Skip to main content
Log in

Continuous low plasma concentrations of everolimus provides equivalent efficacy to oral daily dosing in mouse xenograft models of human cancer

  • Short Communication
  • Published:
Cancer Chemotherapy and Pharmacology Aims and scope Submit manuscript

Abstract

Purpose

Everolimus is a drug used successfully in a number of different oncology indications, but significant on-target toxicities exist. We explored the possibility of improving the therapeutic index (TI) by studying alternative means of administering the drug based upon low continuous dosing.

Methods

All studies were performed using naïve nude mice or nude mice bearing s.c. human renal 786-O tumours or human breast MDA-MB-468 tumours. Everolimus was administered via a standard emulsion, either i.v., p.o., i.p., s.c., or via s.c. osmotic mini-pumps (MP) or via poly-lactic-co-glycolic (PLGA)-microparticles (PLGA-µP) prepared from everolimus powder injected s.c. Total-drug levels in blood, plasma or tissues were quantified ex vivo by LC–MS/MS. Efficacy studies were performed over 2–3 weeks and toxicity assessed by changes in body weight, glucose and white blood cell count. Effects on tumour activity biomarkers were quantified using reverse-phase protein array.

Results

Everolimus administration s.c. in an emulsion decreased the absorption rate but increased the C max and bio-availability of everolimus compared to standard approaches of administration p.o. or i.p. Everolimus administration s.c. via MP or PLGA-µP reduced the C max and provided continuous low concentrations of everolimus in the plasma, which inhibited tumour pS6/S6 to a similar degree to oral administration. Toxicities such as changes in body weight or white blood cell count were unaffected. Provided the everolimus concentration was above the free unbound IC50 for proliferation of the tumour cell line, efficacy could be achieved equivalent to that provided by standard oral administration. However, an overall improvement in the TI could not be demonstrated.

Conclusions

Continuous low plasma concentrations of everolimus can provide strong efficacy in preclinical models, which if translatable to the clinic may reduce on-target toxicities and so increase the TI.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

References

  1. Lebwohl D, Anak O, Sahmoud T, Klimovsky J, Elmroth I, Haas T, Posluszny J, Saletan S, Berg W (2013) Development of everolimus, a novel oral mTOR inhibitor, across a spectrum of diseases. Ann NY Acad Sci 1291:14–32

    Article  CAS  PubMed  Google Scholar 

  2. Tanaka C, O’Reilly T, Kovarik JM, Shand N, Hazell K, Judson I, Raymond E, Zumstein-Mecker S, Stephan C, Boulay A, Hattenberger M, Thomas G, Lane HA (2008) Identifying optimal biologic doses of everolimus (RAD001) in patients with cancer based on the modeling of preclinical and clinical pharmacokinetic and pharmacodynamic data. J Clin Oncol 26:1596–1602

    Article  CAS  PubMed  Google Scholar 

  3. O’Donnell A, Faivre S, Burris HA 3rd, Rea D, Papadimitrakopoulou V, Shand N, Lane HA, Hazell K, Zoellner U, Kovarik JM, Brock C, Jones S, Raymond E, Judson I (2008) Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors. J Clin Oncol 26:1588–1595

    Article  PubMed  Google Scholar 

  4. Ravaud A, Urva SR, Grosch K, Cheung WK, Anak O, Sellami DB (2014) Relationship between everolimus exposure and safety and efficacy: meta-analysis of clinical trials in oncology. Eur J Cancer 50:486–495

    Article  CAS  PubMed  Google Scholar 

  5. Thiery-Vuillemin A, Mouillet G, Nguyen Tan Hon T, Montcuquet P, Maurina T, Almotlak H, Stein U, Montange D, Foubert A, Nerich V, Pivot X, Royer B (2014) Impact of everolimus blood concentration on its anti-cancer activity in patients with metastatic renal cell carcinoma. Cancer Chemother Pharmacol 73:999–1007

    Article  CAS  PubMed  Google Scholar 

  6. de Wit D, Schneider TC, Moes DJ, Roozen CF, den Hartigh J, Gelderblom H, Guchelaar HJ, van der Hoeven JJ, Links TP, Kapiteijn E, van Erp NP (2016) Everolimus pharmacokinetics and its exposure-toxicity relationship in patients with thyroid cancer. Cancer Chemother Pharmacol 78:63–71

    Article  PubMed  PubMed Central  Google Scholar 

  7. Patel JK, Kobashigawa JA (2006) Everolimus: an immunosuppressive agent in transplantation. Expert Opin Pharmacother 7:1347–1355

    Article  CAS  PubMed  Google Scholar 

  8. Deppenweiler M, Falkowski S, Saint-Marcoux F et al (2017) Towards therapeutic drug monitoring of everolimus in cancer? Results of an exploratory study of exposure-effect relationship. Pharmacol Res 121:138–144

    Article  CAS  PubMed  Google Scholar 

  9. Tabernero J, Rojo F, Calvo E et al (2008) Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors. J Clin Oncol 26:1603–1610

    Article  CAS  PubMed  Google Scholar 

  10. FDA (2003) Guidance for industry: exposure-response relationships, p. 14. https://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm064982.htm. Accessed 03 Aug 2017

  11. Aapro M, Andre F, Blackwell K et al (2014) Adverse event management in patients with advanced cancer receiving oral everolimus: focus on breast cancer. Ann Oncol 25:763–773

    Article  CAS  PubMed  Google Scholar 

  12. Shi J, Kantoff PW, Wooster R, Farokhzad OC (2017) Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer 17:20–37

    Article  CAS  PubMed  Google Scholar 

  13. Smith DA, Di L, Kerns EH (2010) The effect of plasma protein binding on in vivo efficacy: misconceptions in drug discovery. Nat Rev Drug Discov 9:929–939

    Article  CAS  PubMed  Google Scholar 

  14. Petersen H, Bizec JC, Schuetz H, Delporte ML (2011) Pharmacokinetic and technical comparison of Sandostatin® LAR® and other formulations of long-acting octreotide. BMC Res Notes 9:344

    Article  Google Scholar 

  15. O’Reilly T, McSheehy PM, Kawai R, Kretz O, McMahon L, Brueggen J, Bruelisauer A, Gschwind HP, Allegrini PR, Lane HA (2010) Comparative pharmacokinetics of RAD001 (everolimus) in normal and tumor-bearing rodents. Cancer Chemother Pharmacol 65:625–639

    Article  PubMed  Google Scholar 

  16. Royce ME, Osman D (2015) Everolimus in the treatment of metastatic breast cancer. Breast Cancer Basic Clin Res 9:73–79

    Article  Google Scholar 

  17. O’Reilly T, McSheehy PM (2010) Biomarker development for the clinical activity of the mTOR inhibitor everolimus (RAD001): Processes, limitations, and further proposals. Transl Oncol 3:65–79

    Article  PubMed  PubMed Central  Google Scholar 

  18. O’Reilly T, Wartmann M, Brueggen J et al (2008) Pharmacokinetic profile of the microtubule stabilizer patupilone in tumor-bearing rodents and comparison of anti-cancer activity with other MTS in vitro and in vivo. Cancer Chemother Pharm 62:1045–1054

    Article  Google Scholar 

  19. Tunçay M, Caliş S, Kaş HS, Ercan MT, Peksoy I, Hincal AA (2000) Diclofenac sodium incorporated PLGA (50:50) microspheres: formulation considerations and in vitro/in vivo evaluation. Int J Pharm 195:179–188

    Article  PubMed  Google Scholar 

  20. Pacifici GM, Viani A (1992) Methods of determining plasma and tissue binding of drugs. Clin Pharmacokinet 23:449–468

    Article  CAS  PubMed  Google Scholar 

  21. Pawlak M, Schick E, Bopp MA, Schneider MJ, Oroszlan P, Ehrat M (2002) Zeptosens’ protein microarrays: a novel high performance microarray platform for low abundance protein analysis. Proteomics 2:383–393

    Article  CAS  PubMed  Google Scholar 

  22. van Oostrum J, Calonder C, Rechsteiner D, Ehrat M, Mestan J, Fabbro D, Voshol H (2009) Tracing pathway activities with kinase inhibitors and reverse phase protein arrays. Proteom Clin Appl 3:412–422

    Article  Google Scholar 

  23. Lane HA, Wood JM, McSheehy PM et al (2009) mTOR inhibitor RAD001 (everolimus) has antiangiogenic/vascular properties distinct from a VEGFR tyrosine kinase inhibitor. Clin Cancer Res 15:1612–1622

    Article  CAS  PubMed  Google Scholar 

  24. Apryshkina O, Manukyants A, Romen F, Elzenga C, Collins L, Morgan (2015) Everolimus novartis investigator’s brochure, edn 14. Tables 5.2 and 5.3, p. 65

  25. O’Reilly T, McSheehy PM, Wartmann M, Lassota P, Brandt R, Lane HA (2011) Evaluation of the mTOR inhibitor, everolimus, in combination with cytotoxic antitumor agents using human tumor models in vitro and in vivo. Anticancer Drugs 22:58–78

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

It is a pleasure to thank Dr Michael Jensen for the provision of in vivo facilities, and Dr. Terry O’Reilly for statistical advice.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Paul McSheehy.

Ethics declarations

All procedures involving animals were conducted in strict adherence to the Swiss law for animal care and handling.

Conflict of interest

All authors are employees and/or share-holders of Novartis.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOC 74 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Laborde, L., Oz, F., Ristov, M. et al. Continuous low plasma concentrations of everolimus provides equivalent efficacy to oral daily dosing in mouse xenograft models of human cancer. Cancer Chemother Pharmacol 80, 869–878 (2017). https://doi.org/10.1007/s00280-017-3407-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00280-017-3407-5

Keywords

Navigation