A new pragmatic design for dose escalation in phase 1 clinical trials using an adaptive continual reassessment method

Bernard North, Hemant Mahendrakumar Kocher, Peter Sasieni, Bernard North, Hemant Mahendrakumar Kocher, Peter Sasieni

Abstract

Background: A key challenge in phase I trials is maintaining rapid escalation in order to avoid exposing too many patients to sub-therapeutic doses, while preserving safety by limiting the frequency of toxic events. Traditional rule-based designs require temporarily stopping recruitment whilst waiting to see whether enrolled patients develop toxicity. This can be both inefficient and introduces logistic challenges to recruitment in the clinic. We describe a novel two-stage dose assignment procedure designed for a phase I clinical trial (STARPAC), where a good estimation of prior was possible.

Methods: The STARPAC design uses rule-based design until the first patient has a dose limiting toxicity (DLT) and then switches to a modified CRM, with rules to handle patient recruitment during follow-up of earlier patients. STARPAC design is compared via simulations with the TITE-CRM and 3 + 3 methods in various toxicity estimate (T1-5), rate of recruitment (R1-2), and DLT events timing (DT1-4), scenarios using several metrics: accuracy of maximum tolerated dose (MTD), numbers of DLTs, number of patients enrolled and those missed; duration of trial; and proportion of patients treated at the therapeutic dose or MTD.

Results: The simulations suggest that STARPAC design performed well in MTD estimation and in treating patients at the highest possible therapeutic levels. STARPAC and TITE-CRM were comparable in the number of patients required and DLTs incurred. The 3 + 3 design often had fewer patients and DLTs although this is due to its low escalation rate leading to poor MTD estimation. For the numbers of declined patients and MTD estimation 3 + 3 is uniformly worse, with STARPAC being better in those metrics for high toxicity scenarios and TITE-CRM better with low toxicity. In situations including doses with toxicities both above and below 30%, the STARPAC design outperformed TITE-CRM with respect to every metric.

Conclusion: When considering doses with toxicities both above and below the target of 30% toxicities, the two-stage STARPAC dose escalation design provides a more efficient phase I trial design than either the traditional 3 + 3 or the TITE-CRM design. Trialists should model various designs via simulation to adopt the most efficient design for their clinical scenario.

Trial registration: Clinical Trials NCT03307148 (11 October 2017).

Keywords: 3 + 3 design; Adaptive design; Bayesian adaptive; CRM; Dose-finding; Maximal tolerated dose; Model-based design; Rule-based design; Simulation.

Conflict of interest statement

HMK received funding for Investigator Initiated trial from Celgene Sarl (AX-CL-Panc-PI-003922).

Figures

Fig. 1
Fig. 1
Flowchart of STARPAC design
Fig. 2
Fig. 2
Schematic of recruitment of patients in STARPAC, 3 + 3 and TITE-CRM designs of a hypothetical scenario where the true toxicity is T1, recruitment rate is R1 and DLT timings are DT4. Missed patients are with dashed boxes, recruited patients are with solid boxes and patients with DLT (which may happen anytime between days 7 to 21 as for DT4) are shown in grey boxes

References

    1. Dixon WJ, Mood AM. A method for obtaining and analyzing sensitivity data. J Am Stat Assoc. 1948;43(241):109–126. doi: 10.1080/01621459.1948.10483254.
    1. Storer BE. Design and analysis of phase I clinical trials. Biometrics. 1989;45(3):925–937. doi: 10.2307/2531693.
    1. O’Quigley John, Conaway Mark. Continual Reassessment and Related Dose-Finding Designs. Statistical Science. 2010;25(2):202–216. doi: 10.1214/10-STS332.
    1. Goodman SN, Zahurak ML, Piantadosi S. Some practical improvements in the continual reassessment method for phase I studies. Stat Med. 1995;14(11):1149–1161. doi: 10.1002/sim.4780141102.
    1. Babb J, Rogatko A, Zacks S. Cancer phase I clinical trials: efficient dose escalation with overdose control. Stat Med. 1998;17(10):1103–1120. doi: 10.1002/(SICI)1097-0258(19980530)17:10<1103::AID-SIM793>;2-9.
    1. Iasonos A, O'Quigley J. Interplay of priors and skeletons in two-stage continual reassessment method. Stat Med. 2012;31(30):4321–4336. doi: 10.1002/sim.5559.
    1. O'Quigley J, Shen LZ. Continual reassessment method: a likelihood approach. Biometrics. 1996;52(2):673–684. doi: 10.2307/2532905.
    1. Iasonos A. A comprehensive comparison of the continual reassessment method to the standard 3 + 3 dose escalation scheme in phase I dose-finding studies. Clin Trials. 2008;5(5):465–477. doi: 10.1177/1740774508096474.
    1. Reiner E, Paoletti X, O'Quigley J. Operating characteristics of the standard phase I clinical trial design. Comput Stat Data Anal. 1999;30(3):303–315. doi: 10.1016/S0167-9473(98)00095-4.
    1. Council MR: “A quick guide why not to use a+B designs” adaptive designs working group of the MRC network of hubs for Trials Methodology Research 2016.
    1. Simon R, Freidlin B, Rubinstein L, Arbuck SG, Collins J, Christian MC. Accelerated titration designs for phase I clinical trials in oncology. J Natl Cancer Inst. 1997;89(15):1138–1147. doi: 10.1093/jnci/89.15.1138.
    1. Skolnik JM, Barrett JS, Jayaraman B, Patel D, Adamson PC. Shortening the timeline of pediatric phase I trials: the rolling six design. J Clin Oncol. 2008;26(2):190–195. doi: 10.1200/JCO.2007.12.7712.
    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. 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. Froeling FE, Kocher HM. Homeostatic restoration of desmoplastic stroma rather than its ablation slows pancreatic cancer progression. Gastroenterology. 2015;148(4):849–850. doi: 10.1053/j.gastro.2015.02.043.
    1. Ene-Obong A, Clear AJ, Watt J, Wang J, Fatah R, Riches JC, Marshall JF, Chin-Aleong J, Chelala C, Gribben JG, et al. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology. 2013;145(5):1121–1132. doi: 10.1053/j.gastro.2013.07.025.
    1. Froeling FE, Feig C, Chelala C, Dobson R, Mein CE, Tuveson DA, Clevers H, Hart IR, Kocher HM. Retinoic acid-induced pancreatic stellate cell quiescence reduces paracrine Wnt-beta-catenin signaling to slow tumor progression. Gastroenterology. 2011;141(4):1486–1497. doi: 10.1053/j.gastro.2011.06.047.
    1. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–1703. doi: 10.1056/NEJMoa1304369.
    1. Arrieta O, Gonzalez-De la Rosa CH, Arechaga-Ocampo E, Villanueva-Rodriguez G, Ceron-Lizarraga TL, Martinez-Barrera L, Vazquez-Manriquez ME, Rios-Trejo MA, Alvarez-Avitia MA, Hernandez-Pedro N, et al. Randomized phase II trial of all-trans-retinoic acid with chemotherapy based on paclitaxel and cisplatin as first-line treatment in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2010;28(21):3463–3471. doi: 10.1200/JCO.2009.26.6452.
    1. Budd GT, Adamson PC, Gupta M, Homayoun P, Sandstrom SK, Murphy RF, McLain D, Tuason L, Peereboom D, Bukowski RM, et al. Phase I/II trial of all-trans retinoic acid and tamoxifen in patients with advanced breast cancer. Clin Cancer Res. 1998;4(3):635–642.
    1. Chen GQ, Shen ZX, Wu F, Han JY, Miao JM, Zhong HJ, Li XS, Zhao JQ, Zhu J, Fang ZW, et al. Pharmacokinetics and efficacy of low-dose all-trans retinoic acid in the treatment of acute promyelocytic leukemia. Leukemia. 1996;10(5):825–828.
    1. Conley BA, Egorin MJ, Sridhara R, Finley R, Hemady R, Wu S, Tait NS, Van Echo DA. Phase I clinical trial of all-trans-retinoic acid with correlation of its pharmacokinetics and pharmacodynamics. Cancer Chemother Pharmacol. 1997;39(4):291–299. doi: 10.1007/s002800050575.
    1. Moore DF, Jr, Pazdur R, Sugarman S, Jones D, 3rd, Lippman SM, Bready B, Abbruzzese JL. Pilot phase II trial of 13-cis-retinoic acid and interferon-alpha combination therapy for advanced pancreatic adenocarcinoma. Am J Clin Oncol. 1995;18(6):525–527. doi: 10.1097/00000421-199512000-00013.
    1. Brembeck FH, Schoppmeyer K, Leupold U, Gornistu C, Keim V, Mossner J, Riecken EO, Rosewicz S. A phase II pilot trial of 13-cis retinoic acid and interferon-alpha in patients with advanced pancreatic carcinoma. Cancer. 1998;83(11):2317–2323. doi: 10.1002/(SICI)1097-0142(19981201)83:11<2317::AID-CNCR11>;2-P.
    1. Michael A, Hill M, Maraveyas A, Dalgleish A, Lofts F. 13-cis-retinoic acid in combination with gemcitabine in the treatment of locally advanced and metastatic pancreatic cancer--report of a pilot phase II study. Clin Oncol (R Coll Radiol) 2007;19(2):150–153. doi: 10.1016/j.clon.2006.11.008.
    1. Von Hoff DD, Ramanathan RK, Borad MJ, Laheru DA, Smith LS, Wood TE, Korn RL, Desai N, Trieu V, Iglesias JL, et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol. 2011;29(34):4548–4554. doi: 10.1200/JCO.2011.36.5742.
    1. Iasonos A, O’Quigley J. Interplay of priors and skeletons in two-stage continual reassessment method. Stat Med. 2012;31(30):4321–4336. doi: 10.1002/sim.5559.
    1. Sweeting M, Mander A, Sabin T: bcrm: Bayesian continual reassessment method designs for phase I dose-finding trials. 2013 54(13):26.
    1. Neuenschwander B, Branson M, Gsponer T. Critical aspects of the Bayesian approach to phase I cancer trials. Stat Med. 2008;27(13):2420–2439. doi: 10.1002/sim.3230.
    1. Iasonos A, O'Quigley J. Adaptive dose-finding studies: a review of model-guided phase I clinical trials. J Clin Oncol. 2014;32(23):2505–2511. doi: 10.1200/JCO.2013.54.6051.
    1. R Core Team (2014). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. .
    1. Cheung YK: Dfcrm: dose-finding by the continual reassessment method. R package version 0.2-2. 2013.
    1. Iasonos A, Gonen M, Bosl GJ. Scientific review of phase I protocols with novel dose-escalation designs: how much information is needed? J Clin Oncol. 2015;33(19):2221–2225. doi: 10.1200/JCO.2014.59.8466.
    1. Fu H, Manner D. Bayesian adaptive dose-finding studies with delayed responses. J Biopharm Stat. 2010;20(5):1055–1070. doi: 10.1080/10543400903315740.
    1. Lin R, Yin G: Nonparametric overdose control with late-onset toxicity in phase I clinical trials. Biostatistics 2017, 18(1):180–194. doi: 10.1093/biostatistics/kxw1038. Epub 2016 Aug 1022.
    1. Lin R, Yuan Y. Time-to-event model-assisted designs for dose-finding trials with delayed toxicity. Biostatistics. 2019(5460289):15.
    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: 10.1080/10543406.10542013.10789892.

Source: PubMed

3
Sottoscrivi