eISSN: 2084-9869
ISSN: 1233-9687
Polish Journal of Pathology
Current issue Archive Manuscripts accepted About the journal Supplements Editorial board Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
4/2017
vol. 68
 
Share:
Share:
Review paper

Review of hereditary leiomyomatosis renal cell carcinoma with focus on clinical and pathobiological aspects of renal tumors

Naoto Kuroda
,
Chisato Ohe
,
Ikuma Kato
,
Mitsuko Furuya
,
Masaya Baba
,
Yoji Nagashima
,
Yukio Nakatani
,
Ichiro Murakami
,
Ming Zhou
,
Michal Michal
,
Ondrej He
,
Mahul B. Amin

Pol J Pathol 2017; 68 (4): 284-290
Online publish date: 2018/03/06
Article file
- Review.pdf  [0.39 MB]
Get citation
 
PlumX metrics:
 

Introduction and history

Kloepfer et al. for the first time described a hereditary form of multiple cutaneous leiomyomata (MCL) [1]. Reed et al. reported a hereditary syndrome of cutaneous leiomyomas and uterine leiomyomas and/or leiomyosarcoma inherited in an autosomal dominant fashion [2]. After then, this disease was designated as Reed syndrome. In 2001, Launonen et al. and Kiuru et al. suggested that RCC with papillary architecture can occur in patients with Reed syndrome and this syndrome was designated as hereditary leiomyomatosis renal cell cancer (carcinoma) (HLRCC). They found that MCL, Reed syndrome and HLRCC are single disease with a variable phenotype [3, 4]. Additionally, Launonen et al. found that the responsible gene for HLRCC is mapped to chromosome 1q42.3-q43 [3]. In 2002, Tomlinson identified that germline mutation of FH gene mapped to this chromosome in HLRCC neoplasms [5]. Of HLRCC patients. In 2013, HLRCC-associated renal tumors has been incorporated into the classification of renal tumors in International Society of Urologic Pathology (ISUP) [6]. In this article, we review HLRCC with focus on clinical and pathobiological aspects of renal tumors.

Definition/diagnostic criteria of the disease entity

The major criteria (high likelihood of HLRCC) is multiple cutaneous piloleiomyomas with at least biopsy proven and histologically confirmed. The minor criterion (suspicious for HLRCC) contains three items. One is multiple symptomatic uterine leiomyomas before age 40. The other is papillary RCC, type 2 in early onset before age 40. The remaining one is family history of HLRCC plus solitary cutaneous leiomyoma or at-first-degree family member who meets one of the above-described criteria. The diagnosis of HLRCC is likely when a proband meets the major criterion and may be suspected when a proband meets at least two minor criteria [7, 8]. For the definitive diagnosis, positive results of germline FH-mutation analysis will be required [8].

Epidemiology

Although the majority of HLRCC female patients is associated with cutaneous and/or uterine leiomyomas [9], only a minority (15-35%) of HLRCC develop RCC [10, 11, 12, 13, 14]. There is no sex predominance [15, 16]. According to the study of Wong et al., the difference in age at the diagnosis of RCC between the first and second generation, and between the first and third generation are –18.6 and –36.2 years, respectively. These results suggest that RCC tend to occur at the younger age in persons with family history of HLRCC renal cancer in their father/mother or grandfather/grandmother [17]. About 7% of HLRCC patients are diagnosed with RCC before 20 years [15, 18, 19, 20]. The risk of RCC in patients with HLRCC is higher with 6.5 fold than that of general populations [21]. Renal cysts are found in 42% of FH gene mutation-positive patients [10, 21, 22, 23, 24, 25].

Clinical symptoms

Patients with HLRCC renal tumors present with hematuria, abdominal/flank mass, abdominal/flank pain, abdominal discomfort, fatigue or weight loss [16, 25, 26, 27, 28]. Rare cases may be incidentally found [16].

Other clinical features

The association of male infertility, adrenocortical hyperplasia/tumor, thyroid follicular carcinoma, cutaneous basal cell carcinoma, bladder cancer, liver hemangioma, Leydig cell tumor, ovarian cystadenoma, gastrointestinal stromal tumor, breast cancer, leukemia, cutis verticis gyrate, eruptive collagenoma and Charcot-Marie-Tooth disease has been previously reported [3, 7, 21, 28, 29, 30, 31, 32, 33].

