Next Article in Journal
The ‘Ins and Outs’ of Early Preclinical Models for Brain Tumor Research: Are They Valuable and Have We Been Doing It Wrong?
Next Article in Special Issue
Rising Serum Uric Acid Level Is Negatively Associated with Survival in Renal Cell Carcinoma
Previous Article in Journal
Follow-Up Liver Stiffness Measurements after Liver Resection Influence Oncologic Outcomes of Hepatitis-B-Associated Hepatocellular Carcinoma with Liver Cirrhosis
Previous Article in Special Issue
Ghrelin Upregulates Oncogenic Aurora A to Promote Renal Cell Carcinoma Invasion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Renal Cell Carcinoma with Sarcomatoid Features: Finally New Therapeutic Hope?

1
Department of Urology, Medical University Innsbruck, A-6020 Innsbruck, Austria
2
Department of Pathology, Hôspital Tenon, HUEP, Sorbonne University, 75005 Paris, France
3
Department of Urology, Royal Bournemouth Hospital, Bournemouth BH7 7DW, UK
4
Division of Clinical Oncology, Internal Medicine, Medical University of Graz, 8036 Graz, Austria
5
Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX 77030, USA
6
Department of Urology, St Vincent’s Hospital of Linz, 4010 Linz, Austria
7
Department of Urology & Andrology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
8
Clinical Division of Oncology, Department of Medicine I & Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
*
Author to whom correspondence should be addressed.
Cancers 2019, 11(3), 422; https://doi.org/10.3390/cancers11030422
Submission received: 7 March 2019 / Revised: 21 March 2019 / Accepted: 22 March 2019 / Published: 25 March 2019
(This article belongs to the Special Issue Renal Cell Carcinoma)

Abstract

:
Renal cell carcinoma (RCC) with sarcomatoid differentiation belongs to the most aggressive clinicopathologic phenotypes of RCC. It is characterized by a high propensity for primary metastasis and limited therapeutic options due to its relative resistance to established systemic targeted therapy. Most trials report on a poor median overall survival of 5 to 12 months. Sarcomatoid RCC can show the typical features of epithelial-mesenchymal transition (EMT) and may contain epithelial and mesenchymal features on both the morphological and immunhistochemical level. On the molecular level, next-generation sequencing confirmed differences in driver mutations between sarcomatoid RCC and non-sarcomatoid RCC. In contrast, mutational profiles within the epithelial and sarcomatoid components of sarcomatoid RCC were shown to be identical, with TP53 being the most frequently altered gene. These data suggest that both epithelial and sarcomatoid components of RCC originate from the same progenitor cell, segregating primarily according to the underlying histologic epithelial subtype of RCC (clear cell, papillary or chromophobe). Current studies have shown that sarcomatoid RCC express programmed death 1 (PD-1) and its ligand (PD-L1) at a much higher level than non-sarcomatoid RCC, suggesting that blockade of the PD-1/PD-L1 axis may be an attractive new therapeutic strategy. Preliminary results of clinical trials evaluating checkpoint inhibitors in patients with sarcomatoid RCC showed encouraging survival data and objective response and complete response rates of up to 62% and 18%, respectively. These findings may establish a new standard of care in the management of patients with sarcomatoid RCC.

1. Introduction

Renal cell carcinoma (RCC) with sarcomatoid differentiation (sRCC) is a highly aggressive form of RCC. Histologically, sRCC shows loss of characteristic epithelial components and contains features such as spindle cells, high cellularity, and cellular atypia. These features are found in 5–8% of clear-cell RCC (ccRCC), 8–9% of chromophobe RCC, and 2–3% of papillary RCC [1,2,3,4]. About 75% of patients with sRCC present with metastatic disease [5,6] and outcomes are generally modest. Therapeutic strategies include vascular endothelial growth factor (VEGF)-targeted monotherapy [7], and combined strategies with sunitinib plus gemcitabine [8] or gemcitabine plus doxorubicin [9]. The majority of studies report on a poor median overall survival, ranging from 5 to 12 months [7,8,9].

2. The sRCC

The sRCC is not a distinct morphogenetic subtype of RCC [3,10]. It originates from epithelial-mesenchymal transition (EMT), and therefore contains both epithelial (carcinoma) and mesenchymal (sarcomatoid) features on both the morphological and immunhistochemical level [11], which is distinctive from primary sarcoma of the kidney [3,10]. The presence of even a small component of sarcomatoid differentiation was shown to independently predict poor survival compared to RCC without sarcomatoid features; thus, its description needs to be included in the surgical pathology report [3]. To which extent this would be necessary in tumors showing only 1% of sarcomatoid features is questionable, since pathology reports are extremely dependent on gross sampling.
Genomic profiling on paired epithelial and sarcomatoid areas of sRCC by next-generation sequencing confirmed different driver mutations between sRCC and ccRCC. However, the epithelial and sarcomatoid components of sRCC showed identical mutational profiles, with TP53 (42%), VHL (35%), CDKN2A (27%), and NF2 (19%) being the most frequently altered genes [12]. These findings have been confirmed by Wang et al. [13]: The epithelioid and sarcomatoid components of sRCC did not show differences in mutational load amongst cancer-related genes, whereas sRCC had a completely different molecular pathogenesis and distinctive mutational and transcriptional profiles compared to ccRCC. Indeed, the authors found fewer deletions at 3p21-25, a lower rate of two-hit loss of VHL and PBRM1, but more mutations in TP53, PTEN, and RELN [13]. Moreover, mutations in known cancer drivers, such as AT-rich interaction domain 1A (ARID1A) and BRCA1 associated protein 1 (BAP1), were significantly mutated in sarcomatoid patterns and mutually exclusive with TP53 and each other [14]. These data corroborate the hypothesis that both epithelial and sarcomatoid components of RCC may originate from the same progenitor cell, but clonal divergence occurs during tumor progression. This implicates that specific genes are involved in this process, leading to unique genetic alterations based on the observed EMT [1,15].
Induction of EMT may upregulate the expression of PD-L1 and other targetable immune checkpoint molecules in various cancer entities, such as claudin-low breast cancer [16], non-small cell lung cancer [17], or RCC [18] in vivo and in vitro. Interestingly, sRCC has been shown to express programmed death 1 (PD-1) and its ligand (PD-L1) at a much higher level than RCC without sarcomatoid elements [19], as seen in Figure 1 and Figure 2. As higher tumoral PD-L1 expression seems to correlate with higher Fuhrmann grade [20], it is essential to compare the PD-L1 status between sRCC and grade 4 non-sarcomatoid ccRCC specifically [19]. Although sRCC is defined as grade 4 RCC, tumoral PD-L1 expression in the epithelioid component of sRCC was even higher than in non-sarcomatoid grade 4 ccRCC [19]. These results may suggest a biologic distinctiveness of sRCC compared to non-sarcomatoid ccRCC at the level of immune markers [19]. In this regard, tumoral PD-L1 and PD-1 expression was found in 54% and 96% of sRCC, compared to 17% and 62% of ccRCC specimens [21]. Moreover, the co-expression of both PD-L1 on tumor cells and PD-1 positive tumor-infiltrating lymphocytes was confirmed in 50% of all sRCC cases, compared to only one case (3%) with ccRCC [21]. These findings suggest that blockade of the PD-1/PD-L1 axis could be an attractive therapeutic approach in EMT-derived tumors, such as sRCC.
A small retrospective study by Ross et al., 2018, on response to checkpoint inhibitors in RCC patients with sarcomatoid differentiation presented as an abstract at American Society of Clinical Oncology (ASCO) Annual Meeting 2018 showed promising outcomes, with durable complete responses (CR) in up to 15% of patients, and an objective response rate (ORR) of 62% [22]. The genomic biomarker analyses of the phase III IMmotion151 study on bevacizumab plus atezolizumab versus sunitinib correlated angiogenesis and immune gene expression signatures with clinical outcomes from 832 RCC patients, focusing on sarcomatoid histology. Interestingly, the PD-L1 prevalence was higher in sRCC (63%), compared to non-sRCC (39%), whereas angiogenesis gene signature was lower in sRCC (34% vs. 65%) [23]. These results may explain why sRCC patients (n = 86) in the PD-L1+ study group showed the greatest therapeutic benefit with atezolizumab plus bevacizumab (progression-free survival: HR, 0.56 (95% CI: 0.38–0.83)) compared to sunitinib monotherapy [23,24]. In addition, the retrospective subgroup analysis of 112 sRCC intermediate or poor-risk patients from the CheckMate214 study [25] confirmed a higher rate of PD-L1 expression (≥1%) in sRCC than in non-sRCC (47–53% vs. 26–29%). More importantly, immunotherapy with nivolumab plus ipilimumab achieved an unprecedently high ORR of 57%, with a CR rate of 18% and a median overall Survival (OS) of 31 months compared to vascular endothelial growth factor (VEGF)-targeted monotherapy with sunitinib (ORR, CR, median OS: 19%, 0% and 14 months) [26].

