J Urol Oncol > Volume 22(1); 2024 > Article
Hong, Kwon, Kang, Seo, and Park: Clinical Characteristics and Outcomes of TFE3-Rearranged/TFEB-Altered Renal Cell Carcinoma with Systemic Therapies, Including Tyrosine Kinase Inhibitors or Immune Checkpoint Inhibitors: An Observational Study

Abstract

Purpose

TFE3-rearranged/TFEB-altered renal cell carcinoma (RCC) is a rare subtype of RCC. Due to its rarity, there is an unmet medical need for effective therapies in advanced settings. The study aims to investigate the clinical and histopathological characteristics of patients with microphthalmia transcription factor family/ transcription factor E (MiTF/TFE) translocation RCC and the clinical outcomes of systemic therapies, including tyrosine kinase inhibitors (TKIs) or immune checkpoint inhibitors (ICIs).

Materials and Methods

This was a single-center, retrospective study. We identified 32 eligible patients among a total of 37 patients diagnosed with MiTF/TFE translocation RCC between January 2004 and September 2021, and the study included 9 patients who were treated with systemic therapies. We collected data on clinical characteristics, targeted sequencing, and clinical outcomes.

Results

The median age of the 32 patients was 45.5 years. Histologically, 26 patients (81.3%) had TFE3-rearranged RCC, and only 1 patient (3.1%) had TFEB-altered RCC. Curative or cytoreductive nephrectomy was performed in all 27 patients (84.4%), and 4 patients (12.6%) were diagnosed with metastatic disease at the time of the initial diagnosis. Nine patients (28.1%) were treated with systemic therapy with TKIs, 2 (6.3%) of whom received simultaneous TKI and ICI treatment. The response to systemic therapy (TKI or ICI) and duration of response ranged from complete response to progressive disease. Excluding 1 patient who was treated with a TKI in the adjuvant setting, the overall response rate in 8 metastatic patients was 50% and the complete response rate was 37.5%. The median follow-up period was 29 months. The median progression-free survival was 21 months, median overall survival was not achieved, and 2 deaths occurred.

Conclusions

Our findings suggest that TKI for treatment for metastatic TFE3-rearranged RCC is efficacious, with an overall response rate of 50% and a median progression-free survival of 21 months.

INTRODUCTION

In 2020, there were more than 430,000 new cases of kidney cancer globally and 179,000 deaths globally [1]. Smoking, obesity, and hypertension are established risk factors for renal cell carcinoma (RCC), which is a heterogeneous disease comprised of several histological subtypes with different genetic and clinicopathological characteristics. Among the histologic subtypes of RCC, clear cell carcinoma is the most common, accounting for 75% to 90% of total kidney cancers [2]. The remaining 20% include non-clear cell RCCs, such as papillary, chromophobe, and other rare subtypes. A single patient with RCC can sometimes harbor more than one subtype.
The benefits of vascular endothelial growth factor receptor (VEGFR)-targeted therapies for advanced RCC have long been known in palliative settings. Although the therapeutic options for advanced RCC have expanded in recent years to include immune checkpoint inhibitors (ICIs), such as pembrolizumab and nivolumab, VEGFR-targeted tyrosine kinase inhibitors (TKIs) remain the backbone of most current treatment guidelines [3-6]. However, novel therapeutic strategies have primarily focused on clear cell RCC, and few studies have evaluated ICIs in non-clear cell RCC.
In the latest (2022) World Health Organization (WHO) Classification of Urinary and Male Genital Tumors, rare subtypes of RCC were newly categorized according to their molecular features [7]. Most notably, the 2022 WHO classification introduced a new category of molecularlydefined renal tumors in addition to a morphology-based classification of renal tumors.
Microphthalmia transcription factor family (MiTF) translocation RCC was first described as an Xp11 translocation RCC by the WHO classification in 2004 [2]. Xp11 translocation RCC is characterized by chromosomal translocations involving the TFE3 transcription factor gene located at the chromosome Xp11.2 locus. The fusions include PRCC, ASPL, and SFPQ/PSF as partner genes [8,9]. Meanwhile, t(6;11) translocation RCC is characterized by fusion between TFEB on chromosome 6p21.2 and Alpha/MALAT1 on chromosome 11q13 [10]. Because of the rarity of t(6;11) translocation RCC, and because it was believed that translocation RCCs with TFE3 or TFEB rearrangements share clinical and histopathological features, these tumors were previously grouped as MiTF/TFE translocation RCC [11,12]. However, as described above, TFE3-rearranged RCC and TFEB-altered RCC were separated into 2 distinct molecularlydefined subtypes in the 2022 WHO classification. MiTF/TFE translocation RCC represents up to 40% of all pediatric and adolescent RCCs and 1% to 4% of adult RCCs [13]. Due to the morphological overlap with more common subtypes, the frequency of translocation RCC in adults is probably underestimated in the absence of specific molecular studies [14].
Although more than a decade has passed since MiTF/TFE translocation RCC was recognized, effective therapies for these tumors represent an unmet medical need. Radical or nephron-sparing nephrectomy is considered for localized tumors, but there are few studies of systemic therapies in advanced settings. The available treatment options have all been based on the extrapolation of data from studies conducted almost exclusively in more common types of RCC. Due to the lack of a full understanding of molecular carcinogenesis, as well as the rarity of the disease, there have only been retrospective studies on VEGFR-targeted agents, mammalian target of rapamycin (mTOR) inhibitors, and ICIs [15-19].
Considering the difficulties in conducting a prospective study, we designed the present study to describe clinical and histopathological characteristics of patients with MiTF/TFE translocation RCC. We also investigated the clinical outcomes of existing systemic therapies.

MATERIALS AND METHODS

This was a single-center, retrospective study. Patients with MiTF/TFE translocation RCC were identified through an electronic medical record search of the patient database for the period between January 2004 and September 2021 in Samsung Medical Center (Seoul, Korea). We identified 32 eligible patients among a total of 37 patients diagnosed with MiTF/TFE translocation RCC, and 9 of those patients who were treated with systemic therapies were included in the present retrospective study (Fig. 1). Eligibility criteria were adult patients (20 years or older) with MiTF/TFE translocation RCC diagnosed by a dedicated genitourinary pathologist utilizing immunohistochemistry (IHC) and/or fluorescent in situ hybridization (FISH) and treatment with at least one dose of TKIs or ICIs.
The medical records of the patients were reviewed, and information on patient death was obtained from census data. The demographic, histological, and clinical characteristics of patients at diagnosis were described and used for the analysis. Treatment and clinical outcomes of the patients were obtained from medical records. The data cutoff date was July 2022. Targeted sequencing of primary tumors using TruSight Oncology 500 (Illumina, San Diego, CA, USA) was performed in 6 of the 9 eligible patients. Tumor samples from 3 patients who were lost to follow-up were not available for targeted sequencing.
We collected tumor samples from archival tissues obtained from surgery. Genomic DNA was extracted and the DNA quality and quantity were assessed in a similar manner to a previous study [20]. DNA libraries were prepared using the hybrid capture-based TruSight Oncology 500 Library Preparation Kit (Illumina) following the manufacturer’s instructions. Because this study was retrospective in nature, matched normal tissues were not available. The tumor mutational burden, microsatellite instability calls, germline variants, and called variants were generated and filtered in a similar manner to another study [21].
Because the sample was small, the endpoints of the present study were mainly descriptive in nature. Data were collected on patients’ baseline characteristics, including sex, age, International Metastatic RCC Database Consortium (IMDC) risk score, performance status, American Joint Committee on Cancer TNM stage, and metastatic sites, as well as primary treatment were collected. In addition, indices for clinical outcomes such as types of therapy, clinical responses, and survival status were also collected. Progression-free survival (PFS) and overall survival (OS) were calculated using the Kaplan-Meier method. Statistical analyses were performed with R for Windows v4.2.0 software (R Core Team, Vienna, Austria; http://www.Rproject.org).

RESULTS

1. Demographic and Clinical Characteristics of Patients

As described in the Methods section, 32 patients diagnosed with MiTF/TFE translocation RCC between January 2004 and September 2021 were eligible for this study (Table 1). Their median age was 45.5 years (range, 20-67 years). Histologically, 26 patients (81.3%) had TFE3-rearranged RCC and only 1 patient (3.1%) had TFEB-altered RCC. The others (15.6%) were morphologically diagnosed with MiTF/TFE translocation RCC. Except for 5 patients (15.6%) whose electronic medical records were not available, curative or cytoreductive nephrectomy was performed in all 27 patients (84.4%), and 4 patients (12.6%) were diagnosed with metastatic disease at the time of initial diagnosis. Nine patients (28.1%) were treated with systemic therapy with TKIs, 2 (6.3%) of whom received simultaneous TKI and ICI treatment.

2. Patients Treated With TKIs or ICIs

All 9 MiTF/TFE translocation RCC patients treated with TKIs had TFE3-translocation RCC, with 8 patients having positive IHC staining for TFE3. The baseline characteristics before TKI or ICI administration are shown in Table 2. Five patients were men and 4were women. The median patient age was 47 years. Among the 9 patients, 4 were IMDC intermediate-risk, 3 were favorable-risk, and the other 2 were poor-risk before TKI or ICI administration. Five patients had an Eastern Cooperative Oncology Group (ECOG) performance status of 0, and the other 4 patients had an ECOG performance status of 1. Eight patients had distant metastases before TKI or ICI administration, and only 1 patient exhibited a complete response (CR) after left radical nephrectomy with lymph node dissection. This patient underwent adjuvant TKI treatment for more than 10 regional lymph node metastases in surgical specimens, which indicated a high risk of relapse. Regarding metastasis sites before TKI or ICI administration, 4 patients had distant lymph node metastasis to the mediastinal or supraclavicular lymph nodes. There were also liver, lung, bone, soft tissue, and peritoneal seeding metastases. All 9 patients underwent nephrectomy with curative or cytoreductive intent. Among them, 7 underwent curative or cytoreductive nephrectomy, radiofrequency ablation, or metastasectomy before TKI administration. The other 2 underwent interim cytoreductive nephrectomy while receiving TKIs in combination with ICIs because of partial response (PR) to treatment.

3. Efficacy of TKIs and ICIs

Treatment with TKIs and ICIs and the resulting clinical outcomes are shown in Table 3. Among 9 patients, 6 were treated with sunitinib, 2 with axitinib and pembrolizumab, and the remaining patient received pazopanib as the first TKI exposure. Eight patients were treated with systemic therapy in a palliative setting, and 1 patient was treated with a TKI as adjuvant therapy. Seven received their first TKI or ICI exposure as first-line therapy, while the other 2 patients received their first TKI exposure as second-line treatment after an mTOR inhibitor or bevacizumab with interferon-α. The response to systemic therapy (TKI or ICI) and the duration of response were variable, ranging from CR to progressive disease (Fig. 2). Five of the 9 patients achieved an overall response to the TKI or ICI to which they were first exposed, with 4 exhibiting CR and 1 showing PR. Excluding 1 patient who was treated with TKI in the adjuvant setting, the overall response rate in 8 metastatic patients was 50% and the CR rate was 37.5%. During the median follow-up of 29 months, the median PFS was 21 months, the median OS was not reached, and 2 deaths occurred (Table 4, Fig. 3). The median PFS was not reached and 5 months for the CR group (3 patients) and non-CR group (5 patients), respectively (p=0.041). The median OS was not reached and 29 months for the CR group and non-CR group, respectively (p=0.32).

4. Tumor Mutational Profiles With Targeted Sequencing

Tumor samples from 6 of the 9 patients were available for targeted sequencing analysis with TruSight Oncology 500 (Illumina). The top 10 mutated genes among the 221 genes were FAT1, FANCA, SPTA1, ANKRD26, GEN1, NUTM1, ALK, FGFR4, MSH3, and EML4 (Fig. 4A). Each of these mutations existed in all 6 samples. While missense mutations were most common, there were also in-frame deletions and insertions, nonsense mutations, frameshift deletions and insertions, and splice site mutations (Fig. 4B).

DISCUSSION

Until recently, MiTF/TFE translocation RCC was defined as kidney cancers harboring gene fusions involving members of the MiT family of transcription factors, including TFE3 and TFEB. Subsequently, the latest WHO classification (2022) separated TFE3-rearranged RCC and TFEB-altered RCC as 2 distinct molecularly-defined entities. These entities may overlap with each other and with other RCC subtype morphologies [22]. These overlapping morphological features may lead to misdiagnosis if specific IHC analyses are missing.
TFE3-rearranged RCC comprises 20%-75% of RCCs in children and 1%-4% of adult RCCs, with a median age of onset of 33 years [23]. As explained above, the incidence of TFE3-rearranged RCC in adults may be underestimated due to their morphological overlap with more common RCC subtypes such as papillary and clear cell carcinoma. Similar to other RCC subtypes, one-third of all TFE3-rearranged RCC patients are asymptomatic and often incidentally diagnosed. TFEB-altered RCC is a much rarer subtype, accounting for only 0.02% of all kidney tumors and comprising 6p21.1 translocated RCC and 6p21.1 amplified RCC [23]. The t(6;11) translocation fuses the gene for TFEB, located on chromosome 6, resulting in overexpression of TFEB. As a TFE3-rearranged tumor, TFEB-altered RCC does not have distinctive microscopic findings. Clinically, most cases with TFE3-rearranged and TFEB-altered RCCs are found incidentally, with median PFS and OS of 72 and 198 months, respectively [24]. Retrospective studies have shown that age and T stage at presentation and the presence of metastases were associated with aggressive behavior [24,25].
In metastatic settings, although no consensus exists regarding the optimal systemic therapy for TFE3-rearranged/TFEB-altered RCC, clinicians often extrapolate from treatment guidelines for clear cell RCC. The juvenile RCC network reported a series of 11 patients treated with sunitinib in the first-line setting, with a median PFS of 8.2 months [17]. Choueiri et al. [16] reported another series of 15 adult patients with metastatic Xp11 translocation RCC who received sunitinib, with 3 responders (20%) and a median PFS of 7.1 months. Subsequently, several attempts have been made to explore the efficacy of systemic therapies in patients with non-clear cell disease [26-28]. In these prospective studies, although sunitinib appeared to have clinically meaningful activity in non-clear cell RCC, each study included a diverse mix of histologic subtypes and no information was available on the clinical benefits for each distinct subtype. Although our small sample size poses a limitation for evaluating the efficacy of firstexposure systemic therapy of TKI and ICI, the median PFS in 9 patients with metastatic TFE3-rearranged RCC was 21 months.
There is no consensus regarding predictive factors for choosing the best systemic therapy for an individual patient. Interestingly, activation of the NRF2 pathway, which has recently been identified as a hallmark of TFE3-rearranged RCC, was previously shown to be associated with resistance to VEGFR-targeted TKIs [29]. Moreover, MiTF/TFE translocation RCCs are known to harbor strong expression of MET. Cabozantinib, a TKI with activity against VEGFR-2 and MET, has been evaluated in patients with clear cell RCC and, more recently, in patients with MiTF/TFE translocation RCC [30-32]. In 2 retrospective studies of patients with TFE3-rearranged/TFEB-altered RCC treated with cabozantinib, the promising disease control rates of 63%-82% suggest a therapeutic role for this TKI [31,32]. Prospective and larger studies are warranted to confirm these results. In addition to MET, PD-L1 expression by tumor cells and tumor-infiltrating mononuclear cells was reported in 30% and 90% of TFE3-rearranged/TFEB-altered RCC cases, respectively, suggesting that ICIs may be beneficial in these populations [33]. In addition, programmed cell death ligand 1 (PD-L1) expression in MiTF/TFE translocation RCC has been reported to be associated with a poor prognosis [34,35]. Boilève et al. [15] reported the efficacy of ICIs in 24 MiTF/TFE translocation RCC patients, with a response rate of 17% and median PFS of 2.5 months. They also reported that mutations in bromodomain-containing genes (PBRM1 and BRD8) might be associated with clinical benefit for ICIs, consistent with a previous report on clear cell RCC [36]. In parallel with advancement of the standard of care for metastatic clear cell RCC, several combinations involving ICIs and TKIs are being investigated in non-clear cell RCC, including TFE3-rearranged/TFEB-altered RCC [3,4].
The small number of patients and retrospective nature are limitations of the present study. Four of 9 patients were diagnosed less than 2 years before the data search, which indicates that MiTF/TFE translocation RCC was diagnosed recently. This might have been due to greater clinical alertness to this rare disease, and vigorous suspicion might have led to a larger number of diagnoses. In addition, all 9 patients were diagnosed with TFE3-rearranged RCC rather than TFEB-altered RCC, and FISH was not performed routinely for diagnosis. This might be due to the much lower prevalence of TFEB-altered RCC than TFE3-rearranged RCC or the frequency of misdiagnosis of TFEB-altered RCC as conventional RCC or TFE3-rearranged RCC. In addition to IHC for several markers, performing FISH for TFE3 or TFEB gene rearrangements or gene sequencing for identifying gene amplification can help differentiate diagnoses. In addition, TKI treatment was not uniform among patients, although 6 of 9 patients were treated with sunitinib. The absence of normal samples for comparison is another limitation of our analysis of mutational profiles. Whole genome sequencing or whole exome sequencing might give more information. Moreover, the small number of tumor samples analyzed was insufficient to draw solid conclusions.

CONCLUSION

Despite these limitations, we demonstrated the efficacy of TKI or ICI treatment for MiTF/TFE translocation RCC, a rare disease. With the further development of novel TKIs in combination with ICIs in other RCCs—mainly the clear cell type—these treatments could be expanded to MiTF/TFE translocation RCC. Further studies evaluating the efficacy of this therapeutic strategy and identifying predictive markers for treatment are warranted.

NOTES

Funding/Support

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Research Ethics

This study was performed in accordance with the Declaration of Helsinki and was approved by the Institutional Review Board of Samsung Medical Center (approval no. 2021-11-088-001; November 22, 2021). While consent to participate was waived by Institutional Review Board of Samsung Medical Center due to its retrospective nature, consent to perform next-generation sequencing was performed.

Conflicts of Interest

The authors have nothing to disclose.

Author Contribution

Conceptualization: JHH, SHP; Data curation: JHH, GYK, MYK, SIS, SHP; Formal analysis: JHH; Methodology: JHH, SHP; Project administration: SHP; Visualization: JHH; Writing - original draft: JHH; Writing - review & editing: JHH, GYK, MYK, SIS, SHP.

Fig. 1.
Flowchart of patient inclusion. TFE3, transcription factor E3; TFEB, transcription factor EB.
juo-234600660033f1.jpg
Fig. 2.
Duration of clinical benefits. CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease.
juo-234600660033f2.jpg
Fig. 3.
Kaplan-Meier curves for progression-free survival (PFS) (A) and overall survival (OS) (B).
juo-234600660033f3.jpg
Fig. 4.
Tumor mutational profiles based on targeted sequencing. The top 10 mutated genes (A) and variant classification (B).
juo-234600660033f4.jpg
Table 1.
Baseline characteristics of 32 patients with MiTF/TFE translocation renal cell carcinoma
Variable Value
Age (yr) 45.5 (20-67)
Sex
 Male 13 (40.6)
 Female 19 (59.4)
Stage at diagnosis
 Stage 1 15 (46.9)
 Stage 2 3 (9.4)
 Stage 3 5 (15.6)
 Stage 4 4 (12.5)
Not available 5 (15.6)
Subtypes
 TFE3 26 (81.3)
 TFEB 1 (3.1)
 Not available 5 (15.6)
Surgical treatment
 Curative nephrectomy 22 (68.8)
 Cytoreductive nephrectomy 5 (15.6)
 Not available 5 (15.6)
Systemic treatment
 Tyrosine kinase inhibitors 9 (28.1)
 Immune checkpoint inhibitors 2 (6.3)
 Not performed 18 (56.3)
 Not available 5 (15.6)

Values are presented as median (range) or number (%).

MiTF/TFE, microphthalmia transcription factor family/transcription factor E; TFE3, transcription factor E3; TFEB, transcription factor EB.

Table 2.
Histological and clinical characteristics* of 9 patients treated with systemic therapy (TKI or ICI)
Case No. Age (yr) Sex ECOG PS IMDC Risk IHC Treatments before systemic therapy (TKI or ICI) TNM stage, AJCC 8th
1 61 Female 0 Intermediate TFE3 Curative radical nephrectomy pT3aN1 (cM0)
2 51 Male 0 Intermediate TFE3 Cytoreductive nephrectomy pT3aN1 (cM1)
3 20 Male 1 Intermediate TFE3 Interim cytoreductive nephrectomy cT3aN0M1
4 55 Male 1 Poor TFE3 Interim cytoreductive nephrectomy cT1aN0M1
5 29 Female 0 Poor TFE3 Cytoreductive nephrectomy pT3a (cT3bN0M1)
6 40 Male 0 Favorable TFE3 Curative nephrectomy → metastasectomy → temsirolimus pT3aN0 (cM0)
7 60 Female 1 Favorable Morphologically diagnosed Curative nephrectomy → metastasectomy pT1a (cN0M0)
8 47 Female 1 Intermediate TFE3 Cytoreductive nephrectomy → bevacizumab+interferon-α pT1aM1 (cN0)
9 20 Male 0 Favorable TFE3 Curative nephrectomy → metastasectomy → RFA → RFA → metastasectomy pT1b (cN0M0)

