Article Data

  • Views 1096
  • Dowloads 152

Reviews

Open Access Special Issue

Germline multigene panel testing in gynecological cancer

  • Sayaka Ueno1,*,
  • Akira Hirasawa1

1Department of Clinical Genomic Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, 700-8558 Okayama, Japan

DOI: 10.22514/ejgo.2022.008 Vol.43,Issue 3,June 2022 pp.25-35

Submitted: 20 March 2022 Accepted: 26 April 2022

Published: 15 June 2022

*Corresponding Author(s): Sayaka Ueno E-mail: sayaka-u@okayama-u.ac.jp

Abstract

Multi-gene panel testing (MGPT) has become widely used in clinical practice. MGPT allows a set of genes to be tested simultaneously, making it a powerful, time- and cost-effective tool for detecting genetic variants. The purposes of identifying germline pathogenic variants is threefold: diagnosis, prevention, and treatment. Germline variants in certain genes cause hereditary tumor syndromes. Diagnosing hereditary tumors enable to predict the types of cancer that may develop in the future. The blood relatives of those diagnosed may also suffer from the same hereditary tumor syndrome. For such individuals, medical intervention tailored to their condition can reduce the incidence of cancer or help in cancer detection in the early stages. Diagnosis of hereditary tumors may also change the cancer treatment strategy for the diagnosed patient. To date, more than 100 genes have been found to have a predisposition to cancer. The type of cancer one develops, the risk of developing it, and the possible preventive strategy differs among genes. The association of some genes with cancer predisposition has not yet been fully confirmed. Nowadays, various types of MGPTs are available, and the genes included differ among the tests. In addition, no consensus has been reached on which genes to be included in MGPT. This review is aimed to summarize the advantages and limitations of MGPT along with some practical considerations while performing MGPT and the gynecological tumors associated with genes commonly included in MGPT.


Keywords

multi-gene panel testing (MGPT); hereditary gynecological cancer; cancer predisposition genes; Lynch syndrome; Peutz-Jeghers syndrome; PTEN hamartoma tumor syndrome; BRCA-related breast/ovarian cancer syndrome; DICER1 syndrome; rhabdoid tumor predisposition syndrome; Gorlin-Goltz syndrome


Cite and Share

Sayaka Ueno,Akira Hirasawa. Germline multigene panel testing in gynecological cancer. European Journal of Gynaecological Oncology. 2022. 43(3);25-35.

References

[1] Huang KL, Mashl RJ, Wu Y, Ritter DI, Wang J, Oh C, et al. Pathogenic Germline Variants in 10,389 Adult Cancers. Cell. 2018; 173: 355–370. e314.

[2] Stadler ZK, Maio A, Chakravarty D, Kemel Y, Sheehan M, Salo-Mullen E, et al. Therapeutic Implications of Germline Testing in Patients with Advanced Cancers. Journal of Clinical Oncology. 2021; 39: 2698–2709.

[3] Conley JM, Cook-Deegan R, Lázaro-Muñoz G. MYRIAD AFTER MYRIAD: THE PROPRIETARY DATA DILEMMA. North Carolina Journal of Law & Technology. 2014; 15: 597–637.

[4] Cook-Deegan R, Niehaus A. After Myriad: Genetic Testing in the Wake of Recent Supreme Court Decisions about Gene Patents. Current Genetic Medicine Reports. 2014; 2: 223–241.

[5] Kurian AW, Ward KC, Hamilton AS, Deapen DM, Abrahamse P, Bondarenko I, et al. Uptake, Results, and Outcomes of Germline Multiple-Gene Sequencing after Diagnosis of Breast Cancer. JAMA Oncology. 2018; 4: 1066.

[6] Lancaster JM, Powell CB, Chen LM, Richardson DL. Society of Gynecologic Oncology statement on risk assessment for inherited gynecologic cancer predispositions. Gynecologic Oncology. 2015; 136: 3–7.

[7] Owens DK, Davidson KW, Krist AH, Barry MJ, Cabana M, Caughey AB, et al. Risk Assessment, Genetic Counseling, and Genetic Testing for BRCA-Related Cancer: US Preventive Services Task Force Recommendation Statement. The Journal of the American Medical Association. 2019; 322: 652–665.

[8] Practice Bulletin No 182: Hereditary Breast and Ovarian Cancer Syndrome. Obstetrics & Gynecology. 2017; 130: e110–e126.

[9] Hampel H, Bennett RL, Buchanan A, Pearlman R, Wiesner GL. A practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: referral indications for cancer predisposition assessment. Genetics in Medicine. 2015; 17: 70–87.

[10] Bashford MT, Kohlman W, Everett J, Parrott A, Pollin TI. Addendum: a practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: referral indications for cancer predisposition assessment. Genetics in Medicine. 2019; 21: 2844.

[11] Beitsch PD, Whitworth PW, Hughes K, Patel R, Rosen B, Compagnoni G, et al. Underdiagnosis of Hereditary Breast Cancer: are Genetic Testing Guidelines a Tool or an Obstacle? Journal of Clinical Oncology. 2019; 37: 453–460.

[12] Yadav S, Hu C, Hart SN, Boddicker N, Polley EC, Na J, et al. Evaluation of Germline Genetic Testing Criteria in a Hospital-Based Series of Women with Breast Cancer. Journal of Clinical Oncology. 2020; 38: 1409–1418.

[13] Yang S, Axilbund JE, O’Leary E, Michalski ST, Evans R, Lincoln SE, et al. Underdiagnosis of Hereditary Breast and Ovarian Cancer in Medicare Patients: Genetic Testing Criteria Miss the Mark. Annals of Surgical Oncology. 2018; 25: 2925–2931.

[14] Ward, DNP, APRN, AGNP-BC M, Elder, PhD, RN B, Habtemariam, DNP, APRN M. Current Testing Guidelines: a Retrospective Analysis of a Community-Based Hereditary Cancer Program. Journal of the Advanced Practitioner in Oncology. 2021; 12: 693–701.

[15] Daly MB, Pal T, Berry MP, Buys SS, Dickson P, Domchek SM, et al. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network. 2021; 19: 77–102.

[16] Konstantinopoulos PA, Norquist B, Lacchetti C, Armstrong D, Grisham RN, Goodfellow PJ, et al. Germline and Somatic Tumor Testing in Epithelial Ovarian Cancer: ASCO Guideline. Journal of Clinical Oncology. 2020; 38: 1222–1245.

[17] Weiss JM, Gupta S, Burke CA, Axell L, Chen LM, Chung DC, et al. NCCN Guidelines® Insights: Genetic/Familial High-Risk Assessment: Colorectal, Version 1.2021. Journal of the National Comprehensive Cancer Network. 2021; 19: 1122–1132.

[18] LaDuca H, Polley EC, Yussuf A, Hoang L, Gutierrez S, Hart SN, et al. A clinical guide to hereditary cancer panel testing: evaluation of gene-specific cancer associations and sensitivity of genetic testing criteria in a cohort of 165,000 high-risk patients. Genetics in Medicine. 2020; 22: 407–415.

[19] Mersch J, Brown N, Pirzadeh-Miller S, Mundt E, Cox HC, Brown K, et al. Prevalence of Variant Reclassification Following Hereditary Cancer Genetic Testing. The Journal of the American Medical Association. 2018; 320: 1266.

[20] David KL, Best RG, Brenman LM, Bush L, Deignan JL, Flannery D, et al. Patient re-contact after revision of genomic test results: points to consider—a statement of the American College of Medical Genetics and Genomics (ACMG). Genetics in Medicine. 2019; 21: 769–771.

[21] Slavin TP, Manjarrez S, Pritchard CC, Gray S, Weitzel JN. The effects of genomic germline variant reclassification on clinical cancer care. Oncotarget. 2019; 10: 417–423.

[22] Lincoln SE, Hambuch T, Zook JM, Bristow SL, Hatchell K, Truty R, et al. One in seven pathogenic variants can be challenging to detect by NGS: an analysis of 450,000 patients with implications for clinical sensitivity and genetic test implementation. Genetics in Medicine. 2021; 23: 1673–1680.

[23] Slavin TP, Coffee B, Bernhisel R, Logan J, Cox HC, Marcucci G, et al. Prevalence and characteristics of likely-somatic variants in cancer susceptibility genes among individuals who had hereditary pan-cancer panel testing. Cancer Genetics. 2019; 235-236: 31–38.

[24] Chao EC, Astbury C, Deignan JL, Pronold M, Reddi HV, Weitzel JN. Incidental detection of acquired variants in germline genetic and genomic testing: a points to consider statement of the American College of Medical Genetics and Genomics (ACMG). Genetics in Medicine. 2021; 23: 1179–1184.

[25] Coffee B, Cox HC, Bernhisel R, Manley S, Bowles K, Roa BB, et al. A substantial proportion of apparently heterozygous TP53 pathogenic variants detected with a next-generation sequencing hereditary pan-cancer panel are acquired somatically. Human Mutation. 2020; 41: 203–211.

[26] Norquist BM, Harrell MI, Brady MF, Walsh T, Lee MK, Gulsuner S, et al. Inherited Mutations in Women with Ovarian Carcinoma. JAMA Oncology. 2016; 2: 482.

[27] Carter NJ, Marshall ML, Susswein LR, Zorn KK, Hiraki S, Arvai KJ, et al. Germline pathogenic variants identified in women with ovarian tumors. Gynecologic Oncology. 2018; 151: 481–488.

[28] Hirasawa A, Imoto I, Naruto T, Akahane T, Yamagami W, Nomura H, et al. Prevalence of pathogenic germline variants detected by multigene sequencing in unselected Japanese patients with ovarian cancer. Oncotarget. 2017; 8: 112258–112267.

[29] Kuchenbaecker KB, Hopper JL, Barnes DR, Phillips KA, Mooij TM, Roos-Blom MJ, et al. Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. The Journal of the American Medical Association. 2017; 317: 2402–2416.

[30] Zhang S, Royer R, Li S, McLaughlin JR, Rosen B, Risch HA, et al. Frequencies of BRCA1 and BRCA2 mutations among 1,342 unselected patients with invasive ovarian cancer. Gynecologic Oncology. 2011; 121: 353–357.

[31] Alsop K, Fereday S, Meldrum C, deFazio A, Emmanuel C, George J, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. Journal of Clinical Oncology. 2012; 30: 2654–2663.

[32] Carter BA. Pathology of breast and ovarian cancers among BRCA1 and BRCA2 mutation carriers: results from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). Yearbook of Pathology and Laboratory Medicine. 2012; 2013: 34–37.

[33] Lynch HT, de la Chapelle A. Hereditary Colorectal Cancer. New England Journal of Medicine. 2003; 348: 919–932.

[34] Ligtenberg MJL, Kuiper RP, Chan TL, Goossens M, Hebeda KM, Voorendt M, et al. Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3′ exons of TACSTD1. Nature Genetics. 2009; 41: 112–117.

[35] Dominguez-Valentin M, Sampson JR, Seppälä TT, Ten Broeke SW, Plazzer JP, Nakken S, et al. Cancer risks by gene, age, and gender in 6350 carriers of pathogenic mismatch repair variants: findings from the Prospective Lynch Syndrome Database. Genetics in Medicine. 2020; 22: 15–25.

[36] Pal T, Akbari MR, Sun P, Lee J, Fulp J, Thompson Z, et al. Frequency of mutations in mismatch repair genes in a population-based study of women with ovarian cancer. British Journal of Cancer. 2012; 107: 1783–1790.

[37] Helder-Woolderink JM, Blok EA, Vasen HFA, Hollema H, Mourits MJ, De Bock GH. Ovarian cancer in Lynch syndrome; a systematic review. European Journal of Cancer. 2016; 55: 65–73.

[38] Engel C, Loeffler M, Steinke V, Rahner N, Holinski-Feder E, Dietmaier W, et al. Risks of less Common Cancers in Proven Mutation Carriers with Lynch Syndrome. Journal of Clinical Oncology. 2012; 30: 4409–4415.

[39] Watson P, Bützow R, Lynch HT, Mecklin J, Järvinen HJ, Vasen HFA, et al. The Clinical Features of Ovarian Cancer in Hereditary Nonpolyposis Colorectal Cancer. Gynecologic Oncology. 2001; 82: 223–228.

[40] Kurian AW, Ward KC, Howlader N, Deapen D, Hamilton AS, Mariotto A, et al. Genetic Testing and Results in a Population-Based Cohort of Breast Cancer Patients and Ovarian Cancer Patients. Journal of Clinical Oncology. 2019; 37: 1305–1315.

[41] Suszynska M, Klonowska K, Jasinska AJ, Kozlowski P. Large-scale meta-analysis of mutations identified in panels of breast/ovarian cancer-related genes — Providing evidence of cancer predisposition genes. Gynecologic Oncology. 2019; 153: 452–462.

[42] Ramus SJ, Song H, Dicks E, Tyrer JP, Rosenthal AN, Intermaggio MP, et al. Germline Mutations in the BRIP1, BARD1, PALB2, and NBN Genes in Women With Ovarian Cancer. Journal of the National Cancer Institute. 2015; 107: djv214..

[43] Kurian AW, Hughes E, Handorf EA, Gutin A, Allen B, Hartman A, et al. Breast and Ovarian Cancer Penetrance Estimates Derived from Germline Multiple-Gene Sequencing Results in Women. JCO Precision Oncology. 2017; 10: 1–12.

[44] Antoniou AC, Casadei S, Heikkinen T, Barrowdale D, Pylkäs K, Roberts J, et al. Breast-cancer risk in families with mutations in PALB2. The New England Journal of Medicine. 2014; 371: 497–506.

[45] Lilyquist J, LaDuca H, Polley E, Davis BT, Shimelis H, Hu C, et al. Frequency of mutations in a large series of clinically ascertained ovarian cancer cases tested on multi-gene panels compared to reference controls. Gynecologic Oncology. 2017; 147: 375–380.

[46] Song H, Dicks E, Ramus SJ, Tyrer JP, Intermaggio MP, Hayward J, et al. Contribution of Germline Mutations in the RAD51B, RAD51C, and RAD51D Genes to Ovarian Cancer in the Population. Journal of Clinical Oncology. 2015; 33: 2901–2907.

[47] Beggs AD, Latchford AR, Vasen HFA, Moslein G, Alonso A, Aretz S, et al. Peutz-Jeghers syndrome: a systematic review and recommendations for management. Gut. 2010; 59: 975–986.

[48] Giardiello FM, Brensinger JD, Tersmette AC, Goodman SN, Petersen GM, Booker SV, et al. Very high risk of cancer in familial Peutz–Jeghers syndrome. Gastroenterology. 2000; 119: 1447–1453.

[49] Jenne DE, Reomann H, Nezu J, Friedel W, Loff. S, Jeschke R, et al. Peutz-Jeghers syndrome is caused by mutations in a novel serine threoninekinase. Nature Genetics. 1998; 18: 38–43.

[50] Hemminki A, Markie D, Tomlinson I, Avizienyte E, Roth S, Loukola A, et al. A serine/threonine kinase gene defective in Peutz–Jeghers syndrome. Nature. 1998; 391: 184–187.

[51] Young RH, Welch WR, Dickersin GR, Scully RE. Ovarian sex cord tumor with annular tubules. Review of 74 cases including 27 with Peutz-Jeghers syndrome and four with adenoma malignum of the cervix. Cancer. 1982; 50: 1384–1402.

[52] Gorlin RJ. Nevoid basal cell carcinoma (Gorlin) syndrome. Genetics in Medicine. 2004; 6: 530–539.

[53] Gorlin RJ, Goltz RW. Multiple Nevoid Basal-Cell Epithelioma, Jaw Cysts and Bifid Rib. New England Journal of Medicine. 1960; 262: 908–912.

[54] Kimonis VE, Mehta SG, Digiovanna JJ, Bale SJ, Pastakia B. Radiological features in 82 patients with nevoid basal cell carcinoma (NBCC or Gorlin) syndrome. Genetics in Medicine. 2004; 6: 495–502.

[55] Marsh A, Wicking C, Wainwright B, Chenevix-Trench G. DHPLC analysis of patients with Nevoid Basal Cell Carcinoma Syndrome reveals novel PTCH missense mutations in the sterol-sensing domain. Human Mutation. 2005; 26: 283.

[56] Soufir N, Gerard B, Portela M, Brice A, Liboutet M, Saiag P, et al. PTCH mutations and deletions in patients with typical nevoid basal cell carcinoma syndrome and in patients with a suspected genetic predisposition to basal cell carcinoma: a French study. British Journal of Cancer. 2006; 95: 548–553.

[57] Smith MJ, Beetz C, Williams SG, Bhaskar SS, O’Sullivan J, Anderson B, et al. Germline mutations in SUFU cause Gorlin syndrome-associated childhood medulloblastoma and redefine the risk associated with PTCH1 mutations. Journal of Clinical Oncology. 2014; 32: 4155–4161.

[58] Slade I, Bacchelli C, Davies H, Murray A, Abbaszadeh F, Hanks S, et al. DICER1 syndrome: clarifying the diagnosis, clinical features and management implications of a pleiotropic tumour predisposition syndrome. Journal of Medical Genetics. 2011; 48: 273–278.

[59] Schultz KAP, Stewart DR, Kamihara J, Bauer AJ, Merideth MA, Stratton P, et al. DICER1 Tumor Predisposition. In Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Gripp KW, et al. (eds.) GeneReviews((R)): Seattle (WA).1993.

[60] Hill DA, Ivanovich J, Priest JR, Gurnett CA, Dehner LP, Desruisseau D, et al. DICER1 mutations in familial pleuropulmonary blastoma. Science. 2009; 325: 965.

[61] Foulkes WD, Priest JR, Duchaine TF. DICER1: mutations, microRNAs and mechanisms. Nature Reviews Cancer. 2014; 14: 662–672.

[62] Schultz KAP, Williams GM, Kamihara J, Stewart DR, Harris AK, Bauer AJ, et al. DICER1 and Associated Conditions: Identification of At-risk Individuals and Recommended Surveillance Strategies. Clinical Cancer Research. 2018; 24: 2251–2261.

[63] Stewart DR, Best AF, Williams GM, Harney LA, Carr AG, Harris AK, et al. Neoplasm Risk Among Individuals With a Pathogenic Germline Variant in DICER1. Journal of Clinical Oncology. 2019; 37: 668–676.

[64] de Kock L, Terzic T, McCluggage WG, Stewart CJR, Shaw P, Foulkes WD, et al. DICER1 Mutations are Consistently Present in Moderately and Poorly Differentiated Sertoli-Leydig Cell Tumors. American Journal of Surgical Pathology. 2017; 41: 1178–1187.

[65] Jelinic P, Mueller JJ, Olvera N, Dao F, Scott SN, Shah R, et al. Recurrent SMARCA4 mutations in small cell carcinoma of the ovary. Nature Genetics. 2014; 46: 424–426.

[66] Ramos P, Karnezis AN, Craig DW, Sekulic A, Russell ML, Hendricks WPD, et al. Small cell carcinoma of the ovary, hypercalcemic type, displays frequent inactivating germline and somatic mutations in SMARCA4. Nature Genetics. 2014; 46: 427–429.

[67] Witkowski L, Carrot-Zhang J, Albrecht S, Fahiminiya S, Hamel N, Tomiak E, et al. Germline and somatic SMARCA4 mutations characterize small cell carcinoma of the ovary, hypercalcemic type. Nature Genetics. 2014; 46: 438–443.

[68] Witkowski L, Goudie C, Ramos P, Boshari T, Brunet J, Karnezis AN, et al. The influence of clinical and genetic factors on patient outcome in small cell carcinoma of the ovary, hypercalcemic type. Gynecologic Oncology. 2016; 141: 454–460.

[69] Sredni ST, Tomita T. Rhabdoid Tumor Predisposition Syndrome. Pediatric and Developmental Pathology. 2015; 18: 49–58.

[70] Tischkowitz M, Huang S, Banerjee S, Hague J, Hendricks WPD, Huntsman DG, et al. Small-Cell Carcinoma of the Ovary, Hypercalcemic Type–Genetics, New Treatment Targets, and Current Management Guidelines. Clinical Cancer Research. 2020; 26: 3908–3917.

[71] Long B, Lilyquist J, Weaver A, Hu C, Gnanaolivu R, Lee KY, et al. Cancer susceptibility gene mutations in type i and II endometrial cancer. Gynecologic Oncology. 2019; 152: 20–25.

[72] Ring KL, Bruegl AS, Allen BA, Elkin EP, Singh N, Hartman A, et al. Germline multi-gene hereditary cancer panel testing in an unselected endometrial cancer cohort. Modern Pathology. 2016; 29: 1381–1389.

[73] Susswein LR, Marshall ML, Nusbaum R, Vogel Postula KJ, Weissman SM, Yackowski L, et al. Pathogenic and likely pathogenic variant prevalence among the first 10,000 patients referred for next-generation cancer panel testing. Genetics in Medicine. 2016; 18: 823–832.

[74] Rossi L, Le Frere-Belda M, Laurent-Puig P, Buecher B, De Pauw A, Stoppa-Lyonnet D, et al. Clinicopathologic Characteristics of En-dometrial Cancer in Lynch Syndrome: A French Multicenter Study. International Journal of Gynecological Cancer. 2017; 27: 953–960.

[75] Nelen MR, Padberg GW, Peeters EAJ, Lin AY, Helm BVD, Frants RR, et al. Localization of the gene for Cowden disease to chromosome 10q22–23. Nature Genetics. 1996; 13: 114–116.

[76] Tan MH, Mester JL, Ngeow J, Rybicki LA, Orloff MS, Eng C. Lifetime cancer risks in individuals with germline PTEN mutations. Clinical Cancer Research. 2012; 18: 400–407.

[77] Segev Y, Iqbal J, Lubinski J, Gronwald J, Lynch HT, Moller P, et al. The incidence of endometrial cancer in women with BRCA1 and BRCA2 mutations: an international prospective cohort study. Gynecologic Oncology. 2013; 130: 127–131.

[78] Shu CA, Pike MC, Jotwani AR, Friebel TM, Soslow RA, Levine DA, et al. Uterine Cancer After Risk-Reducing Salpingo-oophorectomy Without Hysterectomy in Women With BRCA Mutations. JAMA Oncology. 2016; 2: 1434–1440.

[79] Yoshihama T, Hirasawa A, Sugano K, Yoshida T, Ushiama M, Ueki A, et al. Germline multigene panel testing revealed a BRCA2 pathogenic variant in a patient with suspected Lynch syndrome. International Cancer Conference Journal. 2021; 10: 6–10.

[80] Le DT, Kim TW, Van Cutsem E, Geva R, Jäger D, Hara H, et al. Phase II Open-Label Study of Pembrolizumab in Treatment-Refractory, Microsatellite Instability–High/Mismatch Repair–Deficient Metastatic Colorectal Cancer: KEYNOTE-164. Journal of Clinical Oncology. 2020; 38: 11–19.

[81] Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. The New England Journal of Medicine. 2015; 372: 2509–2520.

[82] Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017; 357: 409–413.

[83] Balmaña J, Balaguer F, Cervantes A, Arnold D. Familial risk-colorectal cancer: ESMO Clinical Practice Guidelines. Annals of Oncology. 2013; 24: vi73–vi80.

[84] Giardiello FM, Allen JI, Axilbund JE, Boland CR, Burke CA, Burt RW, et al. Guidelines on Genetic Evaluation and Management of Lynch Syndrome: a Consensus Statement by the us Multi-Society Task Force on Colorectal Cancer. Gastroenterology. 2014; 147: 502–526.

[85] Stoffel EM, Mangu PB, Gruber SB, Hamilton SR, Kalady MF, Lau MWY, et al. Hereditary Colorectal Cancer Syndromes: American Society of Clinical Oncology Clinical Practice Guideline Endorsement of the Familial Risk–Colorectal Cancer: European Society for Medical Oncology Clinical Practice Guidelines. Journal of Clinical Oncology. 2015; 33: 209–217.

[86] Rubenstein JH, Enns R, Heidelbaugh J, Barkun A, Adams MA, Dorn SD, et al. American Gastroenterological Association Institute Guideline on the Diagnosis and Management of Lynch Syndrome. Gastroenterology. 2015; 149: 777–782.

[87] Lindor NM, Petersen GM, Hadley DW, Kinney AY, Miesfeldt S, Lu KH, et al. Recommendations for the Care of Individuals with an Inherited Predisposition to Lynch Syndrome: A Systematic Review. The Journal of the American Medical Association. 2006; 296: 1507.

[88] Syngal S, Brand RE, Church JM, Giardiello FM, Hampel HL, Burt RW. ACG Clinical Guideline: Genetic Testing and Management of Hereditary Gastrointestinal Cancer Syndromes. American Journal of Gastroenterology. 2015; 110: 223–262.

[89] McGarrity TJ, Amos CI, Baker MJ. Peutz-Jeghers Syndrome. In Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Gripp KW, et al.(eds.) GeneReviews((R)): Seattle (WA). 1993.

[90] Gilks CB, Young RH, Aguirre P, DeLellis RA, Scully RE. Adenoma Malignum (Minimal Deviation Adenocarcinoma) of the Uterine Cervix. A clinicopathological and immunohistochemical analysis of 26 cases. The American Journal of Surgical Pathology. 1989; 13: 717–729.

[91] Chen KTK. Female genital tract tumors in Peutz-Jeghers syndrome. Human Pathology. 1986; 17: 858–861.

[92] Mikami Y, Kiyokawa T, Hata S, Fujiwara K, Moriya T, Sasano H, et al. Gastrointestinal immunophenotype in adenocarcinomas of the uterine cervix and related glandular lesions: a possible link between lobular endocervical glandular hyperplasia/pyloric gland metaplasia and ‘adenoma malignum’. Modern Pathology. 2004; 17: 962–972.

[93] Hirasawa A, Akahane T, Tsuruta T, Kobayashi Y, Masuda K, Banno K, et al. Lobular endocervical glandular hyperplasia and peritoneal pigmentation associated with Peutz–Jeghers syndrome due to a germline mutation of STK11. Annals of Oncology. 2012; 23: 2990–2992.

[94] Ito M, Minamiguchi S, Mikami Y, Ueda Y, Sekiyama K, Yamamoto T, et al. Peutz–Jeghers syndrome-associated atypical mucinous proliferation of the uterine cervix: a case of minimal deviation adenocarcinoma (‘adenoma malignum’) in situ. Pathology - Research and Practice. 2012; 208: 623–627.

[95] Kobayashi Y, Masuda K, Kimura T, Nomura H, Hirasawa A, Banno K, et al. A tumor of the uterine cervix with a complex histology in a Peutz-Jeghers syndrome patient with genomic deletion of the STK11 exon 1 region. Future Oncology. 2014; 10: 171–177.

[96] Takei Y, Fujiwara H, Nagashima T, Takahashi Y, Takahashi S, Suzuki

M. Successful pregnancy in a Peutz-Jeghers syndrome patient with lobular endocervical glandular hyperplasia. Journal of Obstetrics and Gynaecology Research. 2015; 41: 468–473.

