Next Article in Journal
Adjuvant and Neoadjuvant Small-Molecule Targeted Therapy in High-Risk Renal Cell Carcinoma
Previous Article in Journal
Adverse Events Associated with Anti-EGFR Therapies for the Treatment of Metastatic Colorectal Cancer
 
 
Current Oncology is published by MDPI from Volume 28 Issue 1 (2021). Previous articles were published by another publisher in Open Access under a CC-BY (or CC-BY-NC-ND) licence, and they are hosted by MDPI on mdpi.com as a courtesy and upon agreement with Multimed Inc..
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Targeted Therapies for Renal Cell Carcinoma: More Gains from Using Them Again

Princess Margaret Hospital, Department of Medical Oncology and Hematology, University of Toronto, 610 University Avenue, Toronto, ON M5G 2M9, Canada
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2009, 16(s1), 45-51; https://doi.org/10.3747/co.v16i0.404
Submission received: 1 July 2009 / Revised: 1 July 2009 / Accepted: 1 July 2009 / Published: 1 July 2009

Abstract

:
The development of molecularly targeted agents that inhibit pathways critical to the development of renal cell carcinoma has significantly improved outcomes in patients with these cancers. Compelling scientific and phase III data have made the use of molecularly targeted agents the standard of care in first-line treatment. Now, available data show that re-treating patients with other tyrosine kinase inhibitors after they progress on sunitinib or sorafenib, or both, is beneficial. A large phase III trial recently showed that, as compared with placebo, treatment with everolimus, an inhibitor of the mammalian target of rapamycin (mTOR), almost halved the risk of progression (37% vs. 65%) and doubled the median progression-free survival (4 months vs. 2 months). Overall survival was not improved in that study, likely reflecting treatment crossover in the placebo arm, but these data position everolimus as the second-line standard of care. A consistent and growing body of literature also suggests that re-treatment with other kinase inhibitors that the patient has not previously encountered is a reasonable option. Outcomes of initial treatment with sunitinib or sorafenib (or both) should not deter the use of second-line targeted therapy, because the first-line use of targeted agents does not appear to be predictive of outcomes with second-line therapy. However, in view of poor absolute outcomes after second-line treatment and the benefits seen with rationally developed targeted agents in the first-line setting, enrolment of secondand subsequent-line patients in further trials would be preferable.

1. Introduction

In 2005, renal cell carcinoma (RCC) was the ninth most common cancer in Canada, with an incidence of 1 per 10,000 population [1], and in that year, the disease was responsible for 2% of the total cost of cancer ($17 billion) [2]. Until recently, patients with metastatic disease had limited treatment options, and their 5-year survival was approximately 20% [3]. Moreover, tumours in RCC rarely responded to chemotherapy and radiotherapy [3]. Cytokine therapy (interferon alfa or interleukin 2) had modest efficacy, with a response rate of 12% and a median survival of 13 months in a recent meta-analysis [4]. However, since the development of molecularly targeted therapies (Table 1) that inhibit the pathways important in the pathogenesis of RCC (Figure 1), the outlook has improved.

2. Discussion

2.1. Epidemiology and Biology of RCC

Loss of the von Hippel–Lindau (VHL) tumour suppressor is a pivotal event in both hereditary and sporadic RCC (Figure 1) [3,11]. The resultant overactivity of the hypoxiainducible factor (HIF) pathway leads to overexpression of a set of hypoxia-related genes [for example, vascular endothelial growth factor, platelet-derived growth factor B (PDGFRB)] and activation of a number of pathways [for example, the phosphoinositide-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and ras signal transduction protein (Ras)/protein encoded by the murine leukemia viral oncogene homolog (Raf)/mitogen-activated protein kinase (Mek)/extracellular signal-regulated kinase (Erk) pathways] [12]. Signalling through mTOR is present in most clear-cell RCC [13,14]. In addition to effects on tumour proliferation, angiogenesis, and apoptosis, activation of mTOR may also potentiate the activity of HIF 12. Inhibition of mTOR results in tumour inhibition in vitro [13] and in vivo [15].
Interestingly, the epithelial growth factor recep-tor (EGFR) pathway is also important to renal oncogenesis. Expression of EGFR occurs at high frequency in RCC (70%–90%) [16,17,18], and loss of VHL causes increased expression of transforming growth factor α [19] through HIF-2α–dependent mechanisms [20,21,22]. Inhibition of this dysregulated autocrine loop is sufficient to reduce or abolish tumour growth in multiple VHL–/– RCC cell lines [20,21].

2.2. First-Line Treatment with a Targeted Agent

Treatment with a number of targeted agents (Table 1) is now accepted as the standard of care in patients with metastatic RCC. In untreated patients, sunitinib [23], temsirolimus [24], and bevacizumab (when combined with interferon alfa) [25] were all found to be superior to interferon alfa alone. For example, Motzer et al. and Cella et al. [23,26] showed that patients treated with sunitinib experienced improved progression-free survival [PFS: 11 months vs. 5 months; hazard ratio (HR): 0.42; 95% confidence interval (CI): 0.32 to 0.54; p < 0.001), an improved objective response rate (ORR: 31% vs. 6%; p < 0.001), and improved quality of life. Similar benefits were also seen in patients who had received prior cytokine therapy [27]. In that group, treatment with sorafenib was superior to placebo with regard to PFS (6 months vs. 3 months; HR: 0.44; 95% CI: 0.35 to 0.55; p < 0.01) and the disease control rate (84% vs. 55%; p < 0.001). Quality of life was also improved in patients receiving sorafenib as compared with those receiving placebo [28]. Only the EGFR inhibitors failed to deliver substantial therapeutic benefit, either as single agents [17,18,29,30,31] or in combination with agents such as bevacizumab [32].

2.3. Second-Line Treatment After Failure with a Firstline Targeted Agent

Until recently, little evidence was available to guide therapy once patients had progressed on first-line treatment with a targeted agent. There is certainly a need to offer treatments to this patient population, not uncommonly encountered in clinical practice and often of appropriate performance status to tolerate more therapy. Re-treatment with another targeted agent has become commonplace practice despite the lack of prospective data [33], and a number of retrospective studies have now been published about this approach (Table 2). In all but two reports, the clinical benefit rate (complete response/partial response/stable disease) exceeded 50%, and in most cases, it reached 70% or better. In most reports, the median duration of benefit was 6 months or more. Re-treatment was generally well tolerated, with the most common grade 3 toxicities being fatigue, hypertension, and hand– foot syndrome.
Prior response to first-line targeted treatment did not predict response to second-line treatment [34,35,36,37]. As with other targeted therapies [38], this lack of crossresistance is thought to indicate incomplete suppression of tumour pathways with initial therapy. Further evidence of activity has been provided by the large prospective expanded-access programs of sorafenib [37] and sunitinib [39], which reported on 2488 and 2341 patients respectively. These programs showed that clinical benefit rates in these populations (84% and 52% respectively) were similar to those reported in phase III trials. A substantial minority of patients previously exposed to targeted therapies also appeared to derive benefit from sunitinib and sorafenib in these programs.
The best evidence to date is provided by Motzer et al. [47], who reported a positive phase III trial in this clinical setting. Patients who had progressed on sunitinib or sorafenib (or both) were randomized to either the mTOR inhibitor everolimus (10 mg orally, once daily) or to placebo. Although patients from all prognostic groups were enrolled into the trial, approximately half the patients had an intermediate prognosis. The primary endpoint was PFS as determined by independent reviewers. The trial was halted early when an interim analysis indicated a substantial and significant difference in PFS in favour of the everolimus group. The progression rate was 37% in the everolimus group as compared with 65% in the placebo group (HR: 0.30; 95% CI: 0.22 to 0.40; p < 0.0001). Median PFS was 4.0 months (95% CI: 3.7 to 5.5 months) in the everolimus group as compared with 1.9 months (95% CI: 1.8 to 1.9 months) in the control group. Median overall survival (OS) had not been reached in the everolimus arm (in excess of 10 months as compared with 8.8 months in the placebo group). The difference in OS did not reach statistical significance, likely as a result of the planned crossover from placebo to everolimus on study. Although more stomatitis (40% vs. 8%), rash (25% vs. 4%), and fatigue (20% vs. 16%) occurred in the everolimus group, these side effects were mostly mild or moderate in severity. Pneumonitis (any grade) was detected in 22 patients in the everolimus group (8%), but only 8 patients [3] had grade 3 pneumonitis. Quality of life was equivalent in both study arms.
At this time, everolimus can reasonably be con-sidered to be the preferred second-line treatment after initial failure of sunitinib or sorafenib. Although OS is always preferable as the primary endpoint in phase III trials, the use of PFS in the Motzer study is an acceptable surrogate, and the overall case supporting the efficacy of everolimus as a second-line treatment is strong [48]. Although everolimus is not yet approved in Canada for metastatic RCC, temsirolimus could be considered in this setting, given that it is currently approved in Canada and has a similar mechanism of activity. Once completed, two phase III trials currently in progress may affect this choice. The first randomizes patients to temsirolimus or sorafenib as second-line therapy after progression on sunitinib (search for “NCT00474786” at www.clinicaltrials. gov/ct2/search). The second randomizes patients who failed a previous systemic treatment, which may be a tyrosine kinase inhibitor, to either sorafenib or axitinib (search for “NCT00678392” at www.clinicaltrials. gov/ct2/search).

3. Conclusions

Inhibitors of multiple kinases such as sunitinib and sorafenib are now established as standard first-line therapy in patients with RCC. When disease progression occurs after such therapy, there is clearly more benefit to be gained by re-treating these patients with another targeted agent. Everolimus is the drug of choice at the present time, and we expect it to obtain approval for this indication in Canada soon. Until such time as approval is forthcoming, temsirolimus (if available) could be substituted, given its similar mechanism of action. It would also not be unreasonable to use any of the targeted agents in Table 2 (if available) to treat patients with progressive RCC who have not previously been exposed to those agents. However, the benefits seen with rationally-developed targeted agents in the first-line setting strongly suggest that it is more appropriate to enrol those patients into clinical trials. Research priorities include the evaluation of predictive biomarkers to allow for patient enrichment, optimization of drug sequencing [concurrent vs. sequential, and simultaneous blockade at several points of the same pathway (vertical blockade) vs. blockade of several collateral pathways (horizontal blockade)], and identification of other effective drugs—for example, histone deacetylase [49,50].

References

  1. Statistics Canada, Health Statistics Division. Cancer Incidence in Canada: 2004 to 2005; Minister of Industry: Ottawa, 2007; Available online: https://publications.gc.ca/site/archivee-archived.html?url=https://publications.gc.ca/collections/collection_2007/%20statcan/82-231-X/82-231-XIE2007001.pdf (accessed on 19 January 2009).
  2. Abugaber, A.; Lang, K.; Danchenko, N.; et al. Cost of illness of renal cell carcinoma in Canada [abstract 15560]. Proc Am Soc Clin Oncol 2007, 25. Available online: www.asco.org/ASCO/ Abstracts+%26+Virtual+Meeting/Abstracts?&vmview=abst_ detail_view&confID=47&abstractID=32312 (accessed on 19 January 2009).
  3. Cohen, H.T.; McGovern, F.J. Renal-cell carcinoma. N Engl J Med 2005, 353, 2477–2490. [Google Scholar] [CrossRef] [PubMed]
  4. Coppin, C. Immunotherapy for renal cell cancer in the era of targeted therapy. Exp Rev Anticancer Ther 2008, 8, 907–919. [Google Scholar] [CrossRef] [PubMed]
  5. Mendel, D.B.; Laird, A.D.; Xin, X.; et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 2003, 9, 327–337. [Google Scholar]
  6. Wilhelm, S.M.; Carter, C.; Tang, L.; et al. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the Raf/Mek/ Erk pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res 2004, 64, 7099–7109. [Google Scholar] [CrossRef]
  7. Carlomagno, F.; Anaganti, S.; Guida, T.; et al. BAY 43-9006 inhibition of oncogenic RET mutants. J Natl Cancer Inst 2006, 98, 326–334. [Google Scholar] [CrossRef]
  8. Le Tourneau, C.; Faivre, S.; Serova, M.; Raymond, E. mTOR C1 inhibitors: is temsirolimus in renal cancer telling us how they really work? Br J Cancer 2008, 99, 1197–1203. [Google Scholar] [CrossRef]
  9. Sonpavde, G.; Hutson, T.E.; Rini, B.I. Axitinib for renal cell carcinoma. Expert Opin Investig Drugs 2008, 17, 741–748. [Google Scholar] [CrossRef]
  10. Presta, L.G.; Chen, H.; O’Connor, S.J.; et al. Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res 1997, 57, 4593–4599. [Google Scholar]
  11. Kaelin, W.G., Jr. The von Hippel–Lindau tumor suppressor protein and clear cell renal carcinoma. Clin Cancer Res 2007, 13, 680s–4s. [Google Scholar] [CrossRef]
