Assessment of the consistency and robustness of results from a multicenter trial of remission maintenance therapy for acute myeloid leukemia

Marc Buyse, Pierre Squifflet, Kathryn J Lucchesi, Mats L Brune, Sylvie Castaigne, Jacob M Rowe, Marc Buyse, Pierre Squifflet, Kathryn J Lucchesi, Mats L Brune, Sylvie Castaigne, Jacob M Rowe

Abstract

Background: Data from a randomized multinational phase 3 trial of 320 adults with acute myeloid leukemia (AML) demonstrated that maintenance therapy with 3-week cycles of histamine dihydrochloride plus low-dose interleukin-2 (HDC/IL-2) for up to 18 months significantly improved leukemia-free survival (LFS) but lacked power to detect an overall survival (OS) difference.

Purpose: To assess the consistency of treatment benefit across patient subsets and the robustness of data with respect to trial centers and endpoints.

Methods: Forest plots were constructed with hazard ratios (HRs) of HDC/IL-2 treatment effects versus no treatment (control) for prospectively defined patient subsets. Inconsistency coefficients (I²) and interaction tests (X²) were used to detect any differences in benefit among subsets. Robustness of results to the elimination of individual study centers was performed using "leave-one-center-out" analyses. Associations between treatment effects on the endpoints were evaluated using weighted linear regression between HRs for LFS and OS estimated within countries.

Results: The benefit of HDC/IL-2 over controls was statistically consistent across all subsets defined by baseline prognostic variables. I² and P-values of X² ranged from 0.00 to 0.51 and 0.14 to 0.91, respectively. Treatment effects were statistically significant in 14 of 28 subsets analyzed. The "leave-one-center-out" analysis confirmed that no single center dominated (P-values ranged from 0.004 to 0.020 [mean 0.009]). The HRs representing the HDC/IL-2 effects on LFS and OS were strongly correlated at the country level (R² = 0.84).

Limitations: Small sample sizes in some of the subsets analyzed.

Conclusions: These analyses confirm the consistency and robustness of the HDC/IL-2 effect as compared with no treatment. LFS may be an acceptable surrogate for OS in future AML trials. Analyses of consistency and robustness may aid interpretation of data from multicenter trials, especially in populations with rare diseases, when the size of randomized clinical trials is limited.

Trial registration: ClinicalTrials.gov: NCT00003991.

Figures

Figure 1
Figure 1
Forest plots of leukemia-free survival (LFS) hazard ratios (HR) and their confidence intervals (CI) by baseline characteristics. O/N = event rate per arm where O is the number of observed events (relapse or death) and N is the sample size. HR = hazard ratio, CI = confidence interval, CR = complete remission, CR1 = first complete remission, WBC = white blood cell, SWOG = Southwest Oncology Group, AML = acute myeloid leukemia, HiDAC = high-dose cytosine arabinoside.
Figure 2
Figure 2
Forest plots of leukemia-free survival (LFS) hazard ratios (HR) and their confidence intervals (CI) by country. O/N = event rate per arm where O is the number of observed events (relapse or death) and N is the sample size.
Figure 3
Figure 3
Distribution of P-values for the treatment effect on leukemia-free survival in a "leave-one-center-out" cross-validation. N = sample size.
Figure 4
Figure 4
Correlation between treatment effects on leukemia-free survival and overall survival (R2 = coefficient of determination). The size of each circle is proportional to the number of patients in the corresponding country.

References

    1. Harmonised Tripartite Guideline: Statistical Principles for Clinical Trials E9.
    1. Estey EH. Treatment of acute myeloid leukemia. Haematologica. 2009;94:10–16. doi: 10.3324/haematol.2008.001263.
    1. Redaelli A, Lee JM, Stephens JM, Pashos CL. Epidemiology and clinical burden of acute myeloid leukemia. Expert Rev Anticancer Ther. 2003;3:695–710. doi: 10.1586/14737140.3.5.695.
    1. Burchill SA. What do, can and should we learn from models to evaluate potential anticancer agents? Future Oncol. 2006;2:201–211. doi: 10.2217/14796694.2.2.201.
    1. Maurillo L, Buccisano F, Del Principe MI, Del PG, Spagnoli A, Panetta P, Ammatuna E, Neri B, Ottaviani L, Sarlo C, Venditti D, Quaresima M, Cerretti R, Rizzo M, de FP, Lo CF, Arcese W, Amadori S, Venditti A. Toward optimization of postremission therapy for residual disease-positive patients with acute myeloid leukemia. J Clin Oncol. 2008;26:4944–4951. doi: 10.1200/JCO.2007.15.9814.
    1. Buyse M, Burzykowski T, Carroll K, Michiels S, Sargent DJ, Miller LL, Elfring GL, Pignon JP, Piedbois P. Progression-free survival is a surrogate for survival in advanced colorectal cancer. J Clin Oncol. 2007;25:5218–5224. doi: 10.1200/JCO.2007.11.8836.
    1. Buyse M, Burzykowski T, Michiels S, Carroll K. Individual- and trial-level surrogacy in colorectal cancer. Stat Methods Med Res. 2008;17:467–475. doi: 10.1177/0962280207081864.
    1. Burzykowski T, Buyse M, Yothers G, Sakamoto J, Sargent D. Exploring and validating surrogate endpoints in colorectal cancer. Lifetime Data Anal. 2008;14:54–64. doi: 10.1007/s10985-007-9079-4.
    1. Buyse ME, Squifflet P, Allard SE, Bhagwat D, Rowe JM. Leukemia-free survival (LFS) as a surrogate for overall survival (OS) in AML patients in remission: a trial of a novel immunotherapy with histamine dihydrochloride plus low-dose IL-2 (HDC/IL-2) [abstract] Haematologica. 2008;93(Suppl 1):209–210.
    1. Yothers G. Toward progression-free survival as a primary end point in advanced colorectal cancer. J Clin Oncol. 2007;25:5153–5154. doi: 10.1200/JCO.2007.13.6796.
    1. Rowe JM. Optimal induction and post-remission therapy for AML in first remission. Hematology Am Soc Hematol Educ Program. 2009. pp. 396–405.
    1. Deschler B, Lubbert M. Acute myeloid leukemia: epidemiology and etiology. Cancer. 2006;107:2099–2107. doi: 10.1002/cncr.22233.
    1. Brune M, Castaigne S, Catalano J, Gehlsen K, Ho AD, Hofmann WK, Hogge DE, Nilsson B, Or R, Romero AI, Rowe JM, Simonsson B, Spearing R, Stadtmauer EA, Szer J, Wallhult E, Hellstrand K. Improved leukemia-free survival after postconsolidation immunotherapy with histamine dihydrochloride and interleukin-2 in acute myeloid leukemia: results of a randomized phase 3 trial. Blood. 2006;108:88–96. doi: 10.1182/blood-2005-10-4073.
    1. Wallhult EA, Whisnant JK, Nilsson BI, Bhagwat D, Hellstrand K, Brune ML. Quality of life during remission maintenance immunotherapy in acute myeloid leukemia: a prospective assessment using EORTC QLQ-C30 in a randomized phase III trial of histamine dihydrochloride plus low-dose interleukin-2 [abstract] Haematologica. 2008;93(Suppl 1):316.
    1. Whitehead A, Whitehead J. A general parametric approach to the meta-analysis of randomized clinical trials. Stat Med. 1991;10:1665–1677. doi: 10.1002/sim.4780101105.
    1. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ. 2003;327:557–560. doi: 10.1136/bmj.327.7414.557.
    1. Marschner IC. Regional differences in multinational clinical trials: anticipating chance variation. Clin Trials. 2010;7:147–156. doi: 10.1177/1740774510361974.
    1. Buyse M, Molenberghs G, Burzykowski T, Renard D, Geys H. The validation of surrogate endpoints in meta-analyses of randomized experiments. Biostatistics. 2000;1:49–67. doi: 10.1093/biostatistics/1.1.49.
    1. Burzykowski T, Molenberghs G, Buyse M. The Evaluation of Surrogate Endpoints. New York, NY: Springer; 2005.
    1. National Cancer Institute. SEER Survival Data 1988-2005 (SEER 9)

Source: PubMed

3
S'abonner