[97] Minard-Colin V, Walterhouse D, Bisogno G, Martelli H, Anderson J, Rodeberg DA, et al. Localized vaginal/uterine rhabdomyosarcoma-results of a pooled analysis from four international cooperative groups. Pediatric Blood & Cancer. 2018; 65: e27096.

[98] Dehner LP, Jarzembowski JA, Hill DA. Embryonal rhabdomyosarcoma of the uterine cervix: a report of 14 cases and a discussion of its unusual clinicopathological associations. Modern Pathology. 2012; 25: 602–614.

[99] de Kock L, Yoon JY, Apellaniz-Ruiz M, Pelletier D, McCluggage WG, Stewart CJR, et al. Significantly greater prevalence of DICER1 alterations in uterine embryonal rhabdomyosarcoma compared to adenosarcoma. Modern Pathology. 2020; 33: 1207–1219.

[100] Meric-Bernstam F, Brusco L, Daniels M, Wathoo C, Bailey AM, Strong L, et al. Incidental germline variants in 1000 advanced cancers on a prospective somatic genomic profiling protocol. Annals of Oncolog. 2016; 27: 795–800.

[101] Mandelker D, Zhang L, Kemel Y, Stadler ZK, Joseph V, Zehir A, et al. Mutation Detection in Patients With Advanced Cancer by Universal Sequencing of Cancer-Related Genes in Tumor and Normal DNA vs Guideline-Based Germline Testing. The Journal of the American Medical Association. 2017; 318: 825–835.

[102] Cobain EF, Wu YM, Vats P, Chugh R, Worden F, Smith DC, et al. Assessment of Clinical Benefit of Integrative Genomic Profiling in Advanced Solid Tumors. JAMA Oncology. 2021; 7: 525–533.

[103] Samadder NJ, Riegert-Johnson D, Boardman L, Rhodes D, Wick M, Okuno S, et al. Comparison of Universal Genetic Testing vs Guideline-Directed Targeted Testing for Patients With Hereditary Cancer Syndrome. JAMA Oncology. 2021; 7: 230–237.

[104] Levine MD, Pearlman R, Hampel H, Cosgrove C, Cohn D, Chassen A, et al. Up-Front Multigene Panel Testing for Cancer Susceptibility in Patients With Newly Diagnosed Endometrial Cancer: A Multicenter Prospective Study. JCO Precision Oncology. 2021; 5: 1588–1602.



Abstracted / indexed in

Science Citation Index Expanded (SciSearch) Created as SCI in 1964, Science Citation Index Expanded now indexes over 9,500 of the world’s most impactful journals across 178 scientific disciplines. More than 53 million records and 1.18 billion cited references date back from 1900 to present.

Biological Abstracts Easily discover critical journal coverage of the life sciences with Biological Abstracts, produced by the Web of Science Group, with topics ranging from botany to microbiology to pharmacology. Including BIOSIS indexing and MeSH terms, specialized indexing in Biological Abstracts helps you to discover more accurate, context-sensitive results.

Google Scholar Google Scholar is a freely accessible web search engine that indexes the full text or metadata of scholarly literature across an array of publishing formats and disciplines.

JournalSeek Genamics JournalSeek is the largest completely categorized database of freely available journal information available on the internet. The database presently contains 39226 titles. Journal information includes the description (aims and scope), journal abbreviation, journal homepage link, subject category and ISSN.

Current Contents - Clinical Medicine Current Contents - Clinical Medicine provides easy access to complete tables of contents, abstracts, bibliographic information and all other significant items in recently published issues from over 1,000 leading journals in clinical medicine.

BIOSIS Previews BIOSIS Previews is an English-language, bibliographic database service, with abstracts and citation indexing. It is part of Clarivate Analytics Web of Science suite. BIOSIS Previews indexes data from 1926 to the present.

Journal Citation Reports/Science Edition Journal Citation Reports/Science Edition aims to evaluate a journal’s value from multiple perspectives including the journal impact factor, descriptive data about a journal’s open access content as well as contributing authors, and provide readers a transparent and publisher-neutral data & statistics information about the journal.

Submission Turnaround Time

Conferences

Top