Imaging findings

Abdominal computed tomography (CT) scan show the hypodensity lesion in the kidney [9, 20, 26]. The contrast enhanced CT shows homogenous or inhomogenous and less enhanced mass [18, 34, 35, 36].

Pathological findings

Macroscopic findings

Grossly, most tumors were solid, but half of tumors show the cystic area partially [15]. The size of the tumor ranges from 2.3 to 20 cm [15]. There is no predilection in laterality [15, 16]. The tumors frequently invade capsular and perinephric adipose tissue as well as renal vein and vena cava [15, 16]. HLRCC renal tumors generally present as solitary and unilateral lesion, but some tumors can occur multifocally or bilaterally [8, 15, 16, 17, 37, 38, 39].

Microscopic findings

Microscopically, the tumor is composed of neoplastic cells with various morphological architectures such as papillary, tubulopapillary, solid, cystic tubulocystic, vacuolated/cribriform or mixed pattern [8, 15, 16, 22, 40]. Regarding the histological subtype, papillary RCC, type 2 is most frequent, but collecting duct carcinoma may be seen [3, 7, 9, 11, 17, 18, 22, 35, 36, 37, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50]. Clear cell RCC, unclassified RCC, oncocytic tumor, cystic tumor, angiomyolipoma or Wilms tumor has been described [7, 37, 39, 49, 50, 51, 52, 53]. The most important hallmark of HLRCC renal cancer is prominent eosinophilic nucleoli and perinucleolar clearing/haloes resembling cytomegaloviral inclusion [13, 15, 16, 25, 27, 34, 40]. Rhabdoid features or multinucleated tumor giant cells may be noted [25]. Fuhrman nuclear grade generally correspond to grade 3 or 4 [17]. Renal cysts or tubular cells with hobnail patterns in the renal parenchyma adjacent to the main tumor may be present [10]. Renal cyst can occur solitarily or multifocally [22, 24, 25]. Renal tumor can develop independently or within renal cysts [22].

Immunohistochemical findings

The application of S-(2-succino)-cysteine (2SC) may be useful for the detection for HLRCC renal tumors, but primary antibody against 2SC is not commercially available [16]. Regarding 2SC staining pattern, both cytoplasmic and nuclear staining is significant for the accurate detection of HLRCC renal tumors, but some sporadic type renal tumors may show the only cytoplasmic positivity for 2SC [16]. The tubular cells with hobnail pattern adjacent to the dominant tumor showed the positive reaction for 2SC. This finding suggests that hobnail tubular cells may be precursor lesions of HLRCC renal tumors [25, 39]. Except for 2SC, there is no characteristic marker of HLRCC [15]. The positivity for PAX8, CD10, vimentin, cytokeratin 8/18, HIF-1a and GLUT1 has been reported [25, 54]. The increased nuclear accumulation of p53 protein has been described [25]. The Ki-67 index is high [35]. TFE3 is negative [15].

Differential diagnosis

Pathologists should differentiate HLRCC renal tumors from papillary RCC, type 2, collecting duct carcinoma (CDC), clear cell RCC, tubulocystic carcinoma, mucinous tubular and spindle cell carcinoma (MTSCC), Xp11.2 RCC, ALK renal cancer and renal oncocytoma. Among them, the distinction from papillary RCC and CDC is most important. The presence of prominent eosinophilic nucleoli and perinucleolar halo may be diagnostic clue to HLRCC renal tumors [13, 15, 16, 25, 27, 34, 40]. However, the association of cutaneous or uterine leiomyomas are the presence of family history of cutaneous and/or uterine leiomyomas, particularly occurring at the young age, or renal tumors are more vital diagnostic clues for the identification of HLRCC [55, 56, 57, 58, 59]. Tubulocystic carcinoma with high grade carcinoma foci may be indistinguishable from HLRCC renal tumors. In this setting, FH gene germline mutation testing is an accurate diagnostic tool [60]. The abundant mucin deposition on the background stroma and the presence of elongated/anastomosing tubules may suggest the possibility of MTSCC. The break part fluorescence in situ hybridization for TFE3 and ALK gene leads to the final diagnosis of Xp11.2 RCC and ALK renal cancer, respectively.

