Skip to main content

Advertisement

Log in

Accelerating drug development by efficiently using emerging PK/PD data from an adaptable entry-into-human trial: example of lumretuzumab

  • Original Article
  • Published:
Cancer Chemotherapy and Pharmacology Aims and scope Submit manuscript

Abstract

Purpose

This study aimed at evaluating if pharmacokinetic and pharmacodynamic data from the first few patients treated with an investigational monoclonal antibody in a dose-escalation study can be used to guide the early initiation of potentially more efficacious combination regimens.

Methods

Emerging pharmacokinetic and pharmacodynamic data from the first nine patients treated with lumretuzumab (a glycoengineered anti-HER3 monoclonal antibody) monotherapy at doses from 100 to 400 mg q2w were used along with a pharmacokinetic model that incorporated target-mediated drug disposition to guide the selection of the starting dose for use in combination regimens.

Results

The dose-escalation study investigated lumretuzumab doses up to 2000 mg q2w and a maximum tolerated dose was not reached. However, the model described in this report predicted linear lumretuzumab pharmacokinetics and >95% target saturation at doses ≥400 mg q2w. These data, along with safety data, contributed to the decision to begin dose-escalation studies in combination with cetuximab and erlotinib using a starting dose of 400 mg lumretuzumab. Pharmacokinetic data from patients treated with lumretuzumab 400–2000 mg q2w in combination regimens were consistent with the model predictions.

Conclusion

PK/PD modelling of emerging clinical data might accelerate development programs by enabling additional parts of a trial to commence before completion of the monotherapy part. The dose and schedule of lumretuzumab were optimised for concomitant therapy at doses substantially below the highest dose investigated.

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
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Barrett JS, Gupta M, Mondick JT (2007) Model-based drug development applied to oncology. Expert Opin Drug Discov 2:185–209

    Article  CAS  PubMed  Google Scholar 

  2. Dostalek M, Gardner I, Gurbaxani BM, Rose RH, Chetty M (2013) Pharmacokinetics, pharmacodynamics and physiologically-based pharmacokinetic modelling of monoclonal antibodies. Clin Pharmacokinet 52:83–124

    Article  CAS  PubMed  Google Scholar 

  3. Mager DE (2006) Target-mediated drug disposition and dynamics. Biochem Pharmacol 72:1–10

    Article  CAS  PubMed  Google Scholar 

  4. Mager DE, Jusko WJ (2001) General pharmacokinetic model for drugs exhibiting target-mediated drug disposition. J Pharmacokinet Pharmacodyn 28:507–532

    Article  CAS  PubMed  Google Scholar 

  5. Cao Y, Jusko WJ (2014) Incorporating target-mediated drug disposition in a minimal physiologically-based pharmacokinetic model for monoclonal antibodies. J Pharmacokinet Pharmacodyn 41:375–387

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Luu KT, Bergqvist S, Chen E, Hu-Lowe D, Kraynov E (2012) A model-based approach to predicting the human pharmacokinetics of a monoclonal antibody exhibiting target-mediated drug disposition. J Pharmacol Exp Ther 341:702–708

    Article  CAS  PubMed  Google Scholar 

  7. Amin DN, Campbell MR, Moasser MM (2010) The role of HER3, the unpretentious member of the HER family, in cancer biology and cancer therapeutics. Semin Cell Dev Biol 21:944–950

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mirschberger C, Schiller CB, Schräml M, Dimoudis N, Friess T, Gerdes CA, Reiff U, Lifke V, Hoelzlwimmer G, Kolm I, Hopfner K-P, Niederfellner G, Bossenmaier B (2013) RG7116, a therapeutic antibody that binds the inactive HER3 receptor and is optimized for immune effector activation. Can Res 73:5183–5194

    Article  CAS  Google Scholar 

  9. Meneses-Lorente G, Friess T, Kolm I, Holzlwimmer G, Bader S, Meille C, Thomas M, Bossenmaier B (2015) Preclinical pharmacokinetics, pharmacodynamics, and efficacy of RG7116: a novel humanized, glycoengineered anti-HER3 antibody. Cancer Chemother Pharmacol 75:837–850

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Meulendijks D, Jacob W, Martinez-Garcia M, Taus A, Lolkema MP, Voest EE, Langenberg MH, Fleitas Kanonnikoff T, Cervantes A, De Jonge MJ, Sleijfer S, Soerensen MM, Thomas M, Ceppi M, Meneses-Lorente G, James I, Adessi C, Michielin F, Abiraj K, Bossenmaier B, Schellens JH, Weisser M, Lassen UN (2016) First-in-human phase I study of lumretuzumab, a glycoengineered humanized anti-HER3 monoclonal antibody, in patients with metastatic or advanced HER3-positive solid tumors. Clin Can Res 22:877–885

    Article  CAS  Google Scholar 

  11. Lassen UN, Cervantes-Ruiperez A, Fleitas T, Meulendijks D, Schellens JH, Lolkema MP, De Jonge MJ, Sleijfer S, Mau Soerensen M, Taus A, Adessi C, Keelarar A, Michielin F, Bossenmaier B, Meneses-Lorente G, James I, Jacob W, Weisser M, Marti M (2014) Phase Ib trial trial of RG7116, a glycoengineered monoclonal antibody targeting HER3, in combination with cetuximab or erlotinib in patients with advanced/metastatic tumors of epithelial cell origin expressing HER3 protein. Ann Oncol 25(Suppl_4):abstract 4440

    Article  Google Scholar 

  12. Meulendijks D, Lolkema MPJK, Voest EE, De Jonge MJ, Sleijfer S, Schellens JHM, Fleitas T, Cervantes-Ruiperez A, Martinez-Garcia M, Taus A, Mau Sorensen M, Thomas M, Meneses-Lorente G, Adessi C, Di Scala L, Keelarar A, Jacob W, Lassen UN (2013) A first-in-human trial of RG7116, a glycoengineered monoclonal antibody targeting HER3, in patients with advanced/metastatic tumors of epithelial cell origin expressing HER3 protein. J Clin Oncol 31(15_suppl):abstract 2522

    Google Scholar 

Download references

Acknowledgements

The authors would like to thank the entire BP27771 study team at Penzberg for their assistance in the preparation of this manuscript. Support for third-party writing assistance for this article, furnished by Jamie Ashman, PhD, was provided by Prism Ideas and funded by Roche Diagnostics GmbH, Penzberg.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Georgina Meneses-Lorente.

Ethics declarations

Funding

This study was funded by Roche Diagnostics GmbH, Penzberg, Germany.

Conflict of interest

All authors are employees of F. Hoffmann-La Roche Ltd.

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. This article does not contain any studies with animals performed by any of the authors.

Informed consent

Informed consent was obtained from all individual participants included in the study.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 61 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meneses-Lorente, G., McIntyre, C., Hsu, J.C. et al. Accelerating drug development by efficiently using emerging PK/PD data from an adaptable entry-into-human trial: example of lumretuzumab. Cancer Chemother Pharmacol 79, 1239–1247 (2017). https://doi.org/10.1007/s00280-017-3328-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00280-017-3328-3

Keywords

Navigation