Rationale and design of the 2 by 2 factorial design GnG-trial: a randomized phase-III study to compare two schedules of gemtuzumab ozogamicin as adjunct to intensive induction therapy and to compare double-blinded intensive postremission therapy with or without glasdegib in older patients with newly diagnosed AML

Sonia Jaramillo, Johannes Krisam, Lucian Le Cornet, Markus Kratzmann, Lukas Baumann, Tim Sauer, Martina Crysandt, Andreas Rank, Dirk Behringer, Lino Teichmann, Martin Görner, Ralf-Ulrich Trappe, Christoph Röllig, Stefan Krause, Maher Hanoun, Olaf Hopfer, Gerhard Held, Sebastian Buske, Lars Fransecky, Sabine Kayser, Christoph Schliemann, Kerstin Schaefer-Eckart, Yousef Al-Fareh, Jörg Schubert, Thomas Geer, Martin Kaufmann, Arne Brecht, Dirk Niemann, Meinhard Kieser, Martin Bornhäuser, Uwe Platzbecker, Hubert Serve, Claudia D Baldus, Carsten Müller-Tidow, Richard F Schlenk, Sonia Jaramillo, Johannes Krisam, Lucian Le Cornet, Markus Kratzmann, Lukas Baumann, Tim Sauer, Martina Crysandt, Andreas Rank, Dirk Behringer, Lino Teichmann, Martin Görner, Ralf-Ulrich Trappe, Christoph Röllig, Stefan Krause, Maher Hanoun, Olaf Hopfer, Gerhard Held, Sebastian Buske, Lars Fransecky, Sabine Kayser, Christoph Schliemann, Kerstin Schaefer-Eckart, Yousef Al-Fareh, Jörg Schubert, Thomas Geer, Martin Kaufmann, Arne Brecht, Dirk Niemann, Meinhard Kieser, Martin Bornhäuser, Uwe Platzbecker, Hubert Serve, Claudia D Baldus, Carsten Müller-Tidow, Richard F Schlenk

Abstract

Background: Overall survival remains poor in older patients with acute myeloid leukemia (AML) with less than 10% being alive after 5 years. In recent studies, a significant improvement in event-free, relapse-free and overall survival was shown by adding gemtuzumab ozogamicin (GO), a humanized antibody-drug conjugate directed against CD33, to intensive induction therapy once or in a sequential dosing schedule. Glasdegib, the small-molecule inhibitor of smoothened (SMO), also showed improved overall survival in patients not eligible for intensive chemotherapy when combined with low-dose cytarabine compared to low-dose cytarabine alone. These findings warrant further investigations in the phase III GnG trial.

Methods/design: This is a randomized phase III trial with measurable residual disease (MRD) after induction therapy and event-free survival (EFS) as primary endpoints. The two research questions are addressed in a 2 by 2 factorial design. Patients age 60 years and older are upfront randomized 1:1 in one of the two induction arms: GO administered to intensive induction therapy on days 1,4, and 7 versus GO administered once on day 1 (GO-147 versus GO-1), and double-blinded 1:1 in one of the subsequent treatment arms glasdegib vs. placebo as adjunct to consolidation therapy and as single-agent maintenance therapy for six months. Chemotherapy backbone for induction therapy consists of standard 7 + 3 schedule with cytarabine 200 mg/m2 continuously days 1 to 7, daunorubicin 60 mg/m2 days 1, 2, and 3 and high-dose cytarabine (1 g/m2, bi-daily, days 1, 2, and 3) for consolidation therapy. Addressing two primary endpoints, MRD-negativity after induction therapy and event-free survival (EFS), 252 evaluable patients are needed to reject each of the two null hypotheses at a two-sided significance level of 2.5% with a power of at least 85%.

Ethics and dissemination: Ethical approval and approvals from the local and federal competent authorities were granted. Trial results will be reported via peer-reviewed journals and presented at conferences and scientific meetings.

Trial status: Protocol version: 1st version 20.10.2020, no amendments yet. Study initiation on February 16, 2021. First patient was recruited on April 1st.

Trial registration: ClinicalTrials.gov NCT04093505 ; EudraCT 2019-003913-32. Registered on October 30, 2018.

Keywords: acute myeloid leukemia; gemtuzumab ozogamicin; glasdegib; measurable residual disease.

Conflict of interest statement

All authors declare no support from any organization for the submitted work, no financial relationships with any organizations that might have an interest in the submitted work in the previous 3 years, and no other relationships or activities that could appear to have influenced the submitted work.