Characteristics of cutaneous leiomyoma

The tumors are usually multiple and the size of the tumor generally ranges from 0.2 to 2.0cm [1, 7, 61, 62]. The tumor present as papules and/or nodules in groups, dermatomal or linear arrangement, and range from flesh, erythematous to pink-brown in color [1, 7, 61, 62, 63, 64, 65, 66]. The location of the tumor involves extremities, shoulders and trunk, but face or neck can be distributed [1, 7, 8, 61, 62, 63, 64, 65, 66]. With the age, the lesions tend to increase in size and number [7, 62]. Most patients present with pain or itching in response to touching or temperature [7, 8, 62]. Histologically, the tumor display the form of piloleiomyoma, but cytologic atypia is generally absent [1, 7, 61, 62, 63, 64, 65, 66]. Rarely, the occurrence of leiomyosarcoma has been described [9].

Characteristics of uterine leiomyoma

The risk that women with HLRCC develop uterine leiomyomas is higher in 8~9 to 71 fold than that of general population [21, 67]. The mean age of HLRCC patients at diagnosis of uterine leiomyoma is 28 years, namely 10 years younger than the general population and surgical resection is frequently carried out on women before 30 years of age because of severe symptoms such as abdominal pain, menorrhagia and metrorrhagia [7, 8, 14, 55, 57, 58, 59]. Tumors generally occur in a multiple form, and occurrence at the younger age and more than seven tumor in number may be speculated as HLRCC uterine leiomyomas [55, 57, 58, 59]. Uterine leiomyoma with patients with HLRCC have characteristic of cellular morphology, prominent eosinophilic nucleoli, perinucleolar haloes, cytoplasmic eosinophilic globules [56, 57, 59]. Atypia, multinucleated giant cells, fibrillary cytoplasm, epithelioid growth pattern, schwannoma-like growth pattern, “Orphan Annie nuclei” with optical clearing, and hemangiopericytomatous blood vessels can be observed [56, 57, 59, 68]. In some cases, leiomyoma may progress to leiomyosarcoma [57]. Immunohistochemically, tumor cells are positive for 2SC [59, 67].

Molecular genetic findings

Biallelic inactivation of FH gene occurs in renal tumors of HLRCC patients [5, 9, 16]. In most tumors, germline mutation occurs in one allele and loss of heterozygosity (LOH) at FH gene locus or somatic mutation of FH gene in the other allele [5, 9, 16]. Pathogenic germline mutation of FH gene have been detected in 76-100% of families with suggestive clinical features [5, 7, 9, 11, 21, 22, 35, 36, 41, 44, 47, 49]. LOH at FH gene locus is observed in 80% of HLRCC renal cancer [3, 15, 37, 42]. Epigenetic alteration seems to be rare. Germline mutations consists of missense (57%), nonsense & frameshift (27%), large deletion (4%) including whole gene deletion, small deletion (4%), duplication (2%), splice-site (6%) alterations [5, 9, 44, 69, 70, 71]. HLRCC-associated mutation occur in the 5’ of the gene, whereas FH-deficiency mutations, autosomal recessive hereditary form with neurological impairment, tend to occur in the 3’ end [41, 69, 71, 72]. Biallelic inactivation (somatic mutation plus LOH) of FH gene occurs in nonsyndromic uterine leiomyomas but is rare in other tumors [73]. Sporadic uterine leiomyosarcoma and cutaneous leiomyoma may show germline mutation rarely [74].

Chromosomal changes

The loss of chromosome 1q was observed as expected findings compatible with tumor suppressor role of FH gene [5, 8, 15, 22, 42, 75]. Additionally, gains in chromosomes 2, 7 and 17 and losses in chromosomes 13q12.3-q21.1, 14, 18 and X were identified in three of analyzed eleven HLRCC renal tumors with papillary type 2 morphology using array comparative genomic hybridization [76].

Phenotype-genotype correlation

There seems to be no relationship between phenotype and genotype in HLRCC to date [9, 69]. There is no evidence of a genetic modifier for RCC risk in HLRCC [77]. The environmental factor may modify the development of RCC [78].

