Open Access

Renal Epithelioid Angiomyolipomas: Clinicopathological Features, Diagnosis, and Management

KATHRINE S. RALLIS 1

1Barts Cancer Institute, Queen Mary University of London, London, U.K.

Cancer Diagnosis & Prognosis Nov-Dec; 5(6): 788-795 DOI: 10.21873/cdp.10495
Received 06 September 2025 | Revised 23 September 2025 | Accepted 24 September 2025
Corresponding author
Kathrine Rallis, MBBS, MSc (ORCiD: 0000-0003-1011-3220), Barts Cancer Institute, Queen Mary University of London, London EC1M 6AU, U.K. Tel: +44 07546272233, e-mail: ha16849@qmul.ac.uk
Download PDF pdf image icon

Abstract

Renal angiomyolipomas (AMLs) are typically benign, fat-rich mesenchymal tumours composed of vascular, smooth muscle, and adipose elements. However, rare fat-poor subtypes, particularly epithelioid angiomyolipoma (EAML), pose significant diagnostic and therapeutic challenges due to their malignant potential and morphological overlap with renal cell carcinoma (RCC). EAML represents less than 1% of AMLs but differs markedly in its biological behaviour, with a propensity for local invasion, lymphadenopathy, and distant metastasis, most commonly to the lungs and liver. Preoperative diagnosis is often delayed or incorrect, as imaging may fail to detect fat-deficient lesions, making histopathological evaluation necessary. EAML is defined by sheets of polygonal epithelioid cells with eosinophilic or clear cytoplasm, prominent nucleoli, and frequent mitotic figures. Genetic alterations in TSC1/TSC2 and activation of the mTOR pathway are central to pathogenesis, particularly in tuberous sclerosis complex-associated cases. Management hinges on surgical resection, yet recurrence and metastasis may occur years after nephrectomy. mTOR inhibitors have demonstrated efficacy in tumour reduction, especially in TSC-linked cases, while VEGF pathway inhibitors and immune checkpoint blockade represent promising but underexplored avenues. Prognostic features include tumour size >9 cm, necrosis, vascular invasion, and high epithelioid cell proportion. Given the rarity and aggressive potential of EAML, long-term surveillance and multidisciplinary care are essential. Continued research into molecular drivers and targeted therapies will be critical to improving diagnostic precision and patient outcomes.
Keywords: Angiomyolipoma, renal cell carcinoma, tyrosine kinase inhibitors, immunotherapy, targeted therapy, review

Introduction

Renal angiomyolipomas (AMLs) are benign neoplasms of the kidney typically composed of a triphasic morphology of vascular, smooth muscle, and adipose tissue, though rare atypical types may be of monophasic or epithelioid histology (1,2). Once considered hamartomas and choristomas, AMLs are currently classified amongst the family of perivascular epithelioid cell tumours (PEComa) (3-5).

AMLs are the most common benign renal tumours associated with an overall prevalence of 0.44% and a strong predisposition to females who represent twice as many cases compared to males (0.60% females versus 0.28% males) (6). A more recent retrospective surgical cohort (2001-2021) found that 70.5% of AML cases were women (female-to-male ratio ≈ 1:2.4); 42% were diagnosed incidentally (7). Sporadic cases comprise 80% of AMLs, while 20% may present as part of tuberous sclerosis (TSC) or rarely pulmonary lymphangioleiomyomatosis (LAM), a condition observed almost exclusively in women (4,8,9).

Different types of renal AMLs exist forming a heterogeneous group of neoplasms which display different pathological, radiological, and clinical features (10). Imaging is crucial in the diagnosis and management of renal AMLs with the detection of adipose tissue constituting an essential diagnostic criterion (8). However, despite having a classic triphasic morphology, certain AMLs display poor fat content on macroscopic imaging, thus rendering it difficult to preoperatively distinguish them from renal cell carcinoma (RCC) to prevent unnecessary nephrectomy for a benign tumour (5,11-16).

AML with epithelial cysts (AMLEC) is an extremely rare subtype of benign, fat-poor AML that was only classified in 2017 (10) following the publication of two case series in the literature (17,18). In contrast, epithelioid AML (EAML) is a rare distinct variant (1%) of fat poor AML of mesenchymal origin, which may undergo malignant transformation (19,20). EAML is characterised by p53 mutations (21) and local aggressiveness, including lymphadenopathy and distal metastases to the lung, liver, mediastinum, and vertebrae (22,23) in a third of cases (9,21,24-26). Size correlates to risk of spread (10,27) and over half of EAMLs are associated with TSC (28). This malignant phenotype contradicts that of typical AML, which has historically proven benign in hundreds of cases, except for two reports, in which high-grade leiomyosarcoma arose from typical angiomyolipoma (29,30).

Diagnosis

EAML poses a diagnostic challenge given its clinico-pathological resemblance and similar epithelioid morphology to RCC (31), sarcoma, and medullary carcinoma (32,33). Additionally, EAML may coexist simultaneously with clear cell RCC (33,34). Therefore, diagnosis often depends on postoperative histopathological assessment (35).

Histologically, EAML is characterised by predominant proliferation of atypical, polymorphic, round to polygonal epithelioid cells featuring abundant clear or densely stained eosinophilic cytoplasm, enlarged vesicular nuclei and often prominent nucleoli similarly to ganglion cells. Short spindle cells and multinucleated giant cells may also be present (32). Some authors suggest a requirement of at least 5% (36) or 10% (37) epithelioid histology for the diagnosis of EAML, though >80% is often quoted as an appropriate cut-off for malignant transformation (38,39), while others report nearly pure epithelioid composition (40,41).

Immunohistochemical staining for human melanoma black (HMB)-45-positive epithelioid cells, in the absence of adipocytes or minimal adult-appearing fat tissue, serves as a useful marker for EAML diagnosis (42). Moreover, CD-68 positivity and cytokeratin negativity are paramount in aiding EAML identification. Positive immunostaining for Melan-A, muscle-specific actin and, rarely, smooth muscle actin may also be featured with negative reactivity against epithelial membrane antigen (32).

Treatment

Malignant AML is always managed by partial or radical nephrectomy followed by adjuvant chemotherapy (2,43). Surgical decision-making is guided by the diagnosis and radiology-derived nephrometric scoring when partial nephrectomy is planned (43). Recently, an increasing number of AMLs are managed with selective arterial embolization and percutaneous ablation, which are associated with less morbidity compared to traditional surgery (43) Integrated approaches, including surgery, chemotherapy, and molecular-targeted therapy, should be considered upon EAML diagnosis. Follow-up is indicated in EAMLs with necrosis, >9 cm tumour size, venous thrombosis, >70% epithelioid cells or >60% cellular atypia (35). Surgery remains the most effective approach for EAML, although still often insufficient. Recurrence or distant metastasis occur within 1.5-9 years postoperatively (23,28,44-48).

Chemotherapy and targeted therapy. Since EAML is a PEComa, which has similar histological features with to lymphagioleiomyomas and clear cell “sugar” tumours of the lung and pancreas (3), it may respond to chemotherapies such as doxorubicin, cyclophosphamide, and cisplatin on long-term follow-up (25,49).

Furthermore, mTOR inhibitors rapamycin/sirolimus (50), everolimus (51), and temsirolimus can achieve tumour regression and reduce gross tumour burden in patients with TSC-associated (52-58) and sporadic renal AMLs (59). A single institution retrospective study on advanced PEComas, including epithelioid variants, demonstrated that mTOR inhibitors offered similar survival and PFS outcomes compared to chemotherapy, but with potentially less morbidity, suggesting preferential use (60). Moreover, one study of 15 PEComa cases showed that the mTOR cascade was always activated, further supporting the use of mTOR inhibitors (61). This can be explained since TSC1/TSC2 genes are disrupted in patients with TSC and sporadic AML (62), thus encoding an abnormal TSC1/TSC2 protein complex that inappropriately regulates mTOR complex 1 (mTORC1) (Figure 1) (63). Dysregulated cell growth and increased VEGF expression occur from mTORC1-dependent HIF activation (64,65). Indeed, a multicentre phase 2 trial showed that sirolimus was successful in inducing renal AML regression and decreasing baseline elevated serum VEGF-D which was shown to correlate to AML size (Spearman correlation coefficient 0.54, p=0.001, at baseline) (66).

Moreover, although the multi-kinase inhibitor sorafenib, prescribed as first/second line treatment for advanced and/or metastatic RCC, was ineffective in treating EAML according to one case report (67), sunitinib, a multi-kinase inhibitor used as second line treatment, was effective in achieving tumour regression according to a paediatric EAML case report (68). The same case report also showed that axitinib, an active selective inhibitor of VEGFR 1, 2 and 3, may offer therapeutic benefit. Specifically, when administered as third-line treatment, it effectively halted primary tumour growth postoperatively, although thoracic metastasis was observed in later disease stage. Subsequently, a phase 3 trial comparing pembrolizumab plus axitinib versus sunitinib for advanced RCC has proven that the former is superior in achieving significantly longer overall survival, progression-free survival, and a higher objective response rate (52). Further research is needed to determine the effectiveness of pembrolizumab plus axitinib in the treatment of renal AML. The advantage of axitinib and other single-target growth factor receptor inhibitors over multikinase inhibitors is that they have the potential to decrease toxicity while precisely targeting the VHL pathway in clear cell kidney cancer (69).

Prognosis and Future Directions

The prognosis of EAML remains guarded due to its potential for local recurrence and distant metastasis. Histological predictors of poor prognosis include marked nuclear atypia, necrosis, high mitotic rate (>1/50 high power fields), vascular invasion, and the presence of >70% epithelioid components or >60% atypical epithelioid cells. Clinical features such as tumour size >9 cm, involvement of renal vein or inferior vena cava, and non-resectability are also associated with a more aggressive clinical course.

Despite aggressive surgical management, the recurrence rate of EAML is non-negligible, with distant metastases most commonly affecting the lungs, liver, and bones. Metastatic progression may occur several years after primary tumour resection, highlighting the need for long-term surveillance. Advanced EAML is notoriously chemoresistant and lacks a standardized systemic treatment protocol, emphasizing the need for further research into novel therapeutic agents.

Recent genomic and transcriptomic analyses suggest that EAML and other PEComas may benefit from precision oncology approaches, especially with the identification of TSC1/TSC2 mutations and mTOR pathway activation as common oncogenic drivers (61,62). mTOR inhibitors have shown considerable promise in both sporadic and TSC-associated cases; however, not all patients respond, possibly due to molecular heterogeneity and downstream resistance mechanisms.

The potential utility of immune checkpoint inhibitors (ICIs), such as pembrolizumab, remains under investigation. Genomic profiling of 34 malignant renal EAMLs revealed molecular features including microsatellite instability (MSI), tumor mutational burden (TMB), and PD-L1 expression, as well as potential targets for precision therapy (70). Given the immunogenic potential of EAML and its occasional expression of PD-L1, further studies are warranted to assess the efficacy of ICIs alone or in combination with VEGF/VEGFR-targeting agents like axitinib (64,68,69). Clinical trials specifically targeting malignant PEComas - including EAML - are crucial to refine these therapeutic strategies. Moreover, liquid biopsy and circulating tumour DNA (ctDNA) may offer valuable, non-invasive tools for monitoring disease activity, particularly in assessing residual disease and detecting early recurrence.

Conclusion

Renal AMLs represent a spectrum of mesenchymal neoplasms, with EAML forming a rare but potentially malignant variant. Although most AMLs are benign and easily diagnosable through imaging, fat-poor subtypes, especially EAML, can mimic RCC and necessitate histopathological evaluation for accurate diagnosis.

Management of EAML is complex, often requiring surgical intervention and adjunctive systemic therapy. Emerging treatments, particularly mTOR inhibitors and VEGF/VEGFR-directed therapies, have improved outcomes in selected cases. However, recurrent and metastatic EAMLs remain challenging to treat, underscoring the importance of molecular diagnostics, multidisciplinary care, and clinical trial participation.

Further research is required to refine diagnostic criteria, identify molecular biomarkers of aggression, and optimize targeted and immunotherapeutic regimens tailored to the unique biology of EAML.

Conflicts of Interest

The Author has no conflicts of interest to declare.