The funding organizations did not influence the study design nor will they influence the results of this trial.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Overall treatment schedule GnG-Study. Abbreviations: DA, daunorubicin; low-dose cytarabine; GO, gemtuzumab ozogamicin; HiDAC, high-dose cytarabine (1 g/m2); MRD, measurable residual disease; CR, complete remission; CRi, CR with incomplete hematological recovery. In case of bone marrow blast count > 10% or no CR/CRi after on day 15 after induction therapy one cycle of HAM (high-dose cytarabine and mitoxantrone) is allowed. Maintenance is intended in all patients in CR/CRi irrespective of completion of consolidation therapy

References

    1. Short NJ, Rytting ME, Cortes JE. Acute myeloid leukaemia. Lancet. 2018;392(10147):593–606. doi: 10.1016/S0140-6736(18)31041-9.
    1. Dombret H, Gardin C. An update of current treatments for adult acute myeloid leukemia. Blood. 2016;127(1):53–61. doi: 10.1182/blood-2015-08-604520.
    1. Schlenk RF, Döhner H. Genomic applications in the clinic: use in treatment paradigm of acute myeloid leukemia. Hematol Am Soc Hematol Educ Progr. 2013;2013(1):324–330. doi: 10.1182/asheducation-2013.1.
    1. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, Dombret H, Ebert BL, Fenaux P, Larson RA, Levine RL, Lo-Coco F, Naoe T, Niederwieser D, Ossenkoppele GJ, Sanz M, Sierra J, Tallman MS, Tien HF, Wei AH, Löwenberg B, Bloomfield CD. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–447. doi: 10.1182/blood-2016-08-733196.
    1. Nagel G, et al. Epidemiological, genetic, and clinical characterization by age of newly diagnosed acute myeloid leukemia based on an academic population-based registry study (AMLSG BiO) Ann Hematol. 2017;96(12):1993–2003. doi: 10.1007/s00277-017-3150-3.
    1. Schlenk RF. Post-remission therapy for acute myeloid leukemia. Haematologica. 2014;99(11):1663–1670. doi: 10.3324/haematol.2014.114611.
    1. Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, Gundem G, van Loo P, Martincorena I, Ganly P, Mudie L, McLaren S, O’Meara S, Raine K, Jones DR, Teague JW, Butler AP, Greaves MF, Ganser A, Döhner K, Schlenk RF, Döhner H, Campbell PJ. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N Engl J Med. 2016;374(23):2209–2221. doi: 10.1056/NEJMoa1516192.
    1. Gerstung M, Papaemmanuil E, Martincorena I, Bullinger L, Gaidzik VI, Paschka P, Heuser M, Thol F, Bolli N, Ganly P, Ganser A, McDermott U, Döhner K, Schlenk RF, Döhner H, Campbell PJ. Precision oncology for acute myeloid leukemia using a knowledge bank approach. Nat Genet. 2017;49(3):332–340. doi: 10.1038/ng.3756.
    1. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–2405. doi: 10.1182/blood-2016-03-643544.
    1. Larson RA, et al. Antibody-targeted chemotherapy of older patients with acute myeloid leukemia in first relapse using Mylotarg (gemtuzumab ozogamicin) Leukemia. 2002;16(9):1627–1636. doi: 10.1038/sj.leu.2402677.
    1. Petersdorf SH, Kopecky KJ, Slovak M, Willman C, Nevill T, Brandwein J, Larson RA, Erba HP, Stiff PJ, Stuart RK, Walter RB, Tallman MS, Stenke L, Appelbaum FR. A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. Blood. 2013;121(24):4854–4860. doi: 10.1182/blood-2013-01-466706.
    1. Delaunay J, Recher C, Pigneux A, Witz F, Vey N, Blanchet O,et al. Addition of gemtuzumab ozogamycin to chemotherapy improves event-free survival but not overall survival of AML patients with intermediate cytogenetics not eligible for allogeneic transplantation. Results of the GOELAMS AML 2006 IR Study. Blood. 2011;118(21):79. 10.1182/blood.V118.21.79.79.