Prognosis

HLRCC renal cancer, particularly papillary RCC, type 2 and CDC behave in an aggressive fashion and most cases generally die of disease within 5 years since the initial diagnosis [8, 11, 13, 15, 16, 17, 18, 20, 25, 27, 28, 35, 38, 41, 45, 46, 47, 48, 49, 50, 62, 64, 75, 76]. Approximately two thirds of patients show stage III/IV at the diagnosis [7]. Seventy-four percent of patients with HLRCC renal cancer die of metastatic disease [44, 79]. The most frequent metastatic site is regional lymph node, but the metastasis to distant lymph node can occur [15, 16, 35, 36]. Furthermore, distant metastasis to lung, bone and liver has been reported [15, 16, 35, 36, 48]. Dissemination to peritoneum, pleura and meninges can occur [15, 25, 48].

Therapy

When the solid renal tumor at the early stage is discovered, surgical resection with wide resection margin including radical nephrectomy and retroperitoneal lymph node resection because of frequent lymph node metastasis should be promptly performed [45, 80, 81]. Radiofrequency or cryoablation should be not recommended [8]. The patients with advanced stage HLRCC renal tumors have previously received immunotherapy such as Interferon or Interleukin-2, molecular targeted therapy such as multikinase inhibitor (sunitinib, sorafenib and pazopanib) including VEGF pathway antagonist (bevacizumab), mTOR inhibitor (everolimus and temsirolimus) and chemotherapy (cisplatin and gemcitabine) [8, 18, 20, 36, 48, 56]. Radiation therapy has also been performed for metastatic sites [48]. However, these therapies seem to be not so effective for advanced tumors.

Clinical management

Active surveillance is not recommended for HLRCC solid renal tumor [80, 81]. The FH gene mutation analysis and annual surveillance should start at the age of 8 to 10 years or even as early as 5 years [7, 8, 17, 19, 79]. FH gene mutation carriers should be examined using MRI with 1 to 3mm slice [79]. Ultrasound sonography for the kidney cannot detect small tumors because of the low sensitivity. However, as MRI might be distressing in childhood, the surveillance in childhood can be replaces by ultrasound sonography every six months by expert radiologists [7, 20]. Because of the small risk of RCC in HLRCC patients, other researchers recommend that FH gene analysis and surveillance should start at the age of 18 to 20 years [20, 77].

Future perspectives

The association of some diseases except for cutaneous and/or uterine leiomyomas in HLRCC has been reported [3, 7, 21, 28, 29, 30, 31, 32, 33]. Among them, somatic mutation of FH gene was identified in one bladder cancer and three breast carcinoma. Therefore, these disease may be a part of HLRCC syndrome [21]. However, Kiuru et al. confirmed that FH gene is not a major predisposing gene for familial breast cancer [82]. Further examinations in a large scale study will be needed in order to elucidate the relationship between HLRCC and bladder/breast carcinoma or other associated lesions. As the HLRCC renal cancer generally gives rise to a dismal outcome, the early detection/treatment is very important for the management of patients with HLRCC. From this point of view, the extensive research on precursor lesion such as hobnail tubular cells or associated renal cysts may be key target. Regarding the management of HLRCC family, further examination by the accumulation of the HLRCC renal tumors will be needed in the future.

The authors declare no conflicts of interest.

References

1. Kloepfer HW, Krafchuk J, Derbres V, et al. Hereditary multiple leiomyoma of the skin. Am J Hum Genet 1958; 10: 48-52.
2. Reed WB, Walker R, Horowitz R. Cutaneous leiomyomata with uterine leiomyomata. Acta Derm Venereol 1973; 53: 409-416.
3. Launonen V, Vierimaa O, Kiuru M, et al. Inherited susceptibility to uterine leiomyomas and renal cell cancer. Proc Natl Acad Sci USA 2001; 98: 3387-3392.
4. Kiuru M, Launonen V, Hietala M, et al. Familial cutaneous leiomyomatosis is a two-hit condition associated with renal cell cancer of characteristic histopathology. Am J Pathol 2001; 159: 825-829.
5. Tomlinson IP, Alam NA, Rowan AJ, et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet 2002; 30: 406-410.
6. Srigley JR, Delahunt B, Eble JN, et al. International Society of Urologic Pathology (ISUP) Vancouver classification of renal neoplasia. Am J Surg Pathol 2013; 37: 1469-1489.
7. Smit DL, Mensenkamp AR, Badeloe S, et al. Hereditary leiomyomatosis and renal cell cancer in families referred for fumarate hydratase germline mutation analysis. Clin Genet 2011; 79: 49-59.
8. Schmidt LS, Linehan WM. Hereditary leiomyomatosis and renal cell carcinoma. Int J Nephrol Renovasc Dis 2014; 7: 253-260.
9. Toro JR, Nickerson ML, Wei MH, et al. Mutation in the fumarate hydratase gene case hereditary leiomyomatosis and renal cell cancer in families in North America. Am J Hum Genet 2003; 73: 95-106.
10. Pfaffenroth EC, Linehan WM. Genetic basis for kidney cancer. Expert Opin Biol Ther 2008; 8: 779-790.
11. Rosner I, Bratslavsky G, Pinto PA, et al. The clinical implications of the genetics of renal cell carcinoma. Urol Oncol 2009; 27: 131-136.
12. Folkins AK, Longacre TA. Hereditary gynaecological malignancies: advances in screening and treatment. Histopathology 2013; 62: 2-30.
13. Surch B, Linehan WM, Srinivasan R. Aerobic glycolysis: A novel target in kidney cancer. Expert Rev Anticancer Ther 2013; 13: 711-719.
14. Randall JM, Millard F, Kurzrock R. Molecular aberrations, targeted therapy, and renal cell carcinoma: current state-of-the art. Cancer Metastasis Rev 2014; 33: 1109-1124.
15. Merino MJ, Torres-Cabala C, Pinto P, et al. The morphologic spectrum of kidney tumors in hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome. Am J Surg Pathol 2007; 31: 1578-1585.
16. Chen YB, Brannon AR, Toubaji A, et al. Hereditary leiomyomatosis and renal cell carcinoma syndrome-associated renal cancer Recognition of the syndrome by pathologic features and the utility of detecting aberrant succination by immunohistochemistry. Am J Surg Pathol 2014; 38: 627-637.
17. Wong MH, Tan CS, Lee SC, et al. Potential genetic anticipation in hereditary leiomyomatosis-renal cell cancer (HLRCC). Fam Cancer 2014; 13: 281-289.
18. Refae MAI, Wong N, Patenaude F, et al. Hereditary leiomyomatosis and renal cell cancer: an unusual and aggressive form of hereditary renal carcinoma. Nat Clin Prac 2007; 4: 256-261.
19. Alrashdi I, Levine S, Paterson J, et al. Hereditary leiomyomatosis renal cell carcinoma: very early diagnosis of renal cancer in a paediatric patient. Fam Cancer 2010; 9: 239-243.
20. van Spaendonck-Zwarts KY, Badeloe S, Oosting SF, et al. Hereditary leiomyomatosis and renal cell cancer presenting as metastatic kidney cancer at 18 years of age: implications for surveillance. Fam Cancer 2012; 11: 123-129.
21. Lehtonen HJ, Kiuru M, Ylisaukko-oja SK, et al. Increased risk of cancer in patients with fumarate hydratase germline mutation. J Med Genet 2006; 43: 523-526.
22. Linehan WM, Pinto PA, Bratslavsky G, et al. Hereditary kidney cancer. Unique opportunity for disease-based therapy. Cancer 2009; 115: 2252-2261.
23. Lencastre A, Cabete J, Goncalves R, et al. Cutaneous leiomyomatosis in a mother and daughter. An Bras Dermatol 2013; 88: 124-127.
24. Kakar R, Davis JC, Crowe DR. Multiple linear leiomyomas of the forehead as the presenting sign of Reed syndrome. Int J Dermatol 2014; 53: 316-318.
25. Udager AM, Alva A, Chen YB, et al. hereditary leiomyomatosis and renal cell carcinoma (HLRCC). A rapid of autopsy report of metastatic renal cell carcinoma. Am J Surg Pathol 2014; 38: 567-577.
26. Hayedeh G, Fatemeh M, Ahmadreza R, et al. Hereditary leiomyomatosis and renal cell carcinoma syndrome: A case report. Dermatol Online J 2008; 14: 16.
27. Kamai T, Tomosugi N, Abe H, et al. Protein profiling of blood samples from patients with hereditary leiomyomatosis and renal cell cancer by surface-enhanced laser desorption/ionization time-of-flight mass spectrometry. Int J Med Sci 2012; 13 14518-14532.
28. Venables ZC, Ramaiya A, Holden S, et al. Hereditary leiomyomatosis associated with renal cell carcinoma. Clin Exp Dermatol 2014; 40: 99-100.
29. Marque M, Gardie B, Bressac B, et al. Novel FH mutation in a patient with cutaneous leiomyomatosis associated with cutis verticis gyrate, eruptive collagenoma and Charcot-Marie-Tooth disease. Br J Dermatol 2010; 163: 1337-1339.
30. McKelvey KD, Siraj S, Kelsay J, et al. Male infertility associated with hereditary leiomyomatosis and renal cell carcinoma. Fertil Steril 2010; 93: 2075e1-e2.
31. Huller C, Grunow N, Nadler T, et al. Cutaneous and uterine leiomyomatosis and ovarian cystadenoma associated with deficiency of fumarate hydratase. Dermatol Pract Concept 2011; 1: 7.
32. Shuch B, Ricketts CJ, Vocke CD, et al. Adrenal nodular hyperplasia in hereditary leiomyomatosis and renal cell cancer. J Urol 2013; 189: 430-435.
33. Lee JJ, Nambudiri VE, Henneberry J, et al. Post-transplant diagnosis of hereditary leiomyomatosis and renal cell carcinoma syndrome in a kidney donar. Clin Kindey J 2014; 7: 615-616.
34. Mustafa S, Jadidi N, Faraj SF, et al. Case of hereditary papillary renal cell carcinoma. J Comm Hosp Int Med Perspect 2011; 1: 9468.
35. Benes CL, Schlegel C, Shoukier M, et al. Hereditary papillary renal cell carcinoma primarily diagnosed in a cervical lymph nodes: a case report of a 30-year-old woman with multiple metastasis. BMC Urol 2013; 13: 3.
36. Kuwada M, Chihara Y, Lou Y, et al. Novel missense mutation in the FH gene in familial renal cell cancer patients lacking cutaneous leiomyomas. BMC Res Notes 2014; 7: 203.
37. Lehtonen HJ, Blanco I, Piulats JM, et al. Conventional renal cancer in a patient with fumarate hydratase mutation. Hum Pathol 2007; 38: 793-796.
38. Soni SS, Gowrishankar S, Adlkey GK, et al. Hereditary leiomyomatosis with renal cell carcinoma. Indian J Dermatol Venereol Leprol 2008; 74: 63-64.
39. Ristau BT, Kamat SN, Tarin TV. Abnormal cystic tumor in a patient with hereditary leiomyomatosis and renal cell cancer syndrome: evidence of a precursor lesion? Case Rep Urol 2015; 2015: 303872.
40. Crumley S, Divatia M, Troung L, et al. Renal cell carcinoma: evolving and emerging subtypes. World J Clin Cases 2013; 16: 262-275.
41. Eng C, Kiuru M, Fernandez MJ, et al. A role for mitochondrial enzymes in inherited neoplasia and beyond. Nat Rev 2003; 3: 193-202.
42. Linehan WM, Vasselli J, Srinivasan R, et al. Genetic basis of cancer of the kidney: disease-specific approaches to therapy. Clin Cancer Res 2004; 10: 6282s-6289s.
43. Chan I, Wing T, Martinez-Mir A, et al. Familial multiple cutaneous and uterine leiomyomas associated with papillary renal cell cancer. Clin Exp Dermatol 2005; 30: 75-78.
44. Gardie B, Remenieras A, Kattygnarath D, et al. Novel FH mutations in families with hereditary leiomyomatosis and renal cell cancer (HLRCC) and patients with isolated type 2 papillary renal cell carcinoma. J Med Genet 2011; 48: 226-234.
45. Maher ER. Genetics of familial renal cancers. Nephron Exp Nephrol 2011; 118: e21-e26.
46. Singer EA, Bratlavsky G, Middelton L, et al. Impact of genetics on the diagnosis and treatment of renal cancer. Curr Urol Rep 2011; 12: 47-55.
47. Raymond VM, Herron CM, Giordano TJ, et al. Familial renal cancer as an indicator of hereditary leiomyomatosis and renal cell cancer syndrome. Fam Cancer 2012; 11: 115-121.
48. de Velasco G, Munoz C, Sepulveda JM, et al. Sequential treatments in hereditary leiomyomatosis and renal cell carcinoma (HLRCC): Case report and review of the literature. Can Urol Assoc 2015; 9: E243-6.
49. Kiuru M, Kujala M, Aittomäki K. Inherited forms of renal cell carcinoma. Scand J Surg 2004; 93: 103-111.
50. Haas NB, Nathanson KL. Hereditary renal cancer syndromes. Adv Chronic Kidney Dis 2014; 21: 81-90.
51. Badeloe S, van Spaendonck-Zwarts KY, van Steensel MAM, et al. Wilms tumour as a possible early manifestation of hereditary leiomyomatosis and renal cell cancer? Br J Dermatol 2009; 160: 707-709.
52. Harris M, Wallace J, Winship I, et al. Hereditary renal cell carcinoma: the clue can be in the skin. Int Med J 2009; e12-e13.
53. Rongioletti F, Fausti V, Ferrando B, et al. A novel missense mutation in fumarate hydratase in a Italian patient with a diffuse variant of cutaneous leiomyomatosis (Reed’s syndrome). Dermatology 2010; 221: 378-380.
54. Linehan WM, Pinto PA, Srinivasan R, et al. Identification of the genes for kidney cancer: opportunity for disease-specific targeted therapeutics. Clin Cancer Res 2007; 13: 671s-679s.
55. Stewart L, Glenn GM, Stratton P, et al. Association of germline mutations in the fumarate hydratase gene and uterine fibroids in women with hereditary leiomyomatosis and renal cell cancer. Arch Dermatol 2008; 144: 1584-1592.
56. Garg K, Tickoo SK, Soslow RA, et al. Morphologic features of uterine leiomyomas associated with hereditary leiomyomatosis and renal cell carcinoma syndrome: A case report. Am J Surg Pathol 2011; 35: 1235-1237.
57. Sanz-Ortega J, Vocke C, Stratton P, et al. Morphologic and molecular characteristics of uterine leiomyomas in hereditary leiomyomatosis and renal cancer (HLRCC) syndrome. Am J Surg Pathol 2013; 37: 74-80.
58. Mann ML, Ezzati M, Tarnawa ED, et al. Fumarate hydratase mutation in a young woman with uterine leiomyoma and a family history of renal cell cancer. Obstet Gynecol 2015; 126: 90-92.
59. Martinek P, Grossmann P, Hes O, et al. genetic testing of leiomyoma tissue in women younger than 30 years old might provide an effective screening approach for the hereditary leiomyomatosis and renal cell cancer syndrome (HLRCC). Virchows Arch 2015; 467: 185-191.
60. Ulamec M, Skenderi F, Zhou M, et al. Molecular genetic alterations in renal cell carcinoma with tubulocystic pattern: tubulocystic renal cell carcinoma, tubulocystic renal cell carcinoma with heterogeneous component and familial leiomyomatosis-associated renal cell carcinoma. Clinicopathological and molecular genetic analysis of 15 cases. App Immunohistochem Mol Morphol 2016; 24: 521-530.
61. Orseth ML, Redick D, Pinczewski J, et al. Something to Reed about Fibroids, cutaneous leiomyomas, and renal cell carcinoma. Am J Obstet Gynecol 2014; 210: 584e1-2.
62. Stewart L, Glenn G, Toro JR. Cutaneous leiomyomas: a clinical marker of risk for hereditary leiomyomatosis and renal cell cancer. Dematol Nurs 2006; 18: 335-341.
63. Varol A, Stapleton K, Rscioli T. The syndrome of hereditary leiomyomatosis and renal cell cancer (HLRCC): The clinical features of an individual with fumarate hydratase gene mutation. Austral J Dermatol 2006; 47: 274-276.
64. Henley ND, Tokarz VA. Multiple cutaneous and uterine leiomyomatosis in a 36-year-old female, and discussion of hereditary leiomyomatosis and renal cell carcinoma. Int J Dermatol 2012; 51: 1213-1216.