Authors’ Contributions

KSR: Conceptualization, Drafting, Reviewing.

Artificial Intelligence (AI) Disclosure

No artificial intelligence (AI) tools, including large language models or machine learning software, were used in the preparation, analysis, or presentation of this manuscript.

References

1 Oesterling JE Fishman EK Goldman SM & Marshall FF The management of renal angiomyolipoma. J Urol. 135(6) 1121 - 1124 1986. DOI: 10.1016/s0022-5347(17)46013-7
2 Nelson CP & Sanda MG Contemporary diagnosis and management of renal angiomyolipoma. J Urol. 168(4 Pt 1) 1315 - 1325 2002. DOI: 10.1016/S0022-5347(05)64440-0
3 Zamboni G Pea M Martignoni G Zancanaro C Faccioli G Gilioli E Pederzoli P & Bonetti F Clear cell “sugar” tumor of the pancreas. A novel member of the family of lesions characterized by the presence of perivascular epithelioid cells. Am J Surg Pathol. 20(6) 722 - 730 1996. DOI: 10.1097/00000478-199606000-00010
4 Thiravit S Teerasamit W & Thiravit P The different faces of renal angiomyolipomas on radiologic imaging: a pictorial review. Br J Radiol. 91(1084) 20170533 2018. DOI: 10.1259/bjr.20170533
5 Labra A Schiappacasse G Constenla D & Cristi J Renal angiomyolipomas: Typical and atypical features on computed tomography and magnetic resonance imaging. World J Radiol. 17(2) 104282 2025. DOI: 10.4329/wjr.v17.i2.104282
6 Fittschen A Wendlik I Oeztuerk S Kratzer W Akinli AS Haenle MM & Graeter T Prevalence of sporadic renal angiomyolipoma: a retrospective analysis of 61,389 in- and out-patients. Abdom Imaging. 39(5) 1009 - 1013 2014. DOI: 10.1007/s00261-014-0129-6
7 Chacko AZ Prashanthy TAM Gopinathan V George AJP Kumar S & Chacko G Adult renal angiomyolipomas: A retrospective analysis of the histological subtypes and their clinicoradiological correlates. Urol Ann. 14(4) 365 - 371 2022. DOI: 10.4103/ua.ua_129_21
8 Flum AS Hamoui N Said MA Yang XJ Casalino DD McGuire BB Perry KT & Nadler RB Update on the diagnosis and management of renal angiomyolipoma. J Urol. 195(4 Part 1) 834 - 846 2016. DOI: 10.1016/j.juro.2015.07.126
9 Meraj R Wikenheiser-Brokamp K Young L & McCormack F Lymphangioleiomyomatosis: New concepts in pathogenesis, diagnosis, and treatment. Semin Respir Crit Care Med. 33(05) 486 - 497 2012. DOI: 10.1055/s-0032-1325159
10 Jinzaki M Silverman SG Akita H Nagashima Y Mikami S & Oya M Renal angiomyolipoma: a radiological classification and update on recent developments in diagnosis and management. Abdom Imaging. 39(3) 588 - 604 2014. DOI: 10.1007/s00261-014-0083-3
11 Jinzaki M Silverman SG Akita H Mikami S & Oya M Diagnosis of renal angiomyolipomas: classic, fat-poor, and epithelioid types. Semin Ultrasound CT MR. 38(1) 37 - 46 2017. DOI: 10.1053/j.sult.2016.11.001
12 Milner J McNeil B Alioto J Proud K Rubinas T Picken M Demos T Turk T & Perry KT Fat poor renal angiomyolipoma: patient, computerized tomography and histological findings. J Urol. 176(3) 905 - 909 2006. DOI: 10.1016/j.juro.2006.04.016
13 Jinzaki M Tanimoto A Narimatsu Y Ohkuma K Kurata T Shinmoto H Hiramatsu K Mukai M & Murai M Angiomyolipoma: imaging findings in lesions with minimal fat. Radiology. 205(2) 497 - 502 1997. DOI: 10.1148/radiology.205.2.9356635
14 Silverman SG Israel GM Herts BR & Richie JP Management of the incidental renal mass. Radiology. 249(1) 16 - 31 2008. DOI: 10.1148/radiol.2491070783
15 Park BK Renal angiomyolipoma based on new classification: how to differentiate it from renal cell carcinoma. Am J Roentgenol. 212(3) 582 - 588 2019. DOI: 10.2214/AJR.18.20408
16 Liang X Zeng XT Hong ZL Su MJ Yang JC & Wu SS Determinants of conventional and contrast-enhanced ultrasound diagnosis of fat-poor angiomyolipoma <5 cm. Front Oncol. 14 1446801 2024. DOI: 10.3389/fonc.2024.1446801
17 Fine SW Reuter VE Epstein JI & Argani P Angiomyolipoma with epithelial cysts (AMLEC): a distinct cystic variant of angiomyolipoma. Am J Surg Pathol. 30(5) 593 - 599 2006. DOI: 10.1097/01.pas.0000194298.19839.b4
18 Davis CJ Barton JH & Sesterhenn IA Cystic angiomyolipoma of the kidney: a clinicopathologic description of 11 cases. Mod Pathol. 19(5) 669 - 674 2006. DOI: 10.1038/modpathol.3800572
19 Eble JN Amin MB & Young RH Epithelioid angiomyolipoma of the kidney: a report of five cases with a prominent and diagnostically confusing epithelioid smooth muscle component. Am J Surg Pathol. 21(10) 1123 - 1130 1997. DOI: 10.1097/00000478-199710000-00001
20 Eble JN Sauter G Epstein JI & Sesterhenn IA Pathology and genetics of tumours of the urinary system and male genital organs (Vol. 7). Lyon, France, IARC Press.
21 Sato K Ueda Y Tachibana H Miyazawa K Chikazawa I Kaji S Nojima T & Katsuda S Malignant epithelioid angiomyolipoma of the kidney in a patient with tuberous sclerosis: An autopsy case report with p53 gene mutation analysis. Pathol Res Pract. 204(10) 771 - 777 2008. DOI: 10.1016/j.prp.2008.04.008
22 Lau SK Marchevsky AM McKenna RJ & Luthringer DJ Malignant monotypic epithelioid angiomyolipoma of the retroperitoneum. Int J Surg Pathol. 11(3) 223 - 228 2003. DOI: 10.1177/106689690301100313
23 Huang KH Huang CY Chung SD Pu YS Shun CT & Chen J Malignant epithelioid angiomyolipoma of the kidney. J Formos Med Assoc Taiwan Yi Zhi. 106(2) S51 - S54 2007. DOI: 10.1016/s0929-6646(09)60353-3
24 Li J Zhu M & Wang YL Malignant epithelioid angiomyolipoma of the kidney with pulmonary metastases and p53 gene mutation. World J Surg Oncol. 10 213 2012. DOI: 10.1186/1477-7819-10-213
25 Cibas ES Goss GA Kulke MH Demetri GD & Fletcher CDM Malignant epithelioid angiomyolipoma (`sarcoma ex angiomyolipoma’) of the kidney: a case report and review of the literature. Am J Surg Pathol. 25(1) 121 - 126 2001. DOI: 10.1097/00000478-200101000-00014
26 Li H Zhang X Zhao Q Wang J Yang X & Huang S Renal epithelioid angiomyolipoma: A case report. Oncol Lett. 26(3) 409 2023. DOI: 10.3892/ol.2023.13995
27 Halpenny D Snow A McNeill G & Torreggiani W The radiological diagnosis and treatment of renal angiomyolipoma—current status. Clin Radiol. 65(2) 99 - 108 2010. DOI: 10.1016/j.crad.2009.09.014
28 Bjornsson J Short MP Kwiatkowski DJ & Henske EP Tuberous sclerosis-associated renal cell carcinoma. Clinical, pathological, and genetic features. Am J Pathol. 149(4) 1201 - 1208 1996.
29 Ferry JA Malt RA & Young RH Renal angiomyolipoma with sarcomatous transformation and pulmonary metastases. Am J Surg Pathol. 15(11) 1083 - 1088 1991. DOI: 10.1097/00000478-199111000-00008
30 Lowe BA Brewer J Houghton DC Jacobson E & Pitre T Malignant transformation of angiomyolipoma. J Urol. 147(5) 1356 - 1358 1992. DOI: 10.1016/s0022-5347(17)37564-x
31 Xiao A Van Ziffle J & Chan E Epithelioid angiomyolipoma with prominent papillary architecture mimicking renal cell carcinoma: a case report. Int J Surg Pathol. 32(1) 206 - 208 2024. DOI: 10.1177/10668969231171942
32 Svec A & Velenská Z Renal epithelioid angiomyolipoma—a close mimic of renal cell carcinoma. Report of a case and review of the literature. Pathol Res Pract. 200(11-12) 851 - 856 2005. DOI: 10.1016/j.prp.2004.09.004
33 Cao Q Liu F Xiao P Tian X Li B & Li Z Coexistence of renal epithelioid angiomyolipoma and clear cell carcinoma in patients without tuberous sclerosis. Int J Surg Pathol. 20(2) 194 - 198 2012. DOI: 10.1177/1066896911413576
34 Pea M Bonetti F Martignoni G Henske EP Manfrin E Colato C & Bernstein J Apparent renal cell carcinomas in tuberous sclerosis are heterogeneous. Am J Surg Pathol. 22(2) 180 - 187 1998. DOI: 10.1097/00000478-199802000-00005
35 Lei JH Liu LR Wei Q Song TR Yang L Yuan HC Jiang Y Xu H Xiong SH & Han P A four-year follow-up study of renal epithelioid angiomyolipoma: a multi-center experience and literature review. Sci Rep. 5 10030 2015. DOI: 10.1038/srep10030
36 Jayaprakash PG Mathews S Azariah MB & Babu G Pure epitheliod perivascular epitheloid cell tumor (epitheliod angiomyolipoma) of kidney: Case report and literature review. J Cancer Res Ther. 10(2) 404 2014. DOI: 10.4103/0973-1482.136672
37 Aydin H Magi-Galluzzi C Lane BR Sercia L Lopez JI Rini BI & Zhou M Renal angiomyolipoma: clinicopathologic study of 194 cases with emphasis on the epithelioid histology and tuberous sclerosis association. Am J Surg Pathol. 33(2) 289 - 297 2009. DOI: 10.1097/PAS.0b013e31817ed7a6
38 He W Cheville JC Sadow PM Gopalan A Fine SW Al-Ahmadie HA Chen YB Oliva E Russo P Reuter VE & Tickoo SK Epithelioid angiomyolipoma of the kidney: pathological features and clinical outcome in a series of consecutively resected tumors. Mod Pathol. 26(10) 1355 - 1364 2013. DOI: 10.1038/modpathol.2013.72
39 Brimo F Robinson B Guo C Zhou M Latour M & Epstein JI Renal epithelioid angiomyolipoma with atypia: a series of 40 cases with emphasis on clinicopathologic prognostic indicators of malignancy. Am J Surg Pathol. 34(5) 715 - 722 2010. DOI: 10.1097/PAS.0b013e3181d90370
40 Bani MA Laabidi B Rejeb SB Khiari R Bouziani A & Msakni I A rare highly aggressive tumor of the kidney: The pure epithelioid angiomyolipoma. Urol Case Rep. 18 52 - 53 2018. DOI: 10.1016/j.eucr.2018.03.007
41 Nese N Martignoni G Fletcher CD Gupta R Pan CC Kim H Ro JY Hwang IS Sato K Bonetti F Pea M Amin MB Hes O Svec A Kida M Vankalakunti M Berel D Rogatko A Gown AM & Amin MB Pure epithelioid PEComas (So-called epithelioid angiomyolipoma) of the kidney. Am J Surg Pathol. 35(2) 161 - 176 2011. DOI: 10.1097/PAS.0b013e318206f2a9
42 Konosu-Fukaya S Nakamura Y Fujishima F Kasajima A McNamara KM Takahashi Y Joh K Saito H Ioritani N Ikeda Y Arai Y Watanabe M & Sasano H Renal epithelioid angiomyolipoma with malignant features: Histological evaluation and novel immunohistochemical findings. Pathol Int. 64(3) 133 - 141 2014. DOI: 10.1111/pin.12142
43 Razik A Das CJ & Sharma S Angiomyolipoma of the kidneys: current perspectives and challenges in diagnostic imaging and image-guided therapy. Curr Probl Diagn Radiol. 48(3) 251 - 261 2019. DOI: 10.1067/j.cpradiol.2018.03.006
44 Varma S Gupta S Talwar J Forte F & Dhar M Renal epithelioid angiomyolipoma: a malignant disease. J Nephrol. 24(1) 18 - 22 2011. DOI: 10.5301/jn.2010.5451
45 Hardman JA McNicholas TA Kirkham N & Fletcher MS Recurrent renal angiomyolipoma associated with renal carcinoma in a patient with tuberous sclerosis. Br J Urol. 72(6) 983 - 984 1993. DOI: 10.1111/j.1464-410x.1993.tb16321.x
46 Yamamoto T Ito K Suzuki K Yamanaka H Ebihara K & Sasaki A Rapidly progressive malignant epithelioid angiomyolipoma of the kidney. J Urol. 168(1) 190 - 191 2002.
47 Mai KT Perkins DG & Collins JP Epithelioid cell variant of renal angiomyolipoma. Histopathology. 28(3) 277 - 280 1996. DOI: 10.1046/j.1365-2559.1996.d01-421.x
48 Martignoni G Renal epithelioid oxyphilic neoplasm (REON) : A pleomorphic monophasic variant of renal angiomyolipoma. Int J Surg Pathol. 2 539 1995.
49 Park HK Zhang S Wong MK & Kim HL Clinical presentation of epithelioid angiomyolipoma. Int J Urol. 14(1) 21 - 25 2007. DOI: 10.1111/j.1442-2042.2006.01665.x
50 Dun S Wang YY Wan L Wang QH Lu Q Yang XY Zhang Q Chen HM Qiu LP & Zou LP Sirolimus can promote the disappearance of renal angiomyolipoma associated with tuberous sclerosis complex: a prospective cohort study. World J Pediatr WJP. 20(6) 602 - 610 2024. DOI: 10.1007/s12519-023-00751-5
51 Guo G Gu L & Zhang X Everolimus in invasive malignant renal epithelioid angiomyolipoma. Front Oncol. 10 610858 2021. DOI: 10.3389/fonc.2020.610858
52 El-Hashemite N Zhang H Henske EP & Kwiatkowski DJ Mutation in TSC2 and activation of mammalian target of rapamycin signalling pathway in renal angiomyolipoma. Lancet Lond Engl. 361(9366) 1348 - 1349 2003. DOI: 10.1016/S0140-6736(03)13044-9
53 Krischock L Beach R & Taylor J Sirolimus and tuberous sclerosis-associated renal angiomyolipomas. Arch Dis Child. 95(5) 391 - 392 2010. DOI: 10.1136/adc.2009.159210
54 Micozkadioglu H Koc Z Ozelsancak R & Yildiz I Rapamycin therapy for renal, brain, and skin lesions in a tuberous sclerosis patient. Ren Fail. 32(10) 1233 - 1236 2010. DOI: 10.3109/0886022X.2010.517345
55 Sampson JR Therapeutic targeting of mTOR in tuberous sclerosis. Biochem Soc Trans. 37(1) 259 - 264 2009. DOI: 10.1042/BST0370259
56 Bissler JJ McCormack FX Young LR Elwing JM Chuck G Leonard JM Schmithorst VJ Laor T Brody AS Bean J Salisbury S & Franz DN Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis. N Engl J Med. 358(2) 140 - 151 2008. DOI: 10.1056/NEJMoa063564
57 Palavra F Robalo C & Reis F Recent advances and challenges of mTOR inhibitors use in the treatment of patients with tuberous sclerosis complex. Oxid Med Cell Longev. 2017 9820181 2017. DOI: 10.1155/2017/9820181
