scholarly journals Autotaxin–Lysophosphatidic Acid Signaling Axis Mediates Tumorigenesis and Development of Acquired Resistance to Sunitinib in Renal Cell Carcinoma

2013 ◽  
Vol 19 (23) ◽  
pp. 6461-6472 ◽  
Author(s):  
Shih-Chi Su ◽  
Xiaoxiao Hu ◽  
Patrick A. Kenney ◽  
Megan M. Merrill ◽  
Kara N. Babaian ◽  
...  
2014 ◽  
Vol 1 (1) ◽  
pp. 1-11 ◽  
Author(s):  
Christudas Morais

Of the many targeted therapies introduced since 2006, sunitinib has carved its way to become the most commonly used first-line therapy for the treatment of metastatic renal cell carcinoma (RCC). Despite significant improvements in progression-free survival, 30% of the patients are intrinsically resistant to sunitinib and the remaining 70% who respond initially will eventually become resistant in 6–15 months. While the molecular mechanisms of acquired resistance to sunitinib have been unravelling at a rapid rate, the mechanisms of intrinsic resistance remain elusive. Combination therapy, sunitinib-rechallenge and sequential therapy have been investigated as means to overcome resistance to sunitinib. Of these, sequential therapy appears to be the most promising strategy. This mini review summarises our emerging understanding of the molecular mechanisms, and the strategies employed to overcome sunitinib resistance.


Oncotarget ◽  
2017 ◽  
Vol 8 (60) ◽  
pp. 101042-101056 ◽  
Author(s):  
Feifei Chen ◽  
Xin Liu ◽  
Qian Cheng ◽  
Shudong Zhu ◽  
Jin Bai ◽  
...  

Cancers ◽  
2021 ◽  
Vol 13 (17) ◽  
pp. 4401
Author(s):  
María Armesto ◽  
Maitane Marquez ◽  
María Arestin ◽  
Peio Errarte ◽  
Ane Rubio ◽  
...  

The anti-angiogenic therapy sunitinib remains the standard first-line treatment for meta static clear cell renal cell carcinoma (ccRCC). However, acquired resistance develops in nearly all responsive patients and represents a major source of treatment failure. We used an integrated miRNA and mRNA transcriptomic approach to identify miRNA:target gene interactions involved in sunitinib resistance. Through the generation of stably resistant clones in three ccRCC cell lines (786-O, A498 and Caki-1), we identified non-overlapping miRNA:target gene networks, suggesting divergent mechanisms of sunitinib resistance. Surprisingly, even though the genes involved in these networks were different, they shared targeting by multiple members of the miR-17~92 cluster. In 786-O cells, targeted genes were related to hypoxia/angiogenic pathways, whereas, in Caki-1 cells, they were related to inflammatory/proliferation pathways. The immunotherapy target PD-L1 was consistently up-regulated in resistant cells, and we demonstrated that the silencing of this gene resulted in an increase in sensitivity to sunitinib treatment only in 786-O-resistant cells, suggesting that some ccRCC patients might benefit from combination therapy with PD-L1 checkpoint inhibitors. In summary, we demonstrate that, although there are clearly divergent mechanisms of sunitinib resistance in ccRCC subtypes, the commonality of miRNAs in multiple pathways could be targeted to overcome sunitinib resistance.


2015 ◽  
Vol 2 (1) ◽  
pp. 25-29 ◽  
Author(s):  
Christina Kaprilian ◽  
Maria Horti ◽  
Kosmas Kandilaris ◽  
Andreas Skolarikos ◽  
Nikolaos Trakas ◽  
...  

Multidrug resistance correlates with unfavourable treatment outcomes in numerous cancers including renal cell carcinoma. The expression and clinical relevance of Glutathione-S-transferase-pi (GST-pi), a multidrug resistance factor, in kidney tumors remain controversial. We analyzed the expression of GST-pi in 60 formalin-fixed, paraffin-embedded renal cell carcinoma samples by immunohistochemistry and compared them with matched normal regions of the kidney. A significantly higher expression of GST-pi was observed in 87% of clear cell carcinoma and 50% of papillary subtypes. GST-pi expression did not correlate with tumor grade or patient survival. GST-pi is unlikely to be a prognostic factor for renal cell carcinoma. However, further studies with large number of samples are warranted to establish the role of GST-pi, if any, in intrinsic or acquired resistance of renal cell carcinoma to conventional treatments. Supplementary files: The supplementary files of this article are found under 'Article Tools' at the left side bar.


Author(s):  
George K. Philips ◽  
Michael B. Atkins

The vascular endothelial growth factor (VEGF) pathway blockers and mammalian target of rapamycin (mTOR) inhibitors have dramatically improved the treatment options and outcome for patients with advanced renal cell carcinoma (RCC). However, because the vast majority of patients will still succumb to their disease, novel treatment approaches are still necessary. Efforts to identify novel therapeutic target treatments are focused on better understanding unique aspects of tumor cell biology guided the Cancer Genome Atlas analyses and the interaction of the tumor with its microenvironment. Areas of promising investigation include a) the identification of mechanisms of acquired resistance to VEGF pathway inhibition and developing agents targeting these in combination with VEGF receptor (VEGFR) pathway blockade; b) the identification of novel therapeutic targets, particularly for patients with VEGF pathway blocker refractory disease; and c) the development of novel immunotherapies, particularly those involving checkpoint inhibitors used alone or in combination with other immunotherapies of VEGF pathway blockers. Specific targets or agents of interest include angiopoietins (trebaninib), c-Met (cabozantinib), activin receptor-like kinase-1 (ALK-1; dalantercept), interleukin (IL)-8, and HDM2 for acquired resistance to VEGF pathway inhibition; hypoxia inducible factor-2 alpha (HIF-2 alpha), TORC1/2, and the Hippo pathway for novel targets, and PD1 and PDL1 antibodies given either alone or in combination with other checkpoint inhibitors, other immunotherapies, or VEGF pathway blockers for novel immunotherapies. In addition, the application of genetic, immunologic, or other biomarkers developed in the context of this research has the potential to select patients with specific tumor types for therapy targeted to specific vulnerabilities within the tumor or tumor microenvironment. Together, these developments should enable the transition to a new era of rational and more effective therapy for patients with advanced RCC.


Sign in / Sign up

Export Citation Format

Share Document