Abstract

Background

Metastatic RCC with sarcomatoid and/or rhabdoid (S/R) dedifferentiation is an aggressive disease associated with improved response to immune checkpoint therapy (ICT). The outcomes of patients treated with VEGFR-targeted therapies (TT) following ICT progression have not been investigated.

Patients and Methods

Retrospective review of 57 patients with sarcomatoid (S), rhabdoid (R), or sarcomatoid plus rhabdoid (S + R) dedifferentiation who received any TT after progression on ICT at an academic cancer center. Clinical endpoints of interest included time on TT, overall survival (OS) from initiation of TT, and objective response rate (ORR) by RECIST version 1.1. Multivariable models adjusted for epithelial histology, IMDC risk, prior VEGFR TT, and inclusion of cabozantinib in the post-ICT TT regimen.

Results

29/57 patients had S dedifferentiation and 19 had R dedifferentiation. The most frequently used TT was cabozantinib (43.9%) followed by selective VEGFR TT (22.8%). The median time on TT was 6.4 months for all, 6.1 months for those with S dedifferentiation, 15.6 months for R dedifferentiation, and 6.1 months for S + R dedifferentiation. Median OS from initiation of TT was 24.9 months for the entire cohort, and the ORR was 20.0%. Patients with R dedifferentiation had significantly longer time on TT than those with S dedifferentiation (HR 0.44, 95% CI, 0.21-0.94). IMDC risk was associated with OS.

Conclusions

A subset of patients with S/R dedifferentiation derive clinical benefit from TT after they have progressive disease on ICT. Patients with R dedifferentiation appeared to derive more benefit from TT than those with S dedifferentiation.

Implications for Practice

Targeted therapies (TT) only benefit a subset of patients with metastatic RCC with sarcomatoid and/or rhabdoid dedifferentiation whose cancer worsens on ICT. More specifically, those patients with rhabdoid dedifferentiation had significantly longer time on TT than those with sarcomatoid dedifferentiation

Introduction

The prognosis for patients diagnosed with metastatic renal cell carcinoma (RCC) is primarily driven by clinicopathologic factors.1 Clinical factors are currently summarized by the International Metastatic RCC Database Consortium (IMDC) risk score that categorizes patients into favorable, intermediate, or poor risk disease.2 Pathologic determinants of prognosis for metastatic RCC are primarily tumor histology and dedifferentiation status. Approximately, 75% of metastatic RCC cases are clear cell histology (ccRCC), where the clear appearance is driven by accumulation of lipid droplets.3 Variant histologies of RCC include papillary RCC, chromophobe RCC, renal medullary carcinoma (RMC), other molecularly defined RCCs, and unclassified RCCs.4 Each of these histologic subtypes has distinct biologic hallmarks, and any histologic subtype may have dedifferentiation present to varying degrees.5 RCC may have 2 types of dedifferentiation present, sarcomatoid, and/or rhabdoid dedifferentiation.6 Dedifferentiation is also referred to as “differentiation” or “features” in the literature, and patients can have varying degrees of dedifferentiation present, ranging from 5%-100% of the tumor.6 Dedifferentiation is an uncommon event that carries a poor prognosis, and either sarcomatoid and/or rhabdoid dedifferentiation is considered World Health Organization (WHO)/International Society of Urological Pathology (ISUP) grade 4.7 Sarcomatoid dedifferentiation consist of atypical spindle cells that resemble a sarcoma, and it is most commonly observed in ccRCC (5% of ccRCC cases) and chromophobe RCC (8% of chromophobe cases).8-10 The sarcomatoid component of an RCC is thought to arise from a common cell of origin within the epithelial component.11,12 Rhabdoid dedifferentiation is defined by the appearance of cancer cells that resemble rhabdomyoblasts and is thought to be present in ~5% of RCC cases, most commonly in ccRCC.13,14 Tumors also may have both sarcomatoid and rhabdoid dedifferentiation present.

The presence of dedifferentiation has therapeutic implications driven by its unique biology and aggressive clinical course. Historically, sarcomatoid dedifferentiation was associated with poor outcomes using systemic therapies developed for metastatic ccRCC. Between 2005 and 2015, VEGF and mTOR-targeted therapy (TT) were the primary treatments for metastatic RCC.15,16 These agents were investigated in patients with sarcomatoid dedifferentiation alone or in combination with chemotherapy, but clinical outcomes were disappointing with median time to progression of ~5 months and objective response rates (ORR) of ~20%.6,17-19 More recently, immune checkpoint therapy (ICT) meaningfully improved outcomes for patients with metastatic RCC and sarcomatoid dedifferentiation.

In a post hoc analysis of CheckMate-214, nivolumab plus ipilimumab produced an ORR of 61% with 19% of patients experiencing a complete response (CR) in those with metastatic clear cell RCC and sarcomatoid dedifferentiation, and this translated to an improvement in progression-free survival (PFS; HR 0.54, 95% CI, 0.33-0.86, median 26.5 vs. 5.1 months) and overall survival (OS; HR 0.45, 95% CI, 0.30-0.70, median NR vs. 14.2 months).20 First-line combination of ICT plus TT have also improved clinical endpoints when compared to VEGF TT alone.21-24 There is biologic rationale for the dramatic responses observed with ICT in patients who had sarcomatoid dedifferentiation because sarcomatoid dedifferentiation has increased expression of PD-L1 and an inflamed tumor microenvironment.25-29 Less is known about clinical outcomes with systemic therapy for rhabdoid dedifferentiation, but preliminary data suggests that ICT combinations also produce meaningful clinical benefit in rhabdoid dedifferentiation.28,30 Unfortunately, most RCCs with sarcomatoid or rhabdoid dedifferentiation who initially respond to ICT will ultimately develop resistance and require subsequent treatment options.20-23

As more patients with sarcomatoid and/or rhabdoid dedifferentiation experience clinical benefit using first-line ICT combinations, there is a growing need to understand treatment patterns and outcomes after progression on ICT. In this setting, patients with metastatic RCC are typically treated with multikinase TT agents, such as cabozantinib or lenvatinib plus the mTOR inhibitor, everolimus, or selective VEGFR TT. Herein, we report clinical outcomes of patients with metastatic RCC and sarcomatoid and/or rhabdoid dedifferentiation treated with TT after progression on ICT.

