Sutimlimab in patients with cold agglutinin disease: results of the randomized placebo-controlled phase 3 CADENZA trial

Alexander Röth, Sigbjørn Berentsen, Wilma Barcellini, Shirley D'Sa, Bernd Jilma, Marc Michel, Ilene C Weitz, Masaki Yamaguchi, Jun-Ichi Nishimura, Josephine M I Vos, Michael Storek, Nancy Wong, Parija Patel, Xiaoyu Jiang, Deepthi S Vagge, Marek Wardęcki, Frank Shafer, Michelle Lee, Catherine M Broome, Alexander Röth, Sigbjørn Berentsen, Wilma Barcellini, Shirley D'Sa, Bernd Jilma, Marc Michel, Ilene C Weitz, Masaki Yamaguchi, Jun-Ichi Nishimura, Josephine M I Vos, Michael Storek, Nancy Wong, Parija Patel, Xiaoyu Jiang, Deepthi S Vagge, Marek Wardęcki, Frank Shafer, Michelle Lee, Catherine M Broome

Abstract

Sutimlimab, a first-in-class humanized immunoglobulin G4 (IgG4) monoclonal antibody that selectively inhibits the classical complement pathway at C1s, rapidly halted hemolysis in the single-arm CARDINAL study in recently transfused patients with cold agglutinin disease (CAD). CADENZA was a 26-week randomized, placebo-controlled phase 3 study to assess safety and efficacy of sutimlimab in patients with CAD without recent (within 6 months prior to enrollment) transfusion history. Forty-two patients with screening hemoglobin ≤10 g/dL, elevated bilirubin, and ≥1 CAD symptom received sutimlimab (n = 22) or placebo (n = 20) on days 0 and 7 and then biweekly. Composite primary endpoint criteria (hemoglobin increase ≥1.5 g/dL at treatment assessment timepoint [mean of weeks 23, 25, 26], avoidance of transfusion, and study-prohibited CAD therapy [weeks 5-26]) were met by 16 patients (73%) on sutimlimab, and 3 patients (15%) on placebo (odds ratio, 15.9 [95% confidence interval, 2.9, 88.0; P < .001]). Sutimlimab, but not placebo, significantly increased mean hemoglobin and FACIT-Fatigue scores at treatment assessment timepoint. Sutimlimab normalized mean bilirubin by week 1. Improvements correlated with near-complete inhibition of the classical complement pathway (2.3% mean activity at week 1) and C4 normalization. Twenty-one (96%) sutimlimab patients and 20 (100%) placebo patients experienced ≥1 treatment-emergent adverse event. Headache, hypertension, rhinitis, Raynaud phenomenon, and acrocyanosis were more frequent with sutimlimab vs placebo, with a difference of ≥3 patients between groups. Three sutimlimab patients discontinued owing to adverse events; no placebo patients discontinued. These data demonstrate that sutimlimab has potential to be an important advancement in the treatment of CAD. This trial was registered at www.clinicaltrials.gov as #NCT03347422.

© 2022 by The American Society of Hematology.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Effect of sutimlimab on a composite primary endpoint comprising Hb levels, transfusions, and need for CAD medications in patients with CAD. For the composite primary endpoint, sutimlimab was compared with placebo using the Cochran-Mantel-Haenszel method, stratified by baseline Hb (< median vs ≥ median) and geographic region (Asia/Other, North America, and Europe). Hb increase from baseline of ≥1.5 g/dL was analyzed at the treatment assessment timepoint, defined as the mean average of weeks 23, 25, and 26. Requirements for transfusion included Hb < 9 g/dL and patient symptomatic or Hb < 7 g/dL and patient asymptomatic. One patient in the sutimlimab arm discontinued treatment prematurely owing to an adverse event (increased blood IgM) and started rituximab treatment during the 9-week posttreatment follow-up period; 2 patients in the sutimlimab arm discontinued prior to week 23, and their statuses were therefore “unknown” for this analysis. BL, baseline.
Figure 2.
Figure 2.
Effect of sutimlimab on Hb levels from baseline to week 26. Sutimlimab treatment resulted in rapid and sustained increase in Hb levels. B, baseline.
Figure 3.
Figure 3.
Effect of sutimlimab on markers of hemolysis from baseline to week 26. (A) Analysis of the change from baseline in bilirubin excluded patients with either a positive Gilbert syndrome genetic test or no test result. Overall, 21 of 22 patients in the sutimlimab arm and 18 of 20 patients in the placebo arm consented to receive Gilbert syndrome testing. Of these, no patient had a positive result, and 1 patient had unknown result and was excluded from the analyses. The normal range for bilirubin was defined as 5.1 to 20.5 µmol/L. (B) Normalization of haptoglobin was defined as greater than the level of haptoglobin detection. (C) Parameters for reticulocyte normalization were not defined for this study. (D) The normal range for LDH was defined as 120 to 246 U/L. B, baseline.
Figure 4.
Figure 4.
Effect of sutimlimab on FACIT-Fatigue Scale scores from baseline to week 26. FACIT-Fatigue Scale scores range from 0 to 52, with a higher score indicating less fatigue. In CAD, a change of 5 is estimated to be a clinically important change. B, baseline.
Figure 5.
Figure 5.
Effect of sutimlimab on classical complement pathway activity. (A) In the Wieslab Complement System Classical Pathway assay, the normal range in serum is 69% to 129%. (B) The International System of Units reference range for serum C4 is 0.18 to 0.45 g/L. Baseline classical complement pathway activity and total C4 levels below the normal ranges are expected in CAD and are consistent with the nature of this disease. B, baseline; C4, complement component 4; CP, classical complement pathway.

References

    1. Berentsen S. Complement activation and inhibition in autoimmune hemolytic anemia: focus on cold agglutinin disease. Semin Hematol. 2018;55(3): 141-149.
    1. Berentsen S, Ulvestad E, Langholm R, et al. . Primary chronic cold agglutinin disease: a population based clinical study of 86 patients. Haematologica. 2006;91(4): 460-466.
    1. Röth A, Fryzek J, Jiang X, et al. . Complement-mediated hemolysis persists year round in patients with cold agglutinin disease. Transfusion. 2022; 62(1):51-59.
    1. Berentsen S. Cold agglutinin disease. Hematology Am Soc Hematol Educ Program. 2016;2016:226-231.
    1. Randen U, Trøen G, Tierens A, et al. . Primary cold agglutinin-associated lymphoproliferative disease: a B-cell lymphoma of the bone marrow distinct from lymphoplasmacytic lymphoma. Haematologica. 2014;99(3):497-504.
    1. Jäger U, Barcellini W, Broome CM, et al. . Diagnosis and treatment of autoimmune hemolytic anemia in adults: recommendations from the First International Consensus Meeting. Blood Rev. 2020;41:100648.
    1. Berentsen S, Randen U, Tjønnfjord GE. Cold agglutinin-mediated autoimmune hemolytic anemia. Hematol Oncol Clin North Am. 2015;29(3):455-471.
    1. Berentsen S. New insights in the pathogenesis and therapy of cold agglutinin-mediated autoimmune hemolytic anemia. Front Immunol. 2020;11:590.
    1. Pruzanski W, Shumak KH. Biologic activity of cold-reacting autoantibodies (first of two parts). N Engl J Med. 1977;297(10):538-542.
    1. Zanella A, Barcellini W. Treatment of autoimmune hemolytic anemias. Haematologica. 2014;99(10):1547-1554.
    1. Berentsen S, Röth A, Randen U, Jilma B, Tjønnfjord GE. Cold agglutinin disease: current challenges and future prospects. J Blood Med. 2019;10:93-103.
    1. Baines AC, Brodsky RA. Complementopathies. Blood Rev. 2017; 31(4):213-223.
    1. Berentsen S. Role of complement in autoimmune hemolytic anemia. Transfus Med Hemother. 2015;42(5):303-310.
    1. Jaffe CJ, Atkinson JP, Frank MM. The role of complement in the clearance of cold agglutinin-sensitized erythrocytes in man. J Clin Invest. 1976;58(4):942-949.
    1. Berentsen S, Beiske K, Tjønnfjord GE. Primary chronic cold agglutinin disease: an update on pathogenesis, clinical features and therapy. Hematology. 2007;12(5): 361-370.
    1. Jäger U, D’Sa S, Schörgenhofer C, et al. . Inhibition of complement C1s improves severe hemolytic anemia in cold agglutinin disease: a first-in-human trial. Blood. 2019;133(9):893-901.
    1. Gelbenegger G, Schoergenhofer C, Derhaschnig U, et al. . Inhibition of complement C1s in patients with cold agglutinin disease: lessons learned from a named patient program. Blood Adv. 2020;4(6):997-1005.
    1. Shi J, Rose EL, Singh A, et al. . TNT003, an inhibitor of the serine protease C1s, prevents complement activation induced by cold agglutinins. Blood. 2014;123(26):4015-4022.
    1. Broome CM, Cunningham JM, Mullins M, et al. . Increased risk of thrombotic events in cold agglutinin disease: A 10-year retrospective analysis. Res Pract Thromb Haemost. 2020;4(4):628-635.
    1. Bylsma LC, Gulbech Ording A, Rosenthal A, et al. . Occurrence, thromboembolic risk, and mortality in Danish patients with cold agglutinin disease. Blood Adv. 2019;3(20):2980-2985.
    1. Shafi M, Taufiq F, Mehmood H, Afsar S, Badar A. Relation between depressive disorder and iron deficiency anemia among adults reporting to a secondary healthcare facility: a hospital-based case control study. J Coll Physicians Surg Pak. 2018;28(6): 456-559.
    1. Vulser H, Wiernik E, Hoertel N, et al. . Association between depression and anemia in otherwise healthy adults. Acta Psychiatr Scand. 2016;134(2):150-160.
    1. Mahdi N, Al-Ola K, Khalek NA, Almawi WY. Depression, anxiety, and stress comorbidities in sickle cell anemia patients with vaso-occlusive crisis. J Pediatr Hematol Oncol. 2010;32(5):345-349.
    1. Kallich JD, Tchekmedyian NS, Damiano AM, Shi J, Black JT, Erder MH. Psychological outcomes associated with anemia-related fatigue in cancer patients. Oncology (Williston Park). 2002;16(9 suppl 10):117-124.
    1. Patel P, Jiang X, Nicholson G, et al. . Medically attended anxiety or depression is increased among newly diagnosed patients with cold agglutinin disease (CAD) [abstract]. Blood. 2020;136(suppl 1):28. Abstract 1631.
    1. Mullins M, Jiang X, Bylsma LC, et al. . Cold agglutinin disease burden: a longitudinal analysis of anemia, medications, transfusions, and health care utilization. Blood Adv. 2017;1(13):839-848.
    1. Barcellini W, Fattizzo B, Zaninoni A, et al. . Clinical heterogeneity and predictors of outcome in primary autoimmune hemolytic anemia: a GIMEMA study of 308 patients. Blood. 2014;124(19):2930-2936.
    1. Barcellini W, Zaja F, Zaninoni A, et al. . Sustained response to low-dose rituximab in idiopathic autoimmune hemolytic anemia. Eur J Haematol. 2013;91(6):546-551.
    1. Berentsen S, Ulvestad E, Gjertsen BT, et al. . Rituximab for primary chronic cold agglutinin disease: a prospective study of 37 courses of therapy in 27 patients. Blood. 2004;103(8):2925-2928.
    1. Reynaud Q, Durieu I, Dutertre M, et al. . Efficacy and safety of rituximab in auto-immune hemolytic anemia: a meta-analysis of 21 studies. Autoimmun Rev. 2015;14(4):304-313.
    1. Schöllkopf C, Kjeldsen L, Bjerrum OW, et al. . Rituximab in chronic cold agglutinin disease: a prospective study of 20 patients. Leuk Lymphoma. 2006;47(2):253-260.
    1. Berentsen S, Randen U, Oksman M, et al. . Bendamustine plus rituximab for chronic cold agglutinin disease: results of a Nordic prospective multicenter trial. Blood. 2017;130(4):537-541.
    1. Berentsen S, Randen U, Vågan AM, et al. . High response rate and durable remissions following fludarabine and rituximab combination therapy for chronic cold agglutinin disease. Blood. 2010;116(17):3180-3184.
    1. Röth A, Bommer M, Hüttmann A, et al. . Eculizumab in cold agglutinin disease (DECADE): an open-label, prospective, bicentric, nonrandomized phase 2 trial. Blood Adv. 2018;2(19):2543-2549.
    1. Berentsen S. How I treat cold agglutinin disease. Blood. 2021;137(10):1295-1303.
    1. Hill QA, Stamps R, Massey E, Grainger JD, Provan D, Hill A; British Society for Haematology . The diagnosis and management of primary autoimmune haemolytic anaemia. Br J Haematol. 2017;176(3):395-411.
    1. Berentsen S, Tjønnfjord GE. Diagnosis and treatment of cold agglutinin mediated autoimmune hemolytic anemia. Blood Rev. 2012;26(3):107-115.
    1. Röth A, Barcellini W, D’Sa S, et al. . Sutimlimab in cold agglutinin disease. N Engl J Med. 2021;384(14):1323-1334.
    1. Bartko J, Schoergenhofer C, Schwameis M, et al. . A randomized, first-in-human, healthy volunteer trial of sutimlimab, a humanized antibody for the specific inhibition of the classical complement pathway. Clin Pharmacol Ther. 2018;104(4): 655-663.
    1. Colonna L, Parry GC, Panicker S, Elkon KB. Uncoupling complement C1s activation from C1q binding in apoptotic cell phagocytosis and immunosuppressive capacity. Clin Immunol. 2016;163:84-90.
    1. Lewis LA, Panicker S, DeOliveira RB, Parry GC, Ram S. Effect of a C1s inhibitor on the efficacy of anti-capsular antibodies against Neisseria meningitidis and Streptococcus pneumoniae. Immunohorizons. 2019;3(11):519-530.
    1. Hill QA, Röth A, Jilma B, et al. . Clinically important change in FACIT-Fatigue score for patients with cold agglutinin disease: an analysis using the phase 3 Cardinal and Cadenza studies. European Hematology Association Virtual Congress 2021. Abstract EP1179.
    1. Cella D, Yount S, Sorensen M, Chartash E, Sengupta N, Grober J. Validation of the Functional Assessment of Chronic Illness Therapy Fatigue Scale relative to other instrumentation in patients with rheumatoid arthritis. J Rheumatol. 2005;32(5): 811-819.
    1. Cella D, Eton DT, Lai JS, Peterman AH, Merkel DE. Combining anchor and distribution-based methods to derive minimal clinically important differences on the Functional Assessment of Cancer Therapy (FACT) anemia and fatigue scales. J Pain Symptom Manage. 2002;24(6): 547-561.
    1. Escalante CP, Chisolm S, Song J, et al. . Fatigue, symptom burden, and health-related quality of life in patients with myelodysplastic syndrome, aplastic anemia, and paroxysmal nocturnal hemoglobinuria. Cancer Med. 2019;8(2):543-553.
    1. Schrezenmeier H, Röth A, Araten DJ, et al. . Baseline clinical characteristics and disease burden in patients with paroxysmal nocturnal hemoglobinuria (PNH): updated analysis from the International PNH Registry. Ann Hematol. 2020;99(7):1505-1514.
    1. Lai JS, Beaumont JL, Ogale S, Brunetta P, Cella D. Validation of the functional assessment of chronic illness therapy-fatigue scale in patients with moderately to severely active systemic lupus erythematosus, participating in a clinical trial. J Rheumatol. 2011;38(4):672-679.
    1. Reddy S, Bruera E, Pace E, Zhang K, Reyes-Gibby CC. Clinically important improvement in the intensity of fatigue in patients with advanced cancer. J Palliat Med. 2007;10(5):1068-1075.
    1. Nordin Å, Taft C, Lundgren-Nilsson Å, Dencker A. Minimal important differences for fatigue patient reported outcome measures – a systematic review. BMC Med Res Methodol. 2016;16:62.
    1. Weitz IC, Ueda Y, Shafer F, et al. . Inflammation and fatigue in patients with cold agglutinin disease (CAD): analysis from the phase 3 Cardinal study [abstract]. Blood. 2020;136(suppl 1):7-8. Abstract 759.

Source: PubMed

3
Suscribir