  12. Cho, D.; Signoretti, S.; Regan, M.; Mier, J.W.; Atkins, M.B. The role of mammalian target of rapamycin inhibitors in the treatment of advanced renal cancer. Clin Cancer Res 2007, 13, 758s–63s. [Google Scholar] [CrossRef] [PubMed]
  13. Robb, V.A.; Karbowniczek, M.; Klein–Szanto, A.J.; Henske, E.P. Activation of the mTOR signaling pathway in renal clear cell carcinoma. J Urol 2007, 177, 346–352. [Google Scholar] [CrossRef] [PubMed]
  14. Lin, F.; Zhang, P.L.; Yang, X.J.; Prichard, J.W.; Lun, M.; Brown, R.E. Morphoproteomic and molecular concomitants of an overexpressed and activated mTOR pathway in renal cell carcinomas. Ann Clin Lab Sci 2006, 36, 283–293. [Google Scholar] [PubMed]
  15. Sourbier, C.; Lindner, V.; Lang, H.; et al. The phosphoinositide 3-kinase/Akt pathway: a new target in human renal cell carcinoma therapy. Cancer Res 2006, 66, 5130–5142. [Google Scholar] [CrossRef]
  16. Asakuma, J.; Sumitomo, M.; Asano, T.; Asano, T.; Hayakawa, M. Modulation of tumor growth and tumor induced angiogenesis after epidermal growth factor receptor inhibition by ZD1839 in renal cell carcinoma. J Urol 2004, 171, 897–902. [Google Scholar] [CrossRef]
  17. Ravaud, A.; Hawkins, R.; Gardner, J.P.; et al. Lapatinib versus hormone therapy in patients with advanced renal cell carcinoma: a randomized phase III clinical trial. J Clin Oncol 2008, 26, 2285–2291. [Google Scholar] [CrossRef]
  18. Rowinsky, E.K.; Schwartz, G.H.; Gollob, J.A.; et al. Safety, pharmacokinetics, and activity of ABX-EGF, a fully human anti-epidermal growth factor receptor monoclonal antibody in patients with metastatic renal cell cancer. J Clin Oncol 2004, 22, 3003–3015. [Google Scholar] [CrossRef]
  19. Knebelmann, B.; Ananth, S.; Cohen, H.T.; Sukhatme, V.P. Transforming growth factor alpha is a target for the von Hippel–Lindau tumor suppressor. Cancer Res 1998, 58, 226–231. [Google Scholar]
  20. Gunaratnam, L.; Morley, M.; Franovic, A.; et al. Hypoxia inducible factor activates the transforming growth factor-alpha/epidermal growth factor receptor growth stimulatory pathway in VHL(–/–) renal cell carcinoma cells. J Biol Chem 2003, 278, 44966–44974. [Google Scholar] [CrossRef]
  21. Smith, K.; Gunaratnam, L.; Morley, M.; Franovic, A.; Mekhail, K.; Lee, S. Silencing of epidermal growth factor receptor suppresses hypoxia-inducible factor-2–driven VHL–/– renal cancer. Cancer Res 2005, 65, 5221–5230. [Google Scholar] [CrossRef]
  22. de Paulsen, N.; Brychzy, A.; Fournier, M.C.; et al. Role of transforming growth factor-alpha in von Hippel–Lindau (VHL)(–/–) clear cell renal carcinoma cell proliferation: a possible mechanism coupling VHL tumor suppressor inactivation and tumorigenesis. Proc Natl Acad Sci USA 2001, 98, 1387–1392. [Google Scholar] [CrossRef] [PubMed]
  23. Motzer, R.J.; Hutson, T.E.; Tomczak, P.; et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007, 356, 115–124. [Google Scholar] [CrossRef] [PubMed]
  24. Hudes, G.; Carducci, M.; Tomczak, P.; et al. on behalf of the Global ARCC Trial. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007, 356, 2271–2281. [Google Scholar] [CrossRef] [PubMed]
  25. Escudier, B.; Pluzanska, A.; Koralewski, P.; et al. on behalf of the Avoren Trial investigators. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet 2008, 370, 2103–2111. [Google Scholar] [CrossRef]
  26. Cella, D.; Li, J.Z.; Cappelleri, J.C.; et al. Quality of life in patients with metastatic renal cell carcinoma treated with sunitinib or interferon alfa: results from a phase III randomized trial. J Clin Oncol 2008, 26, 3763–3769. [Google Scholar] [CrossRef]
  27. Escudier, B.; Eisen, T.; Stadler, W.M.; et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2007, 356, 125–134. [Google Scholar] [CrossRef]
  28. Bukowski, R.; Cella, D.; Gondek, K.; Escudier, B. ; on behalf of the Sorafenib TARGETS Clinical Trial Group. Effects of sorafenib on symptoms and quality of life: results from a large randomized placebo-controlled study in renal cancer. Am J Clin Oncol 2007, 30, 220–227. [Google Scholar]
  29. Motzer, R.J.; Amato, R.; Todd, M.; et al. Phase II trial of antiepidermal growth factor receptor antibody C225 in patients with advanced renal cell carcinoma. Invest New Drugs 2003, 21, 99–101. [Google Scholar] [CrossRef]
  30. Drucker, B.; Bacik, J.; Ginsberg, M.; et al. Phase II trial of ZD1839 (Iressa) in patients with advanced renal cell carcinoma. Invest New Drugs 2003, 21, 341–345. [Google Scholar] [CrossRef]
  31. Beeram, M.; Rowinsky, E.K.; Weiss, G.R.; et al. Durable disease stabilization and antitumor activity with OSI-774 in renal cell carcinoma: a phase II, pharmacokinetic (PK) and biological correlative study with FDG-PET imaging [abstract 3050]. Proc Am Soc Clin Oncol 2004, 22. Available online: www.asco.org/ASCO/ Abstracts+%26+Virtual+Meeting/Abstracts?&vmview=abst_ detail_view&confID=26&abstractID=3632 (accessed on 19 January 2009).
  32. Bukowski, R.M.; Kabbinavar, F.F.; Figlin, R.A.; et al. Randomized phase II study of erlotinib combined with bevacizumab compared with bevacizumab alone in metastatic renal cell cancer. J Clin Oncol 2007, 25, 4536–4541. [Google Scholar] [CrossRef] [PubMed]
  33. Rini, B.I.; Rathmell, W.K.; Godley, P. Renal cell carcinoma. Curr Opin Oncol 2008, 20, 300–306. [Google Scholar] [CrossRef] [PubMed]
  34. Tamaskar, I.; Garcia, J.A.; Elson, P.; et al. Antitumor effects of sunitinib or sorafenib in patients with metastatic renal cell carcinoma who received prior antiangiogenic therapy. J Urol 2008, 179, 81–86. [Google Scholar] [CrossRef] [PubMed]
  35. Rini, B.I.; Michaelson, M.D.; Rosenberg, J.E.; et al. Antitumor activity and biomarker analysis of sunitinib in patients with bevacizumab-refractory metastatic renal cell carcinoma. J Clin Oncol 2008, 26, 3743–3748. [Google Scholar] [CrossRef]
  36. Rini, B.I.; Hutson, T.E.; Elson, P.; et al. A prospective trial of sorafenib in patients (pts) with metastatic clear cell renal cell carcinoma (mccRCC) refractory to prior sunitinib or bevacizumab [abstract 346]. Am Soc Clin Oncol Genitourin Cancers Symp 2023. Available online: www.asco.org/ASCO/ Abstracts+%26+Virtual+Meeting/Abstracts?&vmview=abst_ detail_view&confID=54&abstractID=20239 (accessed on 19 January 2009).
  37. Knox, J.J.; Figlin, R.A.; Stadler, W.M.; et al. on behalf of the ARCCS Investigators. The Advanced Renal Cell Carcinoma Sorafenib (ARCCS) expanded access trial in NorthAmerica: safety and efficacy [abstract 5011]. Proc Am Soc Clin Oncol 2007, 25. Available online: www.asco.org/ASCO/Abstracts+%26+Virtual+Meeting/ Abstracts?&vmview=abst_detail_view&confID=47&abstractID=34297 (accessed on 19 January 2009).
  38. Matar, P.; Rojo, F.; Cassia, R.; et al. Combined epidermal growth factor receptor targeting with the tyrosine kinase inhibitor gefitinib (ZD1839) and the monoclonal antibody cetuximab (IMC-C225): superiority over single-agent receptor targeting. Clin Cancer Res 2004, 10, 6487–6501. [Google Scholar] [CrossRef]
  39. Gore, M.E.; Porta, C.; Oudard, S.; et al. Sunitinib in metastatic renal cell carcinoma (mRCC): preliminary assessment of toxicity in an expanded access trial with subpopulation analysis [abstract 5010]. Proc Am Soc Clin Oncol 2007, 25. Available online: www.asco.org/ASCO/Abstracts+%26+Virtual+Meeting/ Abstracts?&vmview=abst_detail_view&confID=47&abstrac tID=32318 (accessed on 19 January 2009).
  40. Sablin, M.P.; Bouaita, L.; Balleyguier, C.; et al. Sequential use of sorafenib and sunitinib in renal cancer: retrospective analysis in 90 patients [abstract 5038]. Proc Am Soc Clin Oncol 2007, 25. Available online: www.asco.org/ASCO/ Abstracts+%26+Virtual+Meeting/Abstracts?&vmview=abst_ detail_view&confID=47&abstractID=32197 (accessed on 19 January 2009).
  41. Sepulveda, J.; Maroto, P.; Andres, R.; et al. Sorafenib as a secondline and sequential therapy for patients with metastatic renal cell carcinoma (mRCC): analysis for safety and activity on sunitinib progressive pts [abstract 16100]. Proc Am Soc Clin Oncol, 2008; 26. Available online: www.asco.org/ASCO/ Abstracts+%26+Virtual+Meeting/Abstracts?&vmview=abst_ detail_view&confID=55&abstractID=36069 (accessed on 19 January 2009).
  42. Dudek, A.Z.; Zolnierek, J.; Dham, A.; Lindgren, B.R.; Szczylik, C. Sequential therapy with sorafenib and sunitinib in renal cell carcinoma. Cancer 2009, 115, 61–67. [Google Scholar] [CrossRef]
  43. Eichelberg, C.; Heuer, R.; Chun, F.K.; et al. Sequential use of the tyrosine kinase inhibitors sorafenib and sunitinib in metastatic renal cell carcinoma: a retrospective outcome analysis. Eur Urol 2008, 54, 1373–1378. [Google Scholar] [CrossRef]
  44. Rini, B.I.; Wilding, G.T.; Hudes, G.; et al. Axitinib (AG-013736; AG) in patients (pts) with metastatic renal cell cancer (RCC) refractory to sorafenib [abstract 5032]. Proc Am Soc Clin Oncol 2007, 25. Available online: www.asco.org/ASCO/ Abstracts+%26+Virtual+Meeting/Abstracts?&vmview=abst_ detail_view&confID=47&abstractID=33225 (accessed on 19 January 2009).
  45. Dutcher, J.P.; Wilding, G.; Hudes, G.R.; et al. Sequential axitinib (AG-013736) therapy of patients (pts) with metastatic clear cell renal cell cancer (RCC) refractory to sunitinib and sorafenib, cytokines and sorafenib, or sorafenib alone [abstract 5127]. Proc Am Soc Clin Oncol 2008, 26. Available online: www.asco.org/ASCO/Abstracts+%26+Virtual+Meeting/ Abstracts?&vmview=abst_detail_view&confID=55&abstrac tID=35034 (accessed on 19 January 2009).
  46. Whorf, R.C.; Hainsworth, J.D.; Spigel, D.R.; et al. Phase II study of bevacizumab and everolimus (RAD001) in the treatment of advanced renal cell carcinoma (RCC) [abstract 5010]. Proc Am Soc Clin Oncol 2008, 26. Available online: www.asco.org/ASCO/Abstracts+%26+Virtual+Meeting/ Abstracts?&vmview=abst_detail_view&confID=55&abstrac tID=34991 (accessed on 19 January 2009).
  47. Motzer, R.J.; Escudier, B.; Oudard, S.; et al. on behalf of the RECORD-1 Study Group. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 2008, 372, 449–456. [Google Scholar] [CrossRef]
  48. Knox, J.J. Progression-free survival as endpoint in metastatic RCC? Lancet 2008, 372, 427–429. [Google Scholar] [CrossRef]
  49. Drabkin, H.A. The combination of sorafenib and vorinostat causes synergistic growth inhibition in kidney cancer cells [abstract 402]. Am Soc Clin Oncol Genitourin Cancers Symp 2008. Available online: www.asco.org/ASCO/ Abstracts+%26+Virtual+Meeting/Abstracts?&vmview=abst_ detail_view&confID=54&abstractID=20022 (accessed on 19 January 2009).
  50. Hammers, H.J.; Verheul, H.; Wilky, B.; et al. Phase I safety and pharmacokinetic/pharmacodynamic results of the histone deacetylase inhibitor vorinostat in combination with bevacizumab in patients with kidney cancer [abstract 16094]. Proc Am Soc Clin Oncol 2008, 26. Available online: www.asco.org/ASCO/ Abstracts+%26+Virtual+Meeting/Abstracts?&vmview=abst_ detail_view&confID=55&abstractID=33518 (accessed on 19 January 2009). [CrossRef]
Figure 1. Simplified schema of signalling pathways that contribute to renal oncogenesis. VHL = Von Hippel–Lindau tumour-suppressor protein; EGFR = epidermal growth factor receptor; HDAC = histone deacetylase; Hsp90 = heat shock protein 90; HIF = hypoxia-inducible factor; TGFα = transforming growth factor α; RTKs = receptor tyrosine kinases; IGFR-1 = insulin-like growth factor receptor 1; VEGF = vascular endothelial growth factor; Raf = protein encoded by the murine leukemia viral oncogene homolog; Ras = ras signal transduction protein; PI3K = phosphoinositide-3-kinase; MEK = mitogen-activated protein kinase; PTEN = protein encoded by the phosphatase and tensin homolog gene; ERK = extracellular signal-regulated kinase; Akt = protein kinase B; mTOR = mammalian target of rapamycin.
Figure 1. Simplified schema of signalling pathways that contribute to renal oncogenesis. VHL = Von Hippel–Lindau tumour-suppressor protein; EGFR = epidermal growth factor receptor; HDAC = histone deacetylase; Hsp90 = heat shock protein 90; HIF = hypoxia-inducible factor; TGFα = transforming growth factor α; RTKs = receptor tyrosine kinases; IGFR-1 = insulin-like growth factor receptor 1; VEGF = vascular endothelial growth factor; Raf = protein encoded by the murine leukemia viral oncogene homolog; Ras = ras signal transduction protein; PI3K = phosphoinositide-3-kinase; MEK = mitogen-activated protein kinase; PTEN = protein encoded by the phosphatase and tensin homolog gene; ERK = extracellular signal-regulated kinase; Akt = protein kinase B; mTOR = mammalian target of rapamycin.
Curroncol 16 00045 g001
Table 1. Selected targeted agents in the treatment of renal cell carcinoma (RCC).
Table 1. Selected targeted agents in the treatment of renal cell carcinoma (RCC).
Curroncol 16 00045 i001
Table 2. Retrospective data regarding the efficacy of second-line targeted therapy.
Table 2. Retrospective data regarding the efficacy of second-line targeted therapy.
Curroncol 16 00045 i002

Share and Cite

MDPI and ACS Style

Gan, H.K.; Seruga, B.; Knox, J.J. Targeted Therapies for Renal Cell Carcinoma: More Gains from Using Them Again. Curr. Oncol. 2009, 16, 45-51. https://doi.org/10.3747/co.v16i0.404

AMA Style

Gan HK, Seruga B, Knox JJ. Targeted Therapies for Renal Cell Carcinoma: More Gains from Using Them Again. Current Oncology. 2009; 16(s1):45-51. https://doi.org/10.3747/co.v16i0.404

Chicago/Turabian Style

Gan, H. K., B. Seruga, and J. J. Knox. 2009. "Targeted Therapies for Renal Cell Carcinoma: More Gains from Using Them Again" Current Oncology 16, no. s1: 45-51. https://doi.org/10.3747/co.v16i0.404

APA Style

Gan, H. K., Seruga, B., & Knox, J. J. (2009). Targeted Therapies for Renal Cell Carcinoma: More Gains from Using Them Again. Current Oncology, 16(s1), 45-51. https://doi.org/10.3747/co.v16i0.404

Article Metrics

Back to TopTop