3. Conclusions

In summary, sRCC shows the typical features of EMT. On the molecular level, transcriptional data confirmed that sRCC is not a homogeneous RCC subtype and segregates primarily according to the underlying parental epithelial subtype (ccRCC, papillary or chromophobe RCC). Current biomarker studies have shown that sRCC tumors express PD-1/PD-L1 at a much higher level than non-sarcomatoid RCC. These findings will ultimately lead to a change in the treatment paradigm, shifting therapeutic decisions towards checkpoint inhibitors as first line treatment for sRCC.

Author Contributions

R.P. conceived and designed the work and was responsible for writing the draft. E.C. was responsible for pathological figures and PD-L1 staining. T.K., M.P., W.L., L.L., and M.S. critically revised the manuscript and were responsible for supervision.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jones, T.D.; Eble, J.N.; Wang, M.; Maclennan, G.T.; Jain, S.; Cheng, L. Clonal divergence and genetic heterogeneity in clear cell renal cell carcinomas with sarcomatoid transformation. Cancer 2005, 104, 1195–1203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Cheville, J.C.; Lohse, C.M.; Zincke, H.; Weaver, A.L.; Leibovich, B.C.; Frank, I.; Blute, M.L. Sarcomatoid renal cell carcinoma: An examination of underlying histologic subtype and an analysis of associations with patient outcome. Am. J. Surg. Pathol. 2004, 28, 435–441. [Google Scholar] [CrossRef]
  3. De Peralta-Venturina, M.; Moch, H.; Amin, M.; Tamboli, P.; Hailemariam, S.; Mihatsch, M.; Javidan, J.; Stricker, H.; Ro, J.Y.; Amin, M.B. Sarcomatoid differentiation in renal cell carcinoma: A study of 101 cases. Am. J. Surg. Pathol. 2001, 25, 275–284. [Google Scholar] [CrossRef]
  4. Amin, M.B.; Paner, G.P.; Alvarado-Cabrero, I.; Young, A.N.; Stricker, H.J.; Lyles, R.H.; Moch, H. Chromophobe renal cell carcinoma: Histomorphologic characteristics and evaluation of conventional pathologic prognostic parameters in 145 cases. Am. J. Surg. Pathol. 2008, 32, 1822–1834. [Google Scholar] [CrossRef] [PubMed]
  5. Mian, B.M.; Bhadkamkar, N.; Slaton, J.W.; Pisters, P.W.; Daliani, D.; Swanson, D.A.; Pisters, L.L. Prognostic factors and survival of patients with sarcomatoid renal cell carcinoma. J. Urol. 2002, 167, 65–70. [Google Scholar] [CrossRef]
  6. Merrill, M.M.; Wood, C.G.; Tannir, N.M.; Slack, R.S.; Babaian, K.N.; Jonasch, E.; Pagliaro, L.C.; Compton, Z.; Tamboli, P.; Sircar, K.; et al. Clinically nonmetastatic renal cell carcinoma with sarcomatoid dedifferentiation: Natural history and outcomes after surgical resection with curative intent. Urol. Oncol. 2015, 33, 166.e21-9. [Google Scholar] [CrossRef] [PubMed]
  7. Golshayan, A.R.; George, S.; Heng, D.Y.; Elson, P.; Wood, L.S.; Mekhail, T.M.; Garcia, J.A.; Aydin, H.; Zhou, M.; Bukowski, R.M.; et al. Metastatic sarcomatoid renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy. J. Clin. Oncol. 2009, 27, 235–241. [Google Scholar] [CrossRef] [PubMed]
  8. Michaelson, M.D.; McKay, R.R.; Werner, L.; Atkins, M.B.; Van Allen, E.M.; Olivier, K.M.; Song, J.; Signoretti, S.; McDermott, D.F.; Choueiri, T.K. Phase 2 trial of sunitinib and gemcitabine in patients with sarcomatoid and/or poor-risk metastatic renal cell carcinoma. Cancer 2015, 121, 3435–3443. [Google Scholar] [CrossRef] [Green Version]
  9. Roubaud, G.; Gross-Goupil, M.; Wallerand, H.; de Clermont, H.; Dilhuydy, M.S.; Ravaud, A. Combination of gemcitabine and doxorubicin in rapidly progressive metastatic renal cell carcinoma and/or sarcomatoid renal cell carcinoma. Oncology 2011, 80, 214–218. [Google Scholar] [CrossRef]
  10. Delahunt, B.; Cheville, J.C.; Martignoni, G.; Humphrey, P.A.; Magi-Galluzzi, C.; McKenney, J.; Egevad, L.; Algaba, F.; Moch, H.; Grignon, D.J.; 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, 1490–1504. [Google Scholar] [CrossRef]
  11. Mikami, S.; Katsube, K.; Oya, M.; Ishida, M.; Kosaka, T.; Mizuno, R.; Mukai, M.; Okada, Y. Expression of Snail and Slug in renal cell carcinoma: E-cadherin repressor Snail is associated with cancer invasion and prognosis. Lab. Investig. 2011, 91, 1443–1458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Malouf, G.G.; Ali, S.M.; Wang, K.; Balasubramanian, S.; Ross, J.S.; Miller, V.A.; Stephens, P.J.; Khayat, D.; Pal, S.K.; Su, X.; et al. Genomic Characterization of Renal Cell Carcinoma with Sarcomatoid Dedifferentiation Pinpoints Recurrent Genomic Alterations. Eur. Urol. 2016, 70, 348–357. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, Z.; Kim, T.B.; Peng, B.; Karam, J.; Creighton, C.; Joon, A.; Kawakami, F.; Trevisan, P.; Jonasch, E.; Chow, C.W.; et al. Sarcomatoid Renal Cell Carcinoma Has a Distinct Molecular Pathogenesis, Driver Mutation Profile, and Transcriptional Landscape. Clin. Cancer Res. 2017, 23, 6686–6696. [Google Scholar] [CrossRef]
  14. Bi, M.; Zhao, S.; Said, J.W.; Merino, M.J.; Adeniran, A.J.; Xie, Z.; Nawaf, C.B.; Choi, J.; Belldegrun, A.S.; Pantuck, A.J.; et al. Genomic characterization of sarcomatoid transformation in clear cell renal cell carcinoma. Proc. Natl. Acad. Sci. USA 2016, 113, 2170–2175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Lebacle, C.; Pooli, A.; Bessede, T.; Irani, J.; Pantuck, A.J.; Drakaki, A. Epidemiology, biology and treatment of sarcomatoid RCC: Current state of the art. World J. Urol. 2019, 37, 115–123. [Google Scholar] [CrossRef] [PubMed]
  16. Alsuliman, A.; Colak, D.; Al-Harazi, O.; Fitwi, H.; Tulbah, A.; Al-Tweigeri, T.; Al-Alwan, M.; Ghebeh, H. Bidirectional crosstalk between PD-L1 expression and epithelial to mesenchymal transition: Significance in claudin-low breast cancer cells. Mol. Cancer 2015, 14, 149. [Google Scholar] [CrossRef]
  17. Lou, Y.; Diao, L.; Cuentas, E.R.; Denning, W.L.; Chen, L.; Fan, Y.H.; Byers, L.A.; Wang, J.; Papadimitrakopoulou, V.A.; Behrens, C.; et al. Epithelial-Mesenchymal Transition Is Associated with a Distinct Tumor Microenvironment Including Elevation of Inflammatory Signals and Multiple Immune Checkpoints in Lung Adenocarcinoma. Clin. Cancer Res. 2016, 22, 3630–3642. [Google Scholar] [CrossRef]
  18. Wang, Y.; Wang, H.; Zhao, Q.; Xia, Y.; Hu, X.; Guo, J. PD-L1 induces epithelial-to-mesenchymal transition via activating SREBP-1c in renal cell carcinoma. Med. Oncol. 2015, 32, 212. [Google Scholar] [CrossRef]
  19. Kawakami, F.; Sircar, K.; Rodriguez-Canales, J.; Fellman, B.M.; Urbauer, D.L.; Tamboli, P.; Tannir, N.M.; Jonasch, E.; Wistuba, I.I.; Wood, C.G.; et al. Programmed cell death ligand 1 and tumor-infiltrating lymphocyte status in patients with renal cell carcinoma and sarcomatoid dedifferentiation. Cancer 2017, 123, 4823–4831. [Google Scholar] [CrossRef] [PubMed]
  20. Thompson, R.H.; Dong, H.; Kwon, E.D. Implications of B7-H1 expression in clear cell carcinoma of the kidney for prognostication and therapy. Clin Cancer Res. 2007, 13, 709s–715s. [Google Scholar] [CrossRef] [PubMed]
  21. Joseph, R.W.; Millis, S.Z.; Carballido, E.M.; Bryant, D.; Gatalica, Z.; Reddy, S.; Bryce, A.H.; Vogelzang, N.J.; Stanton, M.L.; Castle, E.P.; et al. PD-1 and PD-L1 Expression in Renal Cell Carcinoma with Sarcomatoid Differentiation. Cancer Immunol. Res. 2015, 3, 1303–1307. [Google Scholar] [CrossRef] [PubMed]
  22. Ross, J.A.; McCormick, B.Z.; Gao, J.; Msaouel, P.; Campbell, M.T.; Zurita, A.J.; Shah, A.Y.; Jonasch, E.; Matin, S.F.; Wood, C.G.; et al. Outcomes of patients (pts) with metastatic renal cell carcinoma (mRCC) and sarcomatoid dedifferentiation (sRCC) after treatment with immune checkpoint inhibitors (ICI): A single-institution retrospective study. J Clin Oncol. 2018, 36. [Google Scholar] [CrossRef]
  23. Rini, B.I.; Huseni, M.; Atkins, M.B.; McDermott, M.F.; Powles, T.; Escudier, B.; Banchereau, R.; Liu, L.; Leng, N.; Fan, J.; et al. Molecular correlates differentiate response to atezolizumab (atezo) + bevacizumab (bev) vs sunitinib (sun): Results from a Phase III study (IMmotion151) in untreated metastatic renal cell carcinoma (mRCC). In Proceedings of the ESMO 2018, Munich, Germany, 19–23 October 2018. [Google Scholar]
  24. Motzer, R.J.; Powles, T.; Atkins, M.B.; Escudier, B.; McDermott, D.F.; Suarez, C.; Bracarda, S.; Stadler, W.M.; Donskov, F.; Lee, J.L.; et al. IMmotion151: A Randomized Phase III Study of Atezolizumab Plus Bevacizumab vs Sunitinib in Untreated Metastatic Renal Cell Carcinoma (mRCC). J. Clin. Oncol. 2018, 36. [Google Scholar] [CrossRef]
  25. Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Arén Frontera, O.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.; Barthélémy, P.; Porta, C.; George, S.; et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2018, 378, 1277–1290. [Google Scholar] [CrossRef] [PubMed]
  26. McDermott, D.F.; Motzer, R.J.; Rini, B.I.; Aren Frontera, O.; George, S.; Powles, T.; Donskov, F.; Harrison, M.; Rodriguez-Cid, J.; Ishii, Y.; et al. ChechMate214 retrospective analyses of nivolumab plus ipilimumab or sunitinib in IMDC intermediate/poor-risk patients with previously untreated advanced renal cell carcinoma with sarcomatoid features. In Proceedings of the Seventeenth International Kidney Cancer Symposium, Miami, FL, USA, 2–3 November 2018. [Google Scholar]
Figure 1. (A) Hematoxylin/eosin and phloxin staining for clear-cell RCC (ccRCC) with sarcomatoid features (50× magnification), considered as grade 4 according to the International Society of Urologic Pathologists (ISUP). (B) High PD-L1 (100× magnification) (E1L3N XP Rabbit mAB) cytoplasmatic staining of tumor cells in sRCC (>50% of tumor cells).
Figure 1. (A) Hematoxylin/eosin and phloxin staining for clear-cell RCC (ccRCC) with sarcomatoid features (50× magnification), considered as grade 4 according to the International Society of Urologic Pathologists (ISUP). (B) High PD-L1 (100× magnification) (E1L3N XP Rabbit mAB) cytoplasmatic staining of tumor cells in sRCC (>50% of tumor cells).
Cancers 11 00422 g001
Figure 2. (A) Hematoxylin/eosin and phloxin staining for ccRCC without sarcomatoid features (400× magnification). (B) PD-L1 (E1L3N XP Rabbit mAB) cytoplasmatic staining of tumor cells, showing low PD-L1 expression (<5%), 100× magnification.
Figure 2. (A) Hematoxylin/eosin and phloxin staining for ccRCC without sarcomatoid features (400× magnification). (B) PD-L1 (E1L3N XP Rabbit mAB) cytoplasmatic staining of tumor cells, showing low PD-L1 expression (<5%), 100× magnification.
Cancers 11 00422 g002

Share and Cite

MDPI and ACS Style

Pichler, R.; Compérat, E.; Klatte, T.; Pichler, M.; Loidl, W.; Lusuardi, L.; Schmidinger, M. Renal Cell Carcinoma with Sarcomatoid Features: Finally New Therapeutic Hope? Cancers 2019, 11, 422. https://doi.org/10.3390/cancers11030422

AMA Style

Pichler R, Compérat E, Klatte T, Pichler M, Loidl W, Lusuardi L, Schmidinger M. Renal Cell Carcinoma with Sarcomatoid Features: Finally New Therapeutic Hope? Cancers. 2019; 11(3):422. https://doi.org/10.3390/cancers11030422

Chicago/Turabian Style

Pichler, Renate, Eva Compérat, Tobias Klatte, Martin Pichler, Wolfgang Loidl, Lukas Lusuardi, and Manuela Schmidinger. 2019. "Renal Cell Carcinoma with Sarcomatoid Features: Finally New Therapeutic Hope?" Cancers 11, no. 3: 422. https://doi.org/10.3390/cancers11030422

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop