Practicalities in running early-phase trials using the time-to-event continual reassessment method (TiTE-CRM) for interventions with long toxicity periods using two radiotherapy oncology trials as examples

Erik van Werkhoven, Samantha Hinsley, Eleni Frangou, Jane Holmes, Rosemarie de Haan, Maria Hawkins, Sarah Brown, Sharon B Love, Erik van Werkhoven, Samantha Hinsley, Eleni Frangou, Jane Holmes, Rosemarie de Haan, Maria Hawkins, Sarah Brown, Sharon B Love

Abstract

Background: Awareness of model-based designs for dose-finding studies such as the Continual Reassessment Method (CRM) is now becoming more commonplace amongst clinicians, statisticians and trial management staff. In some settings toxicities can occur a long time after treatment has finished, resulting in extremely long, interrupted, CRM design trials. The Time-to-Event CRM (TiTE-CRM), a modification to the original CRM, accounts for the timing of late-onset toxicities and results in shorter trial duration. In this article, we discuss how to design and deliver a trial using this method, from the grant application stage through to dissemination, using two radiotherapy trials as examples.

Methods: The TiTE-CRM encapsulates the dose-toxicity relationship with a statistical model. The model incorporates observed toxicities and uses a weight to account for the proportion of completed follow-up of participants without toxicity. This model uses all available data to determine the next participant's dose and subsequently declare the maximum tolerated dose. We focus on two trials designed by the authors to illustrate practical issues when designing, setting up, and running such studies.

Results: In setting up a TiTE-CRM trial, model parameters need to be defined and the time element involved might cause complications, therefore looking at operating characteristics through simulations is essential. At the grant application stage, we suggest resources to fund statisticians' time before funding is awarded and make recommendations for the level of detail to include in funding applications. While running the trial, close contact of all involved staff is required as a dose decision is made each time a participant is recruited. We suggest ways of capturing data in a timely manner and give example code in R for design and delivery of the trial. Finally, we touch upon dissemination issues while the trial is running and upon completion.

Conclusion: Model-based designs can be complex. We hope this paper will help clinical trial teams to demystify the conduct of TiTE-CRM trials and be a starting point for using this methodology in practice.

Trial registration: ClinicalTrials.gov NCT03641547 NCT01562210.

Keywords: Adaptive trial design; Clinical trial design; Dose-finding; Late toxicity; Phase I; TiTE-CRM.

Conflict of interest statement

RdH declares research grant funding from AstraZeneca to support 3 trials, including the NCT01562210 example used in this paper. All other authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Graphic illustration of the TiTECRM method. The first plot shows the recruited participants over time. We illustrate the observation window T for each recruit; y3 has experienced a toxicity hence omitting presenting the full observation window. The dose allocated for participant 5, at the current time point, is decided by accounting for all of the available data, which includes the toxicity status and weights of participants 1–4. These will be accounted for in the calculation of the updated dose-toxicity curve. The table shows the weight that each participant contributes in updating the dose-toxicity curve when participant 5 is recruited onto the trial. Although participant 3 has not completed the observation window, they have experienced a toxicity event, so their contributed weight to the model is 1. The second plot presents the dose allocation for each participant. In this scenario, once a toxicity is observed, the model recommends de-escalating to dose level 2. As participant 4 was not on the trial for long when participant 5 was recruited, the model recommends the same dose for participant 5. This figure reflects the example code and (fictitious) data presented in the Additional file 1

References

    1. Love SB, Brown S, Weir CJ, et al. Embracing model-based designs for dose-finding trials. Br J Cancer. 2017;117(3):332–339. doi: 10.1038/bjc.2017.186.
    1. Wheeler GM, Mander AP, Bedding A, et al. How to design a dose-finding study using the continual reassessment method. BMC Med Res Methodol. 2019;19(1):18. doi: 10.1186/s12874-018-0638-z.
    1. O'Quigley J, Pepe M, Fisher L. Continual reassessment method: a practical design for phase 1 clinical trials in cancer. Biometrics. 1990;46(1):33–48. doi: 10.2307/2531628.
    1. Cheung YK, Chappell R. Sequential designs for phase I clinical trials with late-onset toxicities. Biometrics. 2000;56(4):1177–1182. doi: 10.1111/j.0006-341X.2000.01177.x.
    1. Sharma RA, Plummer R, Stock JK, et al. Clinical development of new drug-radiotherapy combinations. Nat Rev Clin Oncol. 2016;13(10):627–642. doi: 10.1038/nrclinonc.2016.79.
    1. Normolle D, Lawrence T. Designing dose-escalation trials with late-onset toxicities using the time-to-event continual reassessment method. J Clin Oncol. 2006;24(27):4426–4433. doi: 10.1200/JCO.2005.04.3844.
    1. Zhao L, Lee J, Mody R, Braun TM. The superiority of the time-to-event continual reassessment method to the rolling six design in pediatric oncology phase I trials. Clin Trials. 2011;8(4):361–369. doi: 10.1177/1740774511407533.
    1. Ben-Josef E, Schipper M, Francis IR, et al. A phase I/II trial of intensity modulated radiation (IMRT) dose escalation with concurrent fixed-dose rate gemcitabine (FDR-G) in patients with unresectable pancreatic cancer. Int J Radiat Oncol Biol Phys. 2012;84(5):1166–1171. doi: 10.1016/j.ijrobp.2012.02.051.
    1. Brown DR, Normolle D, Junck LR, et al. Phase 1 TITE-CRM dose escalation study of concurrent temozolomide and intensity modulated radiation therapy in newly diagnosed glioblastoma multiforme. Int J Radiat Oncol Biol Phys. 2009;75(3):S124. doi: 10.1016/j.ijrobp.2009.07.298.
    1. Chugh R, Griffith KA, Davis EJ, et al. Doxorubicin plus the IGF-1R antibody cixutumumab in soft tissue sarcoma: a phase I study using the TITE-CRM model. Ann Oncol. 2015;26(7):1459–1464. doi: 10.1093/annonc/mdv171.
    1. Frangou E, Holmes J, Love S, McGregor N, Hawkins M. Challenges in implementing model-based phase I designs in a grant-funded clinical trials unit. Trials. 2017;18(1):620. doi: 10.1186/s13063-017-2389-2.
    1. Kim MM, Parmar H, Cao Y, et al. Whole brain radiotherapy and RRx-001: two partial responses in radioresistant melanoma brain metastases from a phase I/II clinical trial: a TITE-CRM phase I/II clinical trial. Transl Oncol. 2016;9(2):108–113. doi: 10.1016/j.tranon.2015.12.003.
    1. Kyriakopoulos CE, Heath EI, Eickhoff JC, et al. A multicenter phase 1/2a dose-escalation study of the antioxidant moiety of vitamin E 2,2,5,7,8-pentamethyl-6-chromanol (APC-100) in men with advanced prostate cancer. Investig New Drugs. 2016;34(2):225–230. doi: 10.1007/s10637-016-0334-y.
    1. de Haan R, van Werkhoven E, van den Heuvel MM, et al. Study protocols of three parallel phase 1 trials combining radical radiotherapy with the PARP inhibitor olaparib. BMC Cancer. 2019;19(1):901. doi: 10.1186/s12885-019-6121-3.
    1. Lao CD, Friedman J, Tsien CI, et al. Concurrent whole brain radiotherapy and bortezomib for brain metastasis. Radiat Oncol. 2013;8:204. doi: 10.1186/1748-717X-8-204.
    1. Lepeak LM, Wilding G, Eickhoff JC, et al. Phase I study of continuous MKC-1 (cMKC-1) in patients (pts) with advanced or metastatic solid malignancies using a modified time-to-event continual reassessment method (TITE-CRM) for dose assignment. J Clin Oncol. 2010;28(15_suppl):e13001. doi: 10.1200/jco.2010.28.15_suppl.e13001.
    1. Muler JH, McGinn CJ, Normolle D, et al. Phase I trial using a time-to-event continual reassessment strategy for dose escalation of cisplatin combined with gemcitabine and radiation therapy in pancreatic cancer. J Clin Oncol. 2004;22(2):238–243. doi: 10.1200/JCO.2004.03.129.
    1. Schneider BJ, Kalemkerian GP, Bradley D, et al. Phase I study of vorinostat (suberoylanilide hydroxamic acid, NSC 701852) in combination with docetaxel in patients with advanced and relapsed solid malignancies. Investig New Drugs. 2012;30(1):249–257. doi: 10.1007/s10637-010-9503-6.
    1. Tevaarwerk A, Wilding G, Eickhoff J, et al. Phase I study of continuous MKC-1 in patients with advanced or metastatic solid malignancies using the modified time-to-event continual reassessment method (TITE-CRM) dose escalation design. Investig New Drugs. 2012;30(3):1039–1045. doi: 10.1007/s10637-010-9629-6.
    1. Zhen DB, Griffith KA, Ruch JM, et al. A phase I trial of cabozantinib and gemcitabine in advanced pancreatic cancer. Investig New Drugs. 2016;34(6):733–739. doi: 10.1007/s10637-016-0376-1.
    1. Braun TM. Generalizing the TITE-CRM to adapt for early- and late-onset toxicities. Stat Med. 2006;25(12):2071–2083. doi: 10.1002/sim.2337.
    1. Braun TM, Levine JE, Ferrara JL. Determining a maximum tolerated cumulative dose: dose reassignment within the TITE-CRM. Control Clin Trials. 2003;24(6):669–681. doi: 10.1016/S0197-2456(03)00094-1.
    1. Huang B, Kuan PF. Time-to-event continual reassessment method incorporating treatment cycle information with application to an oncology phase I trial. Biom J. 2014;56(6):933–946. doi: 10.1002/bimj.201300261.
    1. Wages NA, Conaway MR, O'Quigley J. Using the time-to-event continual reassessment method in the presence of partial orders. Stat Med. 2013;32(1):131–141. doi: 10.1002/sim.5491.
    1. Palma G, Monti S, Conson M, Pacelli R, Cella L. Normal tissue complication probability (NTCP) models for modern radiation therapy. Semin Oncol. 2019;46:210–218. doi: 10.1053/j.seminoncol.2019.07.006.
    1. Polley MY. Practical modifications to the time-to-event continual reassessment method for phase I cancer trials with fast patient accrual and late-onset toxicities. Stat Med. 2011;30(17):2130–2143. doi: 10.1002/sim.4255.
    1. Bekele BN, Ji Y, Shen Y, Thall PF. Monitoring late-onset toxicities in phase I trials using predicted risks. Biostatistics. 2008;9(3):442–457. doi: 10.1093/biostatistics/kxm044.
    1. Cheung K. dfcrm: dose-finding by the continual reassessment method. R package version 0.2–2.1. 2019.
    1. Salter A, Morgan C, Aban IB. Implementation of a two-group likelihood time-to-event continual reassessment method using SAS. Comput Methods Prog Biomed. 2015;121(3):189–196. doi: 10.1016/j.cmpb.2015.06.001.
    1. Sharon E, Polley MY, Bernstein MB, Ahmed M. Immunotherapy and radiation therapy: considerations for successfully combining radiation into the paradigm of immuno-oncology drug development. Radiat Res. 2014;182(2):252–257. doi: 10.1667/RR13707.1.
    1. ICH E6 (R2) Good clinical practice guideline. Accessed 18 Apr 2019.
    1. Harrington KJ, Billingham LJ, Brunner TB, et al. Guidelines for preclinical and early phase clinical assessment of novel radiosensitisers. Br J Cancer. 2011;105(5):628–639. doi: 10.1038/bjc.2011.240.
    1. Thompson MK, Poortmans P, Chalmers AJ, et al. Practice-changing radiation therapy trials for the treatment of cancer: where are we 150 years after the birth of Marie Curie? Br J Cancer. 2018;119(4):389–407. doi: 10.1038/s41416-018-0201-z.
    1. Ivanova A, Wang Y, Foster MC. The rapid enrollment design for phase I clinical trials. Stat Med. 2016;35(15):2516–2524. doi: 10.1002/sim.6886.
    1. Jia N, Braun TM. The adaptive accelerated biased coin design for phase I clinical trials AU. J Appl Stat. 2011;38(12):2911–2924. doi: 10.1080/02664763.2011.573540.
    1. Lin R, Yin G. Nonparametric overdose control with late-onset toxicity in phase I clinical trials. Biostatistics. 2016;18(1):180–194. doi: 10.1093/biostatistics/kxw038.
    1. Mauguen A, Le Deley MC, Zohar S. Dose-finding approach for dose escalation with overdose control considering incomplete observations. Stat Med. 2011;30(13):1584–1594. doi: 10.1002/sim.4128.
    1. Yin G, Zheng S, Xu J. Fractional dose-finding methods with late-onset toxicity in phase I clinical trials. J Biopharm Stat. 2013;23(4):856–870. doi: 10.1080/10543406.2013.789892.

Source: PubMed

3
Iratkozz fel