Clinical pharmacology of alemtuzumab, an anti-CD52 immunomodulator, in multiple sclerosis

Z Li, S Richards, H K Surks, A Jacobs, M A Panzara, Z Li, S Richards, H K Surks, A Jacobs, M A Panzara

Abstract

Alemtuzumab, a humanized anti-CD52 monoclonal antibody, is approved for treatment of relapsing multiple sclerosis (MS). In the Phase II/III trials, patients received 12 or 24 mg/day of alemtuzumab in two treatment courses (5 days for course 1 and 3 days for course 2), 12 months apart. Serum concentrations of alemtuzumab peaked on the last day of dosing in each course and mostly fell below the limit of quantitation by day 30. Alemtuzumab rapidly depleted circulating T and B lymphocytes, with the lowest observed values occurring within days. Lymphocytes repopulated over time, with B cell recovery usually complete within 6 months. T lymphocytes recovered more slowly and generally did not return to baseline by 12 months post-treatment. Approximately 40 and 80% of patients had total lymphocyte counts, reaching the lower limit of normal by 6 and 12 months after each course, respectively. The clearance of alemtuzumab is dependent on circulating lymphocyte count. A majority of treated patients tested positive for anti-alemtuzumab antibodies, including inhibitory antibodies, during the 2-year studies, and a higher proportion of patients tested positive in course 2 than in course 1. The presence of anti-alemtuzumab antibody appeared to be associated with slower clearance of alemtuzumab from the circulation but had no impact on the pharmacodynamics. No effects of age, race or gender on the pharmacokinetics or pharmacodynamics were observed. Together, the pharmacokinetics, pharmacodynamics and immunogenicity results support the continued development and use of alemtuzumab for the treatment of MS, and probably explain its sustained effects beyond the dosing interval.

Trial registration: ClinicalTrials.gov NCT00530348 NCT00548405 NCT00050778.

Keywords: MS; PK/PD; alemtuzumab; immunogenicity; lymphocyte depletion and repopulation.

© 2018 The Authors. Clinical & Experimental Immunology published by John Wiley & Sons Ltd on behalf of British Society for Immunology.

Figures

Figure 1
Figure 1
Mean (± standard deviation) alemtuzumab serum concentration over time after intravenous (i.v.) administration at 12 mg/day or 24 mg/day for 5 days in courses 1 and 3 days in course 2 with 12 months apart between courses (CARE‐MS II). Shown are profiles for course 1 of the 12‐mg cohort (upper left), course 1 of the 24‐mg cohort (upper right), course 2 of the 12‐mg cohort (lower left) and course 2 of the 24‐mg cohort (lower right). Serum concentrations increased with each daily administration within a treatment course, with the highest observed concentrations occurring following the last dose. The mean maximum serum concentration (Cmax) values were comparable between courses. Serum concentrations became low or undetectable (LOQ = 60 ng/ml) within approximately 30 days following each treatment course in the 12‐mg dose group and within approximately 90 days following each treatment course in the 24‐mg dose group. D = day, M = month.
Figure 2
Figure 2
Mean total lymphocyte counts over time (pooled data from CARE‐MS I and CARE‐MS II). Alemtuzumab 12 mg (grey line with solid circles) or 24 mg (black line with solid circles) depleted circulating lymphocytes after each course, and repopulated after depletion. Dashed lines shown are upper limit of normal (ULN) and lower limit of normal (LLN) as marked. Mean total lymphocyte counts were reduced by 88% from baseline at month 1, were at the LLN by month 6 and were 133% of the LLN and 51% of baseline 12 months after course 1. Following the second course of alemtuzumab, mean total lymphocyte count decreased from the month 12 precourse value by 72% at month 13, which corresponded to 38% of the LLN. Mean total lymphocyte counts were at the LLN at month 18. The month 24 mean value was 139% of the LLN and 54% of baseline.
Figure 3
Figure 3
Median anti‐alemtuzumab antibody (ADA) and inhibitory ADA titres over time (pooled data from CARE‐MS I and CARE‐MS II). For ADA (upper panel), in the 12‐mg group (dashed line with open circles) and the 24‐mg group (solid line with filled circle), the median titres were similar and peaked at 1 month post‐dose after each treatment course (months 1 and 13). Peak ADA titres were higher following course 2 than course 1. For inhibitory ADA (lower panel), for both dose groups and in both treatment courses, titres peaked at 1 month after dose (months 1 and 13 for courses 1 and 2, respectively). Peak inhibitory ADA titres were higher in course 2, as was observed for ADA. The observed difference in median inhibitory ADA titre between dose groups was only one dilution, which is considered within the assay variability.
Figure 4
Figure 4
Mean lymphocyte and subset counts over time after each course of alemtuzumab (12 mg/day) by precourse or within‐course anti‐alemtuzumab antibody status in CARE‐MS I and CARE‐MS II. Throughout two courses of treatment, the presence and titre level of ADA or inhibitory ADA generally had no discernible effects on T or B lymphocyte depletion or repopulation for CD3+, CD4+, CD8+ and CD19+ lymphocyte subsets. The absolute counts of CD3+, CD4+, CD8+ and CD19+ cells were similar for patients with ADA or inhibitory ADA compared with those who were always negative for antibodies. CD3+, CD4+, CD8+ and CD19+ cells were depleted by month 1 for patients, irrespective of their ADA or inhibitory ADA status. ADA or inhibitory ADA‐positive status appeared to be associated with reduced depletion of CD16+CD56+ cells by alemtuzumab. The presence of precourse or within‐course ADA was associated with reduced depletion of CD16+CD56+ cells by alemtuzumab.
Figure 5
Figure 5
Pharmacokinetic–pharmacodynamic model used to model lymphocyte counts. The basic PD model was an indirect response model and was built based on the assumption that alemtuzumab causes cell lysis and acts to increase the rate of lymphocyte degradation through a direct effect of its concentration in plasma. Under this model, lymphocytes are produced at a constant zero‐order rate per day (Kin) and eliminated by a first‐order degradation rate (Kout), and the baseline lymphocyte count prior to treatment is equal to the ratio of Kin to Kout. The drug effect was linear and was only included on Kout, the rate of removal of the lymphocyte subtype. Sampling compartments are denoted by lines with solid circles at the end. V1 = volume of distribution of the central compartment; V2 = volume of distribution of the peripheral compartment; Q = intercompartmental clearance; CL = total systemic clearance; A(1) = the amount of alemtuzumab in the central compartment; Kin = zero‐order rate of production of lymphocytes; Kout = first‐order rate of lymphocyte degradation; Cp = alemtuzumab serum concentration; S = slope of the linear relationship between alemtuzumab serum concentration and its effect on lymphocyte degradation.
Figure 6
Figure 6
Observed total lymphocyte count versus time by observed anti‐alemtuzumab antibody status (pooled CARE‐MS I and CARE‐MS II data) for the first 100 days after dosing. A covariate in the total lymphocyte PD model was ADA where positive ADA status resulted in a slope of the cell loss–alemtuzumab concentration relationship that was less than negative ADA status, indicating a greater susceptibility of some or all lymphocytes to the effects of alemtuzumab when ADA is not present, or of lower availability of active alemtuzumab in the presence of ADA. During lymphocyte repopulation the profiles of the counts are approximately the same, and there was no apparent difference by ADA status.
Figure 7
Figure 7
Total lymphocyte count percentage of baseline area under the effect curve (AUEC) versus alemtuzumab exposure (pooled CARE‐MS I and CARE‐MS II data) [left panel: area under the curve (AUC); right panel: maximum serum concentration (Cmax)]. At each treatment course, there was a decrease in total lymphocyte count upon exposure to alemtuzumab, with no apparent further decrease with increasing AUC or Cmax. Lymphocyte depletion was consistently observed upon exposure or second re‐exposure to alemtuzumab, without correlation to Cmax or AUC. There appears to be no difference in lymphocyte depletion or repopulation across the exposure range evaluated following administration of 12 or 24 mg alemtuzumab.

References

    1. Lassmann H. Neuropathology in multiple sclerosis: new concepts. Mult Scler 1998; 4:93–8.
    1. Lublin F, Reingold S, Cohen J et al Defining the clinical course of multiple sclerosis. The 2013 revisions. Neurology 2013; 2014:278–86.
    1. Riechmann L, Clark M, Waldmann H, Winter G. Reshaping human antibodies for therapy. Nature 1988; 332:323–7.
    1. Hale G, Dyer M, Clark M et al Remission induction in non‐Hodgkin lymphoma with reshaped human monoclonal antibody CAMPATH‐1H. Lancet 1988; 2: 1394–9.
    1. Mathieson PW, Cobbold SP, Hale G et al Monoclonal‐antibody therapy in systemic vasculitis. N Engl J Med 1990; 323:250–4.
    1. Weinblatt M, Maddison P, Bulpitt K et al Campath‐1H, a humanized monoclonal antibody, in refractory rheumatoid arthritis. An intravenous doseescalation study. Arthritis Rheum 1995; 38:1589–94.
    1. Hale G. The CD52 antigen and development of the CAMPATH antibodies. Cytotherapy 2001; 3:137–43.
    1. Kottaridis P, Milligan D, Chopra R et al In vivo CAMPATH‐1H prevents GvHD following nonmyeloablative stem‐cell transplantation. Cytotherapy 2001; 3:197–201.
    1. Keating M, Flinn I, Jain V et al Therapeutic role of alemtuzumab (Campath‐1H) in patients who have failed fludarabine: results of a large international study. Blood 2002; 99:3554–61.
    1. Kanda J, Lopez RD, Rizzieri DA. Alemtuzumab for the prevention and treatment of graft‐versus‐host disease. Int J Hematol 2011; 93:586–93.
    1. Migkou M, Dimopoulos MA, Gavriatopoulou M, Terpos E. Applications of monoclonal antibodies for the treatment of hematological malignancies. Expert Opin Biol Ther 2009; 9:207–20.
    1. Gribben JG, Hallek M. Rediscovering alemtuzumab: current and emerging therapeutic roles. Br J Haematol 2009; 144:818–31.
    1. Ravandi F, O’Brien S. Alemtuzumab in CLL and other lymphoid neoplasms. Cancer Invest 2006; 24:718–25.
    1. Coles A, Deans J, Compston A. Campath‐1H treatment of multiple sclerosis: lessons from the bedside for the bench. Clin Neurol Neurosurg 2004; 106:270–74.
    1. Hu Y, Turner MJ, Shields J et al Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model. Immunology 2009; 128:260–70.
    1. Xia MQ, Hale G, Lifely MR et al Structure of the CAMPATH‐1 antigen, a glycosylphosphatidylinositol‐anchored glycoprotein which is an exceptionally good target for complement lysis. Biochem J 1993; 293:633–40.
    1. Watanabe T, Masuyama J, Sohma Y et al CD52 is a novel costimulatory molecule for induction of CD4+ regulatory T cells. Clin Immunol 2006; 120:247–59.
    1. Masuyama J, Yoshio T, Suzuki K et al Characterization of the 4C8 antigen involved in transendothelial migration of CD26(hi) T cells after tight adhesion to human umbilical vein endothelial cell monolayers. J Exp Med 1999; 189:979–90.
    1. Bindon C, Hale G, Waldmann H. Importance of antigen specificity for complement‐mediated lysis by monoclonal antibodies. Eur J Immunol 1988; 18:1507–14.
    1. Hale C, Bartholomew M, Taylor V, Stables J, Topley P, Tite J. Recognition of CD52 allelic gene products by CAMPATH‐1H antibodies. Immunology 1996; 88:183–90.
    1. Crowe JS, Hall VS, Smith MA, Cooper HJ, Tite JP. Humanized monoclonal antibody CAMPATH‐1H: myeloma cell expression of genomic constructs, nucleotide sequence of cDNA constructs and comparison of effector mechanisms of myeloma and Chinese hamster ovary cell derived material. Clin Exp Immunol 1992; 87:105–10.
    1. Stanglmaier M, Reis S, Hallek M. Rituximab and alemtuzumab induce a nonclassic, caspase independent apoptotic pathway in B‐lymphoid cell lines and in chronic lymphocytic leukemia cells. Ann Hematol 2004; 83:634–45.
    1. Thompson S, Jones J, Cox A, Compston D, Coles A. B‐cell reconstitution and BAFF after alemtuzumab (Campath‐1H) treatment of multiple sclerosis. J Clin Immunol 2010; 30:99–105.
    1. Freedman MS, Kaplan JM, Markovic‐Plese S. Insights into the mechanisms of the therapeutic efficacy of alemtuzumab in multiple sclerosis. J Clin Cell Immunol 2013; 4:1000152.
    1. Jones J, Coles A. Mode of action and clinical studies with alemtuzumab. Exp Neurol 2014; 262:37–43.
    1. Cox A, Thompson S, Jones J et al Lymphocyte homeostasis following therapeutic lymphocyte depletion in multiple sclerosis. Eur J Immunol 2005; 35:3332–42.
    1. Jones J, Phuah C, Cox A et al IL‐21 drives secondary autoimmunity in patients with multiple sclerosis, following therapeutic lymphocyte depletion with alemtuzumab (Campath‐1H). J Clin Invest 2009; 119:2052–61.
    1. Jones J, Anderson J, Phuah C et al Improvement in disability after alemtuzumab treatment of multiple sclerosis is associated with neuroprotective autoimmunity. Brain 2010; 133:2232–47.
    1. Hill‐Cawthorne G, Button T, Tuohy O et al Long term lymphocyte reconstitution after alemtuzumab treatment of multiple sclerosis. J Neurol Neurosurg Psychiatry 2012; 83:298–304.
    1. Baker D, Herrod SS, Alvarez‐Gonzalez C, Giovannoni G, Schmierer K. Interpreting lymphocyte reconstitution data from the pivotal phase 3 trials of alemtuzumab. JAMA Neurol 2017; 74:961–9.
    1. Helliwell CL, Coles AJ. Monoclonal antibodies in multiple sclerosis treatment: current and future steps. Ther Adv Neurol Disord 2009; 2:195–203.
    1. Pineda C, Castañeda Hernández G, Jacobs IA, Alvarez DF, Carini C. Assessing the immunogenicity of biopharmaceuticals. Bio Drugs 2016; 30:195–206.
    1. Baert F, Noman M, Vermeire S et al Influence of immunogenicity on the long‐term efficacy of infliximab in Crohn's disease. N Engl J Med 2003; 348:601–8.
    1. Murdaca G, Spanò F, Contatore M et al Immunogenicity of infliximab and adalimumab: what is its role in hypersensitivity and modulation of therapeutic efficacy and safety? Expert Opin Drug Saf 2016; 15:43–52.
    1. Stüve O, Bennett JL. Pharmacological properties, toxicology and scientific rationale for the use of natalizumab (Tysabri) in inflammatory diseases. CNS Drug Rev 2007; 13:79–95.
    1. Tysabri® . Highlights of prescribing information. Available at: (revised 08/2017) (accessed 20 January 2018).
    1. Vartanian T, Solberg Sorensen P, Rice G. Impact of neutralizing antibodies on the clinical efficacy of interferon beta in multiple sclerosis. J Neurol 2004; 251(Suppl. 2): II25–30.
    1. Isaacs J, Watts R, Hazleman B et al Humanised monoclonal antibody therapy for rheumatoid arthritis. Lancet 1992; 340:748–52.
    1. Isaacs J, Manna V, Rapson N et al Campath‐1H in rheumatoid arthritis – an intravenous dose‐ranging study. Br J Rheumatol 1996; 35:231–40.
    1. Moreau T, Coles A, Wing M et al Transient increase in symptoms associated with cytokine release in patients with multiple sclerosis. Brain 1996; 119:225–37.
    1. Jones D, Goldman M. Alemtuzumab for the treatment of relapsing remitting multiple sclerosis: a review of its clinical pharmacology, efficacy and safety. Exp Rev Clin Immunol 2014; 10:1281–91.
    1. Willis M, Robertson N. Alemtuzumab for the treatment of multiple sclerosis. Ther Clin Risk Manag 2015; 11:525–34.
    1. Boyko A, Havrdova E, King J et al Lymphocyte depletion and repopulation is consistent across alemtuzumab treatment courses in patients with relapsing–remitting multiple sclerosis: 6‐year analysis of patients from the CARE‐MS Studies. Mult Scler 2016; 22:P654.
    1. von Kutzleben S, Pryce G, Giovannoni G, Baker D. Depletion of CD52‐positive cells inhibits the development of central nervous system autoimmune disease, but deletes an immune‐tolerance promoting CD8 T‐cell population. Implications for secondary autoimmunity of alemtuzumab in multiple sclerosis. Immunology 2017; 150:444–55.
    1. Noris M, Casiraghi F, Todeschini M et al Regulatory T cells and T cell depletion: role of immunosuppressive drugs. J Am Soc Nephrol 2007; 18:1007–18.
    1. Zhang X, Tao Y, Chopra M et al Differential reconstitution of T cell subsets following immunodepleting treatment with alemtuzumab (anti‐CD52 monoclonal antibody) in patients with relapsing‐remitting multiple sclerosis. J Immunol 2013; 191:5867–74.
    1. Thomas K, Metz I, Tumani H, Brück W, Ziemssen T. 6‐sulfoLacNAc(+) dendritic cells accumulate in various inflammatory, but not ischemic conditions of the central nervous system. Neuropathol Appl Neurobiol 2016; 42:394–8.
    1. Gross CC, Ahmetspahic D, Ruck T et al Alemtuzumab treatment alters circulating innate immune cells in multiple sclerosis. Neurol Neuroimmunol Neuroinflamm 2016; 3:e289.
    1. Ziemssen T, Thomas K. Alemtuzumab in the long‐term treatment of relapsing‐remitting multiple sclerosis: an update on the clinical trial evidence and data from the real world. Ther Adv Neurol Disord 2017; 10:343–59.
    1. Cohen JA, Coles AJ, Arnold DL et al Alemtuzumab versus interferon beta 1a as first‐line treatment for patients with relapsing–remitting multiple sclerosis: a randomised controlled phase 3 trial. Lancet 2012; 380:1819–28.
    1. Zucchini N, Crozat K, Baranek T, Robbins SH, Altfeld M, Dalod M. Natural killer cells in immunodefense against infective agents. Expert Rev Anti Infect Ther 2008; 6:867–85.
    1. Malmberg KJ, Bryceson YT, Carlsten M et al NK cell‐mediated targeting of human cancer and possibilities for new means of immunotherapy. Cancer Immunol Immunother 2008; 57:1541–52.
    1. Terunuma H, Deng X, Dewan Z, Fujimoto S, Yamamoto N. Potential role of NK cells in the induction of immune responses: implications for NK cell‐based immunotherapy for cancers and viral infections. Int Rev Immunol 2008; 27:93–110.

Source: PubMed

3
Prenumerera