    1. Burnett AK, Hills RK, Milligan D, Kjeldsen L, Kell J, Russell NH, Yin JAL, Hunter A, Goldstone AH, Wheatley K. Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial. J Clin Oncol. 2011;29(4):369–377. doi: 10.1200/JCO.2010.31.4310.
    1. Castaigne S, Pautas C, Terré C, Raffoux E, Bordessoule D, Bastie JN, Legrand O, Thomas X, Turlure P, Reman O, de Revel T, Gastaud L, de Gunzburg N, Contentin N, Henry E, Marolleau JP, Aljijakli A, Rousselot P, Fenaux P, Preudhomme C, Chevret S, Dombret H. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet. 2012;379(9825):1508–1516. doi: 10.1016/S0140-6736(12)60485-1.
    1. Lambert J, Pautas C, Terré C, Raffoux E, Turlure P, Caillot D, Legrand O, Thomas X, Gardin C, Gogat-Marchant K, Rubin SD, Benner RJ, Bousset P, Preudhomme C, Chevret S, Dombret H, Castaigne S. Gemtuzumab ozogamicin for de novo acute myeloid leukemia: final efficacy and safety updates from the open-label, phase III ALFA-0701 trial. Haematologica. 2019;104(1):113–119. doi: 10.3324/haematol.2018.188888.
    1. Burnett AK, Hills RK, Milligan D, Kjeldsen L, Kell J, Russell NH, et al. The Addition of Gemtuzumab Ozogamicin to Intensive Chemotherapy in Older Patients with AML Produces a Significant Improvement in Overall Survival: Results of the UK NCRI AML16 Randomized Trial. Blood. 2011;118(9):582–996. doi: 10.1016/S1470-2045(14)70281-5.
    1. Schlenk RF, Paschka P, Krzykalla J, Weber D, Kapp-Schwoerer S, Gaidzik VI, Leis C, Fiedler W, Kindler T, Schroeder T, Mayer K, Lübbert M, Wattad M, Götze K, Horst HA, Koller E, Wulf G, Schleicher J, Bentz M, Greil R, Hertenstein B, Krauter J, Martens U, Nachbaur D, Abu Samra M, Girschikofsky M, Basara N, Benner A, Thol F, Heuser M, Ganser A, Döhner K, Döhner H. Gemtuzumab Ozogamicin in NPM1-Mutated AML: Early Results from the Prospective Randomized AMLSG 09-09 Phase-III Study. J J Clin Oncol. 2020;38(6):623–632. doi: 10.1200/JCO.19.01406.
    1. Hills RK, Castaigne S, Appelbaum FR, Delaunay J, Petersdorf S, Othus M, Estey EH, Dombret H, Chevret S, Ifrah N, Cahn JY, Récher C, Chilton L, Moorman AV, Burnett AK. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014;15(9):986–996. doi: 10.1016/S1470-2045(14)70281-5.
    1. Burnett A, Cavenagh J, Russell N, Hills R, Kell J, Jones G, Nielsen OJ, Khwaja A, Thomas I, Clark R, on behalf of the UK NCRI AML Study Group Defining the dose of gemtuzumab ozogamicin in combination with induction chemotherapy in acute myeloid leukemia: a comparison of 3 mg/m2 with 6 mg/m2 in the NCRI AML17 Trial. Haematologica. 2016;101(6):724–731. doi: 10.3324/haematol.2016.141937.
    1. Lambert J, et al. MRD assessed by WT1 and NPM1 transcript levels identifies distinct outcomes in AML patients and is influenced by gemtuzumab ozogamicin. Oncotarget. 2014;5(15):6280–6288. doi: 10.18632/oncotarget.2196.
    1. Wadleigh M, Richardson PG, Zahrieh D, Lee SJ, Cutler C, Ho V, Alyea EP, Antin JH, Stone RM, Soiffer RJ, DeAngelo DJ. Prior gemtuzumab ozogamicin exposure significantly increases the risk of veno-occlusive disease in patients who undergo myeloablative allogeneic stem cell transplantation. Blood. 2003;102(5):1578–1582. doi: 10.1182/blood-2003-01-0255.
    1. Hütter-Krönke M-L, et al. Salvage therapy with high-dose cytarabine and mitoxantrone in combination with all-trans retinoic acid and gemtuzumab ozogamicin in acute myeloid leukemia refractory to first induction therapy. Haematologica. 2016;101(7):839–845. doi: 10.3324/haematol.2015.141622.
    1. Magwood-Golston JS, Kessler S, Bennett CL. Evaluation of gemtuzumab ozogamycin associated sinusoidal obstructive syndrome: Findings from an academic pharmacovigilance program review and a pharmaceutical sponsored registry. Leuk Res. 2016;44:61–64. doi: 10.1016/j.leukres.2016.03.004.
    1. Schlenk RF, Jaramillo S, Müller-Tidow C. What's new in consolidation therapy in AML? Semin Hematol. 2019;56(2):96–101. doi: 10.1053/j.seminhematol.2018.08.005.
    1. Löwenberg B, Beck J, Graux C, van Putten W, Schouten HC, Verdonck LF, Ferrant A, Sonneveld P, Jongen-Lavrencic M, von Lilienfeld-Toal M, Biemond BJ, Vellenga E, Breems D, de Muijnck H, Schaafsma R, Verhoef G, Döhner H, Gratwohl A, Pabst T, Ossenkoppele GJ, Maertens J, for the Dutch-Belgian Hemato-Oncology Cooperative Group (HOVON) German Austrian AML Study Group (AMLSG) Swiss Group for Clinical Cancer Research Collaborative Group (SAKK) Gemtuzumab ozogamicin as postremission treatment in AML at 60 years of age or more: results of a multicenter phase 3 study. Blood. 2010;115(13):2586–2591. doi: 10.1182/blood-2009-10-246470.
    1. Thol F, Schlenk RF. Gemtuzumab ozogamicin in acute myeloid leukemia revisited. Expert Opin Biol Ther. 2014;14(8):1185–1195. doi: 10.1517/14712598.2014.922534.
    1. Fukushima N, Minami Y, Kakiuchi S, Kuwatsuka Y, Hayakawa F, Jamieson C, Kiyoi H, Naoe T. Small-molecule Hedgehog inhibitor attenuates the leukemia-initiation potential of acute myeloid leukemia cells. Cancer Sci. 2016;107(10):1422–1429. doi: 10.1111/cas.13019.
    1. Sadarangani, A., Pineda, G., Lennon, K.M., Chun H.J., Shih A., Schairer A.E., Court A.C., Goff D.J., Prashad S.L., Geron I., Wall R., McPherson J.D., Moore R.A., Pu M., Bao L., Jackson-Fisher A., Munchhof M., VanArsdale T., Reya T., Morris S.R., Minden M.D., Messer K., Mikkola H.K.A., Marra M.A., Hudson T.J., Jamieson C.H.M. GLI2 inhibition abrogates human leukemia stem cell dormancy. J Transl Med 2015; doi:10.1186/s12967-015-0453-9. doi: 10.1186/s12967-015-0453-9, 13, 1, 98.
    1. Tauchi T, Okabe S, Katagiri Seiichiro, Yuko Tanaka Y, Tohyama K, Ohyashiki K. Targeting the Hedgehog Signaling Pathway By PF-04449913 Limits the Self-Renewal of MDS-Derived Induced Potent Stem Cells (iPSC): Molecular Mechanisms. Blood. 2015; doi:10.1182/blood.V126.23.791.791. doi: 10.4172/1948-5956.1000462.
    1. Martinelli G, Oehler VG, Papayannidis C,Courtney R, Shaik MN, Zhang X, et al. Treatment with PF-04449913, an oral smoothened antagonist, in patients with myeloid malignancies: a phase 1 safety and pharmacokinetics study. Lancet Haematol. 2015;doi: 10.1016/S2352-3026(15)00096-4.
    1. Savona MR, Pollyea DA, Stock W, Oehler VG, Schroeder MA, Lancet J, McCloskey J, Kantarjian HM, Ma WW, Shaik MN, Laird AD, Zeremski M, O'Connell A, Chan G, Cortes JE. Phase IB study of Glasdegib, a Hedgehog pathway inhibitor, in combination with standard chemotherapy in patients with AML or high-risk MDS. Clin Cancer Res. 2018;24(10):2294–2303. doi: 10.1158/1078-0432.CCR-17-2824.
    1. Cortes JE, Heidel FH, Hellmann A, Fiedler W, Smith BD, Robak T, Montesinos P, Pollyea DA, DesJardins P, Ottmann O, Ma WW, Shaik MN, Laird AD, Zeremski M, O’Connell A, Chan G, Heuser M. Randomized comparison of low dose cytarabine with or without Glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33(2):379–389. doi: 10.1038/s41375-018-0312-9.
    1. Jaramillo S, A Benner, J Krauter, H Martin, T Kindler, et al. Condensed versus standard schedule of high-dose cytarabine consolidation therapy with pegfilgrastim growth factor support in acute myeloid leukemia. Blood Cancer J. 2017; doi:10.1038/bcj.2017.45.
    1. Aaronson NK, Ahmedzai S, Bergman B, Bullinger M, Cull A, Duez NJ, Filiberti A, