You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Systematic Review: An Update on the Spectrum of Urological Malignancies in Lynch Syndrome

Abstract

Background:

Lynch syndrome is an autosomal dominant disorder that predisposes individuals affected to certain malignancies. Colon and endometrial cancers are the malignancies most highly associated with Lynch syndrome. However, growing body of evidence links Lynch syndrome to urological cancers.

Objective:

This review aims to clarify the type of urological malignancies that fall under the Lynch-associated cancer spectrum.

Methods:

Using PRISMA guidelines, a systematic search between January 1990 to February 2018, was conducted using the MEDLINE database with the application of the following MESH terms: colorectal neoplasms, hereditary nonpolyposis; DNA mismatch repair; urologic neoplasms; kidney pelvis; ureteral neoplasms; urinary bladder; carcinoma, transitional cell; prostatic neoplasms; testicular neoplasms.

Results:

Upper tract urothelial cancers are well established under the Lynch spectrum. Increasing evidence supports its association with prostate cancer. However, there is, inconclusive and limited evidence for an association with bladder and testicular cancer.

Conclusions:

The evidence underpinning certain urological malignancies associated with Lynch syndrome has expanded in recent years. Our review may assist in providing a summary of the current standing in literature. However, we recommend further investigations to better clarify associations, particularly with prostate, bladder and testicular cancer.

INTRODUCTION

Lynch syndrome, also known as hereditary nonpolyposis colorectal cancer (HNPCC), is an inherited autosomal dominant disorder that predisposes individuals to a spectrum of malignancies. Although early-onset colorectal and endometrial cancer remain the predominant clinical manifestations of Lynch syndrome [1–4], the risk of other extracolonic malignancies is notable. Potential extracolonic malignancies described in the literature include cancers of the ovary, urinary tract, small bowel, stomach, hepato-biliary tract, skin and brain [5–7].

The evidence pertaining to urological malignancies in Lynch syndrome has expanded in recent years. Currently the increased risk and the underlying genetic mechanism of upper tract urothelial carcinoma (UTUC) in patients with Lynch syndrome is well described in the literature [7–11]. Whilst there is a strong association with UTUC, it is controversial whether there is a link between Lynch syndrome and other urological malignancies, especially those of the bladder, prostate and testis. Previous reviews have suggested a weak association, if any, between these cancers [12]. The question as to whether these should be considered part of the malignant spectrum of Lynch syndrome is explored in this review. We detail recent and seminal studies to explore the types of urological tumors that should be considered Lynch-associated cancers.

EVIDENCE ACQUISITION

A systematic search was conducted using the MEDLINE database using PRISMA guidelines. Papers were selected using the time period from January 1990 to February 2018. The following MESH terms were applied: colorectal neoplasms, hereditary nonpolyposis; DNA mismatch repair; urologic neoplasms; kidney pelvis; ureteral neoplasms; urinary bladder; carcinoma, transitional cell; prostatic neoplasms; testicular neoplasms. No language restrictions were imposed. Hand-searching references from articles was used to identify additional relevant studies. Initially, articles were selected using title and abstract. The full manuscript of these articles were then obtained and considered for eligibility. Overall, 18 articles were selected for upper tract urothelial cancer, 14 for bladder cancer, seven for prostate cancer and 11 for testicular cancer.

EVIDENCE SYNTHESIS

Mechanism of carcinogenesis

Genetics

Lynch syndrome is an autosomal dominant disorder. It is associated with a cancer susceptibility caused by a germline mutation(s) in one or more of the DNA mismatch repair (MMR) genes mutS homlog 2 (MSH2) [13], mutL homolog 1 (MLH1) [14], mutS homolog 6 (MSH6) [15] and postmeiotic segregation increased 2 (PMS2) [16]. It is also associated with the loss of expression of MSH2 due to deletions in the epithelial cell adhesion molecule (EpCAM) gene [17]. Urological tumors in Lynch syndrome are particularly associated with MSH2 mutation and MSH6 [11, 18].

Mismatch repair system

Normally, MMR proteins maintain genomic integrity by detecting and excising DNA mismatches that occur during DNA synthesis. MMR function requires the coordination of multiple MMR gene products, thus mutation of any of the four MMR genes results in the disruption of the overall repair system. MMR dysfunction results from the inactivation of both alleles in any one of the four MMR genes. The first allele inactivation in patients with Lynch syndrome originates from the inherited germline mutation; the second allele is inactivated by another mechanism (somatic mutation, loss of heterozygosity or epigenetic silencing).

Microsatellite instability

The loss of gene repair functioning results in the accumulation of errors due to MMR dysfunction, which usually occurs in the microsatellite regions of DNA. This is referred to as microsatellite instability (MSI). MSI is characteristic of Lynch-associated colorectal cancers [19], and also occurs in approximately 15% of sporadic cancers due to other faulty DNA repair mechanisms [20]. Unstable microsatellite regions are thought to affect cell growth, cell apoptosis and some DNA MMR genes.

MSI in recent years has been studied in regard to its association with colorectal tumor prognosis and management. For unclear reasons, MSI-high colorectal tumors are associated with a more favourable prognosis in both Lynch syndrome and sporadic cases [21]. Furthermore, the presence of MSI appears to be unlikely to derive significant benefit from adjuvant fluoropyrimidine-based therapy [22].

Although MSI is well recognised in colorectal cancer as a prognostic marker and aids in management, the microsatellites that are used for identification of MSI have not been fully validated in other Lynch-associated cancers [23]. Outside of the colon, the link between MSI and IHC becomes weaker, suggesting that microsatellites may be organ-specific [24]. The utility of MSI in directing management is yet to be investigated in extra-colonic cancers associated with Lynch syndrome. The link between MSI and IHC in UTUC has not been well investigated though recent data suggest that a proportion of Lynch syndrome-associated UTUC may be MSI-stable [11] and sporadic tumours may be MSI-high [25, 26].

Identification of individuals at risk of Lynch Syndrome

Several clinicopathologic criteria, such as the Amsterdam criteria [27] (Table 1) and revised Bethesda criteria [28] (Table 2), can be used to identify individuals at risk of Lynch syndrome. As these studies have limited sensitivity, further clinical testing is advised for those who meet criteria. Excised tumors may initially be tested for microsatellite instability and immunohistochemistry as described above. However, the definitive diagnosis of Lynch syndrome requires a referral to a genetic counselor to perform genetic testing (prove a germline gene defect), as well as assessing personal history and outlining a comprehensive family pedigree.

Table 1

Amsterdam II criteria for Lynch syndrome

For identifying individuals at risk of being MMR mutation carriers, where all criteria must be met:
Amsterdam II
•  Three or more relatives with histologically verified Lynch syndrome-related cancers (colorectal, endometrial, small intestinal, renal pelvic, ureter). One of which should be a first-degree relative of the other two.
•  Involvement of two or more successive generations should be affected
•  One or more cancers diagnosed before age 50
•  Exclusion of familial adenomatous polyposis
Table 2

Revised Bethesda criteria

Tumors from individuals should be tested for MSI in the following situations:
Revised Bethesda criteria
•  Colorectal cancer diagnosed at less than 50 years of age.
•  Presence of synchronous or metachronous colorectal, or other Lynch syndrome-associated tumor (endometrial, ovarian, pancreas, stomach, ureter, renal pelvis, biliary tract, brain, sebaceous gland adenomas, keratoacanthomas and small bowel)
•  Colorectal cancer with high microsatellite instability histology (tumor-infiltrating lymphocytes, Crohn’s like lymphocytic reaction, mucinous/signet ring differentiation or medullary growth pattern) diagnosed before the age of 60 years.
•  Patient diagnosed with colorectal cancer with at least one first –degree relative with Lynch-related tumor diagnosed younger than the age of 50 years.
•  Patient diagnosed with colorectal cancer with two or more first or second-degree relatives with Lynch-related tumors at any age.

Lynch-associated urological tumors

An updated understanding of the urological cancers involved in this syndrome is important to improve disease and family classification, risk estimates, and screening and surveillance recommendations in known carriers.

Upper tract urothelial cancer

Upper urinary tract urothelial carcinomas (UTUC) refer to malignancies that arise from the pyelocaliceal cavities and ureter. UTUC is a relatively uncommon, constituting five to ten percent of all urothelial cancers [29, 30]. However, it is well recognised as a Lynch-associated malignancy, especially in individuals with MSH2 and MSH6 gene mutation [18, 31–34]. MMR deficiency is detected in 5–11.3% of UTUC via immunohistochemistry, particularly the loss of expression in MSH6 +/–MSH2 (Table 3) [11, 33, 34]. Universal point of care testing using AMS II and IHC criteria in patients with UTUC identified 13.9% having potential Lynch syndrome and confirmed 5.2% had Lynch syndrome [11]. Histopathology may play a role in identifying tumours for MSI testing. Patterns of inverted growth, intra-tumoral lymphocytes, presence of pushing borders tumour grade and pleomorphism, may be suggestive of Lynch associated UTUC [33, 34]. However, the evidence is still lacking in this area. In patients with Lynch syndrome, the reported lifetime risk of developing UTUC is between 0.4 and 20 percent, which confers up to a 22 times greater risk than that of the general population [9, 12, 32, 35–38]. The median age of onset of UTUC in patients with Lynch syndrome is 56 years of age [37]. Furthermore, UTUC is almost twice as common in men compared with women in the normal population [39]. However, in Lynch syndrome, the rates between sexes appear to be similar [18]. This was detailed in a recent national retrospective cohort study of the Danish HNPCC registry of 288 Lynch syndrome families and included 64 individuals who developed UTUC. Analysis of tumor distribution amongst sex showed a cumulative risk of approximately four percent in males and five percent in females [18]. Overall, consistent evidence continues to support UTUC as a part of the Lynch-tumor spectrum.

Table 3

Loss of mismatch repair proteins in patients with UTUC at universal point of care testing

Author, year (reference)Loss of mismatch repair proteinMSH2 + MSH6MLH 1+PMS2MSH 6MSH2MLH1
Metcalfe et al. [11]11.3%5.2%0%6.0%0.0%0.0%
Harper et al. [33]7.0%5.6%0%1.0%0.0%0.0%
Urakami et al. [34]5.0%3.5%0.7%0.7%0.0%0.0%

There are several areas of unmet need for Lynch syndrome patients in regard to screening for UTUC [40]. Recent review and consensus opinion publications suggest frequent urinalyses in these patients, using the AUA criteria of microhematuria (and any gross hematuria) as triggers for screening [25, 40]. Mork et al acknowledged the high rates of microhematuria in the general public which could lead to a high negative workup, however suggested potential justification in this at-risk population [25]. Urinary cytology alone may miss low-grade tumors and is a poor screening test. Retrograde cystoscopy may be useful, however cystoscopy alone is not recommended as a means for screening [40]. Lynch syndrome patients also frequently have CT scans for follow up of colorectal or endometrial cancer, and thus a coordinated effort to modify these scans to contain a urographic phase may help minimize costs as well as radiation exposure. Pradere et al have recently summarised recommendations for post-radical nephorureterectomy surveillance regime involving cystoscopy, urinary cytology and CT urogram, depending on depth of tumour invasion [40]. It is also suggested that Lynch syndrome-related UTUC may be more sensitive to chemotherapy [41], as has been seen in colorectal cancer. While experience of the authors is consistent, there are no studies that have conclusively demonstrated this finding.

Bladder cancer

Bladder cancer is the most common urinary malignancy in the general population, with environmental exposures accounting for majority of cases. However, studies looking at heredity in bladder cancer suggest a small increase in risk in those with a positive family history [42]. A number of genes have been identified to play a role in its pathogenesis, most prominently the p53 and retinoblastoma genes [43, 44].

There is limited, but growing evidence, describing the relationship between Lynch syndrome and bladder cancer. As such, the inclusion of urothelial bladder cancer within the Lynch-associated tumor spectrum remains controversial. It was previously thought that bladder cancer did not have any relationship to Lynch syndrome due to the lack of reported incidence and differences in its mechanisms of carcinogenesis [12]. This notion was supported by studies that reported a low frequency of MSI in bladder tumors and/or no significant risk association between Lynch syndrome and bladder cancer [7, 45–47].

However, recently published literature supports a relationship [6, 18, 31, 38, 46, 48–50], with an estimated increased risk of bladder cancer in Lynch syndrome between 1–20%, especially amongst males and MSH2 carriers. In particular, Skeldon et al examined the risk of bladder cancer in Canadian cohort of 321 patients with MMR gene mutations. Of those with MSH2 mutations (n = 177), 6.21% of patients had bladder cancer (n = 11). When this was compared with the general Canadian population, the relative risk of bladder cancer in MSH2 carriers was significantly higher (p < 0.001) with earlier age of onset (59.6 years vs >70 years) [49]. Furthermore, a recent study reviewed records of 288 Lynch syndrome families with total of 3235 mutation carriers and first-degree relatives [18]. 54 patients with bladder cancer were identified, with 86% having a MSH2 mutation. This study estimated the increased risks for bladder cancer in this population at 3.3% for men and 2.6% for women for bladder cancer. Moller et al estimate survival rates of those with Lynch associated bladder cancer to be 93% at 5 years and 81% at 10 years [51].

Although a significant increase in risk of bladder cancer in Lynch syndrome has been reported, there are several factors that may undermine these observations. The association of Lynch syndrome with bladder cancer may be confounded by studies not accounting for sporadic cases of bladder cancer and those with higher environmental risk factors. Additionally, patients with prior or concurrent UTUC may confound results, as a result of seeding in the bladder. Furthermore, as previously discussed by Roupret et al. [12], the low rates of MSI reported in bladder cancer specimens may undermine its association with Lynch syndrome [18], however the utility of MSI in extracolonic cancer, as aforementioned, requires further investigation. Phelan et al. [50] explore the difficulty in truly differentiation the role of inheritance or genetic association and environmental risk factor clustering in families.

Overall, we have identified several studies that suggest a potential association between bladder cancer and Lynch syndrome. Despite these reports, it remains difficult to suggest whether bladder cancer is a Lynch-associated cancer, and if it is, the penetrance is much less than UTUC. We recommend further investigation with larger cohort studies taking into account potential confounders to help further clarify the contention in the literature.

Prostate cancer

Prostate cancer is the most common malignancy in men and the second leading cause of cancer related deaths [30]. Most adenocarcinomas of the prostate are sporadic, however a small proportion are attributable to genetic factors, most notably the BRCA2 gene [52]. Although the molecular genetic mechanisms involved in prostate cancer are not well understood, recent epidemiological evidence supports a link between prostate cancer and Lynch syndrome.

The argument for the inclusion of prostate cancer in the Lynch syndrome spectrum has been underpinned by the 2014 systematic review and meta-analysis by Ryan et al. [53]. It identified 23 studies (six molecular studies, 18 risk studies) that analysed data on prostate cancer in MMR gene mutation carriers. It found that 74 percent (95% CI, range 57–85%) of prostate cancers in mutation carriers were MMR-deficient (especially MSH2 mutations). There was a relative risk of 3.67 for prostate cancer developing in those with the gene mutations (95% CI, 2.32–6.67). The authors also reported the epidemiologic risk of prostate cancer was significantly increased by two to three-fold in Lynch syndrome patients. This association was reinforced in a recent study of 188 males diagnosed with Lynch syndrome that had an almost five fold increase in prostate cancer incidence over a ten-year period [54]. These results are in accordance with the 2016 study of 288 Danish Lynch syndrome families, where almost 10% of the cohort developed prostate cancer. MMR–deficiency was identified in 69% of cases [55]. Also of note, current evidence suggests no increased risk of early of tumor onset [53] or advanced staging at diagnosis [54]. However higher-grade tumors (Gleason 8–10) have been reported [55].

The known susceptibility of BRCA mutation carriers to prostate cancer provides another interesting twist to this narrative, since the protein products of BRCA1 and BRCA2 are involved with DNA repair and other associated functions to stabilize DNA, similar to MMR proteins [56]. Given the state of evidence in the literature, we believe that there is a strong argument for the inclusion of prostate cancer in the Lynch syndrome tumor spectrum. However, the clinical implications for screening and surveillance remain unclear. Barrow et al recommended a trial of PSA testing in MSH2 mutation carriers from 40–50 years [47]. Ryan and colleagues suggest a more relevant role once there is clarification in the mutation specific risks [53]. There may also be therapeutic implications for Lynch syndrome patients diagnosed with prostate cancer in regard to therapies such as radiation which may potentiate a mutagenic phenotype.

Testicular cancer

Testicular germ cell tumors are the most frequent malignant neoplasm found in young males (25–35 years of age) [57]. Although familial aggregation of testicular cancer is well recognised [58], there have been no published reports of its occurrence in Lynch syndrome families. Notably, while MMR gene mutations and microsatellite instability in testicular cancer have been examined in contemporary literature, the existing evidence remains conflicting.

The studies that support the association are primarily those that investigate refractory testicular germ cell tumors [59–61]. Mayer et al found 33% of refractory specimens had MSI [59]. Another study proposed the distinct prognostic value of MSI [62]. It showed that MMR deficient tumors (MSH2) with MSI-high phenotype were significantly associated with faster clinical relapse and death following chemotherapy than germ cell tumors with an intact MMR system. The same authors published a later study that correlated MSI with increased tumor recurrence [63].

Conversely, Carcano et al found no microsatellite instability and normal MLH1 and MSH2 expression in all 133 specimens primary testicular germ cell tumors [64]. These results were consistent with several previously published studies [65–67].

This discrepancy in the literature may be explained by differences in methodology, demographics and the subgroup of tumor analysed. The lack of microsatellite instability is more often observed in studies pertaining to primary testicular cancers, whereas, its presence may play a role in refractory or recurrent tumors. Given the uncertain nature of the data, it would be reasonable to systematically explore the association of Lynch syndrome with refractory or recurrent testicular germ cell tumors, keeping in mind the potential for discordant IHC and MSI findings and the potential for tissue-specific microsatellites.

CONCLUSIONS

Determining the urological malignancies that may be Lynch-associated is pertinent to refining the management course for families with Lynch syndrome. Patients with Lynch syndrome would benefit from informed measures for early detection and screening of susceptible urologic cancers. Unfortunately, there is insufficient evidence currently to direct such clinical decision-making. Although UTUC is well supported in the literature as a Lynch-associated cancer, a lack of awareness of this association may exist amongst clinicians, leading to a proportion of UTUC misclassified as sporadic [68]. The opportunity to appropriately screen and manage patients earlier in their disease progression is missed, as well as the missed opportunity to diagnose an inheritable syndrome that would impact family members. There is an emerging role of universal point of care testing for possible Lynch syndrome in those with UTUC [11]. Further studies investigating the utility of screening for prostate cancer in males with Lynch syndrome are required. Similarly, the paucity of consistent evidence for the development of bladder cancer in this syndrome and the inconsistent evidence for testicular cancer means that further evidence is needed to help clarify the potential association and relative importance in the Lynch syndrome spectrum.

DISCLOSURES

None.

CONFLICTS OF INTEREST

The authors have no conflicts of interest to disclose.

Acknowledgments

None.

REFERENCES

[1] 

Aarnio M, Sankila R, Pukkala E, Salovaara R, Aaltonen LA, de la Chapelle A et al., Cancer risk in mutation carriers of DNA-mismatch-repair genes. Int J Cancer (1999) ;81: (2):214–8.

[2] 

Vasen HF, Wijnen JT, Menko FH, Kleibeuker JH, Taal BG, Griffioen G et al., Cancer risk in families with hereditary nonpolyposis colorectal cancer diagnosed by mutation analysis. Erratum appears in Gastroenterology. 1996;111(5):1402. Gastroenterology (1996) ;110: (4):1020–7.

[3] 

Watson P, Vasen HF, Mecklin JP, Bernstein I, Aarnio M, Jarvinen HJ et al., The risk of extra-colonic, extra-endometrial cancer in the Lynch syndrome. International Journal of Cancer (2008) ;123: (2):444–9.

[4] 

Backes FJ, Cohn DELynch syndrome. Clin Obstet Gynecol (2011) ;54: (2):199–214.

[5] 

Watson P, Riley BThe tumor spectrum in the Lynch syndrome. Fam Cancer (2005) ;4: (3):245–8.

[6] 

Win A, Young J, Lindor N, Tucker K, Ahnen D, Young G et al., Colorectal and other cancer risks for carriers and noncarriers from families with a DNA mismatch repair gene mutation: A prospective cohort study. Journal of clinical oncology (2012) ;30: (9):958–64.

[7] 

Bonadona V, Bonaiti B, Olschwang S, Grandjouan S, Huiart L, Longy M et al., Cancer risks associated with germline mutations in MLH1, MSH2, and MSH6 genes in Lynch syndrome. JAMA (2011) ;305: (22):2304–10.

[8] 

Lynch HT, Ens JA, Lynch JFThe Lynch syndrome II and urological malignancies. Journal of Urology (1990) ;143: (1):24–8.

[9] 

Watson P, Vasen HFA, Mecklin JP, Bernstein I, Aarnio M, Jarvinen HJ et al., The risk of extra-colonic, extra-endometrial cancer in the Lynch syndrome. International Journal of Cancer (2008) ;123: (2):444–9.

[10] 

Dowty JG, Win AK, Buchanan DD, Lindor NM, Macrae FA, Clendenning M et al., Cancer risks for MLH1 and MSH2 mutation carriers. Hum Mutat (2013) ;34: (3):490–7.

[11] 

Metcalfe MJ, Petros FG, Rao P, Mork ME, Xiao L, Broaddus RR et al., Universal Point of Care Testing for Lynch Syndrome in Patients with Upper Tract Urothelial Carcinoma. J Urol (2018) ;199: (1):60–5.

[12] 

Roupret M, Yates DR, Comperat E, Cussenot OUpper urinary tract urothelial cell carcinomas and other urological malignancies involved in the hereditary nonpolyposis colorectal cancer (lynch syndrome) tumor spectrum. European Urology (2008) ;54: (6):1226–36.

[13] 

Leach FS, Nicolaides NC, Papadopoulos N, Liu B, Jen J, Parsons R et al., Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer. Cell (1993) ;75: (6):1215–25.

[14] 

Bronner CE, Baker SM, Morrison PT, Warren G, Smith LG, Lescoe MK et al., Mutation in the DNA mismatch repair gene homologue hMLH1 is associated with hereditary non-polyposis colon cancer. Nature. (1994) ;368: (6468):258–61.

[15] 

Miyaki M, Konishi M, Tanaka K, Kikuchi-Yanoshita R, Muraoka M, Yasuno M et al., Germline mutation of MSH6 as the cause of hereditary nonpolyposis colorectal cancer. Nat Genet (1997) ;17: (3):271–2.

[16] 

Nicolaides NC, Papadopoulos N, Liu B, Wei YF, Carter KC, Ruben SM et al., Mutations of two PMS homologues in hereditary nonpolyposis colon cancer. Nature (1994) ;371: (6492):75–80.

[17] 

Kovacs ME, Papp J, Szentirmay Z, Otto S, Olah EDeletions removing the last exon of TACSTD1 constitute a distinct class of mutations predisposing to Lynch syndrome. Hum Mutat (2009) ;30: (2):197–203.

[18] 

Joost P, Therkildsen C, Dominguez-Valentin M, Jonsson M, Nilbert MUrinary Tract Cancer in Lynch Syndrome; Increased Risk in Carriers of MSH2 Mutations. Urology (2015) ;86: (6):1212–7.

[19] 

Halvarsson B, Lindblom A, Rambech E, Lagerstedt K, Nilbert MMicrosatellite instability analysis and/or immunostaining for the diagnosis of hereditary nonpolyposis colorectal cancer? Virchows Arch (2004) ;444: (2):135–41.

[20] 

Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA et al., CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet (2006) ;38: (7):787–93.

[21] 

Gryfe R, Kim H, Hsieh ET, Aronson MD, Holowaty EJ, Bull SB et al., Tumor microsatellite instability and clinical outcome in young patients with colorectal cancer. N Engl J Med (2000) ;342: (2):69–77.

[22] 

Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM et al., Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med (2003) ;349: (3):247–57.

[23] 

Broaddus RR, Lynch PM, Lu KH, Luthra R, Michelson SJUnusual tumors associated with the hereditary nonpolyposis colorectal cancer syndrome. Mod Pathol (2004) ;17: (8):981–9.

[24] 

Kuismanen SA, Moisio AL, Schweizer P, Truninger K, Salovaara R, Arola J et al., Endometrial and colorectal tumors from patients with hereditary nonpolyposis colon cancer display different patterns of microsatellite instability. Am J Pathol (2002) ;160: (6):1953–8.

[25] 

Mork M, Hubosky SG, Roupret M, Margulis V, Raman J, Lotan Y et al., Lynch syndrome: A primer for urologists and panel recommendations. J Urol (2015) ;194: (1):21–9.

[26] 

Hause RJ, Pritchard CC, Shendure J, Salipante SJClassification and characterization of microsatellite instability across 18 cancer types. Nat Med (2016) ;22: (11):1342–50.

[27] 

Vasen HF, Watson P, Mecklin JP, Lynch HTNew clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the International Collaborative group on HNPCC. Gastroenterology (1999) ;116: (6):1453–6.

[28] 

Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Ruschoff J et al., Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst (2004) ;96: (4):261–8.

[29] 

Roupret M, Babjuk M, Comperat E, Zigeuner R, Sylvester RJ, Burger M et al., European Association of Urology Guidelines on Upper Urinary Tract Urothelial Carcinoma: 2017 Update. Eur Urol. (2017) .

[30] 

Siegel RL, Miller KD, Jemal ACancer Statistics, 2017. CA Cancer J Clin. (2017) ;67: (1):7–30.

[31] 

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: (35):4409–15.

[32] 

Vasen HF, Stormorken A, Menko FH, Nagengast FM, Kleibeuker JH, Griffioen G et al., MSH2 mutation carriers are at higher risk of cancer than MLH1 mutation carriers: A study of hereditary nonpolyposis colorectal cancer families. J Clin Oncol (2001) ;19: (20):4074–80.

