Abstract
An article in a recent issue of The Oncologist reports the results of a phase I clinical trial of BEZ235 in patients with advanced renal cell carcinoma. The study was terminated early due to toxicity and a lack of clinical efficacy. The rapid and concise publication of this type of clinical result is necessary for effective collaboration and, ultimately, better outcomes for cancer patients.
In the July 2016 issue of The Oncologist, Carlo et al. report the results of a phase I clinical trial [1] of BEZ235 in patients with advanced renal cell carcinoma (RCC). BEZ235 is a dual pan-class PI3K and mTOR inhibitor currently undergoing phase I/II clinical trials as a single agent or in combination in solid tumors and hematologic malignancies. The authors are to be congratulated for reporting the early termination of the BEZ235 trial due to toxicity and a lack of clinical efficacy.
PI3Ks are a family of kinases that regulate multiple hallmarks of cancer, including survival, proliferation, tumor metabolism, autophagy, and angiogenesis [2]. Many of these hallmarks are regulated through PI3K mediated regulation of the mTOR pathway [3, 4]. The genetic and molecular rationale of targeting the PI3K/Akt/mTOR pathway in RCC is supported by the existence of recurrent mutations of the pathway in clear cell RCC and elevation of the PI3K/Akt/mTOR proteomic signature in both clear cell and papillary RCC [5, 6]. Consistent with this, two mTOR inhibitors, temsirolimus and everolimus, have received U.S. Food and Drug Administration (FDA) approval in renal cell carcinoma. However, resistance to mTOR inhibitors is acquired early, possibly due to feedback activation of PI3K although upstream receptor tyrosine kinase signaling, providing the rationale for combined inhibition of PI3K and mTOR [7–9].
In the current study, Carlo et al. reported increased frequency of grade 3–4 adverse effects with BEZ235 in 50% of patients (5 of 10) without objective responses in the evaluable patients [1]. Most of the dose-limiting toxicities, including fatigue, diarrhea, nausea, and mucositis have been described with both PI3K and mTOR inhibitors, and thus, it is not surprising that combined pan-PI3K and mTOR blockade resulted in a high frequency of adverse events. Additionally, poor tolerability and modest clinical activity have been previously reported with BEZ235 in other solid tumors. In a phase II trial of advanced pancreatic neuroendocrine tumors (PNETs) comparing everolimus and BEZ235, adverse events led to treatment discontinuation in 39% and 16% of BEZ235- and everolimus-treated patients, respectively [10]. In another phase II trial in patients with PNETs whose disease progressed on everolimus, poor tolerance of BEZ235 with only modest activity led to study termination [11]. Finally, in metastatic RCC, apitolisib (GDC-0980), another dual pan-PI3K and mTOR inhibitor, also demonstrated a high incidence of grade 3–4 adverse effects and worrisomely shorter progression-free survival with a trend toward shorter overall survival compared with everolimus [12].
Overall, data from the aforementioned clinical trials make us question whether daily concurrent pan-PI3K and mTOR inhibition offers a viable strategy for treating cancer. It appears that full blockade of class I PI3K and mTOR cannot be tolerated due to multiple on-target adverse effects. While a pan-PI3K inhibitor aims to abolish total PI3K activity in the tumor, it fails to exploit dependency to a specific isoform that in turn could avoid unnecessary adverse effects from the inhibition of other “nonrelevant isoforms.” Can we do better?
Isoform-specific PI3K inhibitors are currently under development. Early phase studies of PI3Kα and PI3Kβ inhibitors appear to show a more favorable adverse effect profile with preliminary evidence of responses in tumors harboring activating PIK3CA mutations or PTEN loss [13, 14]. Ongoing clinical trials are evaluating BYL719, an isoform specific PI3Kα inhibitor in combination with everolimus in patients with advanced RCC and PNETs [15]. Adverse events are likely to remain an issue, leaving open the question of how or if tolerability might be improved. In this regard, consideration will have to be given to the mTOR inhibitors and how critical is their use in these combinations, especially at the “approved” doses. Few drugs have proven more difficult to administer on a daily regimen than inhibitors of mTOR, and their activity, even in diseases for which regulatory approvals have been granted, have been modest [9, 16–19]. As regards toxicity, for example, even in pivotal trials everolimus has been discontinued or reduced by a high percentage of patients [17–21]. Even in the most recent everolimus approval, discontinuation for adverse events occurred in 29% of patients, and dose reductions or delays occurred in 70%, indicating that despite widespread experience, administration of everolimus remains very difficult and, in a majority of patients, not possible at a 10-mg dose [19, 22]. Temsirolimus, originally explored in patients with advanced RCC and shown in that pivotal trial to be relatively well tolerated, has also encountered tolerability issues in subsequent studies and has not been well tolerated at higher doses [23–28]. Thus, the issue of tolerability emerges in our minds as critical to the future of these drugs in any combination. We must ask whether the optimal drug or optimal schedule for mTOR inhibition has been explored or come to recognize that the mTOR pathway, shown in countless preclinical studies to be central to so many functions, cannot be inhibited continuously without inflicting intolerable toxicities [29–31]. A recent FDA approval of everolimus, in combination with lenvatinib, in RCC suggests that tolerability may be mitigated by lower doses, while possibly retaining activity, but the validity of this, especially in other diseases, and its clinical importance remain to be determined [32]. But might intermittent mTOR inhibition be better tolerated? While the temsirolimus clinical data suggest intermittent (weekly) administration can also be problematic, an intermittent schedule, especially in combination regimens, might improve tolerability. After all, do we have firm data in tumors that continuous 24/7 inhibition of mTOR is essential?
If we accept the limitations of our preclinical models and the even greater limitations of our speculations, then we realize only clinical trials in humans can give us answers. In the end, we must test our hypotheses, obtain results, and move forward. As the article by Carlo et al. shows, we can quickly establish tolerability and even a level of efficacy everyone would consider valuable—not one that is marginal in quantity [33]. And once we establish this, we must submit it for publication, rapidly and concisely, so that others can learn from it. Only by doing this, can we aspire to move forward to hopefully better outcomes for the many patients who still struggle with cancer.
Disclosures
The authors indicated no financial relationships.
References
- 1.Carlo MI, Molina AM, Lakhman Y, et al. A phase Ib study of BEZ235, a dual inhibitor of phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR), in patients with advanced renal cell carcinoma. The Oncologist. 2016;21:787–788. doi: 10.1634/theoncologist.2016-0145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 3.Bhat M, Robichaud N, Hulea L, et al. Targeting the translation machinery in cancer. Nat Rev Drug Discov. 2015;14:261–278. doi: 10.1038/nrd4505. [DOI] [PubMed] [Google Scholar]
- 4.Dibble CC, Cantley LC. Regulation of mTORC1 by PI3K signaling. Trends Cell Biol. 2015;25:545–555. doi: 10.1016/j.tcb.2015.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Chen F, Zhang Y, Şenbabaoğlu Y, et al. Multilevel genomics-based taxonomy of renal cell carcinoma. Cell Reports. 2016;14:2476–2489. doi: 10.1016/j.celrep.2016.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Cancer Genome Atlas Research Network Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499:43–49. doi: 10.1038/nature12222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Rodrik-Outmezguine VS, Chandarlapaty S, Pagano NC, et al. mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov. 2011;1:248–259. doi: 10.1158/2159-8290.CD-11-0085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.O’Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66:1500–1508. doi: 10.1158/0008-5472.CAN-05-2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Motzer RJ, Escudier B, Oudard S, et al. 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. doi: 10.1016/S0140-6736(08)61039-9. [DOI] [PubMed] [Google Scholar]
- 10.Salazar R, Verslype C, Baudin E, et al. Phase II studies of BEZ235 in patients with advanced pancreatic neuroendocrine tumors (pNET) J Clin Oncol. 2015;33(suppl):4102a. [Google Scholar]
- 11.Fazio N, Buzzoni R, Baudin E, et al. A phase II study of BEZ235 in patients with everolimus-resistant, advanced pancreatic neuroendocrine tumours. Anticancer Res. 2016;36:713–719. [PMC free article] [PubMed] [Google Scholar]
- 12.Powles T, Lackner MR, Oudard S, et al. Randomized open-label phase II trial of apitolisib (GDC-0980), a novel inhibitor of the PI3K/mammalian target of rapamycin pathway, versus everolimus in patients with metastatic renal cell carcinoma. J Clin Oncol. 2016;34:1660–1668. doi: 10.1200/JCO.2015.64.8808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gonzalez-Angulo AM, Juric D, Argilés G, et al. Safety, pharmacokinetics, and preliminary activity of the α-specific PI3K inhibitor BYL719: Results from the first-in-human study. J Clin Oncol. 2013;31(suppl):2531a. [Google Scholar]
- 14.Arkenau H-T, Mateo J, Lemech CR, et al. A phase I/II, first-in-human dose-escalation study of GSK2636771 in patients (pts) with PTEN-deficient advanced tumors. J Clin Oncol. 2014;32(suppl):2514a. [Google Scholar]
- 15.ClinicalTrials.gov. Study of safety and efficacy of alpelisib with everolimus or alpelisib with everolimus and exemestane in advanced breast cancer patients, renal cell cancer and pancreatic tumors (NCT02077933). Memorial Sloan Kettering Cancer Center SC. Available at https://clinicaltrials.gov/ct2/show/NCT02077933. Accessed May 22, 2016.
- 16.Motzer RJ, Escudier B, Oudard S, et al. RECORD‐1 Study Group Phase 3 trial of everolimus for metastatic renal cell carcinoma : final results and analysis of prognostic factors. Cancer. 2010;116:4256–4265. doi: 10.1002/cncr.25219. [DOI] [PubMed] [Google Scholar]
- 17.Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012;366:520–529. doi: 10.1056/NEJMoa1109653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yao JC, Shah MH, Ito T, et al. RAD001 in Advanced Neuroendocrine Tumors, Third Trial (RADIANT-3) Study Group Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–523. doi: 10.1056/NEJMoa1009290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yao JC, Fazio N, Singh S, et al. RAD001 in Advanced Neuroendocrine Tumours, Fourth Trial (RADIANT-4) Study Group Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet. 2016;387:968–977. doi: 10.1016/S0140-6736(15)00817-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.U.S. Food and Drug Administration. Highlights of prescribing information. AFINITOR (everolimus) tablets for oral administration. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/022334s016lbl.pdf. Accessed May 22, 2016.
- 21.U.S. Food and Drug Administration. Everolimus 2012. Available at http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm313008.htm. Accessed May 22, 2016.
- 22.U.S. Food and Drug Administration. Everolimus (Afinitor). Available at http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm488028.htm. Accessed May 22, 2016.
- 23.Hess G, Herbrecht R, Romaguera J, et al. Phase III study to evaluate temsirolimus compared with investigator’s choice therapy for the treatment of relapsed or refractory mantle cell lymphoma. J Clin Oncol. 2009;27:3822–3829. doi: 10.1200/JCO.2008.20.7977. [DOI] [PubMed] [Google Scholar]
- 24.Wolff AC, Lazar AA, Bondarenko I, et al. Randomized phase III placebo-controlled trial of letrozole plus oral temsirolimus as first-line endocrine therapy in postmenopausal women with locally advanced or metastatic breast cancer. J Clin Oncol. 2013;31:195–202. doi: 10.1200/JCO.2011.38.3331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Rini BI, Bellmunt J, Clancy J, et al. Randomized phase III trial of temsirolimus and bevacizumab versus interferon alfa and bevacizumab in metastatic renal cell carcinoma: INTORACT trial. J Clin Oncol. 2014;32:752–759. doi: 10.1200/JCO.2013.50.5305. [DOI] [PubMed] [Google Scholar]
- 26.Hutson TE, Escudier B, Esteban E, et al. Randomized phase III trial of temsirolimus versus sorafenib as second-line therapy after sunitinib in patients with metastatic renal cell carcinoma. J Clin Oncol. 2014;32:760–767. doi: 10.1200/JCO.2013.50.3961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dreyling M, Jurczak W, Jerkeman M, et al. Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. Lancet. 2016;387:770–778. doi: 10.1016/S0140-6736(15)00667-4. [DOI] [PubMed] [Google Scholar]
- 28.Hudes G, Carducci M, Tomczak P, et al. Global ARCC Trial Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007;356:2271–2281. doi: 10.1056/NEJMoa066838. [DOI] [PubMed] [Google Scholar]
- 29.Dancey J. mTOR signaling and drug development in cancer. Nat Rev Clin Oncol. 2010;7:209–219. doi: 10.1038/nrclinonc.2010.21. [DOI] [PubMed] [Google Scholar]
- 30.Lamming DW, Sabatini DM. A Central role for mTOR in lipid homeostasis. Cell Metab. 2013;18:465–469. doi: 10.1016/j.cmet.2013.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhang Y, Nicholatos J, Dreier JR, et al. Coordinated regulation of protein synthesis and degradation by mTORC1. Nature. 2014;513:440–443. doi: 10.1038/nature13492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.U.S. Food and Drug Administration. Lenvatinib in combination with Everolimus. Available at http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm501070.htm. Accessed May 22, 2016.
- 33.Ellis LM, Bernstein DS, Voest EE, et al. American Society of Clinical Oncology perspective: Raising the bar for clinical trials by defining clinically meaningful outcomes. J Clin Oncol. 2014;32:1277–1280. doi: 10.1200/JCO.2013.53.8009. [DOI] [PubMed] [Google Scholar]