65. Choudhary S, McLeod M, Torchia D, et al. Multiple cutaneous and uterine leiomyomatosis syndrome: A review. J Clin Aesthet Dermatol 2013; 6: 16-21.
66. Frey MK, Worley MJ, Heyman KP, et al. A case report of hereditary leiomyomatosis and renal cell cancer. Am J Obstet Gynecol 2010; 68: e8-e9.
67. Joseph NM, Solomon DA, Frizzell N, et al. Morphology and immunohistochemistry for 2SC and FH aid in detection for fumarate hydratase gene aberrations in uterine leiomyomas from young patients. Am J Surg Pathol 2015; 39: 1529-1539.
68. Reyes C, Karamurzin Y, Frizzell N, et al. Uterine smooth muscle tumors with features suggesting fumarate hydratase aberration: detailed morphologic analysis and correlation with S-(s-scuccino)-cysteine immunohistochemistry. Mod Pathol 2014; 27: 1020-1027.
69. Alam NA, Rowan AJ, Wortham NC, et al. Genetic and functional analyses of FH mutations in multiple cutaneous and uterine leiomyomatosis, hereditary leiomyomatosis and renal cancer, and fumarate hydratase deficiency. Hum Mol Genet 2003; 12: 1241-1252.
70. Ahvenainen T, Lehtonen HJ, Lehtonen R, et al. Mutation screening of fumarate hydratase by multiplex ligation-dependent probe amplification: detection of exonic deletion in a patient with leiomyomatosis and renal cell cancer. Cancer Genet Cytogenet 2008; 183: 83-88.
71. Bayley JP, Launonen V, Tomlinson IPM. The FH mutation database: an online database of fumarate hydratase mutations involved in the MCUL (HLRCC) tumor syndrome and congenital fumarase deficiency. BMC Med Gent 2008; 9: 20.
72. Coughlin EM, Christensen E, Krisnamoorthy KS, et al. Molecular analysis and prenatal diagnosis of human fumarase deficiency. Mol Genet Metab 1998; 63: 254-262.
73. Lehtonen R, Kiuru M, Vanharanta S, et al. Biallelic inactivation of fumarate hydratase (FH) occurs in nonsyndromic uterine leiomyomas but is rare in other tumors. Am J Pathol 2004; 164: 17-22.
74. Kiuru M, Lehtonen R, Arola J, et al. Few FH mutations in sporadic counterparts of tumor types observed in hereditary leiomyomatosis and renal cell cancer families. Cancer Res 2002; 62: 4554-4557.
75. Barrisford GW, Singer EA, Rosner IL, et al. Familial renal cancer: Molecular genetics and surgical treatment. Int J Surg Oncol 2011; 2011: 658767.
76. Koski TA, Lehtonen HJ, Jee KJ, et al. Array comparative genomic hybridization identifies as a distinct DNA copy number profile in renal cell cancer associated with hereditary leiomyomatosis and renal cell cancer. Genes Chromsom Cancer 2009; 48: 544-551.
77. Vahteristo P, Koski TA, Näätsaari L, et al. No evidence for a genetic modifier for renal cell cancer risk in HLRCC syndrome. Fam Cancer 2010; 9: 245-251.
78. Alam NA, Olpin S, Rowan A, et al. Missense mutations in fumarate hydratase in multiple cutaneous and uterine leiomyomatosis and renal cell cancer. J Mol Diagn 2005; 7: 437-443.
79. Menko FH, Maher E, Schmidt LS, et al. Hereditary leiomyomatosis and renal cell cancer (HLRCC). Renal cancer risk, surveillance and treatment. Fam Cancer 2014; 13: 637-644.
80. Linehan WM, Bratslavsky G, Pinto PA, et al. Molecular diagnosis and therapy of kidney cancer. Annu Rev Med 2010; 61: 329-343.
81. Metwalli AR, Linehan WM. Nephron-sparing surgery for multifocal and hereditary renal tumors. Curr Opin Urol 2014; 24: 466-473.
82. Kiuru M, Lehtonen R, Eerola H, et al. No germline FH mutations in familial breast cancer patients. Eur J Hum Genet 2005; 13: 506-509.

Address for correspondences

Naoto Kuroda, MD
Department of Diagnostic Pathology,
Kochi Red Cross Hospital,
Shin-honmachi 2-13-51, Kochi City,
Kochi 780-8562, Japan
Tel. +81-88-822-1201
Fax. +81-88-822-1056
e-mail: kurochankcohi@yahoo.co.jp
Copyright: © 2018 Polish Association of Pathologists and the Polish Branch of the International Academy of Pathology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.