[33] 

Harper HL, McKenney JK, Heald B, Stephenson A, Campbell SC, Plesec T et al., Upper tract urothelial carcinomas: Frequency of association with mismatch repair protein loss and lynch syndrome. Mod Pathol (2017) ;30: (1):146–56.

[34] 

Urakami S, Inoshita N, Oka S, Miyama Y, Nomura S, Arai M et al., Clinicopathological characteristics of patients with upper urinary tract urothelial cancer with loss of immunohistochemical expression of the DNA mismatch repair proteins in universal screening. Int J Urol (2018) ;25: (2):151–6.

[35] 

Crockett DG, Wagner DG, Holmang S, Johansson SL, Lynch HTUpper urinary tract carcinoma in Lynch syndrome cases. Journal of Urology (2011) ;185: (5):1627–30.

[36] 

Sijmons RH, Kiemeney LA, Witjes JA, Vasen HFUrinary tract cancer and hereditary nonpolyposis colorectal cancer: Risks and screening options. J Urol (1998) ;160: (2):466–70.

[37] 

Watson P, Lynch HTThe tumor spectrum in HNPCC. Anticancer Research (1994) ;14: (4B):1635–9.

[38] 

van der Post RS, Kiemeney LA, Ligtenberg MJ, Witjes JA, Hulsbergen-van de Kaa CA, Bodmer D et al., Risk of urothelial bladder cancer in Lynch syndrome is increased, in particular among MSH2 mutation carriers. Journal of Medical Genetics (2010) ;47: (7):464–70.

[39] 

Raman JD, Messer J, Sielatycki JA, Hollenbeak CSIncidence and survival of patients with carcinoma of the ureter and renal pelvis in the USA, 1973-2005. BJU Int (2011) ;107: (7):1059–64.

[40] 

Pradere B, Lotan Y, Roupret MLynch syndrome in upper tract urothelial carcinoma: Significance, screening, and surveillance. Curr Opin Urol (2017) ;27: (1):48–55.

[41] 

Hollande C, Colin P, de La Motte Rouge T, Audenet F, Yates DR, Phe V et al., Hereditary-like urothelial carcinomas of the upper urinary tract benefit more from adjuvant cisplatin-based chemotherapy after radical nephroureterectomy than do sporadic tumours. BJU Int (2014) ;113: (4):574–80.

[42] 

Plna K, Hemminki KFamilial bladder cancer in the National Swedish Family Cancer Database. J Urol (2001) ;166: (6):2129–33.

[43] 

Malats N, Bustos A, Nascimento CM, Fernandez F, Rivas M, Puente D et al., P53 as a prognostic marker for bladder cancer: A meta-analysis and review. Lancet Oncol (2005) ;6: (9):678–86.

[44] 

Cordon-Cardo C, Wartinger D, Petrylak D, Dalbagni G, Fair WR, Fuks Z et al., Altered expression of the retinoblastoma gene product: Prognostic indicator in bladder cancer. J Natl Cancer Inst (1992) ;84: (16):1251–6.

[45] 

Bermejo JL, Eng C, Hemminki KCancer characteristics in Swedish families fulfilling criteria for hereditary nonpolyposis colorectal cancer. Gastroenterology (2005) ;129: (6):1889–99.

[46] 

Geary J, Sasieni P, Houlston R, Izatt L, Eeles R, Payne SJ et al., Gene-related cancer spectrum in families with hereditary non-polyposis colorectal cancer (HNPCC). Familial Cancer (2008) ;7: (2):163–72.

[47] 

Barrow PJ, Ingham S, O’Hara C, Green K, McIntyre I, Lalloo F et al., The spectrum of urological malignancy in Lynch syndrome. Fam Cancer (2013) ;12: (1):57–63.

[48] 

van der Post RS, Kiemeney LA, Ligtenberg MJL, Witjes JA, Hulsbergen-van de Kaa CA, Bodmer D et al., Risk of urothelial bladder cancer in Lynch syndrome is increased, in particular among MSH2 mutation carriers. Journal of Medical Genetics (2010) ;47: (7):464–70.

[49] 

Skeldon SC, Semotiuk K, Aronson M, Holter S, Gallinger S, Pollett A et al., Patients with Lynch syndrome mismatch repair gene mutations are at higher risk for not only upper tract urothelial cancer but also bladder cancer. European Urology (2013) ;63: (2):379–85.

[50] 

Phelan A, Lopez-Beltran A, Montironi R, Zhang S, Raspollini MR, Cheng M et al., Inherited forms of bladder cancer: A review of Lynch syndrome and other inherited conditions. Future Oncol (2018) ;14: (3):277–90.

[51] 

Moller P, Seppala TT, Bernstein I, Holinski-Feder E, Sala P, Gareth Evans D et al., Cancer risk and survival in path_MMR carriers by gene and gender up to 75 years of age: A report from the Prospective Lynch Syndrome Database. Gut (2018) ;67: (7):1306–16.

[52] 

Levy-Lahad E, Friedman ECancer risks among BRCA1 and BRCA2 mutation carriers. Br J Cancer (2007) ;96: (1):11–5.

[53] 

Ryan S, Jenkins MA, Win AKRisk of prostate cancer in Lynch syndrome: A systematic review and meta-analysis. Cancer Epidemiology, Biomarkers & Prevention (2014) ;23: (3):437–49.

[54] 

Haraldsdottir S, Hampel H, Wei L, Wu C, Frankel W, Bekaii-Saab T et al., Prostate cancer incidence in males with Lynch syndrome. Genetics in Medicine (2014) ;16: (7):553–7.

[55] 

Dominguez-Valentin M, Joost P, Therkildsen C, Jonsson M, Rambech E, Nilbert MFrequent mismatch-repair defects link prostate cancer to Lynch syndrome. BMC Urology (2016) ;16: , 15.

[56] 

Venkitaraman AR. Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell (2002) ;108: (2):171–82.

[57] 

Bray F, Ferlay J, Devesa SS, McGlynn KA, Moller HInterpreting the international trends in testicular seminoma and nonseminoma incidence. Nat Clin Pract Urol (2006) ;3: (10):532–43.

[58] 

Garner MJ, Turner MC, Ghadirian P, Krewski DEpidemiology of testicular cancer: An overview. Int J Cancer (2005) ;116: (3):331–9.

[59] 

Mayer F, Wermann H, Albers P, Stoop H, Gillis AJ, Hartmann JT et al., Histopathological and molecular features of late relapses in non-seminomas. BJU International (2011) ;107: (6):936–43.

[60] 

Mayer F, Gillis AJ, Dinjens W, Oosterhuis JW, Bokemeyer C, Looijenga LHMicrosatellite instability of germ cell tumors is associated with resistance to systemic treatment. Cancer Res (2002) ;62: (10):2758–60.

[61] 

Honecker F, Wermann H, Mayer F, Gillis AJ, Stoop H, van Gurp RJ et al., Microsatellite instability, mismatch repair deficiency, and BRAF mutation in treatment-resistant germ cell tumors. Journal of Clinical Oncology (2009) ;27: (13):2129–36.

[62] 

Velasco A, Riquelme E, Schultz M, Wistuba II, Villarroel L, Koh MS et al., Microsatellite instability and loss of heterozygosity have distinct prognostic value for testicular germ cell tumor recurrence. Cancer Biol Ther. ((2004) );3: (11):1152–8; discussion 9-61.

[63] 

Velasco A, Corvalan A, Wistuba II, Riquelme E, Chuaqui R, Majerson A et al., Mismatch repair expression in testicular cancer predicts recurrence and survival. International Journal of Cancer (2008) ;122: (8):1774–7.

[64] 

Carcano FM, Lengert AH, Vidal DO, Scapulatempo Neto C, Queiroz L, Marques H et al., Absence of microsatellite instability and BRAF (V600E) mutation in testicular germ cell tumors. Andrology (2016) ;4: (5):866–72.

[65] 

Faulkner SW, Friedlander MLMicrosatellite instability in germ cell tumors of the testis and ovary. Gynecol Oncol (2000) ;79: (1):38–43.

[66] 

Olasz J, Mandoky L, Geczi L, Bodrogi I, Csuka O, Bak MInfluence of hMLH1 methylation, mismatch repair deficiency and microsatellite instability on chemoresistance of testicular germ-cell tumors. Anticancer Res (2005) ;25: (6B):4319–24.

[67] 

Vladusic T, Hrascan R, Kruslin B, Pecina-Slaus N, Perica K, Bicanic A et al., Histological groups of human postpubertal testicular germ cell tumours harbour different genetic alterations. Anticancer Research (2014) ;34: (8):4005–12.

[68] 

Audenet F, Colin P, Yates DR, Ouzzane A, Pignot G, Long JA et al., A proportion of hereditary upper urinary tract urothelial carcinomas are misclassified as sporadic according to a multi-institutional database analysis: Proposal of patient-specific risk identification tool. BJU International (2012) ;110: (11 Pt B):E583–9.