Methods

We retrospectively reviewed the records of patients with mRCC and sarcomatoid, rhabdoid, or sarcomatoid plus rhabdoid dedifferentiation who experienced progressive disease on an ICT-based treatment regimen and subsequently received a TT at the University of Texas MD Anderson Cancer Center (MDACC) between November 2015 and April 2021. Patients were identified by electronic medical record search for “sarcomatoid” or “rhabdoid” and “renal cell carcinoma,” and eligible patients had metastatic disease and progressive disease on an ICT-based regimen. In order to define presence of dedifferentiation, a patient’s pathology had to be reviewed by one of the genitourinary pathologists at MDACC for routine clinical care and reported as having dedifferentiation present. Patients with any degree of dedifferentiation were included in the cohort, and over the years included, our pathologists have variably used categorical presence (yes/no) and degree of dedifferentiation when reporting clinical pathology for dedifferentiation. Given the retrospective nature of this study, the samples were not rereviewed by independent, blinded pathologists. The type of TT after receipt of ICT was categorized as cabozantinib monotherapy, lenvatinib plus everolimus, selective VEGFR targeted therapy such as axitinib, pazopanib, or sunitinib, or ICT plus a TT such as cabozantinib, lenvatinib, or axitinib. The institutional review board approved this study. Clinical data were collected by individual chart review from the electronic medical record systemic. Demographic characteristics, histological subtype, International Metastatic RCC Database Consortium (IMDC) risk score at initiation of TT, sites of disease, and prior systemic therapies were recorded.

The clinical endpoints of interest were time on TT and overall survival (OS) from initiation of TT. Time on TT was calculated as the time from TT initiation until discontinuation for any reason, and patients who remained on TT at the time of analysis were censored using the date of last follow up. OS was calculated as the time from TT initiation until death or last follow up, if a patient was still living at the time of data collection. Two board-certified radiologists (D.S.S. and T.K.B.), blinded to clinical data, assessed radiographic tumor response using RECIST version 1.1.31 For each patient, one of these radiologist performed RECIST measures, so they were not confirmed by the other radiologist. Patients without baseline scans or without measurable lesions at baseline were considered not evaluable (NE). Directed acyclic graphs (DAGs) were used to avoid collider bias and identify potential confounders and mediators to be adjusted in multivariable Cox regression models when estimating the total and directed effects of exposures of interest.32 We prespecified type of dedifferentiation and IMDC risk score, as the 2 exposures of interest to assess their impact on OS and time on TT (Supplementary Fig. S1A-S1B). The multivariable analysis for type of dedifferentiation adjusted for histology as a confounding variable, and it did not adjust for mediators in Supplementary Fig. S1A. The multivariable analysis for IMDC risk score adjusted for histology and type of dedifferentiation as confounding variables, and it did not adjust for mediators in Supplementary Fig. S1B. Median survival times were calculated using the Kaplan-Meier method. Follow-up time was calculated based on the reverse Kaplan-Meier method. Hazard ratios (HR) and 95% confidence intervals (CI) were estimated using multivariable Cox regression models.

Results

Between November 2015 and April 2021, 57 patients initiated a TT after progressing on an ICT-based regimen. The median follow-up time was 49.9 months. Sarcomatoid was the most common type of dedifferentiation present (n = 29) followed by rhabdoid (n = 19) and then mixed sarcomatoid and rhabdoid dedifferentiation (n = 9). Histologic subtype by type of dedifferentiation is reported in Table 1. Some patients with sarcomatoid dedifferentiation had chromophobe (n = 2/29) or unclassified histology (n = 3/29), but every patient with rhabdoid or mixed sarcomatoid and rhabdoid dedifferentiation had clear cell histology. Prior to receipt of TT, 48 patients (84.2%) had previously underwent a nephrectomy, and 34 patients (59.6%) received metastasis-directed therapy. At initiation of TT, 54.4% of patients had 2 or 3 anatomic sites with metastases present, and most patients had IMDC intermediate (66.7%) or poor risk (28.1%) disease. Prior to receipt of TT, the most common ICT-based regimen was nivolumab plus ipilimumab (42.1%) followed by ICT monotherapy (31.6%) and ICT plus VEGF TT (21.1%). Most patients (93%) discontinued ICT due to progressive disease, but 4 patients discontinued ICT due to immune-related adverse effects. The ORR with an ICT-based regimen was 36.4% (20/55) and an additional 25.5% (14/55) had stable disease as best response to ICT-based therapy. Median time on ICT-based regimen was 5.7 months (interquartile range 3.5-10.4 months).

Table 1.

Baseline characteristics and treatment information for all patients and by type of dedifferentiation.

Metastatic sarcomatoid RCC
(n = 29)
Metastatic rhabdoid RCC
(n = 19)
Metastatic S + R RCC
(n = 9)
Full cohort
(n = 57)
Histology
 Clear cell24 (82.8%)19 (100%)9 (100%)52 (91.2%)
 Chromophobe2 (6.9%)0 (0%)0 (0%)2 (3.5%)
 Unclassified3 (10.3%)0 (0%)0 (0%)3 (5.3%)
# metastases at ICT
 11 (3.4%)4 (21.1%)2 (22.2%)7 (12.3%)
 2-314 (48.3%)12 (63.1%)5 (55.6%)31 (54.4%)
 ≥ 414 (48.3%)3 (15.8%)1 (11.1%)18 (31.6%)
 N/A0 (0%)0 (0%)1 (11.1%)1 (1.8%)
Prior nephrectomy
 Yes23 (79.3%)16 (84.2%)9 (100%)48 (84.2%)
 No6 (20.7%)3 (15.8%)0 (0%)9 (15.8%)
Metastasis-directed therapy
 Yes22 (75.9%)10 (52.6%)2 (22.2%)34 (59.6%)
 No7 (24.1%)9 (47.4%)7 (77.8%)23 (40.4%)
Prior VEGF TT
 Yes14 (48.3%)8 (42.1%)4 (44.4%)26 (45.6%)
 No15 (51.7%)11 (57.9%)5 (55.6%)31 (54.4%)
Type of prior ICT
 Nivo + ipi12 (41.4%)9 (47.4%)3 (33.3%)24 (42.1%)
 ICT monotherapy12 (41.4%)3 (15.8%)3 (33.3%)18 (31.6%)
 ICT + VEGF TT4 (13.8%)6 (31.6%)2 (22.2%)12 (21.1%)
 ICT + other1 (3.4%)1 (5.2%)1 (1.1%)3 (5.2%)
Reason for ICT d/c
 Progressive disease26 (89.7%)18 (94.7%)9 (100%)53 (93.0%)
 Immune toxicity3 (10.3%)1 (5.3%)0 (0%)4 (7.0%)
IMDC risk score at TT
 Favorable1 (3.4%)0 (0%)1 (11.1%)2 (3.5%)
 Intermediate20 (69.0%)13 (68.4%)5 (55.6%)38 (66.7%)
 Poor8 (27.6%)6 (31.6%)2 (22.2%)16 (28.1%)
 N/A0 (0%)0 (0%)1 (11.1%)1 (1.1%)
Type of TT post-ICT
 Cabozantinib16 (55.2%)3 (15.8%)6 (66.7%)25 (43.9%)
 Other VEGFR TT7 (24.1%)5 (26.3%)1 (11.1%)13 (22.8%)
 Len + eve2 (6.9%)4 (21.1%)1 (11.1%)7 (12.2%)
 ICT + TT4 (13.8%)7 (36.8%)1 (11.1%)12 (21.1%)
Metastatic sarcomatoid RCC
(n = 29)
Metastatic rhabdoid RCC
(n = 19)
Metastatic S + R RCC
(n = 9)
Full cohort
(n = 57)
Histology
 Clear cell24 (82.8%)19 (100%)9 (100%)52 (91.2%)
 Chromophobe2 (6.9%)0 (0%)0 (0%)2 (3.5%)
 Unclassified3 (10.3%)0 (0%)0 (0%)3 (5.3%)
# metastases at ICT
 11 (3.4%)4 (21.1%)2 (22.2%)7 (12.3%)
 2-314 (48.3%)12 (63.1%)5 (55.6%)31 (54.4%)
 ≥ 414 (48.3%)3 (15.8%)1 (11.1%)18 (31.6%)
 N/A0 (0%)0 (0%)1 (11.1%)1 (1.8%)
Prior nephrectomy
 Yes23 (79.3%)16 (84.2%)9 (100%)48 (84.2%)
 No6 (20.7%)3 (15.8%)0 (0%)9 (15.8%)
Metastasis-directed therapy
 Yes22 (75.9%)10 (52.6%)2 (22.2%)34 (59.6%)
 No7 (24.1%)9 (47.4%)7 (77.8%)23 (40.4%)
Prior VEGF TT
 Yes14 (48.3%)8 (42.1%)4 (44.4%)26 (45.6%)
 No15 (51.7%)11 (57.9%)5 (55.6%)31 (54.4%)
Type of prior ICT
 Nivo + ipi12 (41.4%)9 (47.4%)3 (33.3%)24 (42.1%)
 ICT monotherapy12 (41.4%)3 (15.8%)3 (33.3%)18 (31.6%)
 ICT + VEGF TT4 (13.8%)6 (31.6%)2 (22.2%)12 (21.1%)
 ICT + other1 (3.4%)1 (5.2%)1 (1.1%)3 (5.2%)
Reason for ICT d/c
 Progressive disease26 (89.7%)18 (94.7%)9 (100%)53 (93.0%)
 Immune toxicity3 (10.3%)1 (5.3%)0 (0%)4 (7.0%)
IMDC risk score at TT
 Favorable1 (3.4%)0 (0%)1 (11.1%)2 (3.5%)
 Intermediate20 (69.0%)13 (68.4%)5 (55.6%)38 (66.7%)
 Poor8 (27.6%)6 (31.6%)2 (22.2%)16 (28.1%)
 N/A0 (0%)0 (0%)1 (11.1%)1 (1.1%)
Type of TT post-ICT
 Cabozantinib16 (55.2%)3 (15.8%)6 (66.7%)25 (43.9%)
 Other VEGFR TT7 (24.1%)5 (26.3%)1 (11.1%)13 (22.8%)
 Len + eve2 (6.9%)4 (21.1%)1 (11.1%)7 (12.2%)
 ICT + TT4 (13.8%)7 (36.8%)1 (11.1%)12 (21.1%)

Abbreviations: RCC: renal cell carcinoma; S + R: sarcomatoid plus rhabdoid; #: number of; ICT: immune checkpoint therapy; N/A: not available; VEGF: vascular endothelial growth factor; TT: targeted therapy; nivo: nivolumab; ipi: ipilimumab; d/c: discontinuation; IMDC: International Metastatic RCC Database Consortium; VEGFR: vascular endothelial growth factor receptor; len: lenvatinib; eve: everolimus.

Table 1.

Baseline characteristics and treatment information for all patients and by type of dedifferentiation.

Metastatic sarcomatoid RCC
(n = 29)
Metastatic rhabdoid RCC
(n = 19)
Metastatic S + R RCC
(n = 9)
Full cohort
(n = 57)
Histology
 Clear cell24 (82.8%)19 (100%)9 (100%)52 (91.2%)
 Chromophobe2 (6.9%)0 (0%)0 (0%)2 (3.5%)
 Unclassified3 (10.3%)0 (0%)0 (0%)3 (5.3%)
# metastases at ICT
 11 (3.4%)4 (21.1%)2 (22.2%)7 (12.3%)
 2-314 (48.3%)12 (63.1%)5 (55.6%)31 (54.4%)
 ≥ 414 (48.3%)3 (15.8%)1 (11.1%)18 (31.6%)
 N/A0 (0%)0 (0%)1 (11.1%)1 (1.8%)
Prior nephrectomy
 Yes23 (79.3%)16 (84.2%)9 (100%)48 (84.2%)
 No6 (20.7%)3 (15.8%)0 (0%)9 (15.8%)
Metastasis-directed therapy
 Yes22 (75.9%)10 (52.6%)2 (22.2%)34 (59.6%)
 No7 (24.1%)9 (47.4%)7 (77.8%)23 (40.4%)
Prior VEGF TT
 Yes14 (48.3%)8 (42.1%)4 (44.4%)26 (45.6%)
 No15 (51.7%)11 (57.9%)5 (55.6%)31 (54.4%)
Type of prior ICT
 Nivo + ipi12 (41.4%)9 (47.4%)3 (33.3%)24 (42.1%)
 ICT monotherapy12 (41.4%)3 (15.8%)3 (33.3%)18 (31.6%)
 ICT + VEGF TT4 (13.8%)6 (31.6%)2 (22.2%)12 (21.1%)
 ICT + other1 (3.4%)1 (5.2%)1 (1.1%)3 (5.2%)
Reason for ICT d/c
 Progressive disease26 (89.7%)18 (94.7%)9 (100%)53 (93.0%)
 Immune toxicity3 (10.3%)1 (5.3%)0 (0%)4 (7.0%)
IMDC risk score at TT
 Favorable1 (3.4%)0 (0%)1 (11.1%)2 (3.5%)
 Intermediate20 (69.0%)13 (68.4%)5 (55.6%)38 (66.7%)
 Poor8 (27.6%)6 (31.6%)2 (22.2%)16 (28.1%)
 N/A0 (0%)0 (0%)1 (11.1%)1 (1.1%)
Type of TT post-ICT
 Cabozantinib16 (55.2%)3 (15.8%)6 (66.7%)25 (43.9%)
 Other VEGFR TT7 (24.1%)5 (26.3%)1 (11.1%)13 (22.8%)
 Len + eve2 (6.9%)4 (21.1%)1 (11.1%)7 (12.2%)
 ICT + TT4 (13.8%)7 (36.8%)1 (11.1%)12 (21.1%)
Metastatic sarcomatoid RCC
(n = 29)
Metastatic rhabdoid RCC
(n = 19)
Metastatic S + R RCC
(n = 9)
Full cohort
(n = 57)
Histology
 Clear cell24 (82.8%)19 (100%)9 (100%)52 (91.2%)
 Chromophobe2 (6.9%)0 (0%)0 (0%)2 (3.5%)
 Unclassified3 (10.3%)0 (0%)0 (0%)3 (5.3%)
# metastases at ICT
 11 (3.4%)4 (21.1%)2 (22.2%)7 (12.3%)
 2-314 (48.3%)12 (63.1%)5 (55.6%)31 (54.4%)
 ≥ 414 (48.3%)3 (15.8%)1 (11.1%)18 (31.6%)
 N/A0 (0%)0 (0%)1 (11.1%)1 (1.8%)
Prior nephrectomy
 Yes23 (79.3%)16 (84.2%)9 (100%)48 (84.2%)
 No6 (20.7%)3 (15.8%)0 (0%)9 (15.8%)
Metastasis-directed therapy
 Yes22 (75.9%)10 (52.6%)2 (22.2%)34 (59.6%)
 No7 (24.1%)9 (47.4%)7 (77.8%)23 (40.4%)
Prior VEGF TT
 Yes14 (48.3%)8 (42.1%)4 (44.4%)26 (45.6%)
 No15 (51.7%)11 (57.9%)5 (55.6%)31 (54.4%)
Type of prior ICT
 Nivo + ipi12 (41.4%)9 (47.4%)3 (33.3%)24 (42.1%)
 ICT monotherapy12 (41.4%)3 (15.8%)3 (33.3%)18 (31.6%)
 ICT + VEGF TT4 (13.8%)6 (31.6%)2 (22.2%)12 (21.1%)
 ICT + other1 (3.4%)1 (5.2%)1 (1.1%)3 (5.2%)
Reason for ICT d/c
 Progressive disease26 (89.7%)18 (94.7%)9 (100%)53 (93.0%)
 Immune toxicity3 (10.3%)1 (5.3%)0 (0%)4 (7.0%)
IMDC risk score at TT
 Favorable1 (3.4%)0 (0%)1 (11.1%)2 (3.5%)
 Intermediate20 (69.0%)13 (68.4%)5 (55.6%)38 (66.7%)
 Poor8 (27.6%)6 (31.6%)2 (22.2%)16 (28.1%)
 N/A0 (0%)0 (0%)1 (11.1%)1 (1.1%)
Type of TT post-ICT
 Cabozantinib16 (55.2%)3 (15.8%)6 (66.7%)25 (43.9%)
 Other VEGFR TT7 (24.1%)5 (26.3%)1 (11.1%)13 (22.8%)
 Len + eve2 (6.9%)4 (21.1%)1 (11.1%)7 (12.2%)
 ICT + TT4 (13.8%)7 (36.8%)1 (11.1%)12 (21.1%)

Abbreviations: RCC: renal cell carcinoma; S + R: sarcomatoid plus rhabdoid; #: number of; ICT: immune checkpoint therapy; N/A: not available; VEGF: vascular endothelial growth factor; TT: targeted therapy; nivo: nivolumab; ipi: ipilimumab; d/c: discontinuation; IMDC: International Metastatic RCC Database Consortium; VEGFR: vascular endothelial growth factor receptor; len: lenvatinib; eve: everolimus.

For type of TT after ICT, the most common treatment was cabozantinib monotherapy (43.9%) followed by selective VEGFR TT (22.8%), a TT plus ICT after progression on ICT (21.1%), and lenvatinib plus everolimus (12.2%). Selective VEGFR TT included axitinib (n = 10/13), pazopanib (n = 2/13), and sunitinib (n = 1/13). The TT plus ICT after progression cohort included the following treatments: cabozantinib plus nivolumab, lenvatinib plus pembrolizumab, and pembrolizumab plus axitinib. For the full cohort, the median time on TT was 6.4 months (95% CI, 4.4 to 8.5 months) and median OS from initiation of TT was 24.9 months (95% CI, 20.0 to 29.8 months). By RECIST version 1.1 criteria, ORR was noted in 9/45 patients (20.0%) with only one patient achieving a CR (Fig. 1). Stable disease was achieved in 26/45 patients (57.8%), whereas 10/45 patients (22.2%) experienced primary progressive disease. Due to lack of available imaging studies in this retrospective real-world study, 12/57 patients were NE by RECIST criteria. Since 26 patients had received TT either prior to ICT or in combination with ICT, we also evaluated ORR based on prior receipt of TT. In patients who received prior TT, the ORR was 14.3% (3/21), whereas, the ORR was 25.0% (6/24) in those who did not have prior exposure to TT.

Waterfall plot of confirmed overall response. Best % change = best radiographic response of target lesion per RECIST version 1.1, as measured by a board-certified radiologist specializing in genitourinary cancers. Color represents histology. Orange is clear cell, green is chromophobe, and blue is unclassified, Shading represents type of dedifferentiation. Black diagonal is sarcomatoid, no shading is rhabdoid, and blue perpendicular is sarcomatoid and rhabdoid dedifferentiation. # is cabozantinib, % is VEGFR TT, $ is lenvatinib plus everolimus, and ^ is targeted therapy (TT) plus immune checkpoint therapy (ICT).
Figure 1.

Waterfall plot of confirmed overall response. Best % change = best radiographic response of target lesion per RECIST version 1.1, as measured by a board-certified radiologist specializing in genitourinary cancers. Color represents histology. Orange is clear cell, green is chromophobe, and blue is unclassified, Shading represents type of dedifferentiation. Black diagonal is sarcomatoid, no shading is rhabdoid, and blue perpendicular is sarcomatoid and rhabdoid dedifferentiation. # is cabozantinib, % is VEGFR TT, $ is lenvatinib plus everolimus, and ^ is targeted therapy (TT) plus immune checkpoint therapy (ICT).

Little is known about treatment outcomes with metastatic ccRCC and rhabdoid dedifferentiation, and the morphological and biological distinctions led us to hypothesize that outcomes will differ by type of dedifferentiation. In multivariable analysis, patients with metastatic RCC and rhabdoid dedifferentiation had improved time on TT after progression on ICT when compared to sarcomatoid dedifferentiation [Supplementary Table S1, hazard ratio (HR) 0.44, 95% CI, 0.21-0.94, 15.6 vs. 6.1 months, Fig. 2]. Metastatic RCC with rhabdoid dedifferentiation was not significantly associated with improved OS from TT initiation compared with sarcomatoid dedifferentiation (Supplementary Table S1). In contrast, patients with sarcomatoid plus rhabdoid dedifferentiation did not have a significant difference in time on TT or OS when compared to sarcomatoid dedifferentiation (Supplementary Table S1, Fig. 2). In a multivariable analysis, IMDC risk score was associated with OS from TT initiation (HR 2.22, 95% CI, 1.07-4.61, Fig. 3) but not time on TT (HR 1.78, 95% CI, 0.88-3.60).

Adjusted survival curves of the direct effect of sarcomatoid and/or rhabdoid dedifferentiation on time on targeted therapy.
Figure 2.

Adjusted survival curves of the direct effect of sarcomatoid and/or rhabdoid dedifferentiation on time on targeted therapy.

Adjusted survival curves of the direct effect of International Metastatic RCC Database Consortium (IMDC) risk score on overall survival.
Figure 3.

Adjusted survival curves of the direct effect of International Metastatic RCC Database Consortium (IMDC) risk score on overall survival.

Ten patients in our cohort had clinical next generation sequencing performed (Fig. 4). For these patients, the most frequently altered genes were BAP1, TP53, and SETD2, whereas alterations were not observed in genes associated with favorable outcomes, such as PBRM1 or KDM5C. Patients with metastatic ccRCC and rhabdoid dedifferentiation had a high frequency of genomic alterations in BAP1 (n = 3/4) and SETD2 (n = 2/4). The 2 patients with metastatic chromophobe RCC and sarcomatoid dedifferentiation both had genomic alterations in TP53.

Oncoplot of somatic DNA alterations in tumor tissue from 10 patients with clinical next generation sequencing performed.
Figure 4.

Oncoplot of somatic DNA alterations in tumor tissue from 10 patients with clinical next generation sequencing performed.

Discussion

The development of ICT improved outcomes for patients with metastatic RCC and sarcomatoid dedifferentiation, since these therapies have the potential to produce durable clinical responses. However, little is known about the activity of contemporary TT agents after patients with metastatic RCC and sarcomatoid and/or rhabdoid dedifferentiation develop progressive disease on ICT. In our retrospective cohort of 57 patients, a small subset of patients with metastatic RCC and sarcomatoid and/or rhabdoid dedifferentiation derived clinical benefit from TT after progression on ICT with an ORR of 20.0% and median time on TT of only 6.4 months. Despite the small sample size, patients with metastatic RCC and rhabdoid dedifferentiation had improved time on TT after progression on ICT when compared to those with sarcomatoid dedifferentiation, which is notable because the influence of rhabdoid versus sarcomatoid dedifferentiation on systemic therapy outcomes is understudied. Furthermore, we found that the IMDC risk score remains a valuable prognostic tool in patients with sarcomatoid and/or rhabdoid dedifferentiation who have received prior ICT.

Prior to the introduction of ICT, TT was investigated in multiple studies for patients with metastatic RCC and sarcomatoid dedifferentiation. Three retrospective studies found that selective VEGFR TT have limited clinical activity for patients with metastatic RCC and sarcomatoid dedifferentiation.33-35 In a single center study of 43 patients, VEGFR TT produced an ORR of 19% and median PFS of 5.3 months.33 In a subsequent IMDC database analysis, VEGFR TT produced a median PFS of 4.5 months and OS of 10.4 months.34 Finally, an analysis of 199 patients by our group found that survival did not improve in the TT era when compared to patients with sarcomatoid dedifferentiation who received chemotherapy or cytokine therapy.35 Multiple clinical trials evaluated combining VEGFR TT with chemotherapy. One of these trials randomized patients with sarcomatoid dedifferentiation to sunitinib alone versus sunitinib plus gemcitabine, and the sunitinib alone arm produced an ORR of 11.1% and a median PFS of 3.3 months.19 Cabozantinib and lenvatinib are multikinase TT that encompass tyrosine kinases beyond the VEGFR, such as MET, TAM receptors, and FGFR. Given the benefit of these agents verse VEGFR TT in metastatic clear cell RCC, we hoped that clinical outcomes after ICT would outperform historical controls. However, the observed ORR of 20.0% is similar to the 10%-20% reported with VEGFR TT, and the waterfall plot in Fig. 1 shows responders received cabozantinib as well as VEGFR TT. This raises an important question of what should be given to patients with sarcomatoid dedifferentiation who progress on first-line TT plus ICT, and given our findings, nivolumab plus ipilimumab warrants further investigation in this setting. A retrospective, multicenter study reported outcomes with cabozantinib as second-line or third-line therapy after prior VEGFR TT (86%) or nivolumab plus ipilimumab (14%) for patients with metastatic RCC and sarcomatoid dedifferentiation.36 In that study, ORR was higher at 44%-47%, median PFS was similar at 7.6 months, and median OS was shorter at 9.1 months. Our study also included a group who received TT + ICT (21.1%) after progressive disease on prior ICT, and we did not see any distinct pattern of benefit in these patients compared to multi-kinase TT alone, which aligns with the recent findings from CONTACT-03.37

While interest in sarcomatoid dedifferentiation increased after introduction of ICT, less remains known about systemic therapy outcomes for patients with metastatic RCC and rhabdoid dedifferentiation.

Rhabdoid and sarcomatoid dedifferentiation are both considered WHO/ISUP grade 4 tumors, and across all stages, these dedifferentiated tumors have an inferior prognosis.7 Thus, some clinicians view sarcomatoid and rhabdoid dedifferentiation as similar entities once metastatic. In a multiomic study of sarcomatoid and/or rhabdoid dedifferentiation, both sarcomatoid and rhabdoid dedifferentiation were found to have improved responses to ICT.28 Yet, prior studies suggest that sarcomatoid dedifferentiation may harbor distinct mutations from rhabdoid dedifferentiation, and our analysis of somatic DNA alterations in 10 patients demonstrated differences in BAP1, SETD2, and TP53.6,38,39 We observed a notable difference in clinical outcomes with TT after ICT between patients with sarcomatoid versus rhabdoid dedifferentiation. Patients with rhabdoid dedifferentiation had improved time on TT than those with sarcomatoid dedifferentiation and demonstrated the best percent change by RECIST. These differences suggest that the underlying biological differences between rhabdoid and sarcomatoid dedifferentiation impact treatment outcomes and will require therapeutic regimes tailored to each dedifferentiation type. Furthermore, there is an unmet need to investigate and compare the efficacy of available systemic therapies, including ICT, in large cohorts of patients sarcomatoid, rhabdoid, or mixed sarcomatoid plus rhabdoid dedifferentiation. Our findings, coupled with the histologic and biologic distinctions, suggest that sarcomatoid and rhabdoid dedifferentiation should be investigated as distinct entities in future studies.

Our study is limited by its retrospective design, the small number of patients, and the unique practice patterns of a high volume, tertiary cancer center. While our study benefited from pathologists who have expertise in the diagnosis of RCC, dedifferentiation can be heterogeneous across a tumor and at risk for sampling errors.6 Our study included patients who received any TT received after progression on ICT, but the small sample size did not allow us to compare the magnitude of benefit between individual TT agents.

Conclusions

Only a subset of patients with metastatic RCC and sarcomatoid and/or rhabdoid dedifferentiation derive clinical benefit from TT after they progress on ICT. Patients with rhabdoid dedifferentiation may derive more benefit from TT than those with sarcomatoid dedifferentiation. The IMDC risk score remains a useful prognostic tool in this setting.

Supplementary Material

Supplementary material is available at The Oncologist online.

Funding

This work was supported in part by MD Anderson’s Prometheus informatics system. This study was supported in part by the Cancer Center Support Grant to MD Anderson Cancer Center (grant P30 CA016672) from the National Cancer Institute. AWH is supported by the Rob Heyvaert and Paul Heynen Prostate Cancer Foundation Young Investigator Award, an Early Investigator Research Award by the Department of Defense, and philanthropic donations from Michael and Patricia Berns. PM was supported by the Andrew Sabin Family Foundation Fellowship, Gateway for Cancer Research, a Translational Research Partnership Award (KC200096P1) by the United States Department of Defense, an Advanced Discovery Award by the Kidney Cancer Association, a Translational Research Award by the V Foundation, the MD Anderson Physician-Scientist Award, donations from the Renal Medullary Carcinoma Research Foundation in honor of Ryse Williams, as well as philanthropic donations by the Chris “CJ” Johnson Foundation, and by the family of Mike and Mary Allen

Conflict of Interest

Andrew W. Hahn reports advisory board consultation for Janssen, and Intellisphere, honoraria from Medscape, travel support from Dava Oncology, and institutional research funding from Eisai. Amado J. Zurita reports advisory board consultation for Amedco, CancerNet LLC, AstraZeneca, Incyte, Bayer, Pfizer, and Biocept; institutional research funding from Infinity Pharma, Pfizer, ABX, Merck, and Curium; and honoraria from Amedco, Janssen-Cliag, AstraZeneca, Mckesson Specialty Health, CancerNet LLC, and Pfizer. Amishi Y. Shah reports advisory board consultation for Exelixis and Pfizer; institutional research funds from BMS, Eisai, EMD Serono, and 4D Pharma. Nizar M. Tannir reports advisory consultation for BMS, Oncorena, Merck, Eli Lilly, Eisai Medical Research and Nektar Therapeutics; institutional research funding from BMS, Nektar Therapeutics, Arrowhead Pharmaceuticals; Novartis, Calithera Biosciences, and Exelixis; and honoraria from Eisai Medical Research, BMS, Intellisphere, Oncorena, Merck, Neoleukin, Exelixis, and AstraZeneca. Pavlos Msaouel has received honoraria for service on a Scientific Advisory Board for Mirati Therapeutics, Bristol Myers Squibb, and Exelixis; consulting for Axiom Healthcare Strategies; non-branded educational programs supported by Exelixis and Pfizer; and research funding for clinical trials from Takeda, Bristol Myers Squibb, Mirati Therapeutics, Gateway for Cancer Research, and UT MD Anderson Cancer Center. The other authors indicated no financial relationships.

Author Contributions

Conception and design: A.W.H., P.M.; Provision of study material or patients: M.T.C., A.J.Z., A.Y.S., E.J., J.G., S.G., O.A., P.R., K.S., N.M.T., P.M.; Collection/assembly of data: A.W.H., D.S.S., P.V.V., T.K.B., A.J.W.; Analysis and interpretation: A.W.H., P.M.; Manuscript writing: A.W.H., D.S.S., P.V.V., T.K.B., A.J.W., M.T.C., A.J.Z., A.Y.S., E.J., J.G., S.G., O.A., P.R., K.S., N.M.T., P.M.; Final approval of manuscript: All authors.

Data Availability

The identified data underlying this article cannot be shared publicly due to institutional compliance issues. The data will be shared on reasonable request to the corresponding author.

References

1.

Msaouel
P
,
Lee
J
,
Thall
PF.
Making patient-specific treatment decisions using prognostic variables and utilities of clinical outcomes
.
Cancers (Basel)
.
2021
;
13
(
11
):
2741
. https://doi.org/10.3390/cancers13112741

2.

Ernst
MS
,
Navani
V
,
Wells
JC
, et al. .
Outcomes for International Metastatic Renal Cell Carcinoma Database Consortium Prognostic Groups in contemporary first-line combination therapies for metastatic renal cell carcinoma
.
Eur Urol
.
2023
;
84
(
1
):
109
-
11
.

3.

Venkatesh
N
,
Martini
A
,
McQuade
JL
,
Msaouel
P
,
Hahn
AW.
Obesity and renal cell carcinoma: biological mechanisms and perspectives
.
Semin Cancer Biol
.
2023
;
94
:
21
-
33
. https://doi.org/10.1016/j.semcancer.2023.06.001

4.

Moch
H
,
Amin
MB
,
Berney
DM
, 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
(
5
):
458
-
468
. https://doi.org/10.1016/j.eururo.2022.06.016

5.

Zoumpourlis
P
,
Genovese
G
,
Tannir
NM
,
Msaouel
P.
Systemic therapies for the management of non-clear cell renal cell carcinoma: what works, what doesn’t, and what the future holds
.
Clin Genitourin Cancer
.
2021
;
19
(
2
):
103
-
116
. https://doi.org/10.1016/j.clgc.2020.11.005

6.

Hahn
AW
,
Lebenthal
J
,
Genovese
G
, et al. .
The significance of sarcomatoid and rhabdoid dedifferentiation in renal cell carcinoma
.
Cancer Treat Res Commun
.
2022
;
33
:
100640
. https://doi.org/10.1016/j.ctarc.2022.100640

7.

Delahunt
B
,
Cheville
JC
,
Martignoni
G
, et al. .
The International Society of Urological Pathology (ISUP) grading system for renal cell carcinoma and other prognostic parameters
.
Am J Surg Pathol
.
2013
;
37
(
10
):
1490
-
1504
. https://doi.org/10.1097/PAS.0b013e318299f0fb

8.

Reuter
VE.
Sarcomatoid lesions of the urogenital tract
.
Semin Diagn Pathol
.
1993
;
10
(
2
):
188
-
201
.

9.

Cheville
JC
,
Lohse
CM
,
Zincke
H
, et al. .
Sarcomatoid renal cell carcinoma: an examination of underlying histologic subtype and an analysis of associations with patient outcome
.
Am J Surg Pathol
.
2004
;
28
(
4
):
435
-
441
. https://doi.org/10.1097/00000478-200404000-00002

10.

Blum
KA
,
Gupta
S
,
Tickoo
SK
, et al. .
Sarcomatoid renal cell carcinoma: biology, natural history and management
.
Nat Rev Urol
.
2020
;
17
(
12
):
659
-
678
. https://doi.org/10.1038/s41585-020-00382-9

11.

Delahunt
B.
Sarcomatoid renal carcinoma: the final common dedifferentiation pathway of renal epithelial malignancies
.
Pathology (Phila)
.
1999
;
31
(
3
):
185
-
190
. https://doi.org/10.1080/003130299104945

12.

Conant
JL
,
Peng
Z
,
Evans
MF
,
Naud
S
,
Cooper
K.
Sarcomatoid renal cell carcinoma is an example of epithelial--mesenchymal transition
.
J Clin Pathol
.
2011
;
64
(
12
):
1088
-
1092
. https://doi.org/10.1136/jclinpath-2011-200216

13.

Gökden
N
,
Nappi
O
,
Swanson
PE
, et al. .
Renal cell carcinoma with rhabdoid features
.
Am J Surg Pathol
.
2000
;
24
(
10
):
1329
-
1338
. https://doi.org/10.1097/00000478-200010000-00002

14.

Shannon
B
,
Stan Wisniewski
Z
,
Bentel
J
,
Cohen
RJ.
Adult rhabdoid renal cell carcinoma
.
Arch Pathol Lab Med
.
2002
;
126
(
12
):
1506
-
1510
. https://doi.org/10.5858/2002-126-1506-ARRCC

15.

Choueiri
TK
,
Motzer
RJ.
Systemic therapy for metastatic renal-cell carcinoma
.
N Engl J Med
.
2017
;
376
(
4
):
354
-
366
. https://doi.org/10.1056/NEJMra1601333

16.

Adashek
JJ
,
Genovese
G
,
Tannir
NM
,
Msaouel
P.
Recent advancements in the treatment of metastatic clear cell renal cell carcinoma: a review of the evidence using second-generation p-values
.
Cancer Treat Res Commun
.
2020
;
23
:
100166
. https://doi.org/10.1016/j.ctarc.2020.100166

17.

Michaelson
MD
,
McKay
RR
,
Werner
L
, et al. .
Phase 2 trial of sunitinib and gemcitabine in patients with sarcomatoid and/or poor-risk metastatic renal cell carcinoma
.
Cancer
.
2015
;
121
(
19
):
3435
-
3443
. https://doi.org/10.1002/cncr.29503

18.

Maiti
A
,
Nemati-Shafaee
M
,
Msaouel
P
, et al. .
Phase 2 trial of capecitabine, gemcitabine, and bevacizumab in sarcomatoid renal-cell carcinoma
.
Clin Genitourin Cancer
.
2017
;
16
(
1
):
e47
-
e57
. https://doi.org/10.1016/j.clgc.2017.07.028

19.

Haas
NB
,
Puligandla
M
,
McDermott
DF
, et al. .
ECOG 1808: randomized phase II trial of sunitinib with or without gemcitabine in advanced kidney cancer with sarcomatoid features
.
J Clin Oncol
.
2016
;
34
(
15_suppl
):
4511
-
4511
. https://doi.org/10.1200/jco.2016.34.15_suppl.4511

20.

Tannir
NM
,
Signoretti
S
,
Choueiri
TK
, et al. .
Efficacy and safety of nivolumab plus ipilimumab versus sunitinib in first-line treatment of patients with advanced sarcomatoid renal cell carcinoma
.
Clin Cancer Res
.
2021
;
27
(
1
):
78
-
86
. https://doi.org/10.1158/1078-0432.CCR-20-2063

21.

Rini
BI
,
Plimack
ER
,
Stus
V
, et al. .
Pembrolizumab (pembro) plus axitinib (axi) versus sunitinib as first-line therapy for metastatic renal cell carcinoma (mRCC): outcomes in the combined IMDC intermediate/poor risk and sarcomatoid subgroups of the phase 3 KEYNOTE-426 study
.
J Clin Oncol
.
2019
;
37
(
15_suppl
):
4500
-
4500
. https://doi.org/10.1200/jco.2019.37.15_suppl.4500

22.

Choueiri
TK
,
Larkin
J
,
Pal
S
, et al. .
Efficacy and correlative analyses of avelumab plus axitinib versus sunitinib in sarcomatoid renal cell carcinoma: post hoc analysis of a randomized clinical trial
.
ESMO Open
.
2021
;
6
(
3
):
100101
. https://doi.org/10.1016/j.esmoop.2021.100101

23.

Motzer
RJ
,
Choueiri
TK
,
Powles
T
, et al. .
Nivolumab + cabozantinib (NIVO+CABO) versus sunitinib (SUN) for advanced renal cell carcinoma (aRCC): outcomes by sarcomatoid histology and updated trial results with extended follow-up of CheckMate 9ER
.
J Clin Oncol
.
2021
;
39
(
6_suppl
):
308
-
308
. https://doi.org/10.1200/jco.2021.39.6_suppl.308

24.

Choueiri
TK
,
Eto
M
,
Kopyltsov
E
, et al. .
Phase III CLEAR trial in advanced renal cell carcinoma (aRCC): outcomes in subgroups and toxicity update
.
Ann Oncol
.
2021
;
32
(
Suppl 5
):
S678
-
S724
.

25.

Joseph
RW
,
Millis
SZ
,
Carballido
EM
, et al. .
PD-1 and PD-L1 expression in renal cell carcinoma with sarcomatoid differentiation
.
Cancer Immunol Res
.
2015
;
3
(
12
):
1303
-
1307
. https://doi.org/10.1158/2326-6066.CIR-15-0150

26.

Kawakami
F
,
Sircar
K
,
Rodriguez-Canales
J
, et al. .
Programmed cell death ligand 1 and tumor-infiltrating lymphocyte status in patients with renal cell carcinoma and sarcomatoid dedifferentiation
.
Cancer
.
2017
;
123
(
24
):
4823
-
4831
. https://doi.org/10.1002/cncr.30937

27.

Gupta
S
,
Cheville
JC
,
Jungbluth
AA
, et al. .
JAK2/PD-L1/PD-L2 (9p241) amplifications in renal cell carcinomas with sarcomatoid transformation: implications for clinical management
.
Mod Pathol
.
2019
;
32
(
9
):
1344
-
1358
. https://doi.org/10.1038/s41379-019-0269-x

28.

Bakouny
Z
,
Braun
DA
,
Shukla
SA
, et al. .
Integrative molecular characterization of sarcomatoid and rhabdoid renal cell carcinoma
.
Nat Commun
.
2021
;
12
(
1
):
808
. https://doi.org/10.1038/s41467-021-21068-9

29.

Motzer
RJ
,
Banchereau
R
,
Hamidi
H
, et al. .
Molecular subsets in renal cancer determine outcome to checkpoint and angiogenesis blockade
.
Cancer Cell
.
2020
;
38
(
6
):
803
-
817.e4
. https://doi.org/10.1016/j.ccell.2020.10.011

30.

Chahoud
J
,
Msaouel
P
,
Campbell
MT
, et al. .
Nivolumab for the treatment of patients with metastatic non-clear cell renal cell carcinoma (nccRCC): a single-institutional experience and literature meta-analysis
.
Oncologist
.
2020
;
25
(
3
):
252
-
258
. https://doi.org/10.1634/theoncologist.2019-0372

31.

Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
, et al. .
New response evaluation criteria in solid tumours: revised RECIST guideline (version 11)
.
Eur J Cancer
.
2009
;
45
(
2
):
228
-
247
. https://doi.org/10.1016/j.ejca.2008.10.026

32.

Shapiro
DD
,
Msaouel
P.
Causal diagram techniques for urologic oncology research
.
Clin Genitourin Cancer
.
2021
;
19
(
3
):
271.e1
-
271.e7
. https://doi.org/10.1016/j.clgc.2020.08.003

33.

Golshayan
AR
,
George
S
,
Heng
DY
, et al. .
Metastatic sarcomatoid renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy
.
J Clin Oncol
.
2009
;
27
(
2
):
235
-
241
. https://doi.org/10.1200/JCO.2008.18.0000

34.

Kyriakopoulos
CE
,
Chittoria
N
,
Choueiri
TK
, et al. .
Outcome of patients with metastatic sarcomatoid renal cell carcinoma: results from the International Metastatic Renal Cell Carcinoma Database Consortium
.
Clin Genitourin Cancer
.
2015
;
13
(
2
):
e79
-
e85
. https://doi.org/10.1016/j.clgc.2014.08.011

35.

Keskin
SK
,
Msaouel
P
,
Hess
KR
, et al. .
Outcomes of patients with renal cell carcinoma and sarcomatoid dedifferentiation treated with nephrectomy and systemic therapies: comparison between the cytokine and targeted therapy eras
.
J Urol
.
2017
;
198
(
3
):
530
-
537
. https://doi.org/10.1016/j.juro.2017.04.067

36.

Santoni
M
,
Massari
F
,
Grande
E
, et al. .
Cabozantinib in pretreated patients with metastatic renal cell carcinoma with sarcomatoid differentiation: a real-world study
.
Target Oncol
.
2021
;
16
(
5
):
625
-
632
. https://doi.org/10.1007/s11523-021-00828-z

37.

Pal
SK
,
Albiges
L
,
Tomczak
P
, et al. .
Atezolizumab plus cabozantinib versus cabozantinib monotherapy for patients with renal cell carcinoma after progression with previous immune checkpoint inhibitor treatment (CONTACT-03): a multicentre, randomised, open-label, phase 3 trial
.
Lancet
.
2023
;
402
(
10397
):
185
-
195
. https://doi.org/10.1016/s0140-6736(23)00922-4

38.

Malouf
GG
,
Ali
SM
,
Wang
K
, et al. .
Genomic characterization of renal cell carcinoma with sarcomatoid dedifferentiation pinpoints recurrent genomic alterations
.
Eur Urol
.
2016
;
70
(
2
):
348
-
357
. https://doi.org/10.1016/j.eururo.2016.01.051

39.

Wang
Z
,
Kim
TB
,
Peng
B
, et al. .
Sarcomatoid renal cell carcinoma has a distinct molecular pathogenesis, driver mutation profile, and transcriptional landscape
.
Clin Cancer Res
.
2017
;
23
(
21
):
6686
-
6696
. https://doi.org/10.1158/1078-0432.CCR-17-1057

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.