58 Tessone IM Lichtbroun B Srivastava A Tabakin AL Polotti CF Groisberg R Sadimin E Singer EA & Grandhi MS Massive malignant epithelioid angiomyolipoma of the kidney. J Kidney Cancer VHL. 9(2) 13 - 18 2022. DOI: 10.15586/jkcvhl.v9i2.210
59 Wagner AJ Malinowska-Kolodziej I Morgan JA Qin W Fletcher CD Vena N Ligon AH Antonescu CR Ramaiya NH Demetri GD Kwiatkowski DJ & Maki RG Clinical activity of mTOR inhibition with sirolimus in malignant perivascular epithelioid cell tumors: targeting the pathogenic activation of mTORC1 in tumors. J Clin Oncol. 28(5) 835 - 840 2010. DOI: 10.1200/JCO.2009.25.2981
60 Testa S Bui NQ & Ganjoo KN Systemic treatments and molecular biomarkers for perivascular epithelioid cell tumors: a single-institution retrospective analysis. Cancer Res Commun. 3(7) 1212 - 1223 2023. DOI: 10.1158/2767-9764.CRC-23-0139
61 Kenerson H Folpe AL Takayama TK & Yeung RS Activation of the mTOR pathway in sporadic angiomyolipomas and other perivascular epithelioid cell neoplasms. Hum Pathol. 38(9) 1361 - 1371 2007. DOI: 10.1016/j.humpath.2007.01.028
62 Henske EP Neumann HPH Scheithauer BW Herbst EW Short MP & Kwiatkowski DJ Loss of heterozygosity in the tuberous sclerosis (TSC2) region of chromosome band l6p13 occurs in sporadic as well as TSC-associated renal angiomyolipomas. Genes Chromosomes Cancer. 13(4) 295 - 298 1995. DOI: 10.1002/gcc.2870130411
63 Samuels JA Treatment of renal angiomyolipoma and other hamartomas in patients with tuberous sclerosis complex. Clin J Am Soc Nephrol. 12(7) 1196 - 1202 2017. DOI: 10.2215/CJN.08150816
64 Brugarolas JB Vazquez F Reddy A Sellers WR & Kaelin WG TSC2 regulates VEGF through mTOR-dependent and -independent pathways. Cancer Cell. 4(2) 147 - 158 2003. DOI: 10.1016/S1535-6108(03)00187-9
65 Wolff N Kabbani W Bradley T Raj G Watumull L & Brugarolas J Sirolimus and temsirolimus for epithelioid angiomyolipoma. J Clin Oncol. 28(5) e65 - e68 2010. DOI: 10.1200/JCO.2009.26.3061
66 Dabora SL Franz DN Ashwal S Sagalowsky A DiMario FJ Jr Miles D Cutler D Krueger D Uppot RN Rabenou R Camposano S Paolini J Fennessy F Lee N Woodrum C Manola J Garber J & Thiele EA Multicenter phase 2 trial of sirolimus for tuberous sclerosis: kidney angiomyolipomas and other tumors regress and VEGF- D levels decrease. PLoS One. 6(9) e23379 2011. DOI: 10.1371/journal.pone.0023379
67 Guo B Song H Yue J & Li G Malignant renal epithelioid angiomyolipoma: A case report and review of the literature. Oncol Lett. 11(1) 95 - 98 2016. DOI: 10.3892/ol.2015.3846
68 Citak EC Yilmaz EB Yaman E Kaya S Taskinlar H Arpaci RB & Apaydin D Malignant epitheloid angiomyolipoma of the kidney in a child treated with sunitinib, everolimus and axitinib. Can Urol Assoc J. 9(7-8) E542 - E545 2015. DOI: 10.5489/cuaj.2823
69 Linehan WM Targeting VEGF receptors in kidney cancer. Lancet Oncol. 8(11) 956 - 957 2007. DOI: 10.1016/S1470-2045(07)70322-4
70 Sager RA Basin M Jacob JM Basnet A Pavlick DC Lin DI Necchi A Spiess PE Li R Kamat AM Grivas P Daneshvar MA Goldberg H Mollapour M Ross JS & Bratslavsky G Malignant epithelioid angiomyolipoma (eAML)/PEComa of the kidney: A genomic landscape study. J Clin Oncol. 42(4_suppl) 451 - 451 2024. DOI: 10.1200/JCO.2024.42.4_suppl.451