Skip to main content

Unravelling genetic variants of a swedish family with high risk of prostate cancer

Abstract

Background

Prostate cancer is the most prevalent cancer in men worldwide. It is a polygenic disease with a substantial proportion of heritability. Identification of novel candidate biomarkers is crucial for clinical cancer prevention and the development of therapeutic strategies. Here, we describe the analysis of rare and common genetic variants that can predispose to the development of prostate cancer.

Methods

Whole-genome sequencing was performed on germline DNA of five Swedish siblings which were diagnosed with prostate cancer. The high-risk variants were identified setting the minor allele frequency < 0.01, CADD > 10 and if tested in PRACTICAL, OR > 1.5, while the low-risk variants were identified minor allele frequency > 0.01, CADD > 10 and if tested in PRACTICAL, OR > 1.1.

Results

We identified 38 candidate high-risk gene variants and 332 candidate low-risk gene variants, where 2 and 14 variants were in coding regions, respectively, that were shared by the brothers with prostate cancer.

Conclusions

This study expanded the knowledge of potential risk factor candidates involved in hereditary and familial prostate cancer. Our findings can be beneficial when applying targeted screening in families with a high risk of developing the disease.

Peer Review reports

Background

Prostate cancer (PrCa) is the second most frequent cancer in men worldwide and the most common diagnosed cancer among men in the United States and most of the Western world [1]. PrCa is a heterogeneous disease that can be associated with several risk factors including age, race, familial history of PrCa, diet or environmental factors [2]. Epidemiological and twin studies had supported the pivotal role of genetic predisposition in the development of PrCa. A positive familial history of PrCa has been associated with more than a two-fold increased risk of the disease [3]. Furthermore, having a brother with PrCa represents a greater risk to be affected by the disease than being a son of a PrCa patient [4]. In addition, Scandinavia twins studies have demonstrated that the 42% of PrCa risk was due to heritable factors and the probability of having PrCa was 21% for a man with a monozygotic twin affected and 6% for a man with a dizygotic twin affected [5] with 3.7 year of difference between the diagnose of the first and second monozygotic twin [6].

For these reasons, germline mutations are becoming the focus of increased numbers of research to look for inherited variations that are suitable for potential prognosis and treatment. Although the androgen receptor (AR) has a pivotal role in PrCa, mutations in the AR gene account for a small fraction of all case [7], underling that different genetic variations may contribute to the formation of prostate tumors. Evidence has shown that the genetic contribution to inherited PrCa is constituted by a mixture of rare gene variants with high to moderate penetrance and common variants with low penetrance. Linkage studies have identified 8q24 as a significant PrCa risk region [8] and the missense G84E variant in the HOXB13 [9] gene is highly associated with an increased risk of PrCa. It has been shown that variants in DNA repair genes, such as the BRCA1/2, ATM, CHEK2 and NBN genes, are associated with an increased risk of developing PrCa, in particular in men with advanced/metastatic PrCa [10]. Retrospective and association studies have shown that carriers of pathogenic BRCA2 variants are associated with two- to six-times higher relative risks of PrCa while carriers of pathogenic BRCA1 variants are associated with a moderate risk [11, 12]. Moreover, the knowledge of a patient’s germline status has become of emerging importance, in predicting the response to targeted treatments, especially in advanced or metastatic conditions [13].

The polygenic nature of PrCa susceptibility has brought the attention into genome-wide association study (GWAS) and to the formation of large collaborative international consortia that includes thousands of cases and controls to detect PrCa risk variants. Since the first GWAS in 2006 [14], around 170 variants have been identified to be risk factors for PrCa susceptibility. However, they can only explain approximately 38% of the familial relative risk of PrCa [15] and additional evidence is required to include them in routine genetic testing.

Here, we present the results of five brothers with PrCa and previously shown not to be carrying any of the high-risk variants so far identified in the literature. Through whole-genome sequencing on their germline DNA we identified novel high- and low-risk genetic variants that could contribute to the PrCa risk observed in their family.

Materials and methods

Study population

The individuals in this study were five brothers from a Swedish family that had undergone genetic counselling at the Department of Clinical Genetics, Karolinska University Hospital Solna, Sweden. The five brothers were diagnosed with PrCa between the age of 64–80, and they had also a brother diagnosed with a neuroendocrine pancreatic tumor (NET) at the age of 79 years, and a seventh brother who was healthy (at the age of 61 years).

Additional family history of PrCa includes their father and paternal uncle were diagnosed with PrCa. No other cancer was known in the family on the mother´s or father´s side. All seven brothers gave written informed consent to participate to the study and DNA was isolated from peripheral blood.

Whole-genome sequencing of prostate cancer family samples

DNA was quantified using a Qubit Fluorometer (Life Technologies, USA) and converted to sequencing libraries using a PCR-free paired-end protocol (Illumina TruSeq DNA PCR-free for > 1000 ng input). Sequencing was done using the Illumina NovaSeq 6000 platforms aiming at 30x median coverage and performed at Clinical Genomics Stockholm, SciLifeLab.

Bioinformatics workflow

Sequencing reads were aligned to the reference genome GRCh37 using BWA [16] and Picard (http://picard.sourceforge.net) was used to mark PCR-duplicated reads. Variants were called using GATK best practice procedure as implemented at the Broad Institute (www.broadinstitute.org/gatk). Variant annotation was done using Ensembl Variant Effect Predictor (VEP) [17], including RefSeq gene annotation and dbSNP153. Max minor allele frequency (MMAF) was obtained from Genome Aggregation Database [18] (gnomAD), or from the SweGen database [19] when missing from gnomAD. To predict pathogenic effects of the variants, the in silico prediction database CADD (Combined Annotation Dependent Depletion) [20] was used, while to predict the associated risk factor for PrCa the odds ratio (OR) and P values from the PRACTICAL project [21] was used. Variants in the autosomal and sex chromosomes were analysed. Variants located within segmental duplication regions obtained from the UCSC genome browser [22] were excluded.

Known hereditary cancer genes

Variants in 154 genes belonging to the Comprehensive Hereditary Cancer Panel (https://blueprintgenetics.com/) were investigated in the brothers to identify pathogenic variants. That includes the 10 genes (ATM, BRCA1,BRCA2, PALB2, CHEK2, HOXB13, MLH1, MSH2, MSH6, PMS2) recommended for genetic testing by the National Comprehensive Cancer Network guidelines for PrCa [23].

High- and low-risk PrCa predisposing variants

Variants detected in all siblings with PrCa were divided in two groups: rare high-risk variants and common low-risk variants. Rare high-risk variants were considered variants with MMAF (gnomAD/SWEGEN) < 0.01, CADD > 10 and if tested in PRACTICAL [21], OR > 1.5. Common low-risk variants were considered variants with MMAF (gnomAD) > 0.01, CADD > 10 and if tested in PRACTICAL [21], OR > 1.1 and P- values < 0.05. For both our categories risk factors were also considered the variants shared by all brothers with PrCa and not imputed in the custom high-density genotyping array, the OncoArray, from the PRACTICAL project [21]. Further, for all variants a CADD score greater of equal 10 indicates that these are predicted to be the 10% most deleterious substitutions that you can do to the human genome, while a score of greater or equal 20 indicates the 1% most deleterious.

Enrichment analysis

The annotated variants were imported in R and Bioconductor, the disgenet2r package [24] was used for the gene-neoplastic disease association analysis and neoplastic process enrichment analysis, while the clusterProfiler package [25] was used for or the gene ontology enrichment analysis.

Results

Identification of 38 novel candidate high-risk variants in inherited prostate cancer

To identify the PrCa predisposing genes we analysed the whole-genome sequence from five brothers which were diagnosed with PrCa. To test if they inherited genetic susceptibility to cancer we first checked if they had pathogenic variants in genes included in the comprehensive hereditary cancer panel (https://blueprintgenetics.com/tests/panels/hereditary-cancer/comprehensive-hereditarycancer-panel/#panel_content-headin) . We found 4 variants carried by all brothers but none of them had any known pathogenic consequences according to ClinVar (Table 1).

Table 1 Variants in coding regions of the genes included in the Comprehensive Hereditary Cancer carried by all siblings

Next, we searched for genetic variants that were shared by all sibling and we found 69,548 shared variants, of which 68,846 were in non-coding regions and 702 were in coding transcripts. Since the genetic contribution to inherited PrCa includes a variety of rare or common variants, we started our analysis of the shared variants by looking for high-risk candidates. First, we selected variants with MMAF < 0.01 and CADD > 10, to be defined as the most likely high-risk variants. We excluded variants with low risk (OR < 1.5) in the PRACTICAL study [21]. We identified 38 variants in 35 genes suggested to be associated with high PrCa risk. Most variants were intronic variants; however, two variants, rs150518260 and rs139884486, were found to be located within the coding regions of the SGCB and the IRF4 genes, respectively and they were also the most deleterious substitution (CADD > 20) that we found within our high-risk variant (Table 2). Furthermore, we found that eight of the suggested genes (NFIB, FBXW7, PPARA, ETV4, NCOA2, CYP7B1, OPCML, IRF4) have been demonstrated to be involved in neoplastic processes (Supplementary Fig. 1 A) and four of them, the ETV4, NCOA2, CYP7B1 and PPARA genes, have been shown to be associated with malignant neoplasms of the prostate (Supplementary Fig. 1B and Supplementary Table 1). None of the siblings carried the rare high-risk missense variant (G84E) on the HOXB13 gene but they were carrying one upstream variant in the non-coding region of the HOXB3 gene (Table 2).

Table 2 Rare and deleterious variants with MMAF < 0.01, CADD > 10, ODD Ratio > 1.5 shared by the five brothers with prostate cancer

Identification of novel candidate low-risk variants in inherited prostate cancer

Since the five siblings affected by PrCa were sharing a high number of variants, we decided, to study also low-risk PrCa candidates. All variants shared by the five affected brothers with MMAF > 0.01, CADD > 10 and OR > 1.1 for those being tested in the PRACTICAL study, were selected for further analysis.In total, 332 variants in 225 genes (Supplementary Table 2) were suggested as potential low-risk candidates in familial PrCa. We found that most of the variants, 227, were in the non-coding transcript region, while 88 variants were in the intragenic region and 17 in coding regions (Table 3). Interestingly, we found that the selected genes have been described in association with several neoplastic diseases with PrCa among the top enriched neoplastic process (Supplementary Fig. 2 A). Gene ontology analysis showed that these genes were enriched for pathways involved in early development but also for pathways related to cell division, DNA replication and apoptotic signaling (Supplementary Fig. 2B). Fourteen variants with potential pathogenic effect (non-synonymous, frameshift, in-frame deletion) were seen in 12 genes (Table 4), where four of the genes (KRT18, TET2, IL32, SMPD1) have already been shown to be associated with PrCa (Supplementary Fig. 2 C). Moreover, nine of this fourteen variants located in TET2, NOP16, PRIM2, CACNA1B, PSMD13, SMD13, KRT18, MEF18 and KRT10 genes belong to the 1% most deleterious substitution in the human genome with a CADD score > 20 (Table 4). Among the low-risk variants all siblings were carrying two upstream variants in the non-coding regions of the HOXB3 gene and one downstream variant in the intragenic region of the HOXB6 genes (Supplementary Table 2). In addition, we found that two variants in the KRT18 gene (chr12:53343318 C > T and chr12:53343325T > A) were located on the same allele.

Table 3 Overview of low-risk variant consequences with MMAF > 0.01, CADD > 10 and ODD Ratio > 1.1 shared by the brothers with prostate cancer
Table 4 Low risk variants with MMAF > 0.01, CADD > 10, ODD Ratio > 1.1 in coding transcript shared by all brothers with prostate cancer

Discussion

To identify potential variant contribution to hereditary PrCa, we sequenced the whole genome of siblings in a family with a seemingly high risk of PrCa. As expected less than 1% of the shared variants were located within protein-coding regions of the genome. Although the stability, the structure and biochemical function of the proteins are important factors implicated in the development of PrCa, several other mechanisms that regulate the transcriptional and translational level of the protein may have a pivotal role in driving tumorigenesis.

We initially screened for rare variants that could potentially represent high-risk factors in the development of PrCa. As a second approach we searched for variants considered of low-risk impact in the development of the disease. Two candidate high-risk missense variants were found in the SGCB and the IRF4 genes. The SGCB gene has not previously been linked to any neoplastic disease, mutations in this gene have been associated with the development of limb-girdle muscular dystrophy type 2E [26]. Instead, IRF4, an important regulator of the immune response, has been shown to be associated with many lymphoid malignancies with evidence pointing to a pivotal role in multiple myeloma [27]. Since inflammation is a risk factor for prostate carcinogenesis it could be possible that variations of the IFR4 gene may contribute to increased risk of PrCa. Moreover, we found that four of our high-risk variants were situated in genes earlier demonstrated to be involved in the initiation and progression of PrCa. It has been shown that ETV4 may be involved in the initial events of PrCa development when it is fused with the TMPRSS22 locus [28]. The expression of NCOA2 has been demonstrated to promote PrCa metastasis and its inhibition has therapeutic potential [29]. The expression of CYP7B1 has been shown to be upregulated in prostate cancer [30] and it is also been demonstrated that a single polymorphism in the promoter of this gene has an effect on its expression [31]. Furthermore, we observed a variant in the PPARA gene, which is overexpressed in advanced PrCa [32]. Variants in this gene have been associated with pesticide exposure and increased risk of PrCa [33] underling the importance of gene-environment interaction as a contributing factor to increased risk of PrCa.

In our analysis, we did not find pathogenic variants in any of the genes recommended for genetic testing by the National Comprehensive Cancer Network guidelines for PrCa [23], however in our high-risk group we found that they were carrying one novel variant in the upstream non coding region of the HOXB3 gene. Further, in our low-risk category we found that they were carrying two novel variants in the HOXB3 gene and one novel variant in the HOXB6 gene but none of them was carrying the well know high-risk missense variant (G84E) in HOXB13 gene [9]. Although none of the sibling was carrying the rare HOXB13 mutation (G84E) we could suggest, based on our results, that also HOXB3 and HOXB6 genes may be PrCa susceptibility genes and germline mutation on these genes can play a role in predisposing the disease. The variation in the HOXB3 and HOXB6 genes that we found where located in upstream and downstream intragenic region and misregulation of the expression of these genes may lead to changes of the downstream gene expression and signaling pathways that play fundamental roles in the development of PrCa.

We identified several candidates as low-risk variants, as well as two missense variants, rs34402524 and rs28927679 in the TET2 and PSMD13 genes, respectively, previously shown to be associated with PrCa low-risk (OR of 1.1) [21]. It is well known that aside genetic factors also epigenetic factors can contribute to the initiation and progression of PrCa [34]. TET2 is an enzyme involved in DNA demethylation, and it has been shown to be an important player during tumorigenesis [35]. TET2 has been shown to be able to bind to the androgen receptor and modulate its signalling pathway. Downregulation of TET2 drives the PrCa proliferation and is strongly associated with reduced patient survival suggesting that the expression levels of this protein can be used as a biomarker for PrCa progression [36]. DNA methylation studies have demonstrated that DNA methylation patter is often altered in cancer compared to normal tissue [37]. This underline that a better understanding of the role DNA methylases including the TET family could be a promising therapeutic target to classify and predict PrCa clinical outcomes more accurately than clinical parameters alone. Interestingly, we found three variants in the KRT18 gene, previously linked to PrCa. KRT18 is a well-known epithelial marker in diagnostic histopathology [38] and its downregulation is associated with prostate cancer aggressiveness [39].

This study has several limitations that need to be considered. The cohort of patients is limited to a single high-risk family containing five brothers with PrCa. The study does not include other affected or unaffected in the family. Strict criteria to identify high- and low-risk variants using population frequency excludes more common variants that could contribute to increased risk of PrCa. Furthermore, in silico prediction programs may be inaccurate resulting in exclusion of variants of high impact. Our variants are not validated with a secondary method or did we identify the variants in additional high-risk families. Moreover, our study does not include analysis of epigenetic variation that could contribute to the increased risk of PrCa of this particular family.

Conclusions

PrCa is a complex disease which risk can be influenced by several genes and pathways. The identification of risk genes is crucial for genetic counselling of PrCa families and to apply the proper therapeutic strategies. The effect of a single genetic variant on the relative risk is probably low. Our study provided additional support that cumulative variants with low- or moderate effect in the germline of an individual contribute to the risk of PrCa. Further studies are needed to estimate the contribution of these variants and genes to PrCa.

Availability of data and materials

Access to the data is controlled. Variants that fulfilled our selection criteria can be found in the tables and supplementary tables. However, Swedish laws and regulations prohibit the release of individual and personally identifying data. Therefore, the whole data cannot be made publicly available. The data that support the findings of this study are available from the corresponding author upon reasonable request.

Abbreviations

PrCa:

prostate cancer

AR:

androgen receptor

GWAS:

genome-wide association study

HOXB3/6/16 :

Homeobox B3/6/16

BRCA1/2 :

Breast cancer type1/2

ATM :

ATM Serine/Threonine Kinase

CHEK2 :

Checkpoint Kinase 2

NBN :

Nibrin

SGCB :

Sarcoglycan Beta

IRF4 :

Interferon Regulatory Factor 4

NFIB :

Nuclear Factor I B

PPARA :

Peroxisome Proliferator Activated Receptor Alpha

ETV4 :

ETS Variant Transcription Factor 4

NCOA2 :

Nuclear Receptor Coactivator 2

ESRRG :

Estrogen Related Receptor Gamma

OPCML :

Opioid Binding Protein/Cell Adhesion Molecule Like

KRT18 :

Keratin 18

TET2 :

Tet Methylcytosine Dioxygenase 2

IL32 :

Interleukin 32

SMPD1 :

Sphingomyelin Phosphodiesterase 1

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Cancer J Clin. 2018;68:394–424.

  2. Patel AR, Klein EA. Risk factors for prostate cancer. Nat Clin Pract Urol Nat Publishing Group. 2009;6:87–95.

    Article  Google Scholar 

  3. Kiciński M, Vangronsveld J, Nawrot TS. An Epidemiological Reappraisal of the Familial Aggregation of Prostate Cancer: A Meta-Analysis. PLOS ONE Public Library of Science. 2011;6:e27130.

    Article  Google Scholar 

  4. Zeegers MPA, Jellema A, Ostrer H. Empiric risk of prostate carcinoma for relatives of patients with prostate carcinoma. Cancer. 2003;97:1894–903.

    Article  Google Scholar 

  5. Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J, Koskenvuo M, et al. Environmental and Heritable Factors in the Causation of Cancer — Analyses of Cohorts of Twins from Sweden, Denmark, and Finland. N Engl J Med. 2000;343:78–85.

    Article  CAS  Google Scholar 

  6. Mucci LA, Hjelmborg JB, Harris JR, Czene K, Havelick DJ, Scheike T, et al. Familial Risk and Heritability of Cancer Among Twins in Nordic Countries. JAMA. 2016;315:68.

    Article  CAS  Google Scholar 

  7. Marcelli M, Ittmann M, Mariani S, Sutherland R, Nigam R, Murthy L, et al. Androgen Receptor Mutations in Prostate Cancer. Cancer Res Am Assoc Cancer Res. 2000;60:944–9.

    CAS  Google Scholar 

  8. Amundadottir LT, Sulem P, Gudmundsson J, Helgason A, Baker A, Agnarsson BA, et al. A common variant associated with prostate cancer in European and African populations. Nat Genet. 2006;38:652–8.

    Article  CAS  Google Scholar 

  9. Ewing CM, Ray AM, Lange EM, Zuhlke KA, Robbins CM, Tembe WD, et al. Germline Mutations in HOXB13 and Prostate-Cancer Risk [Internet]. https://doi.org/10.1056/NEJMoa1110000. Massachusetts Medical Society; 2012 [cited 2021 Jul 1]. Available from: https://www.nejm.org/doi/10.1056/NEJMoa1110000.

  10. Pritchard CC, Mateo J, Walsh MF, De Sarkar N, Abida W, Beltran H, et al. Inherited DNA-Repair Gene Mutations in Men with Metastatic Prostate Cancer. N Engl J Med. 2016;375:443–53.

    Article  CAS  Google Scholar 

  11. Nyberg T, Frost D, Barrowdale D, Evans DG, Bancroft E, Adlard J, et al. Prostate Cancer Risks for Male BRCA1 and BRCA2 Mutation Carriers: A Prospective Cohort Study. Eur Urol. 2020;77:24–35.

    Article  CAS  Google Scholar 

  12. Oh M, Alkhushaym N, Fallatah S, Althagafi A, Aljadeed R, Alsowaida Y, et al. The association of BRCA1 and BRCA2 mutations with prostate cancer risk, frequency, and mortality: A meta-analysis. Prostate. 2019;79:880–95.

    Article  CAS  Google Scholar 

  13. Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, et al. DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N Engl J Med. 2015;373:1697–708.

    Article  CAS  Google Scholar 

  14. Gudmundsson J, Sulem P, Manolescu A, Amundadottir LT, Gudbjartsson D, Helgason A, et al. Genome-wide association study identifies a second prostate cancer susceptibility variant at 8q24. Nat Genet. 2007;39:631–7.

    Article  CAS  Google Scholar 

  15. Benafif S, Kote-Jarai Z, Eeles RA. A Review of Prostate Cancer Genome-Wide Association Studies (GWAS). Cancer Epidemiol Biomarkers Prev.  2018;27:845–57.

  16. Li H, Durbin R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics. 2009;25:1754–60.

    Article  CAS  Google Scholar 

  17. McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GRS, Thormann A, et al. The Ensembl Variant Effect Predictor Genome Biology. 2016;17:122.

    PubMed  Google Scholar 

  18. Karczewski KJ, Francioli LC, Tiao G, Cummings BB, Alföldi J, Wang Q, et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020;581:434–43.

    Article  CAS  Google Scholar 

  19. Ameur A, Dahlberg J, Olason P, Vezzi F, Karlsson R, Martin M, et al. SweGen: a whole-genome data resource of genetic variability in a cross-section of the Swedish population. Eur J Hum Genet. 2017;25:1253–60.

    Article  CAS  Google Scholar 

  20. Rentzsch P, Schubach M, Shendure J, Kircher M. CADD-Splice—improving genome-wide variant effect prediction using deep learning-derived splice scores. Genome Med. 2021;13:31.

    Article  CAS  Google Scholar 

  21. Schumacher FR, Al Olama AA, Berndt SI, Benlloch S, Ahmed M, Saunders EJ, et al. Association analyses of more than 140,000 men identify 63 new prostate cancer susceptibility loci. Nat Genet. 2018;50:928–36.

    Article  CAS  Google Scholar 

  22. Bailey JA, Gu Z, Clark RA, Reinert K, Samonte RV, Schwartz S, et al. Recent Segmental Duplications in the Human Genome. 297: Science. American Association for the Advancement of Science; 2002. pp. 1003–7.

  23. Mohler JL, Antonarakis ES, Armstrong AJ, D’Amico AV, Davis BJ, Dorff T, et al. Prostate Cancer, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Netw Natl Compr Cancer Netw. 2019;17:479–505.

    Article  CAS  Google Scholar 

  24. Piñero J, Ramírez-Anguita JM, Saüch-Pitarch J, Ronzano F, Centeno E, Sanz F, et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res. 2020;48:D845–55.

    PubMed  Google Scholar 

  25. Yu G, Wang L-G, Han Y, He Q-Y clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. OMICS: A Journal of Integrative Biology. Mary Ann Liebert, Inc., publishers; 2012;16:284–7.

  26. Semplicini C, Vissing J, Dahlqvist JR, Stojkovic T, Bello L, Witting N, et al. Clinical and genetic spectrum in limb-girdle muscular dystrophy type 2E. Neurology. 84: Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology; 2015. pp. 1772–81.

  27. Agnarelli A, Chevassut T, Mancini EJ. IRF4 in multiple myeloma—Biology, disease and therapeutic target. Leuk Res. 2018;72:52–8.

    Article  CAS  Google Scholar 

  28. Tomlins SA, Mehra R, Rhodes DR, Smith LR, Roulston D, Helgeson BE, et al. TMPRSS2:ETV4 Gene Fusions Define a Third Molecular Subtype of Prostate Cancer. Cancer Res American Association for Cancer Research. 2006;66:3396–400.

    CAS  Google Scholar 

  29. Qin J, Lee H-J, Wu S-P, Lin S-C, Lanz RB, Creighton CJ, et al. Androgen deprivation–induced NCoA2 promotes metastatic and castration-resistant prostate cancer. J Clin Invest American Society for Clinical Investigation. 2014;124:5013–26.

    Article  CAS  Google Scholar 

  30. Olsson M, Gustafsson O, Skogastierna C, Tolf A, Rietz BD, Morfin R, et al. Regulation and expression of human CYP7B1 in prostate: overexpression of CYP7B1 during progression of prostatic adenocarcinoma. Prostate. 2007;67:1439–46.

    Article  CAS  Google Scholar 

  31. Jakobsson J, Karypidis H, Johansson J-E, Roh H-K, Rane A, Ekström L. A functional C-G polymorphism in the CYP7B1 promoter region and its different distribution in Orientals and Caucasians. Pharmacogenomics J. 2004;4:245–50.

    Article  CAS  Google Scholar 

  32. Collett GP, Betts AM, Johnson MI, Pulimood AB, Cook S, Neal DE, et al. Peroxisome Proliferator-activated Receptor α Is an Androgen-responsive Gene in Human Prostate and Is Highly Expressed in Prostatic Adenocarcinoma. Clin Cancer Res American Association for Cancer Research. 2000;6:3241–8.

    CAS  Google Scholar 

  33. Andreotti G, Koutros S, Berndt SI, Hughes Barry K, Hou L, Hoppin JA, et al. The Interaction between Pesticide Use and Genetic Variants Involved in Lipid Metabolism on Prostate Cancer Risk. J Cancer Epidemiol Hindawi. 2012;2012:e358076.

    Google Scholar 

  34. Kumaraswamy A, Welker Leng KR, Westbrook TC, Yates JA, Zhao SG, Evans CP, et al. Recent Advances in Epigenetic Biomarkers and Epigenetic Targeting in Prostate Cancer. Eur Urol. 2021;80:71–81.

    Article  CAS  Google Scholar 

  35. Seethy A, Pethusamy K, Chattopadhyay I, Sah R, Chopra A, Dhar R, et al. TETology: Epigenetic Mastermind in Action. Appl Biochem Biotechnol. 2021;193:1701–26.

    Article  CAS  Google Scholar 

  36. Nickerson ML, Das S, Im KM, Turan S, Berndt SI, Li H, et al. TET2 binds the androgen receptor and loss is associated with prostate cancer. Oncogene. 2017;36:2172–83.

    Article  CAS  Google Scholar 

  37. Geybels MS, Zhao S, Wong C-J, Bibikova M, Klotzle B, Wu M, et al. Epigenomic profiling of DNA methylation in paired prostate cancer versus adjacent benign tissue. Prostate. 2015;75:1941–50.

    Article  CAS  Google Scholar 

  38. Moll R, Franke WW, Schiller DL, Geiger B, Krepler R. The catalog of human cytokeratins: patterns of expression in normal epithelia, tumors and cultured cells. Cell. 1982;31:11–24.

    Article  CAS  Google Scholar 

  39. Yin B, Zhang M, Zeng Y, Li Y, Zhang C, Getzenberg RH, et al. Downregulation of cytokeratin 18 is associated with paclitaxel–resistance and tumor aggressiveness in prostate cancer. Int J Oncol. 2016;48:1730–6.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We acknowledge “The PRACTICAL consortium, CRUK, BPC3, CAPS, PEGASUS”, the support from Science for Life Laboratory, the Clinical Genomics Stockholm (funded by the Swedish Research Council), Swedish National Infrastructure for Computing (SNIC) (partially funded by the Swedish Research Council through Grant Agreement No. 2018–05973) and the Uppsala Multidisciplinary Center for Advanced Computational Science (UPPMAX) for assistance with massively parallel sequencing.

Funding

Open access funding provided by Karolinska Institute. This work was funded by the Swedish Research Council, the Swedish Cancer Society and the Cancer Research Funds of Radiumhemmet.

Author information

Authors and Affiliations

Authors

Contributions

A.L. conceived and designed the study. S.B. performed the bioinformatic analysis. All co-authors were involved in data interpretation and discussion. S.B. wrote the manuscript. H.H., and A.L. further edited the manuscript. The author(s) read and approved the final manuscript.

Corresponding authors

Correspondence to Serena Barilla or Hafdis T. Helgadottir.

Ethics declarations

Ethics approval and consent to participate

All patients gave written informed consent to participate in the study and to donate blood samples. The study was approved by the research ethics committee at Karolinska Institutet and the regional ethics committee in Stockholm (2008-125-32, 2014-1324-32, 2019–02333, 2020 − 00953).

Competing interests

The authors declare no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Barilla, S., Lindblom, A. & Helgadottir, H.T. Unravelling genetic variants of a swedish family with high risk of prostate cancer. Hered Cancer Clin Pract 20, 28 (2022). https://doi.org/10.1186/s13053-022-00234-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13053-022-00234-0

Keywords