Ibrutinib-induced lymphocytosis in patients with chronic lymphocytic leukemia: correlative analyses from a phase II study

S E M Herman, C U Niemann, M Farooqui, J Jones, R Z Mustafa, A Lipsky, N Saba, S Martyr, S Soto, J Valdez, J A Gyamfi, I Maric, K R Calvo, L B Pedersen, C H Geisler, D Liu, G E Marti, G Aue, A Wiestner, S E M Herman, C U Niemann, M Farooqui, J Jones, R Z Mustafa, A Lipsky, N Saba, S Martyr, S Soto, J Valdez, J A Gyamfi, I Maric, K R Calvo, L B Pedersen, C H Geisler, D Liu, G E Marti, G Aue, A Wiestner

Abstract

Ibrutinib and other targeted inhibitors of B-cell receptor signaling achieve impressive clinical results for patients with chronic lymphocytic leukemia (CLL). A treatment-induced rise in absolute lymphocyte count (ALC) has emerged as a class effect of kinase inhibitors in CLL and warrants further investigation. Here we report correlative studies in 64 patients with CLL treated with ibrutinib. We quantified tumor burden in blood, lymph nodes (LNs), spleen and bone marrow, assessed phenotypic changes of circulating cells and measured whole-blood viscosity. With just one dose of ibrutinib, the average increase in ALC was 66%, and in>40% of patients the ALC peaked within 24 h of initiating treatment. Circulating CLL cells on day 2 showed increased Ki67 and CD38 expression, indicating an efflux of tumor cells from the tissue compartments into the blood. The kinetics and degree of the treatment-induced lymphocytosis was highly variable; interestingly, in patients with a high baseline ALC the relative increase was mild and resolution rapid. After two cycles of treatment the disease burden in the LN, bone marrow and spleen decreased irrespective of the relative change in ALC. Whole-blood viscosity was dependent on both ALC and hemoglobin. No adverse events were attributed to the lymphocytosis.

Trial registration: ClinicalTrials.gov NCT01500733.

Conflict of interest statement

Conflict-of-Interest Disclosure

C.H.G. obtained research funding from Genzyme/Sanofi and is on advisory boards for Roche, Janssen, Celgene and GlaxoSmithKline. The remaining authors declare no competing financial interests.

Figures

Figure 1
Figure 1
Ibrutinib-induced lymphocytosis develops rapidly, peaks within days, and is highly variable between patients. (a–d) Data on 41 patients with complete counts for the first six months on ibrutinib are depicted. (a) Change in mean ALC over time; vertical lines indicate SEM. (b) Mean relative change of ALC on treatment over baseline; vertical lines indicate SEM. (c) Cumulative proportion of patients reaching their peak ALC at the indicated time point. Note >40% of patients reached the peak ALC on day 2. (d) Box and whisker plots demonstrating distinct inter-patient variability in ALCs.
Figure 2
Figure 2
Variability in development and resolution of ibrutinib-induced lymphocytosis. (a) Hierarchical clustering of change in ALC normalized to baseline (day 1) in individual patients (n=41) reveals three distinct patterns. The degree of change is color coded according to the legend shown. (b–c) The mean for each of the three patient clusters is shown as (b) ALC or (c) relative change of ALC compared to pre-treatment. (d) The pre-treatment ALC for the three patient clusters are significantly different by Student’s T-test.
Figure 3
Figure 3
Ibrutinib rapidly decreased total tumor burden and increases the rate of cell death (a) A graphic representation of the estimated tumor burden in different anatomic compartments pre-treatment is shown for four representative patients (identified by study code). (b–d) Change in tumor burden from pre-treatment (Pre) to 2 and 6 months (MO) on treatment in different tissues. (b) Graphic representation of total lymph node volume computed from whole body CT scans. (c) Graphic representation of spleen volume computed from CT scan. (d) CLL cell infiltration of bone marrow visualized by CD79a staining. (e) Changes in disease burden on treatment is shown for the four representative patients. Also see Supplementary table S1. (f) The viability of circulating CLL cells was measured in fresh whole blood samples using the LIVE/DEAD stain (n=12, each symbol represents a different patient). Ibrutinib doubles the rate of cell death (P=0.03 by paired Student’s T-test.)
Figure 4
Figure 4
Ibrutinib induced lymphocytosis is driven by the release of cells from the lymph node. (a) More CLL cells in the lymph node (LN) express Ki67 (proliferation marker) and CD38 (activation marker) compared to CLL cells in the peripheral blood (PB). All data are from untreated patients, red symbols represent ibrutinib study patients (n=5). (b) Shown is the relative change in the frequency of Ki67 positive CLL cells in the peripheral blood on day 2 (after one dose of ibrutinib) compared to pre-treatment (n=28). (c) Change in the frequency of CD38 positive CLL cells on day 2 compared to pre-treatment (n=11, CD38+ patients only). (d) Efflux of CLL cells from the lymph node increases the fraction of Ki67 positive cells in the blood. Shown is the data for one representative patient. The frequency of Ki67 expressing CLL cells pre-treatment in lymph node (measured in single cell suspension by flow cytometry) and in the the peripheral blood is shown. On day 2, 30% of the ALC consists of “additional cells” and the frequency of Ki67 positive CLL cells has increased to 8.2%. If all the additional cells derive from the lymph node (with Ki67 expression in 12.8%) the predicted frequency of Ki67 positive cells is 8.3%. (e–f) Regression analysis of predicted vs. actual percent Ki67 expression (e) and ALC values (f). Dashed lines indicate 95% confidence intervals (n=8).
Figure 5
Figure 5
Whole blood viscosity is rarely elevated in CLL and is influenced by ALC and hemoglobin. (a) Correlation of whole blood viscosity (normal range 3.6–6 cP, shaded) with ALC (n= 105; R=0.41; P<0.001). (b) Correlation of whole blood viscosity with hemoglobin concentration (n= 105; R=0.59; P<0.001). (c) Whole blood viscosity pre-treatment (pre) and on treatment days 2 and 28. (d) Change in hemoglobin in patients with an initial value ≥ 10 g/dL (n=26). (e) Contour plot of whole blood viscosity as a function of both ALC and hemoglobin. Whole blood viscosity measurements are color coded from low (white) to high (red). Correlations by Pearson’s test.

References

    1. Chiorazzi N, Rai KR, Ferrarini M. Chronic lymphocytic leukemia. N Engl J Med. 2005;352(8):804–15.
    1. NCI. Surveillance Epidemiology and End Results. National Cancer Institute, NIH, Department of Health & Human Services; 2012.
    1. Howlader N, Ries LA, Mariotto AB, Reichman ME, Ruhl J, Cronin KA. Improved estimates of cancer-specific survival rates from population-based data. J Natl Cancer Inst. 2010;102(20):1584–98.
    1. Wierda WG, O’Brien S, Wang X, Faderl S, Ferrajoli A, Do KA, et al. Prognostic nomogram and index for overall survival in previously untreated patients with chronic lymphocytic leukemia. Blood. 2007;109(11):4679–85.
    1. Rosenquist R, Cortese D, Bhoi S, Mansouri L, Gunnarsson R. Prognostic markers and their clinical applicability in chronic lymphocytic leukemia: where do we stand? Leukemia & lymphoma. 2013
    1. Stevenson FK, Krysov S, Davies AJ, Steele AJ, Packham G. B-cell receptor signaling in chronic lymphocytic leukemia. Blood. 2011;118(16):4313–20.
    1. Wiestner A. Emerging role of kinase-targeted strategies in chronic lymphocytic leukemia. Blood. 2012;120(24):4684–91.
    1. Woyach JA, Johnson AJ, Byrd JC. The B-cell receptor signaling pathway as a therapeutic target in CLL. Blood. 2012;120(6):1175–84.
    1. Fais F, Ghiotto F, Hashimoto S, Sellars B, Valetto A, Allen SL, et al. Chronic lymphocytic leukemia B cells express restricted sets of mutated and unmutated antigen receptors. The Journal of clinical investigation. 1998;102(8):1515–25.
    1. Tobin G, Thunberg U, Karlsson K, Murray F, Laurell A, Willander K, et al. Subsets with restricted immunoglobulin gene rearrangement features indicate a role for antigen selection in the development of chronic lymphocytic leukemia. Blood. 2004;104(9):2879–85.
    1. Messmer BT, Albesiano E, Efremov DG, Ghiotto F, Allen SL, Kolitz J, et al. Multiple distinct sets of stereotyped antigen receptors indicate a role for antigen in promoting chronic lymphocytic leukemia. J Exp Med. 2004;200(4):519–25.
    1. Agathangelidis A, Darzentas N, Hadzidimitriou A, Brochet X, Murray F, Yan XJ, et al. Stereotyped B-cell receptors in one-third of chronic lymphocytic leukemia: a molecular classification with implications for targeted therapies. Blood. 2012;119(19):4467–75.
    1. Herishanu Y, Perez-Galan P, Liu D, Biancotto A, Pittaluga S, Vire B, et al. The lymph node microenvironment promotes B-cell receptor signaling, NF-kappaB activation, and tumor proliferation in chronic lymphocytic leukemia. Blood. 2011;117(2):563–74.
    1. Burger JA, Ghia P, Rosenwald A, Caligaris-Cappio F. The microenvironment in mature B-cell malignancies: a target for new treatment strategies. Blood. 2009;114(16):3367–75.
    1. Niemann CU, Wiestner A. B-cell receptor signaling as a driver of lymphoma development and evolution. Semin Cancer Biol. 2013
    1. Young RM, Staudt LM. Targeting pathological B cell receptor signalling in lymphoid malignancies. Nat Rev Drug Discov. 2013;12(3):229–43.
    1. Buggy JJ, Elias L. Bruton Tyrosine Kinase (BTK) and Its Role in B-cell Malignancy. Int Rev Immunol. 2012;31(2):119–32.
    1. Humphries LA, Dangelmaier C, Sommer K, Kipp K, Kato RM, Griffith N, et al. Tec kinases mediate sustained calcium influx via site-specific tyrosine phosphorylation of the phospholipase Cgamma Src homology 2-Src homology 3 linker. J Biol Chem. 2004;279(36):37651–61.
    1. Davis RE, Ngo VN, Lenz G, Tolar P, Young RM, Romesser PB, et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010;463(7277):88–92.
    1. Woyach JA, Bojnik E, Ruppert AS, Stefanovski MR, Goettl VM, Smucker KA, et al. Bruton’s tyrosine kinase (BTK) function is important to the development and expansion of chronic lymphocytic leukemia (CLL) Blood. 2013
    1. Kil LP, de Bruijn MJ, van Hulst JA, Langerak AW, Yuvaraj S, Hendriks RW. Bruton’s tyrosine kinase mediated signaling enhances leukemogenesis in a mouse model for chronic lymphocytic leukemia. Am J Blood Res. 2013;3(1):71–83.
    1. Pan Z, Scheerens H, Li SJ, Schultz BE, Sprengeler PA, Burrill LC, et al. Discovery of selective irreversible inhibitors for Bruton’s tyrosine kinase. ChemMedChem. 2007;2(1):58–61.
    1. Advani RH, Buggy JJ, Sharman JP, Smith SM, Boyd TE, Grant B, et al. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies. J Clin Oncol. 2013;31(1):88–94.
    1. Wiestner A. Targeting B-Cell receptor signaling for anticancer therapy: the Bruton’s tyrosine kinase inhibitor ibrutinib induces impressive responses in B-cell malignancies. J Clin Oncol. 2013;31(1):128–30.
    1. Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369(1):32–42.
    1. Herman SE, Gordon AL, Hertlein E, Ramanunni A, Zhang X, Jaglowski S, et al. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood. 2011;117(23):6287–96.
    1. de Rooij MF, Kuil A, Geest CR, Eldering E, Chang BY, Buggy JJ, et al. The clinically active BTK inhibitor PCI-32765 targets B-cell receptor- and chemokine-controlled adhesion and migration in chronic lymphocytic leukemia. Blood. 2012;119(11):2590–4.
    1. Ponader S, Chen SS, Buggy JJ, Balakrishnan K, Gandhi V, Wierda WG, et al. The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo. Blood. 2012;119(5):1182–9.
    1. de Gorter DJ, Beuling EA, Kersseboom R, Middendorp S, van Gils JM, Hendriks RW, et al. Bruton’s tyrosine kinase and phospholipase Cgamma2 mediate chemokine-controlled B cell migration and homing. Immunity. 2007;26(1):93–104.
    1. Herman SE, Sun X, McAuley EM, Hsieh MM, Pittaluga S, Raffeld M, et al. Modeling tumor-host interactions of chronic lymphocytic leukemia in xenografted mice to study tumor biology and evaluate targeted therapy. Leukemia. 2013;27(8):1769–73.
    1. Davids MS, Brown JR. Targeting the B cell receptor pathway in chronic lymphocytic leukemia. Leukemia & lymphoma. 2012;53(12):2362–70.
    1. Cheson BD, Byrd JC, Rai KR, Kay NE, O’Brien SM, Flinn IW, et al. Novel targeted agents and the need to refine clinical end points in chronic lymphocytic leukemia. J Clin Oncol. 2012;30(23):2820–2.
    1. Friedberg JW, Sharman J, Sweetenham J, Johnston PB, Vose JM, Lacasce A, et al. Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia. Blood. 2010;115(13):2578–85.
    1. Zhang B, Lewis SM. The splenomegaly of myeloproliferative and lymphoproliferative disorders: splenic cellularity and vascularity. European journal of haematology. 1989;43 (1):63–6.
    1. Sambuceti G, Brignone M, Marini C, Massollo M, Fiz F, Morbelli S, et al. Estimating the whole bone-marrow asset in humans by a computational approach to integrated PET/CT imaging. Eur J Nucl Med Mol Imaging. 2012;39(8):1326–38.
    1. Pesic V, Plecas-Solarovic B, Radojevic K, Kosec D, Pilipovic I, Perisic M, et al. Long-term beta-adrenergic receptor blockade increases levels of the most mature thymocyte subsets in aged rats. International immunopharmacology. 2007;7(5):674–86.
    1. Nadler SB, Hidalgo JH, Bloch T. Prediction of blood volume in normal human adults. Surgery. 1962;51(2):224–32.
    1. Woyach JA, Smucker K, Smith LL, Lozanski A, Zhong Y, Ruppert AS, et al. Prolonged lymphocytosis during ibrutinib therapy is associated with distinct molecular characteristics and does not indicate a suboptimal response to therapy. Blood. 2014
    1. Kuse R, Lueb H. Gastrointestinal involvement in patients with chronic lymphocytic leukemia. Leukemia. 1997;11 (Suppl 2):S50–1.
    1. Baumhoer D, Tzankov A, Dirnhofer S, Tornillo L, Terracciano LM. Patterns of liver infiltration in lymphoproliferative disease. Histopathology. 2008;53(1):81–90.
    1. Brandtzaeg P, Farstad IN, Johansen FE, Morton HC, Norderhaug IN, Yamanaka T. The B-cell system of human mucosae and exocrine glands. Immunol Rev. 1999;171:45–87.
    1. Messmer BT, Messmer D, Allen SL, Kolitz JE, Kudalkar P, Cesar D, et al. In vivo measurements document the dynamic cellular kinetics of chronic lymphocytic leukemia B cells. J Clin Invest. 2005;115(3):755–64.
    1. Calissano C, Damle RN, Hayes G, Murphy EJ, Hellerstein MK, Moreno C, et al. In vivo intraclonal and interclonal kinetic heterogeneity in B-cell chronic lymphocytic leukemia. Blood. 2009;114(23):4832–42.
    1. Chang BY, Francesco M, De Rooij MF, Magadala P, Steggerda SM, Huang MM, et al. Egress of CD19+CD5+ cells into peripheral blood following treatment with the Bruton tyrosine kinase inhibitor ibrutinib in mantle cell lymphoma patients. Blood. 2013;122(14):2412–24.
    1. Herman SE, Barr PM, McAuley EM, Liu D, Wiestner A, Friedberg JW. Fostamatinib inhibits B-cell receptor signaling, cellular activation and tumor proliferation in patients with relapsed and refractory chronic lymphocytic leukemia. Leukemia. 2013;27(8):1769–73.
    1. Herman SEM, Farooqui M, Bezabhie R, Aue G, Wiestner A. In Vivo Effects of Ibrutinib On BCR Signaling, Tumor Cell Activation and Proliferation in Blood and Tissue-Resident Cells of Chronic Lymphocytic Leukemia Patients. ASH Annual Meeting Abstracts. 2012;120(21):185.
    1. Cheng S, Ma J, Guo A, Lu P, Leonard JP, Coleman M, et al. BTK inhibition targets in vivo CLL proliferation through its effects on B-cell receptor signaling activity. Leukemia. 2013
    1. Ganzel C, Becker J, Mintz PD, Lazarus HM, Rowe JM. Hyperleukocytosis, leukostasis and leukapheresis: practice management. Blood reviews. 2012;26(3):117–22.
    1. Tam CS, O’Brien S, Wierda W, Kantarjian H, Wen S, Do KA, et al. Long-term results of the fludarabine, cyclophosphamide, and rituximab regimen as initial therapy of chronic lymphocytic leukemia. Blood. 2008;112(4):975–80.
    1. Calissano C, Damle RN, Marsilio S, Yan XJ, Yancopoulos S, Hayes G, et al. Intraclonal complexity in chronic lymphocytic leukemia: fractions enriched in recently born/divided and older/quiescent cells. Mol Med. 2011;17(11–12):1374–82.
    1. Cukierman T, Gatt ME, Libster D, Goldschmidt N, Matzner Y. Chronic lymphocytic leukemia presenting with extreme hyperleukocytosis and thrombosis of the common femoral vein. Leuk Lymphoma. 2002;43(9):1865–8.
    1. Wang ML, Rule S, Martin P, Goy A, Auer R, Kahl BS, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013;369(6):507–16.

Source: PubMed

Подписаться