Adverse event burden in older patients with CLL receiving bendamustine plus rituximab or ibrutinib regimens: Alliance A041202

Amy S Ruppert, Allison M Booth, Wei Ding, Nancy L Bartlett, Danielle M Brander, Steven Coutre, Jennifer R Brown, Sreenivasa Nattam, Richard A Larson, Harry Erba, Mark Litzow, Carolyn Owen, Charles S Kuzma, Jeremy S Abramson, Richard F Little, Scott E Smith, Richard M Stone, John C Byrd, Sumithra J Mandrekar, Jennifer A Woyach, Amy S Ruppert, Allison M Booth, Wei Ding, Nancy L Bartlett, Danielle M Brander, Steven Coutre, Jennifer R Brown, Sreenivasa Nattam, Richard A Larson, Harry Erba, Mark Litzow, Carolyn Owen, Charles S Kuzma, Jeremy S Abramson, Richard F Little, Scott E Smith, Richard M Stone, John C Byrd, Sumithra J Mandrekar, Jennifer A Woyach

Abstract

Ibrutinib has superior progression-free survival compared with bendamustine plus rituximab (BR) in older CLL patients, however, differences in treatment duration, six monthly BR cycles versus continuous ibrutinib, complicate adverse event (AE) comparisons. We introduce the AE burden score (AEsc) to compare AEs, calculated for each patient by summing over products of reporting period length and grade for each all-cause grade 1-4 AE and dividing by the length of time over which AEs are assessed. A total of 176 patients received BR and 361 ibrutinib alone or with six cycles of rituximab. At 38 months median follow-up, 64% remained on ibrutinib. Median AEsc was higher with BR versus ibrutinib in the first six cycles (7.2 versus 4.9, p < 0.0001). Within ibrutinib arms, median AEsc decreased significantly to 3.7 after six cycles (p < 0.0001). 10% and 14% of BR and ibrutinib patients discontinued treatment for AEs. In ibrutinib arms, cumulative incidence of grade 3 or higher atrial fibrillation, hypertension, and infection (AEs of clinical interest) at 12 months was 4.5%, 17.5%, and 12.8%, respectively, and increased more slowly thereafter to 7.7%, 25.4%, and 20.5% at 36 months. Analytical tools including the AEsc and cumulative incidence of AEs can help to better characterize AE burden over time. ClinicalTrials.gov identifier: NCT01886872.

Conflict of interest statement

Competing Interests

All other authors declare no competing interests.

© 2021. The Author(s), under exclusive licence to Springer Nature Limited.

Figures

Figure 1:. Sample calculation of the AE…
Figure 1:. Sample calculation of the AE score for a typical patient receiving bendamustine plus rituximab (BR).
The AE score (AESC) is calculated for an individual patient by summing over the products of reporting period length and grade recorded for each AE and dividing by the length of time over which AEs were assessed. If AEs were assessed during a particular reporting period but the grade for an AE was blank, the grade of the AE was equal to 0. If AEs were not assessed during a particular reporting period, AEs from that reporting period were not used in the calculation of the AESC. For the purposes of this study, the AE reporting length is the difference between reporting period start and end dates divided by 28, to represent the number of 28-day cycles. In general, reporting period lengths are approximately 1 for the first 5 AE assessments, 1 or 3 for AE assessment 6 depending on whether the patient received BR or ibrutinib regimens, and 3 for assessments 7 and higher.
Figure 2:. AE score by treatment group,…
Figure 2:. AE score by treatment group, across all assessments/cycles, the first 6 cycles of treatment, and after 6 cycles of treatment.
Box-and-whisker plots show the distribution of AE scores for patients treated with bendamustine plus rituximab (BR) or ibrutinib regimens (IBR). Across all cycles and in the first 6 cycles of treatment, 176 patients in the BR group had AE scores and 361 patients in the IBR group had AE scores. Among those who completed 6 cycles of therapy, after 6 cycles 117 patients in the BR group had AE scores and 325 patients in the IBR group had AE scores. AE scores for six outliers are not shown in the plots (AE score of 59 with BR across all cycles and during the first 6 cycles of treatment, AE score of 34 with IBR during the first 6 cycles of treatment, AE score of 33 with BR across all cycles and during the first 6 cycles of treatment, and AE score of 29.3 with BR during the first 6 cycles of treatment) to better visualize the AE score distributions.
Figure 3:. Limited to grade 3/4 AEs,…
Figure 3:. Limited to grade 3/4 AEs, AE score by treatment group, across all assessments/cycles, the first 6 cycles, and after 6 cycles.
Box-and-whisker plots show the distribution of AE scores for patients treated with bendamustine plus rituximab (BR) and ibrutinib regimens (IBR). Across all cycles and in the first 6 cycles of treatment, 176 patients in the BR group had AE scores and 361 patients in the IBR group had AE scores. Among those who completed 6 cycles of therapy, after 6 cycles 117 patients in the BR group had AE scores and 325 patients in the IBR group had AE scores. AE scores for two outliers are not shown in the plots (AE score of 41 with BR across all cycles and during the first 6 cycles of treatment) to better visualize the AE score distributions.
Figure 4:. Cumulative incidence of atrial fibrillation,…
Figure 4:. Cumulative incidence of atrial fibrillation, hypertension, and infection, by treatment group.
The cumulative incidence of grade 3 or higher atrial fibrillation occurring in 176 patients treated with bendamustine plus rituximab (BR) and 361 patients treated with ibrutinib regimens (IBR) is depicted (a), all grades of atrial fibrillation (b), grade 3 or higher hypertension (c), all grades of hypertension (d), grade 3 or higher infection (e), all grades of infection (f).
Figure 5:. Progression-free survival from date of…
Figure 5:. Progression-free survival from date of off-treatment for AE.
Kaplan-Meier curves are shown for 18 patients who discontinued treatment with bendamustine plus rituximab (BR) for AE and 52 patients who discontinued treatment with ibrutinib regimens (IBR) for AE (a), and Kaplan-Meier curves for 24 patients who received less 1 year (yr) of IBR and discontinued treatment for AE versus 28 patients who received more than 1 yr of IBR and discontinued treatment for AE (b). Differences in curves were tested using two-sided log-rank tests.

References

    1. Eichhorst B, Dreyling M, Robak T, Montserrat E, Hallek M. Chronic lymphocytic leukemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2011; 22(Supplement 6): vi50–vi54.
    1. Eichhorst BF, Busch R, Stilgenbauer S, Stauch M, Bergmann MA, Ritgen M et al. First-line therapy with fludarabine compared with chlorambucil does not result in a major benefit for elderly patients with advanced chronic lymphocytic leukemia. Blood. 2009; 114(16): 3382–3391.
    1. Eichhorst B, Fink AM, Bahlo J, Busch R, Math D, Kovacs G et al. First-line chemoimmunotherapy with bendamustine and rituximab versus fludarabine, cyclophosphamide, and rituximab in patients with advanced chronic lymphocytic leukaemia (CLL10): an international, open-label, randomised, phase 3, non-inferiority trial. Lancet Oncol. 2016. July; 17(7):928–942.
    1. Burger JA, Tedeschi A, Barr PM, Robak T, Owen C, Ghia P et al. RESONATE-2 Investigators. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia. N Engl J Med. 2015. December 17; 373(25):2425–2437.
    1. Woyach JA, Ruppert AS, Heerema NA, Zhao W, Booth AM, Ding W et al. Ibrutinib Regimens versus Chemoimmunotherapy in Older Patients with Untreated CLL. N Engl J Med. 2018. December 27; 379(26):2517–2528.
    1. Hilal T, Gea-Banacloche JC, Leis JF. Chronic lymphocytic leukemia and infection risk in the era of targeted therapies: Linking mechanisms with infections. Blood Rev. 2018. September; 32(5):387–399.
    1. Barr PM, Robak T, Owen C, Tedeschi A, Bairey O, Bartlett NL et al. Sustained efficacy and detailed clinical follow-up of first-line ibrutinib treatment in older patients with chronic lymphocytic leukemia: extended phase 3 results from RESONATE-2. Haematologica. 2018. September; 103(9):1502–1510.
    1. National Cancer Institute: CTEP. Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0. Bethesda, MD: National Institutes of Health; 2009.
    1. Fine JP and Gray RJ. A proportional hazards model for the subdistribution of a competing risk. J Amer Statist Assoc. 1999; 94(446):496–509.
    1. Thanarajasingam G, Hubbard JM, Sloan JA, Grothey A. The Imperative for a new approach to toxicity analysis in oncology clinical trials. J Natl Cancer Inst. 2015. October; 107(10).
    1. Carbini M, Suárez-Fariñas M, Maki RG. A method to summarize toxicity in cancer randomized cinical trials. Clin Cancer Res. 2018. October 15; 24(20):4968–4975.
    1. Trotti A, Pajak TF, Gwede CK, Paulus R, Cooper J, Forastiere A et al. TAME: development of a new method for summarising adverse events of cancer treatment by the Radiation Therapy Oncology Group. Lancet Oncol. 2007. May 31; 8(7):613–624.
    1. Lee SM, Hershman DL, Martin P, Leonard JP, Cheung YK. Toxicity burden score: a novel approach to summarize multiple toxic effects. Ann Oncol. 2012. February; 23(2):537–541.
    1. Le-Rademacher JG, Hillman S, Storrick E, Mahoney MR, Thall PF, Jatoi A et al. Adverse event burden score – a versatile summary measure for cancer clinical trials. Cancers. 2020. November 4; 12(11): 3251.
    1. Schuurhuizen CS, Verheul HM, Braamse AM, Buffart LM, Bloemendal HJ, Dekker J et al. The predictive value of cumulative toxicity for quality of life in patients with metastatic colorectal cancer during first-line palliative chemotherapy. Cancer Manag Res. 2018. August 29; 10:3015–3021.
    1. Sivendran S, Latif A, McBride RB, Stensland KD, Wisnivesky J, Haines L et al. Adverse event reporting in cancer clinical trial publications. J Clin Oncol. 2014. January 10; 32(2):83–89.
    1. Hillman SL, Mandrekar SJ, Bot B, DeMatteo RP, Perez EA, Ballman KV et al. Evaluation of the value of attribution in the interpretation of adverse event data: a North Central cancer treatment group and american college of surgeons oncology group investigation. J Clin Oncol. 2010. June 20; 28(18):3002–3007.
    1. George GC, Barata PC, Campbell A, Chen A, Cortes JA, Hyman DM et al. Improving attribution of adverse events in oncology clinical trials. Cancer Treat Rev. 2019; 76:33–40.
    1. Bercusson A, Colley T, Shah A, Warris A, Armstrong-James D. Ibrutinib blocks Btk-dependent NF-ĸB and NFAT responses in human macrophages during Aspergillus fumigatus phagocytosis. Blood. 2018. November 1; 132(18):1985–1988.
    1. Rogers KA, Mousa L, Zhao Q, Bhat SA, Byrd JC, El Boghdadly Z et al. Incidence of opportunistic infections during ibrutinib treatment for B-cell malignancies. Leukemia. 2019. October; 33(10):2527–2530.
    1. Shanafelt TD, Wang V, Kay NE, Hanson CA, O’Brien SM, Barrientos JC et al. Ibrutinib and rituximab provides superior clinical outcome compared to FCR in younger patients with chronic lymphocytic leukemia (CLL): extended follow-up from the E1912 trial. Blood. 2019. November 13; 134(Supplement_1):33 (abstract 642).

Source: PubMed

Подписаться