Tolerability, response and outcome of high-risk neuroblastoma patients treated with long-term infusion of anti-GD2 antibody ch14.18/CHO

Ina Mueller, Karoline Ehlert, Stefanie Endres, Lena Pill, Nikolai Siebert, Silke Kietz, Penelope Brock, Alberto Garaventa, Dominique Valteau-Couanet, Evelyne Janzek, Norbert Hosten, Andreas Zinke, Winfried Barthlen, Emine Varol, Hans Loibner, Ruth Ladenstein, Holger N Lode, Ina Mueller, Karoline Ehlert, Stefanie Endres, Lena Pill, Nikolai Siebert, Silke Kietz, Penelope Brock, Alberto Garaventa, Dominique Valteau-Couanet, Evelyne Janzek, Norbert Hosten, Andreas Zinke, Winfried Barthlen, Emine Varol, Hans Loibner, Ruth Ladenstein, Holger N Lode

Abstract

Immunotherapy with short term infusion (STI) of monoclonal anti-GD2 antibody (mAb) ch14.18 (4 × 25 mg/m2/d; 8-20 h) in combination with cytokines and 13-cis retinoic acid (RA) prolonged survival in high-risk neuroblastoma (NB) patients. Here, we investigated long-term infusion (LTI) of ch14.18 produced in Chinese hamster ovary cells (ch14.18/CHO; 10 × 10 mg/m2; 24 h) in combination with subcutaneous (s.c.) interleukin-2 (IL-2) in a single center program and report clinical response, toxicity and survival. Fifty-three high-risk NB patients received up to 6 cycles of 100 mg/m2 ch14.18/CHO (d8-17) as LTI combined with 6 × 106 IU/m2 s.c. IL-2 (d1-5; 8-12) and 160 mg/m2 oral RA (d19-32). Pain toxicity was documented with validated pain scores and intravenous (i.v.) morphine usage. Response was assessed in 37/53 evaluable patients following International Neuroblastoma Risk Group criteria. Progression-free (PFS) and overall survival (OS) was analyzed by the Kaplan-Meier method and compared to a matched historical control group from the database of AIEOP, the "Italian Pediatric Ematology and Oncology Association". LTI of ch14.18/CHO showed acceptable toxicity profile indicated by low pain scores, reduced i.v. morphine usage and low frequency of Grade ≥3 adverse events that allowed outpatient treatment. We observed a best response rate of 40.5% (15/37; 5 CR, 10 PR), 4-year (4 y) PFS of 33.1% (observation 0.1- 4.9 y, mean: 2.2 y) and a 4 y OS of 47.7% (observation 0.27 - 5.20 y, mean: 3.6 y). Survival of the entire cohort (53/53) and the relapsed patients (29/53) was significantly improved compared to historical controls. LTI of ch14.18/CHO thus shows an acceptable toxicity profile, objective clinical responses and a strong signal of clinical efficacy in NB patients.

Keywords: anti-GD2 antibody; ch14.18/CHO; immunotherapy; neuroblastoma.

Figures

Figure 1.
Figure 1.
Treatment schematic, pain assessment and intravenous morphine usage during LTI of ch14.18/CHO. A) Ch14.18/CHO was administered by LTI of 100 mg/m2 (d8–17) (horizontal bar) with 6 × 106 IU/m2 s.c. IL-2 (d1–5; 8–12) (black arrows) and p.o. isotretinoin (160 mg/m2/day d22–35). Pain toxicity of anti-GD2 antibody ch14.18/CHO was evaluated by systematic assessments of pain scores and intravenous morphine usage of 49/53 evaluable patients as described in the “Patients and Methods” section. B) Pain assessment scores were determined three times daily per patient and cycle. Data represent mean ± SEM. C) Usage of i.v. morphine in µg/kg/h was determined daily per patient and cycle and presented as mean ± SEM. When error bars are not visible, they are covered by the symbol. Total morphine usage per cycle ± SEM is indicated in mg/kg/cycle.
Figure 2.
Figure 2.
Analysis of survival and time to progression following LTI of ch14.18/CHO. Patients treated by LTI of 100 mg/m2 ch14.18/CHO in combination with 6 × 106 IU/m2 s.c. IL-2 (d1–5; 8–12) and oral 13-cis RA (d19–32) were analyzed for progression-free survival (PFS) (A) and overall survival (OS) (B) using the Kaplan-Meier method. Patients of the entire cohort (n = 53) and patients with relapsed status at base line (n = 29) were analyzed separately. A) PFS curves of the entire LTI cohort (red) and relapsed patients (blue) (top panel). B) OS of the entire LTI cohort (red) and relapsed patients (blue) was compared to relapsed patients of the AIEOP data base not treated with ch14.18/CHO (green). The starting point of the AIEOP relapsed patients equals to the date of first relapse plus the median time between relapse and start of ch14.18/CHO therapy for the LTI patients (1 y 7 d). Patients in the AIEOP relapsed group who died before the auxiliary starting point were excluded. The difference between LTI relapsed- and AIEOP relapsed- patients was statistically significant (P = 0.002).

References

    1. Cheung NK, Dyer MA. Neuroblastoma: developmental biology, cancer genomics and immunotherapy. Nat Rev Cancer. 2013;13(6):397–411. PMID:23702928
    1. Simon T, Hero B, Faldum A, Handgretinger R, Schrappe M, Klingebiel T, Berthold F. Long term outcome of high-risk neuroblastoma patients after immunotherapy with antibody ch14.18 or oral metronomic chemotherapy. BMC Cancer. 2011;11:21. PMID:21244693
    1. Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, Smith M, Anderson B, Villablanca JG, Matthay KK, et al.. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363(14):1324–1334.
    1. Zeng Y, Fest S, Kunert R, Katinger H, Pistoia V, Michon J, Lewis G, Ladenstein R, Lode HN. Anti-neuroblastoma effect of ch14.18 antibody produced in CHO cells is mediated by NK-cells in mice. Mol Immunol. 2005;42(11):1311–1319.
    1. Ladenstein R, Weixler S, Baykan B, Bleeke M, Kunert R, Katinger D, Pribill I, Glander P, Bauer S, Pistoia V, et al.. Ch14.18 antibody produced in CHO cells in relapsed or refractory Stage 4 neuroblastoma patients: a SIOPEN Phase 1 study. MAbs. 2013;5(5):801–809.
    1. Cheung NK, Lazarus H, Miraldi FD, Abramowsky CR, Kallick S, Saarinen UM, Spitzer T, Strandjord SE, Coccia PF, Berger NA. Ganglioside GD2 specific monoclonal antibody 3F8: a phase I study in patients with neuroblastoma and malignant melanoma. J Clin Oncol. 1987;5(9):1430–1440. PMID:3625258
    1. Yu AL, Uttenreuther-Fischer MM, Huang CS, Tsui CC, Gillies SD, Reisfeld RA, Kung FH. Phase I trial of a human-mouse chimeric anti-disialoganglioside monoclonal antibody ch14.18 in patients with refractory neuroblastoma and osteosarcoma. J Clin Oncol. 1998;16(6):2169–2180. PMID:9626218
    1. Handgretinger R, Anderson K, Lang P, Dopfer R, Klingebiel T, Schrappe M, Reuland P, Gillies SD, Reisfeld RA, Neithammer D. A phase I study of human/mouse chimeric antiganglioside GD2 antibody ch14.18 in patients with neuroblastoma. Eur J Cancer. 1995;31A(2):261–267. PMID:7718335
    1. Xiao WH, Yu AL, Sorkin LS. Electrophysiological characteristics of primary afferent fibers after systemic administration of anti-GD2 ganglioside antibody. Pain. 1997;69(1–2):145–151. PMID:9060025
    1. Navid F, Sondel PM, Barfield R, Shulkin BL, Kaufman RA, Allay JA, Gan J, Hutson P, Seo S, Kim K, et al.. Phase I trial of a novel anti-GD2 monoclonal antibody, Hu14.18K322A, designed to decrease toxicity in children with refractory or recurrent neuroblastoma. J Clin Oncol. 2014;32(14):1445–1452. doi:10.1200/JCO.2013.50.4423. PMID:24711551
    1. Moreno L, Rubie H, Varo A, Le Deley MC, Amoroso L, Chevance A, Garaventa A, Gambart M, Bautista F, Valteau-Couanet D, et al.. Outcome of children with relapsed or refractory neuroblastoma: A meta-analysis of ITCC/SIOPEN European phase II clinical trials. Pediatr Blood Cancer. 2017;64(1):25–31. doi:10.1002/pbc.26192. PMID:27555472
    1. Sorkin LS, Otto M, Baldwin WM III, Vail E, Gillies SD, Handgretinger R, Barfield RC, Ming Yu H, Yu AL. Anti-GD(2) with an FC point mutation reduces complement fixation and decreases antibody-induced allodynia. Pain. 2010;149(1):135–142. doi:10.1016/j.pain.2010.01.024. PMID:20171010
    1. Siebert N, Eger C, Seidel D, Juttner M, Zumpe M, Wegner D, Kietz S, Ehlert K, Veal GJ, Siegmund W, et al.. Pharmacokinetics and pharmacodynamics of ch14.18/CHO in relapsed/refractory high-risk neuroblastoma patients treated by long-term infusion in combination with IL-2. MAbs. 2016;8(3):604–616. doi:10.1080/19420862.2015.1130196. PMID:26785755
    1. London WB, Bagatell R, Weigel B, Fox E, van Ryn C, Naranjo A, et al.. Historical gold standard for time-to-progression (TTP) and progression-free survival (PFS) from relapsed/refractory neuroblastoma modern era (2002–2014) patients. J Clin Oncol. 2014;32(5s):5–6. PMID:24190110
    1. Cohn SL, Pearson AD, London WB, Monclair T, Ambros PF, Brodeur GM, Faldum A, Hero B, Iehara T, Machin D, et al.. The International Neuroblastoma Risk Group (INRG) classification system: an INRG task force report. J Clin Oncol. 2009;27(2):289–297. doi:10.1200/JCO.2008.16.6785. PMID:19047291
    1. Buttner W, Breitkopf L, Miele B, Finke W. [Initial results of the reliability and validity of a German-language scale for the quantitative measurement of postoperative pain in young children]. Anaesthesist. 1990;39(11):593–602. PMID:2288408
    1. Ramsay MA, Savege TM, Simpson BR, Goodwin R. Controlled sedation with alphaxalone-alphadolone. Br Med J. 1974;2(5920):656–659. doi:10.1136/bmj.2.5920.656. PMID:4835444
    1. Zernikow B, Hechler T. Pain therapy in children and adolescents. Dtsch Arztebl Int. 2008;105(28–29):511–521. PMID:19626208
    1. Wilson GA, Doyle E. Validation of three paediatric pain scores for use by parents. Anaesthesia. 1996;51(11):1005–1007. doi:10.1111/j.1365-2044.1996.tb14991.x. PMID:8943588
    1. Mehes G, Luegmayr A, Hattinger CM, Lorch T, Ambros IM, Gadner H, Ambros PF. Automatic detection and genetic profiling of disseminated neuroblastoma cells. Med Pediatr Oncol. 2001;36(1):205–209. doi:10.1002/1096-911X(20010101)36:1%3c205::AID-MPO1050%;2-G. PMID:11464886
    1. Garaventa A, Parodi S, De BB, Dau D, Manzitti C, Conte M, Casale F, Viscardi E, Bianchi M, D'Angelo P, et al.. Outcome of children with neuroblastoma after progression or relapse. A retrospective study of the Italian neuroblastoma registry. Eur J Cancer. 2009;45(16):2835–2842. doi:10.1016/j.ejca.2009.06.010. PMID:19616426

Source: PubMed

3
Suscribir