winqvist, R. Rare copy number variants observed in hereditary breast cancer cases disrupt genes in estrogen signaling and TP53 tumor suppression network. PLoS Genet. 2012, 8, e1002734. [Google Scholar] [CrossRef] [Green Version]
  18. Murphy, C.C.; Gerber, D.E.; Pruitt, S.L. Prevalence of Prior Cancer Among Persons Newly Diagnosed with Cancer: An Initial Report from the Surveillance, Epidemiology, and End Results Program. JAMA Oncol. 2018, 4, 832–836. [Google Scholar] [CrossRef] [Green Version]
  19. Vogt, A.; Schmid, S.; Heinimann, K.; Frick, H.; Herrmann, C.; Cerny, T.; Omlin, A. Multiple primary tumours: Challenges and approaches, a review. ESMO Open 2017, 2, e000172. [Google Scholar] [CrossRef] [Green Version]
  20. La Francis, I.E.; Cooper, R.B. Second primary malignancies associated with primary female breast cancer: A review of the Danbury Hospital experience. Conn. Med. 1992, 56, 411–414. [Google Scholar]
  21. Soerjomataram, I.; Louwman, W.J.; de Vries, E.; Lemmens, V.E.; Klokman, W.J.; Coebergh, J.W. Primary malignancy afer primary female breast cancer in the south of the Netherlands, 1972–2001. Breast Cancer Res. Treat. 2005, 93, 91–95. [Google Scholar] [CrossRef]
  22. Naseem, H.; Boylan, J.; Speake, D.; Leask, K.; Shenton, A.; Lalloo, F.; Hill, J.; Trump, D.; Evans, D.G. Inherited association of breast and colorectal cancer: Limited role of CHEK2 compared with high-penetrance genes. Clin. Genet. 2006, 70, 388–395. [Google Scholar] [CrossRef] [PubMed]
  23. Phelan, C.M.; Iqbal, J.; Lynch, H.T.; Lubinski, J.; Gronwald, J.; Moller, P.; Ghadirian, P.; Foulkes, W.D.; Armel, S.; Eisen, A.; et al. Incidence of colorectal cancer in brca1 and BRCA2 mutation carriers: Results from a follow-up study. Br. J. Cancer 2014, 110, 530–534. [Google Scholar] [CrossRef] [Green Version]
  24. DePristo, M.A.; Banks, E.; Poplin, R.; Garimella, K.V.; Maguire, J.R.; Hartl, C.; Philippakis, A.A.; del Angel, G.; Rivas, M.A.; Hanna, M.; et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 2011, 43, 491–498. [Google Scholar] [CrossRef] [PubMed]
  25. Karczewski, K.J.; Francioli, L.C.; Tiao, G.; Cummings, B.B.; Alföldi, J.; Wang, Q.; Collins, R.L.; Laricchia, K.M.; Ganna, A.; Birnbaum, D.P.; et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 2020, 581, 434–443. [Google Scholar] [CrossRef]
  26. Naslavsky, M.S.; Yamamoto, G.L.; de Almeida, T.F.; Ezquina, S.A.M.; Sunaga, D.Y.; Pho, N.; Bozoklian, D.; Sandberg, T.O.M.; Brito, L.A.; Lazar, M.; et al. Exomic variants of an elderly cohort of Brazilians in the ABraOM database. Hum. Mutat. 2017, 38, 751–763. [Google Scholar] [CrossRef]
  27. Vaser, R.; Adusumalli, S.; Leng, S.N.; Sikic, M.; Ng, P.C. SIFT missense predictions for genomes. Nat. Protoc. 2016, 11, 1–9. [Google Scholar] [CrossRef] [PubMed]
  28. Adzhubei, I.A.; Schmidt, S.; Peshkin, L.; Ramensky, V.E.; Gerasimova, A.; Bork, P.; Kondrashov, A.S.; Sunyaev, S.R. A method and server for predicting damaging missense mutations. Nat. Methods 2010, 7, 248–249. [Google Scholar] [CrossRef] [Green Version]
  29. Schwarz, J.M.; Cooper, D.N.; Schuelke, M.; Seelow, D. MutationTaster2: Mutation prediction for the deep-sequencing age. Nat. Methods 2014, 11, 361–362. [Google Scholar] [CrossRef]
  30. Kumar, P.; Henikoff, S.; Ng, P.C. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat. Protoc. 2009, 4, 1073–1081. [Google Scholar] [CrossRef]
  31. Shihab, H.A.; Gough, J.; Cooper, D.N.; Stenson, P.D.; Barker, G.L.A.; Edwards, K.J.; Day, I.N.M.; Gaunt, T.R. Predicting the functional, molecular, and phenotypic consequences of amino acid substitutions using hidden markov models. Hum. Mutat. 2013, 34, 57–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Gu, Z.; Gu, L.; Eils, R.; Schlesner, M.; Brors, B. Circlize implements and enhances circular visualization in R. Bioinformatics 2014, 30, 2811–2812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Meijers-Heijboer, H.; Wijnen, J.; Vasen, H.; Wasielewski, M.; Wagner, A.; Hollestelle, A.; Elstrodt, F.; van den Bos, R.; de Snoo, A.; Fat, G.T.; et al. The CHEK2 1100delC mutation identifies families with a hereditary breast and colorectal cancer phenotype. Am. J. Hum. Genet. 2003, 72, 1308–1314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sukumar, J.; Kassem, M.; Agnese, D.; Pilarski, R.; Ramaswamy, B.; Sweet, K.; Sardesai, S. Concurrent germline BRCA1, BRCA2, and CHEK2 pathogenic variants in hereditary breast cancer: A case series. Breast Cancer Res. Treat. 2021, 2, 569–575. [Google Scholar] [CrossRef]
  35. Beltrami, C.M.; Canto, L.M.; Villacis, R.A.R.; Petersen, A.H.; Aagaard, M.M.; Cury, S.S.; Formiga, M.N.C.; Junior, S.A.; Rogatto, R.R. The repertoire of germline variants in patients with early-onset rectal cancer. Cancer Commun. 2022, 5, 481–485. [Google Scholar] [CrossRef]
  36. Megid, T.B.C.; Barros-Filho, M.C.; Pisani, J.P.; Achatz, M.I. Double heterozygous pathogenic variants prevalence in a cohort of patients with hereditary breast cancer. Front. Oncol. 2022, 12, 873395. [Google Scholar] [CrossRef]
  37. Agaoglu, N.B.; Doganay, L. Concurrent pathogenic variations in patients with hereditary cancer syndromes. Eur. J. Med. Genet. 2021, 64, 104366. [Google Scholar] [CrossRef]
  38. Lin, D.I.; Huang, R.S.P.; Mata, D.A.; Decker, B.; Danziger, N.; Lechpammer, M.; Hiemenz, M.; Ramkissoon, S.H.; Ross, J.S.; Elvin, J.A. Clinicopathological and genomic characterization of BCORL1-driven high-grade endometrial stromal sarcomas. Mod. Pathol. 2021, 34, 2200–2210. [Google Scholar] [CrossRef]
  39. Min, J.Y.; Lee, G.H.; Khanal, T.; Jin, S.W.; Lee, S.Y.; Kim, H.G.; Hyon, J.Y.; Chung, Y.H.; Ha, S.K.; Han, E.H.; et al. Upregulation of CYP1B1 by hypoxia is mediated by ERα activation in breast cancer cells. Am. J. Cancer Res. 2022, 12, 2798–2816. [Google Scholar]
  40. He, X.F.; Wei, J.; Liu, Z.Z.; Xie, J.J.; Wang, W.; Du, Y.P.; Chen, Y.; Si, H.Q.; Liu, Q.; Wu, L.X.; et al. Association between CYP1A2 and CYP1B1 polymorphisms and colorectal cancer risk: A meta-analysis. PLoS ONE 2014, 9, e100487. [Google Scholar]
  41. Vishnubalaji, R.; Alajez, N.M. Epigenetic regulation of triple negative breast cancer (TNBC) by TGF-β signaling. Sci. Rep. 2021, 11, 15410. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, X.; Wang, F.; Wu, J.; Zhang, T.; Liu, F.; Mao, Y.; Hua, D. Expression of CYP1B1 and B7-H3 significantly correlates with poor prognosis in colorectal cancer patients. Int. J. Clin. Exp. Pathol. 2018, 11, 2654–2664. [Google Scholar] [PubMed]
  43. Solomon, J.P.; Benayed, R.; Hechtman, J.F.; Ladanyi, M. Identifying patients with NTRK fusion cancer. Ann. Oncol. 2019, 30, viii16–viii22. [Google Scholar] [CrossRef] [PubMed]
  44. Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef]
  45. Amatu, A.; Sartore-Bianchi, A.; Siena, S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open. 2016, 1, e000023. [Google Scholar] [CrossRef] [Green Version]
  46. Lieber, S.; Reinartz, S.; Raifer, H.; Finkernagel, F.; Dreyer, T.; Bronger, H.; Jansen, J.M.; Wagner, U.; Worzfeld, T.; Müller, R.; et al. Prognosis of ovarian cancer is associated with effector memory CD8+ T cell accumulation in ascites, CXCL9 levels and activation-triggered signal transduction in T cells. Oncoimmunology 2018, 7, e1424672. [Google Scholar] [CrossRef] [Green Version]
  47. Perret, R.; Escuriol, J.; Velasco, V.; Mayeur, L.; Soubeyran, I.; Delfour, C.; Aubert, S.; Polivka, M.; Karanian, M.; Meurgey, A.; et al. NFATc2-rearranged sarcomas: Clinicopathologic, molecular, and cytogenetic study of 7 cases with evidence of AGGRECAN as a novel diagnostic marker. Mod. Pathol. 2020, 33, 1930–1944. [Google Scholar] [CrossRef]
  48. Lang, T.; Ding, X.; Kong, L.; Zhou, X.; Zhang, Z.; Ju, H.; Ding, S. NFATC2 is a novel therapeutic target for colorectal cancer stem cells. Onco Targets Ther. 2018, 11, 6911–6924. [Google Scholar] [CrossRef] [Green Version]
  49. Vaughn, C.P.; Hart, K.J.; Samowitz, W.S.; Swensen, J.J. Avoidance of pseudogene interference in the detection of 3’ deletions in PMS2. Hum. Mutat. 2011, 32, 1063–1071. [Google Scholar] [CrossRef]
  50. Blount, J.; Prakash, A. The Changing Landscape of Lynch Syndrome due to PMS2 Mutations. Clin. Genet. 2018, 94, 61–69. [Google Scholar] [CrossRef]
  51. Herman, D.S.; Smith, C.; Liu, C.; Vaughn, C.P.; Palaniappan, S.; Pritchard, C.C.; Shirts, B.H. Efficient detection of copy number mutations in PMS2 exons with a close homolog. J. Mol. Diagn. 2018, 20, 512–521. [Google Scholar] [CrossRef] [Green Version]
  52. Niessen, R.C.; Kleibeuker, J.H.; Jager, P.O.J.; Sijmons, R.H.; Hofstra, R.M.W. Getting rid of the PMS2 pseudogenes: Mission impossible? Hum. Mutat. 2007, 28, 414. [Google Scholar] [CrossRef] [PubMed]
  53. Pös, O.; Radvanszky, J.; Buglyó, G.; Pös, Z.; Rusnakova, D.; Nagy, B.; Szemes, T. DNA copy number variation: Main characteristics, evolutionary significance, and pathological aspects. Biomed. J. 2021, 44, 548–559. [Google Scholar] [CrossRef] [PubMed]
  54. Singh, A.K.; Olsen, M.F.; Lavik, L.A.S.; Vold, T.; Drabløs, F.; Sjursen, W. Detecting copy number variation in next generation sequencing data from diagnostic gene panels. BMC Med. Genomics 2021, 14, 214. [Google Scholar] [CrossRef] [PubMed]
  55. Fragoso-Ontiveros, V.; De la Fuente-Hernandez, M.A.; Gonzalez-Osnaya, V.; Gamez-Rosales, M.; Perez-Montiel, M.D.; Isla-Ortiz, D.; Cantu-De Leon, D.F.; Alvarez-Gomez, R.M. A Pathogenic Variant Reclassified to the Pseudogene PMS2P1 in a Patient with Suspected Hereditary Cancer. Int. J. Mol. Sci. 2023, 24, 1398. [Google Scholar] [CrossRef]
  56. Roberts, M.E.; Jackson, S.A.; Susswein, L.R.; Zeinomar, N.; Ma, X.; Marshall, M.L.; Stettner, A.R.; Milewski, B.; Xu, Z.; Solomon, B.D.; et al. MSH6 and PMS2 germline pathogenic variants implicated in Lynch syndrome are associated with breast cancer. Genet. Med. 2018, 20, 1167–1174. [Google Scholar] [CrossRef]
  57. Palles, C.; Cazier, J.B.; Howarth, K.M.; Domingo, E.; Jones, A.M.; Broderick, P.; Kemp, Z.; Spain, S.L.; Almeida, E.G.; Sanguero, I.; et al. Germline mutations in the proof-reading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat. Genet. 2013, 2, 136–144. [Google Scholar] [CrossRef] [Green Version]
  58. Chubb, D.; Broderick, P.; Dobbins, S.E.; Frampton, M.; Kinnersley, B.; Penegar, S.; Price, A.; Ma, Y.P.; Sherborne, A.L.; Palles, C.; et al. Rare disruptive mutations and their contribution to the heritable risk of colorectal cancer. Nat. Commun. 2016, 7, 11883. [Google Scholar] [CrossRef] [Green Version]
  59. Spier, I.; Holzapfel, S.; Altmüller, J.; Zhao, B.; Horpaopan, S.; Vogt, S.; Chen, S.; Morak, M.; Raeder, S.; Kayser, K.; et al. Frequency and phenotypic spectrum of germline mutations in POLE and seven other polymerase genes in 266 patients with colorectal adenomas and carcinomas. Int. J. Cancer 2015, 137, 320–331. [Google Scholar] [CrossRef]
  60. Perea, J.; García, J.L.; Pérez, J.; Rueda, D.; Arriba, M.; Rodríguez, Y.; Urioste, M.; González-Sarmiento, R. NOMO-1 gene is deleted in early-onset colorectal cancer. Oncotarget 2017, 8, 24429–24436. [Google Scholar] [CrossRef] [Green Version]
  61. Pérez-García, J.; Martel-Martel, A.; García-Vallés, P.; Corchete, L.A.; García, J.L.; Gestoso-Uzal, N.; Vidal-Tocino, R.; Blanco, Ó.; Méndez, L.; Sánchez-Martín, M.; et al. Recurrent NOMO1 Gene Deletion Is a Potential Clinical Marker in Early-Onset Colorectal Cancer and Is Involved in the Regulation of Cell Migration. Cancers 2022, 14, 4029. [Google Scholar] [CrossRef] [PubMed]
  62. Meng, T.C.; Fukada, T.; Tonks, N.K. Reversible oxidation and inactivation of protein tyrosine phosphatases in vivo. Mol. Cell 2002, 9, 387–399. [Google Scholar] [CrossRef] [PubMed]
  63. Jiang, Y.; Cao, Y.; Wang, Y.; Li, W.; Liu, X.; Lv, Y.; Li, X.; Mi, J. Cysteine transporter SLC3A1 promotes breast cancer tumorigenesis. Theranostics 2017, 7, 1036–1046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Maldonado, L.Y.; Arsene, D.; Mato, J.M.; Lu, S.C. Methionine adenosyltransferases in cancers: Mechanisms of dysregulation and implications for therapy. Exp. Biol. Med. 2018, 243, 107–117. [Google Scholar] [CrossRef] [PubMed]
  65. Datta, A.; Biswas, K.; Sommers, J.A.; Thompson, H.; Awate, S.; Nicolae, C.M.; Thakar, T.; Moldovan, G.L.; Shoemaker, R.H.; Sharan, S.K.; et al. WRN helicase safeguards deprotected replication forks in BRCA2-mutated cancer cells. Nat. Commun. 2021, 12, 6561. [Google Scholar] [CrossRef]
  66. Frau, M.; Feo, F.; Pascale, R.M. Pleiotropic effects of methionine adenosyltransferases deregulation as determinants of liver cancer progression and prognosis. J. Hepatol. 2013, 59, 830–841. [Google Scholar] [CrossRef] [Green Version]
  67. Frigola, J.; Muñoz, M.; Clark, S.J.; Moreno, V.; Capellà, G.; Peinado, M.A. Hypermethylation of the prostacyclin synthase (PTGIS) promoter is a frequent event in colorectal cancer and associated with aneuploidy. Oncogene 2005, 24, 7320–7326. [Google Scholar] [CrossRef] [Green Version]
  68. Dai, D.; Chen, B.; Feng, Y.; Wang, W.; Jiang, Y.; Huang, H.; Liu, J. Prognostic value of prostaglandin I2 synthase and its correlation with tumor-infiltrating immune cells in lung cancer, ovarian cancer, and gastric cancer. Aging 2020, 12, 9658–9685. [Google Scholar] [CrossRef]
  69. Lichao, S.; Liang, P.; Chunguang, G.; Fang, L.; Zhihua, Y.; Yuliang, R. Overexpression of PTGIS could predict liver metastasis and is correlated with poor prognosis in colon cancer patients. Pathol. Oncol. Res. 2012, 18, 563–569. [Google Scholar] [CrossRef]
  70. Iglesias-Pedraz, J.M.; Fossatti-Jara, D.M.; Valle-Riestra-Felice, V.; Cruz-Visalaya, S.R.; Ayala Felix, J.A.; Comai, L. WRN modulates translation by influencing nuclear mRNA export in HeLa cancer cells. BMC Mol. Cell Biol. 2020, 21, 71. [Google Scholar] [CrossRef]
  71. Yang, L.; Wang, G.; Zhao, X.; Ye, S.; Shen, P.; Wang, W.; Zheng, S. A Novel WRN Frameshift Mutation Identified by Multiplex Genetic Testing in a Family with Multiple Cases of Cancer. PLoS ONE 2015, 10, e0133020. [Google Scholar] [CrossRef]
  72. Zimmer, K.; Puccini, A.; Xiu, J.; Baca, Y.; Spizzo, G.; Lenz, H.J.; Battaglin, F.; Goldberg, R.M.; Grothey, A.; Shields, A.F.; et al. WRN-Mutated Colorectal Cancer Is Characterized by a Distinct Genetic Phenotype. Cancers 2020, 12, 1319. [Google Scholar] [CrossRef] [PubMed]
  73. Norouzi, M.; Shafiei, M.; Abdollahi, Z.; Miar, P.; Galehdari, H.; Emami, M.H.; Zeinalian, M.; Tabatabaiefar, M.A. WRN Germline Mutation Is the Likely Inherited Etiology of Various Cancer Types in One Iranian Family. Front. Oncol. 2021, 11, 648649. [Google Scholar] [CrossRef] [PubMed]
  74. Lietman, C.D.; McKean, M. Targeting GNAQ/11 through PKC inhibition in uveal melanoma. Cancer Gene Ther. 2022, 29, 1809–1813. [Google Scholar] [CrossRef]
  75. Lee, S.H.; Jung, S.H.; Kim, T.M.; Rhee, J.K.; Park, H.C.; Kim, M.S.; Kim, S.S.; An, C.H.; Lee, S.H.; Chung, Y.J. Whole-exome sequencing identified mutational profiles of high-grade colon adenomas. Oncotarget 2017, 8, 6579–6588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Waszak, S.M.; Northcott, P.A.; Buchhalter, I.; Robinson, G.W.; Sutter, C.; Groebner, S.; Grund, K.B.; Brugières, L.; Jones, D.T.W.; Pajtler, K.W.; et al. Spectrum and prevalence of genetic predisposition in medulloblastoma: A retrospective genetic study and prospective validation in a clinical trial cohort. Lancet Oncol. 2018, 19, 785–798. [Google Scholar] [CrossRef]
  77. Yoshida, M.; Nakabayashi, K.; Yang, W.; Sato-Otsubo, A.; Tsujimoto, S.I.; Ogata-Kawata, H.; Kawai, T.; Ishiwata, K.; Sakamoto, M.; Okamura, K.; et al. Prevalence of pathogenic variants in cancer-predisposing genes in second cancer after childhood solid cancers. Cancer Med. 2023, 12, 11264–11273. [Google Scholar] [CrossRef]
  78. Poliani, L.; Greco, L.; Barile, M.; Dal Buono, A.; Bianchi, P.; Basso, G.; Giatti, V.; Genuardi, M.; Malesci, A.; Laghi, L.; et al. Canonical and uncanonical pathogenic germline variants in colorectal cancer patients by next-generation sequencing in a European referral center. ESMO Open 2022, 7, 100607. [Google Scholar] [CrossRef]
  79. Chang, N.C.; Sincennes, M.C.; Chevalier, F.P.; Brun, C.E.; Lacaria, M.; Segalés, J.; Muñoz-Cánoves, P.; Ming, H.; Rudnicki, M.A. The Dystrophin Glycoprotein Complex Regulates the Epigenetic Activation of Muscle Stem Cell Commitment. Cell Stem Cell 2018, 22, 755–768. [Google Scholar] [CrossRef] [Green Version]
  80. Zeng, K.; Wu, Y.; Wang, C.; Wang, S.; Sun, H.; Zou, R.; Sun, G.; Song, H.; Liu, W.; Sun, N.; et al. ASH2L is involved in promotion of endometrial cancer progression via upregulation of PAX2 transcription. Cancer Sci. 2020, 111, 2062–2077. [Google Scholar] [CrossRef]
  81. Qi, J.; Huo, L.; Zhu, Y.T.; Zhu, Y.J. Absent, small or homeotic 2-like protein (ASH2L) enhances the transcription of the estrogen receptor α gene through GATA-binding protein 3 (GATA3). J. Biol. Chem. 2014, 289, 31373–31381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Mungamuri, S.K.; Wang, S.; Manfredi, J.J.; Gu, W.; Aaronson, S.A. Ash2L enables P53-dependent apoptosis by favoring stable transcription pre-initiation complex formation on its pro-apoptotic target promoters. Oncogene 2015, 34, 2461–2470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yu, W.; Huang, W.; Yang, Y.; Qiu, R.; Zeng, Y.; Hou, Y.; Sun, G.; Shi, H.; Leng, S.; Feng, D.; et al. GATA3 recruits UTX for gene transcriptional activation to suppress metastasis of breast cancer. Cell Death Dis. 2019, 10, 832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Representative pedigree of three index patients following the hereditary breast and ovarian cancer syndrome criteria. Index patients 1 (A), 2 (B), and 3 (C). The arrowheads indicate the index patients. The numbers below the symbols represent the age at diagnosis of each tumor type. (D) Graphical representation of PMS2 gene. PMS2 gene spans 15 exons and 862 amino acids in length. Exons 2 to 15 deletion of the PMS2 gene is represented in red (Case 1). In blue, gains of exons 12 to 15 are detected in Case 2. Patient 3 presented the exon 1 loss of PMS2. (E). POLE2 gains were detected in index patients 1 and 3.
Figure 1. Representative pedigree of three index patients following the hereditary breast and ovarian cancer syndrome criteria. Index patients 1 (A), 2 (B), and 3 (C). The arrowheads indicate the index patients. The numbers below the symbols represent the age at diagnosis of each tumor type. (D) Graphical representation of PMS2 gene. PMS2 gene spans 15 exons and 862 amino acids in length. Exons 2 to 15 deletion of the PMS2 gene is represented in red (Case 1). In blue, gains of exons 12 to 15 are detected in Case 2. Patient 3 presented the exon 1 loss of PMS2. (E). POLE2 gains were detected in index patients 1 and 3.
Genes 14 01580 g001
Figure 2. Circus plot showing selected germline genomic variants (SNVs and CNVs) detected in three index patients. PMS2 copy number variations are highlighted in grey.
Figure 2. Circus plot showing selected germline genomic variants (SNVs and CNVs) detected in three index patients. PMS2 copy number variations are highlighted in grey.
Genes 14 01580 g002
Table 1. Clinical characteristics of the three HBOC/HBCC patients included in this study.
Table 1. Clinical characteristics of the three HBOC/HBCC patients included in this study.
PatientClinical FeaturesRelatives with Cancer
1BC (64—left, T1N0M0, IDC-NE)Sister (BC, 68)
BC (64—right, TxN1M0, adenocarcinoma, NI)
CRC (71, TxNxMx, rectal carcinoma, NI)
2BC (55—left, T2N2aM0, IDC-NE—ER+/PR−/HER2−, surgery + CT + RT)Sister (BC, 40 and CRC, 48)
BC (55—right, T2N0M0, IDC-NE—ER+/PR+/HER2+, surgery + CT + RT)Sister (thyroid cancer, 45)
CRC (57, T3N2M0, TA, surgery + CT)Maternal aunt (bone cancer, 52)
3BC (48—right, TxN0M0, IDC-NE, surgery + CT + RT)
CRC (56, T3N2M0, TA, surgery + CT)
Mother (bilateral BC, NI)
Sister (BC, NI and pancreas cancer, NI)
Maternal cousin (CRC, NI)
Paternal cousin (BC, NI)
Paternal cousin (BC, NI)
BC: breast cancer; CRC: colorectal cancer; CT: chemotherapy; ER: estrogen receptor; HBCC: hereditary breast and colorectal cancer syndrome according to [22,33]; HBOC: hereditary breast and ovarian cancer syndrome according to NCCN Guidelines Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic v2.2021; HER2: erb-b2 receptor tyrosine kinase 2; IDC-NE: invasive ductal carcinoma of non-specific type; NI: not informed; PR: progesterone receptor; RT: radiotherapy; TA: Tubular Adenocarcinoma; TNM staging system: T—tumor; N—lymph nodes; M—metastasis.
Table 2. Relevant single nucleotide variants and copy number variations identified in the three index patients.
Table 2. Relevant single nucleotide variants and copy number variations identified in the three index patients.
SINGLE NUCLEOTIDE VARIANTS
PatientGenecDNAProteinType/EffectdbNSFP *GnomAD Variant FrequencyNCBI RefSeqdbSNP 154 v2, NCBIACMG/ClinVar
1CYP1B1c.1159G > Ap.Glu387LysMissense40.00029NM_000104.4rs55989760LP/LP
NTRK1c.16C > Tp.Arg6TrpMissense40.00325NM_001012331.2rs201472270VUS/-
NFATC2c.1952G > Ap.Arg651HisMissense40.00006NM_173091.4rs148347040VUS/-
(+47 VUS)
2SLC3A1c.1400T > Cp.Met467ThrMissense50.00250NM_000341.4rs121912691VUS/LP
MAT1Ac.826dupGp.Ala276Glyfs*76LoF-0.00001NM_000429.3rs763178849LP/-
PTGISc.824G > Ap.Arg275GlnMissense30.00098NM_000961.4rs61734270VUS/LP
WRNc.95A > Gp.Lys32ArgMissense30.00305NM_000553.6rs34477820VUS/-
(+58 VUS)
3GNAQc.286A > Tp.Thr96SerMissense30.00440NM_002072.5rs753716491VUS/-
PTCH1c.140G > Tp.Arg47LeuMissense20NM_000264.5rs775408408VUS/-
(+31 VUS)
COPY NUMBER VARIATIONS
PatientGeneChromStartEndTypeOther GenesACMG/ClinVar
1PMS2757649556045662LossCCZ1, OCM, RNF216, RSPH10BVUS/VUS
POLE2145015485450160008Gain KLHDC1 VUS/-
NOMO1161494628714959012Loss - VUS/-
(+209 CNVs)
2PMS2760130306022622Gain - VUS/-
(+16 CNVs)
3PMS2760486286049129Loss AIMP2 VUS/P
ASH2L83791459937963256Loss EIF4EBP1 VUS/-
POLE2145012087350131392Gain - VUS/-
(+147 CNVs)
Chrom: chromosome; CNVs: copy number variations; LP: likely pathogenic; P: pathogenic; VUS: variant of uncertain significance. * Number of prediction algorithms (total of six) that indicated variants likely deleterious.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Côrtes, L.; Basso, T.R.; Villacis, R.A.R.; Souza, J.d.S.; Jørgensen, M.M.A.; Achatz, M.I.; Rogatto, S.R. Co-Occurrence of Germline Genomic Variants and Copy Number Variations in Hereditary Breast and Colorectal Cancer Patients. Genes 2023, 14, 1580. https://doi.org/10.3390/genes14081580

AMA Style

Côrtes L, Basso TR, Villacis RAR, Souza JdS, Jørgensen MMA, Achatz MI, Rogatto SR. Co-Occurrence of Germline Genomic Variants and Copy Number Variations in Hereditary Breast and Colorectal Cancer Patients. Genes. 2023; 14(8):1580. https://doi.org/10.3390/genes14081580

Chicago/Turabian Style

Côrtes, Luiza, Tatiane Ramos Basso, Rolando André Rios Villacis, Jeferson dos Santos Souza, Mads Malik Aagaard Jørgensen, Maria Isabel Achatz, and Silvia Regina Rogatto. 2023. "Co-Occurrence of Germline Genomic Variants and Copy Number Variations in Hereditary Breast and Colorectal Cancer Patients" Genes 14, no. 8: 1580. https://doi.org/10.3390/genes14081580

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop