Skip to main content

Advertisement

Log in

The RAD51D c.82G>A (p.Val28Met) variant disrupts normal splicing and is associated with hereditary ovarian cancer

  • Brief Report
  • Published:
Breast Cancer Research and Treatment Aims and scope Submit manuscript

Abstract

Purpose

Mutations in RAD51D are associated with a predisposition to primary ovarian, fallopian tube, and peritoneal carcinoma. Our study aims to characterize a RAD51D missense variant in a hereditary ovarian cancer family.

Methods

The effects of the RAD51D c.82G>A (p.Val28Met) variant on mRNA splicing were evaluated and characterized using RT-PCR, cloning and DNA sequencing.

Results

This variant completely disrupts normal splicing and results in the loss of 3′end of 5′UTR and the entire exon 1 (c.-86_c.82), which presumably leads to loss of the RAD51D protein. The RAD51D c.82G>A (p.Val28Met) variant is clinically significant and classified as likely pathogenic.

Conclusions

Our results indicate that the RAD51D c.82G>A (p.Val28Met) variant contributes to cancer predisposition through disruption of normal mRNA splicing. The identification of this variant in an individual affected with high-grade serous fallopian tube cancer suggests that the RAD51D variant may contribute to predisposition to the ovarian cancer in this family.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

References

  1. Pittman DL, Weinberg LR, Schimenti JC (1998) Identification, characterization, and genetic mapping of Rad51d, a new mouse and human RAD51/RecA-related gene. Genomics 49(1):103–111. https://doi.org/10.1006/geno.1998.5226

    Article  CAS  PubMed  Google Scholar 

  2. Kim YM, Choi BS (2011) Structural and functional characterization of the N-terminal domain of human Rad51D. Int J Biochem Cell Biol 43(3):416–422. https://doi.org/10.1016/j.biocel.2010.11.014

    Article  CAS  PubMed  Google Scholar 

  3. Braybrooke JP, Li JL, Wu L, Caple F, Benson FE, Hickson ID (2003) Functional interaction between the Bloom’s syndrome helicase and the RAD51 paralog, RAD51L3 (RAD51D). J Biol Chem 278(48):48357–48366. https://doi.org/10.1074/jbc.M308838200

    Article  CAS  PubMed  Google Scholar 

  4. Masson JY, Tarsounas MC, Stasiak AZ, Stasiak A, Shah R, McIlwraith MJ, Benson FE, West SC (2001) Identification and purification of two distinct complexes containing the five RAD51 paralogs. Genes Dev 15(24):3296–3307. https://doi.org/10.1101/gad.947001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Chun J, Buechelmaier ES, Powell SN (2013) Rad51 paralog complexes BCDX2 and CX3 act at different stages in the BRCA1-BRCA2-dependent homologous recombination pathway. Mol Cell Biol 33(2):387–395. https://doi.org/10.1128/MCB.00465-12

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Tarsounas M, Munoz P, Claas A, Smiraldo PG, Pittman DL, Blasco MA, West SC (2004) Telomere maintenance requires the RAD51D recombination/repair protein. Cell 117(3):337–347

    Article  CAS  Google Scholar 

  7. Reh WA, Nairn RS, Lowery MP, Vasquez KM (2017) The homologous recombination protein RAD51D protects the genome from large deletions. Nucleic Acids Res 45(4):1835–1847. https://doi.org/10.1093/nar/gkw1204

    Article  CAS  PubMed  Google Scholar 

  8. Pittman DL, Schimenti JC (2000) Midgestation lethality in mice deficient for the RecA-related gene, Rad51d/Rad51l3. Genesis 26(3):167–173

    Article  CAS  Google Scholar 

  9. Loveday C, Turnbull C, Ramsay E, Hughes D, Ruark E, Frankum JR, Bowden G, Kalmyrzaev B, Warren-Perry M, Snape K, Adlard JW, Barwell J, Berg J, Brady AF, Brewer C, Brice G, Chapman C, Cook J, Davidson R, Donaldson A, Douglas F, Greenhalgh L, Henderson A, Izatt L, Kumar A, Lalloo F, Miedzybrodzka Z, Morrison PJ, Paterson J, Porteous M, Rogers MT, Shanley S, Walker L, Breast Cancer Susceptibility C, Eccles D, Evans DG, Renwick A, Seal S, Lord CJ, Ashworth A, Reis-Filho JS, Antoniou AC, Rahman N (2011) Germline mutations in RAD51D confer susceptibility to ovarian cancer. Nat Genet 43(9):879–882. https://doi.org/10.1038/ng.893

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Thompson ER, Rowley SM, Sawyer S, Eccles DM, Trainer AH, Mitchell G, James PA, Campbell IG (2013) Analysis of RAD51D in ovarian cancer patients and families with a history of ovarian or breast cancer. PLoS ONE 8(1):e54772. https://doi.org/10.1371/journal.pone.0054772

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wickramanayake A, Bernier G, Pennil C, Casadei S, Agnew KJ, Stray SM, Mandell J, Garcia RL, Walsh T, King MC, Swisher EM (2012) Loss of function germline mutations in RAD51D in women with ovarian carcinoma. Gynecol Oncol 127(3):552–555. https://doi.org/10.1016/j.ygyno.2012.09.009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Osher DJ, De Leeneer K, Michils G, Hamel N, Tomiak E, Poppe B, Leunen K, Legius E, Shuen A, Smith E, Arseneau J, Tonin P, Matthijs G, Claes K, Tischkowitz MD, Foulkes WD (2012) Mutation analysis of RAD51D in non-BRCA1/2 ovarian and breast cancer families. Br J Cancer 106(8):1460–1463. https://doi.org/10.1038/bjc.2012.87

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ollier M, Radosevic-Robin N, Kwiatkowski F, Ponelle F, Viala S, Privat M, Uhrhammer N, Bernard-Gallon D, Penault-Llorca F, Bignon YJ, Bidet Y (2015) DNA repair genes implicated in triple negative familial non-BRCA1/2 breast cancer predisposition. Am J Cancer Res 5(7):2113–2126

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Hirasawa A, Imoto I, Naruto T, Akahane T, Yamagami W, Nomura H, Masuda K, Susumu N, Tsuda H, Aoki D (2017) Prevalence of pathogenic germline variants detected by multigene sequencing in unselected Japanese patients with ovarian cancer. Oncotarget 8(68):112258–112267. https://doi.org/10.18632/oncotarget.22733

    Article  PubMed  PubMed Central  Google Scholar 

  15. Song H, Dicks E, Ramus SJ, Tyrer JP, Intermaggio MP, Hayward J, Edlund CK, Conti D, Harrington P, Fraser L, Philpott S, Anderson C, Rosenthal A, Gentry-Maharaj A, Bowtell DD, Alsop K, Cicek MS, Cunningham JM, Fridley BL, Alsop J, Jimenez-Linan M, Hogdall E, Hogdall CK, Jensen A, Kjaer SK, Lubinski J, Huzarski T, Jakubowska A, Gronwald J, Poblete S, Lele S, Sucheston-Campbell L, Moysich KB, Odunsi K, Goode EL, Menon U, Jacobs IJ, Gayther SA, Pharoah PD (2015) Contribution of germline mutations in the RAD51B, RAD51C, and RAD51D genes to ovarian cancer in the population. J Clin Oncol 33(26):2901–2907. https://doi.org/10.1200/JCO.2015.61.2408

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Norquist BM, Harrell MI, Brady MF, Walsh T, Lee MK, Gulsuner S, Bernards SS, Casadei S, Yi Q, Burger RA, Chan JK, Davidson SA, Mannel RS, DiSilvestro PA, Lankes HA, Ramirez NC, King MC, Swisher EM, Birrer MJ (2016) Inherited mutations in women with ovarian carcinoma. JAMA Oncol 2(4):482–490. https://doi.org/10.1001/jamaoncol.2015.5495

    Article  PubMed  PubMed Central  Google Scholar 

  17. Kraus C, Hoyer J, Vasileiou G, Wunderle M, Lux MP, Fasching PA, Krumbiegel M, Uebe S, Reuter M, Beckmann MW, Reis A (2017) Gene panel sequencing in familial breast/ovarian cancer patients identifies multiple novel mutations also in genes others than BRCA1/2. Int J Cancer 140(1):95–102. https://doi.org/10.1002/ijc.30428

    Article  CAS  PubMed  Google Scholar 

  18. Chen X, Li Y, Ouyang T, Li J, Wang T, Fan Z, Fan T, Lin B, Xie Y (2018) Associations between RAD51D germline mutations and breast cancer risk and survival in BRCA1/2-negative breast cancers. Ann Oncol 29(10):2046–2051. https://doi.org/10.1093/annonc/mdy338

    Article  CAS  PubMed  Google Scholar 

  19. Couch FJ, Hart SN, Sharma P, Toland AE, Wang X, Miron P, Olson JE, Godwin AK, Pankratz VS, Olswold C, Slettedahl S, Hallberg E, Guidugli L, Davila JI, Beckmann MW, Janni W, Rack B, Ekici AB, Slamon DJ, Konstantopoulou I, Fostira F, Vratimos A, Fountzilas G, Pelttari LM, Tapper WJ, Durcan L, Cross SS, Pilarski R, Shapiro CL, Klemp J, Yao S, Garber J, Cox A, Brauch H, Ambrosone C, Nevanlinna H, Yannoukakos D, Slager SL, Vachon CM, Eccles DM, Fasching PA (2015) Inherited mutations in 17 breast cancer susceptibility genes among a large triple-negative breast cancer cohort unselected for family history of breast cancer. J Clin Oncol 33(4):304–311. https://doi.org/10.1200/JCO.2014.57.1414

    Article  CAS  PubMed  Google Scholar 

  20. Couch FJ, Shimelis H, Hu C, Hart SN, Polley EC, Na J, Hallberg E, Moore R, Thomas A, Lilyquist J, Feng B, McFarland R, Pesaran T, Huether R, LaDuca H, Chao EC, Goldgar DE, Dolinsky JS (2017) Associations between cancer predisposition testing panel genes and breast cancer. JAMA Oncol 3(9):1190–1196. https://doi.org/10.1001/jamaoncol.2017.0424

    Article  PubMed  PubMed Central  Google Scholar 

  21. Golmard L, Castera L, Krieger S, Moncoutier V, Abidallah K, Tenreiro H, Lauge A, Tarabeux J, Millot GA, Nicolas A, Lae M, Abadie C, Berthet P, Polycarpe F, Frebourg T, Elan C, de Pauw A, Gauthier-Villars M, Buecher B, Stern MH, Stoppa-Lyonnet D, Vaur D, Houdayer C (2017) Contribution of germline deleterious variants in the RAD51 paralogs to breast and ovarian cancers. Eur J Hum Genet 25(12):1345–1353. https://doi.org/10.1038/s41431-017-0021-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gonzalez-Rivera M, Lobo M, Lopez-Tarruella S, Jerez Y, Del Monte-Millan M, Massarrah T, Ramos-Medina R, Ocana I, Picornell A, Santillan Garzon S, Perez-Carbornero L, Garcia-Saenz JA, Gomez H, Moreno F, Marquez-Rodas I, Fuentes H, Martin M (2016) Frequency of germline DNA genetic findings in an unselected prospective cohort of triple-negative breast cancer patients participating in a platinum-based neoadjuvant chemotherapy trial. Breast Cancer Res Treat 156(3):507–515. https://doi.org/10.1007/s10549-016-3792-1

    Article  CAS  PubMed  Google Scholar 

  23. Gutierrez-Enriquez S, Bonache S, de Garibay GR, Osorio A, Santamarina M, Ramon y Cajal T, Esteban-Cardenosa E, Tenes A, Yanowsky K, Barroso A, Montalban G, Blanco A, Cornet M, Gadea N, Infante M, Caldes T, Diaz-Rubio E, Balmana J, Lasa A, Vega A, Benitez J, de la Hoya M, Diez O (2014) About 1% of the breast and ovarian Spanish families testing negative for BRCA1 and BRCA2 are carriers of RAD51D pathogenic variants. Int J Cancer 134(9):2088–2097. https://doi.org/10.1002/ijc.28540

    Article  CAS  PubMed  Google Scholar 

  24. Hauke J, Horvath J, Gross E, Gehrig A, Honisch E, Hackmann K, Schmidt G, Arnold N, Faust U, Sutter C, Hentschel J, Wang-Gohrke S, Smogavec M, Weber BHF, Weber-Lassalle N, Weber-Lassalle K, Borde J, Ernst C, Altmuller J, Volk AE, Thiele H, Hubbel V, Nurnberg P, Keupp K, Versmold B, Pohl E, Kubisch C, Grill S, Paul V, Herold N, Lichey N, Rhiem K, Ditsch N, Ruckert C, Wappenschmidt B, Auber B, Rump A, Niederacher D, Haaf T, Ramser J, Dworniczak B, Engel C, Meindl A, Schmutzler RK, Hahnen E (2018) Gene panel testing of 5589 BRCA1/2-negative index patients with breast cancer in a routine diagnostic setting: results of the German Consortium for Hereditary Breast and Ovarian Cancer. Cancer Med 7(4):1349–1358. https://doi.org/10.1002/cam4.1376

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, Mortimer P, Swaisland H, Lau A, O’Connor MJ, Ashworth A, Carmichael J, Kaye SB, Schellens JH, de Bono JS (2009) Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 361(2):123–134. https://doi.org/10.1056/NEJMoa0900212

    Article  CAS  PubMed  Google Scholar 

  26. Ashworth A (2008) A synthetic lethal therapeutic approach: poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. J Clin Oncol 26(22):3785–3790. https://doi.org/10.1200/JCO.2008.16.0812

    Article  CAS  PubMed  Google Scholar 

  27. Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, Weitzel JN, Friedlander M, Arun B, Loman N, Schmutzler RK, Wardley A, Mitchell G, Earl H, Wickens M, Carmichael J (2010) Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet 376(9737):235–244. https://doi.org/10.1016/S0140-6736(10)60892-6

    Article  CAS  PubMed  Google Scholar 

  28. Brown JS, Kaye SB, Yap TA (2016) PARP inhibitors: the race is on. Br J Cancer 114(7):713–715. https://doi.org/10.1038/bjc.2016.67

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kaufman B, Shapira-Frommer R, Schmutzler RK, Audeh MW, Friedlander M, Balmana J, Mitchell G, Fried G, Stemmer SM, Hubert A, Rosengarten O, Steiner M, Loman N, Bowen K, Fielding A, Domchek SM (2015) Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol 33(3):244–250. https://doi.org/10.1200/JCO.2014.56.2728

    Article  CAS  PubMed  Google Scholar 

  30. Dal Molin GZ, Omatsu K, Sood AK, Coleman RL (2018) Rucaparib in ovarian cancer: an update on safety, efficacy and place in therapy. Ther Adv Med Oncol. https://doi.org/10.1177/1758835918778483

    Article  Google Scholar 

  31. Shroff RT, Hendifar A, McWilliams RR, Geva R, Epelbaum R, Rolfe L, Goble S, Lin KK, Biankin AV, Giordano H, Vonderheide RH, Domchek SM (2018) Rucaparib monotherapy in patients with pancreatic cancer and a known deleterious BRCA mutation. JCO Precis Oncol. https://doi.org/10.1200/PO.17.00316

    Article  PubMed  PubMed Central  Google Scholar 

  32. Robson M, Im SA, Senkus E, Xu B, Domchek SM, Masuda N, Delaloge S, Li W, Tung N, Armstrong A, Wu W, Goessl C, Runswick S, Conte P (2017) Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med 377(6):523–533. https://doi.org/10.1056/NEJMoa1706450

    Article  CAS  Google Scholar 

  33. Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh DY, Reinacher-Schick A, Tortora G, Algul H, O’Reilly EM, McGuinness D, Cui KY, Schlienger K, Locker GY, Kindler HL (2019) Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med 381(4):317–327. https://doi.org/10.1056/NEJMoa1903387

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, Lisyanskaya A, Floquet A, Leary A, Sonke GS, Gourley C, Banerjee S, Oza A, Gonzalez-Martin A, Aghajanian C, Bradley W, Mathews C, Liu J, Lowe ES, Bloomfield R, DiSilvestro P (2018) Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med 379(26):2495–2505. https://doi.org/10.1056/NEJMoa1810858

    Article  CAS  PubMed  Google Scholar 

  35. Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, Nava Rodrigues D, Robinson D, Omlin A, Tunariu N, Boysen G, Porta N, Flohr P, Gillman A, Figueiredo I, Paulding C, Seed G, Jain S, Ralph C, Protheroe A, Hussain S, Jones R, Elliott T, McGovern U, Bianchini D, Goodall J, Zafeiriou Z, Williamson CT, Ferraldeschi R, Riisnaes R, Ebbs B, Fowler G, Roda D, Yuan W, Wu YM, Cao X, Brough R, Pemberton H, A’Hern R, Swain A, Kunju LP, Eeles R, Attard G, Lord CJ, Ashworth A, Rubin MA, Knudsen KE, Feng FY, Chinnaiyan AM, Hall E, de Bono JS (2015) DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med 373(18):1697–1708. https://doi.org/10.1056/NEJMoa1506859

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ray-Coquard I, Pautier P, Pignata S, Perol D, Gonzalez-Martin A, Berger R, Fujiwara K, Vergote I, Colombo N, Maenpaa J, Selle F, Sehouli J, Lorusso D, Guerra Alia EM, Reinthaller A, Nagao S, Lefeuvre-Plesse C, Canzler U, Scambia G, Lortholary A, Marme F, Combe P, de Gregorio N, Rodrigues M, Buderath P, Dubot C, Burges A, You B, Pujade-Lauraine E, Harter P, Investigators P (2019) Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N Engl J Med 381(25):2416–2428. https://doi.org/10.1056/NEJMoa1911361

    Article  CAS  PubMed  Google Scholar 

  37. Takata M, Sasaki MS, Tachiiri S, Fukushima T, Sonoda E, Schild D, Thompson LH, Takeda S (2001) Chromosome instability and defective recombinational repair in knockout mutants of the five Rad51 paralogs. Mol Cell Biol 21(8):2858–2866. https://doi.org/10.1128/MCB.21.8.2858-2866.2001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Swisher EM, Lin KK, Oza AM, Scott CL, Giordano H, Sun J, Konecny GE, Coleman RL, Tinker AV, O’Malley DM, Kristeleit RS, Ma L, Bell-McGuinn KM, Brenton JD, Cragun JM, Oaknin A, Ray-Coquard I, Harrell MI, Mann E, Kaufmann SH, Floquet A, Leary A, Harding TC, Goble S, Maloney L, Isaacson J, Allen AR, Rolfe L, Yelensky R, Raponi M, McNeish IA (2017) Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol 18(1):75–87. https://doi.org/10.1016/S1470-2045(16)30559-9

    Article  CAS  PubMed  Google Scholar 

  39. Agarwal N, Kutlar F, Mojica-Henshaw MP, Ou CN, Gaikwad A, Reading NS, Bailey L, Kutlar A, Prchal JT (2007) Missense mutation of the last nucleotide of exon 1 (G->C) of beta globin gene not only leads to undetectable mutant peptide and transcript but also interferes with the expression of wild allele. Haematologica 92(12):1715–1716. https://doi.org/10.3324/haematol.11543

    Article  CAS  PubMed  Google Scholar 

  40. Ozkara HA, Sandhoff K (2003) A new point mutation (G412 to A) at the last nucleotide of exon 3 of hexosaminidase alpha-subunit gene affects splicing. Brain Dev 25(3):203–206

    Article  Google Scholar 

  41. Zhang L, Chen L, Bacares R, Ruggeri JM, Somar J, Kemel Y, Stadler ZK, Offit K (2011) BRCA1 R71K missense mutation contributes to cancer predisposition by increasing alternative transcript levels. Breast Cancer Res Treat 130(3):1051–1056. https://doi.org/10.1007/s10549-011-1732-7

    Article  CAS  PubMed  Google Scholar 

  42. Vettore S, De Rocco D, Gerber B, Scandellari R, Bianco AM, Balduini CL, Pecci A, Fabris F, Savoia A (2010) A G to C transversion at the last nucleotide of exon 25 of the MYH9 gene results in a missense mutation rather than in a splicing defect. Eur J Med Genet 53(5):256–260. https://doi.org/10.1016/j.ejmg.2010.06.010

    Article  PubMed  Google Scholar 

  43. Kanai N, Yanai F, Hirose S, Nibu K, Izuhara K, Tani T, Kubota T, Mitsudome A (1999) A G to A transition at the last nucleotide of exon 6 of the gamma c gene (868G–>A) may result in either a splice or missense mutation in patients with X-linked severe combined immunodeficiency. Hum Genet 104(1):36–42. https://doi.org/10.1007/s004390050907

    Article  CAS  PubMed  Google Scholar 

  44. Rivera B, Di Iorio M, Frankum J, Nadaf J, Fahiminiya S, Arcand SL, Burk DL, Grapton D, Tomiak E, Hastings V, Hamel N, Wagener R, Aleynikova O, Giroux S, Hamdan FF, Dionne-Laporte A, Zogopoulos G, Rousseau F, Berghuis AM, Provencher D, Rouleau GA, Michaud JL, Mes-Masson AM, Majewski J, Bens S, Siebert R, Narod SA, Akbari MR, Lord CJ, Tonin PN, Orthwein A, Foulkes WD (2017) Functionally null RAD51D missense mutation associates strongly with ovarian carcinoma. Cancer Res 77(16):4517–4529. https://doi.org/10.1158/0008-5472.CAN-17-0190

    Article  CAS  PubMed  Google Scholar 

  45. Daly MB, Pilarski R, Yurgelun MB, Berry MP, Buys SS, Dickson P, Domchek SM, Elkhanany A, Friedman S, Garber JE, Goggins M, Hutton ML, Khan S, Klein C, Kohlmann W, Kurian AW, Laronga C, Litton JK, Mak JS, Menendez CS, Merajver SD, Norquist BS, Offit K, Pal T, Pederson HJ, Reiser G, Shannon KM, Visvanathan K, Weitzel JN, Wick MJ, Wisinski KB, Dwyer MA, Darlow SD (2020) NCCN guidelines insights: genetic/familial high-risk assessment: breast, ovarian, and pancreatic, version 1.2020. J Natl Compr Cancer Netw 18(4):380–391. https://doi.org/10.6004/jnccn.2020.0017

    Article  Google Scholar 

Download references

Funding

This study was funded by Department of Pathology, Memorial Sloan Kettering Cancer Center.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Liying Zhang.

Ethics declarations

Conflict of interest

Z.K.S. reports that an immediate family member holds consulting/advisory roles with Allergan, Adverum, Genentech/Roche, Gyroscope Tx, Novartis, Neurogene, Optos Plc, Regeneron, Regenxbio. L.Z. reports honoraria (Future Technology Research LLC, BGI, Illumina); honoraria and Travel and accommodation expenses (Roche Diagnostics Asia Pacific). Family members hold leadership position and ownership interests of Decipher Medicine.

Informed consent

Informed consent was obtained from all individual participants included in the study.

Additional information

Publisher's Note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, C., Arnold, A.G., Catchings, A. et al. The RAD51D c.82G>A (p.Val28Met) variant disrupts normal splicing and is associated with hereditary ovarian cancer. Breast Cancer Res Treat 185, 869–877 (2021). https://doi.org/10.1007/s10549-020-06066-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10549-020-06066-7

Keywords

Navigation