TKI, tyrosine kinase inhibitor; ICI, immune checkpoint inhibitor; ECOG PS, Eastern Cooperative Oncology Group performance status; IMDC, International Metastatic RCC Database Consortium; IHC, immunohistochemistry; AJCC, American Joint Committee on Cancer; TFE3, transcription factor E3; RFA, radiofrequency ablation.

* Before TKI or ICI administration.

Initial stage.

Table 3.
Treatment and clinical outcomes of 9 patients treated with systemic therapy (TKI or ICI)
Case No. Age (yr) Sex TKI or ICI Lines of treatment Setting Best response Last response Subsequent treatment PFS (mo) OS (mo) Status
1 61 Female Pazopanib 1 Adjuvant CR PD RT → sorafenib → everolimus 18 33 Deceased
2 51 Male Sunitinib 1 Palliative SD PD Cabozantinib and RT 5 32 Alive
3 20 Male Axitinib/pembrolizumab 1 Palliative CR CR Nephrectomy 15 15 Alive
4 55 Male Axitinib/pembrolizumab 1 Palliative PR PR Nephrectomy 15 15 Alive
5 29 Female Sunitinib 1 Palliative SD PD Cabozantinib 5 8 Lost to follow-up
6 40 Male Sunitinib 2 Palliative CR CR Off 63 63 Alive
7 60 Female Sunitinib 1 Palliative CR CR Off 79 79 Alive
8 47 Female Sunitinib 2 Palliative SD PD Everolimus and RT 24 29 Deceased
9 20 Male Sunitinib 1 Palliative PD PD HD IL-2 → everolimus 2 14 Lost to follow-up

TKI, tyrosine kinase inhibitor; ICI, immune checkpoint inhibitor; PFS, progression-free survival; OS, overall survival; CR, complete response; PR, partial response; PD, progressive disease; RT, radiotherapy; SD, stable disease; HD IL-2, high-dose interleukin-2.

Table 4.
Clinical outcomes of systemic therapy (TKI or ICI)
Variable Value
Best response of first-exposed TKI or ICI
 CR 4 (44.4)
 PR 1 (11.1)
 SD 3 (33.3)
 PD 1 (11.1)
Overall response
 CR 3 (33.3)
 PR 1 (11.1)
 PD 5 (55.5)
Progression-free survival (mo) 21 (2-79)
Overall survival (mo), median Not reached

Values are presented as number (%) or median (range).

TKI, tyrosine kinase inhibitor; ICI, immune checkpoint inhibitor; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease.

REFERENCES

1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209-49.
crossref pmid pdf
2. Lopez-Beltran A, Scarpelli M, Montironi R, Kirkali Z. 2004 WHO classification of the renal tumors of the adults. Eur Urol 2006;49:798-805.
crossref pmid
3. Ljungberg B, Albiges L, Abu-Ghanem Y, Bedke J, Capitanio U, Dabestani S, et al. European Association of Urology guidelines on renal cell carcinoma: the 2022 update. Eur Urol 2022;82:399-410.
pmid
4. Motzer RJ, Jonasch E, Agarwal N, Alva A, Baine M, Beckermann K, et al. Kidney cancer, version 3.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2022;20:71-90.
pmid pmc
5. Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med 2018;378:1277-90.
pmid pmc
6. Powles T, Plimack ER, Soulières D, Waddell T, Stus V, Gafanov R, et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol 2020;21:1563-73.
crossref pmid
7. Moch H, Amin MB, Berney DM, Compérat EM, Gill AJ, Hartmann A, et al. The 2022 World Health Organization classification of tumours of the urinary system and male genital organs-part a: renal, penile, and testicular tumours. Eur Urol 2022;82:458-68.
crossref pmid
8. Sun G, Chen J, Liang J, Yin X, Zhang M, Yao J, et al. Integrated exome and RNA sequencing of TFE3-translocation renal cell carcinoma. Nat Commun 2021;12:5262.
crossref pmid pmc pdf
9. Wang XT, Xia QY, Ye SB, Wang X, Li R, Fang R, et al. RNA sequencing of Xp11 translocation-associated cancers reveals novel gene fusions and distinctive clinicopathologic correlations. Mod Pathol 2018;31:1346-60.
crossref pmid pdf
10. Davis IJ, Hsi BL, Arroyo JD, Vargas SO, Yeh YA, Motyckova G, et al. Cloning of an alpha-TFEB fusion in renal tumors harboring the t(6;11)(p21;q13) chromosome translocation. Proc Natl Acad Sci U S A 2003;100:6051-6.
crossref pmid pmc
11. Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO classification of tumours of the urinary system and male genital organs—Part A: renal, penile, and testicular tumours. Eur Urol 2016;70:93-105.
crossref pmid
12. Srigley JR, Delahunt B, Eble JN, Egevad L, Epstein JI, Grignon D, et al. The International Society of Urological Pathology (ISUP) Vancouver classification of renal neoplasia. Am J Surg Pathol 2013;37:1469-89.
crossref pmid
13. Ellati RT, Abukhiran I, Alqasem K, Jasser J, Khzouz J, Bisharat T, et al. Clinicopathologic features of translocation renal cell carcinoma. Clin Genitourin Cancer 2017;15:112-6.
crossref pmid
14. Malouf GG, Su X, Yao H, Gao J, Xiong L, He Q, et al. Nextgeneration sequencing of translocation renal cell carcinoma reveals novel RNA splicing partners and frequent mutations of chromatin-remodeling genes. Clin Cancer Res 2014;20:4129-40.
crossref pmid pmc pdf
15. Boilève A, Carlo MI, Barthélémy P, Oudard S, Borchiellini D, Voss MH, et al. Immune checkpoint inhibitors in MITF family translocation renal cell carcinomas and genetic correlates of exceptional responders. J Immunother Cancer 2018;6:159.
pmid pmc
16. Choueiri TK, Lim ZD, Hirsch MS, Tamboli P, Jonasch E, McDermott DF, et al. Vascular endothelial growth factortargeted therapy for the treatment of adult metastatic Xp11.2 translocation renal cell carcinoma. Cancer 2010;116:5219-25.
crossref pmid pmc
17. Malouf GG, Camparo P, Oudard S, Schleiermacher G, Theodore C, Rustine A, et al. Targeted agents in metastatic Xp11 translocation/TFE3 gene fusion renal cell carcinoma (RCC): a report from the Juvenile RCC Network. Ann Oncol 2010;21:1834-8.
crossref pmid
18. Parikh J, Coleman T, Messias N, Brown J. Temsirolimus in the treatment of renal cell carcinoma associated with Xp11.2 translocation/TFE gene fusion proteins: a case report and review of literature. Rare tumors 2009;1:e53.
crossref pmid pmc pdf
19. Rua Fernández OR, Escala Cornejo R, Navarro Martín M, García Muñoz M, Antunez Plaza P, García Dominguez AR, et al. Renal cell carcinoma associated with Xp11.2 translocation/TFE3 Gene-fusion: a long response to mammalian target of rapamycin (mTOR) Inhibitors. Urology 2018;117:41-3.
crossref pmid
20. Lee JY, Kim K, Sung HH, Jeon HG, Jeong BC, Seo SI, et al. Molecular characterization of urothelial carcinoma of the bladder and upper urinary tract. Transl Oncol 2018;11:37-42.
crossref pmid pmc
21. Pestinger V, Smith M, Sillo T, Findlay JM, Laes JF, Martin G, et al. Use of an integrated pan-cancer oncology enrichment NGS assay to measure tumour mutational burden and detect clinically actionable variants. Mol Diagn Ther 2020;24:339-49.
crossref pmid pmc pdf
22. Klatte T, Streubel B, Wrba F, Remzi M, Krammer B, de Martino M, et al. Renal cell carcinoma associated with transcription factor E3 expression and Xp11.2 translocation: incidence, characteristics, and prognosis. Am J Clin Pathol 2012;137:761-8.
pmid
23. Caliò A, Segala D, Munari E, Brunelli M, Martignoni G. MiT family translocation renal cell carcinoma: from the early descriptions to the current knowledge. Cancers (Basel) 2019;11:1110.
crossref pmid pmc
24. Wu Y, Chen S, Zhang M, Liu K, Jing J, Pan K, et al. Factors associated with survival from Xp11.2 translocation renal cell carcinoma diagnosis—a systematic review and pooled analysis. Pathol Oncol Res 2021;27:610360.
crossref pmid pmc
25. Caliò A, Brunelli M, Segala D, Pedron S, Remo A, Ammendola S, et al. Comprehensive analysis of 34 MiT family translocation renal cell carcinomas and review of the literature: investigating prognostic markers and therapy targets. Pathology 2020;52:297-309.
crossref pmid
26. Armstrong AJ, Halabi S, Eisen T, Broderick S, Stadler WM, Jones RJ, et al. Everolimus versus sunitinib for patients with metastatic non-clear cell renal cell carcinoma (ASPEN): a multicentre, open-label, randomised phase 2 trial. Lancet Oncol 2016;17:378-88.
crossref pmid pmc
27. Motzer RJ, Barrios CH, Kim TM, Falcon S, Cosgriff T, Harker WG, et al. Phase II randomized trial comparing sequential first-line everolimus and second-line sunitinib versus first-line sunitinib and second-line everolimus in patients with metastatic renal cell carcinoma. J Clin Oncol 2014;32:2765-72.
crossref pmid pmc
28. Tannir NM, Jonasch E, Albiges L, Altinmakas E, Ng CS, Matin SF, et al. Everolimus versus sunitinib prospective evaluation in metastatic non-clear cell renal cell carcinoma (ESPN): a randomized multicenter phase 2 trial. Eur Urol 2016;69:866-74.
crossref pmid pmc
29. Bakouny Z, Sadagopan A, Ravi P, Metaferia NY, Li J, Abu-Hammad S, et al. Integrative clinical and molecular characterization of translocation renal cell carcinoma. Cell Rep 2022;38:110190.
crossref pmid pmc
30. Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. Lancet Oncol 2016;17:917-27.
crossref pmid
31. Martínez Chanzá N, Xie W, Asim Bilen M, Dzimitrowicz H, Burkart J, Geynisman DM, et al. Cabozantinib in advanced non-clear-cell renal cell carcinoma: a multicentre, retrospective, cohort study. Lancet Oncol 2019;20:581-90.
pmid pmc
32. Thouvenin J, Alhalabi O, Carlo M, Carril-Ajuria L, Hirsch L, Martinez-Chanza N, et al. Efficacy of cabozantinib in metastatic MiT family translocation renal cell carcinomas. Oncologist 2022;27:1041-7.
crossref pmid pmc pdf
33. Choueiri TK, Fay AP, Gray KP, Callea M, Ho TH, Albiges L, et al. PD-L1 expression in nonclear-cell renal cell carcinoma. Ann Oncol 2014;25:2178-84.
crossref pmid pmc
34. Chang K, Qu Y, Dai B, Zhao JY, Gan H, Shi G, et al. PD-L1 expression in Xp11.2 translocation renal cell carcinoma: indicator of tumor aggressiveness. Scie Rep 2017;7:2074.
crossref pmid pmc pdf
35. Chipollini J, Azizi M, Peyton CC, Tang DH, Dhillon J, Spiess PE. Implications of programmed death ligand-1 positivity in non-clear cell renal cell carcinoma. J Kidney Cancer VHL 2018;5:6-13.
crossref pmid pmc pdf
36. Miao D, Margolis CA, Gao W, Voss MH, Li W, Martini DJ, et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 2018;359:801-6.
crossref pmid pmc


Editorial Office
Department of Urology, Seoul National University Hospital, Seoul National University College of Medicine,
101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
TEL: +82-2-2072-0817,   FAX: +82-2-742-4665   Email: journal@e-juo.org
Korean Urological Oncology Society
50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
Tel: +82-2-704-8574, E-mail: kspe.editor@gmail.com

Copyright © The Korean Urological Oncology Society.

Developed in M2PI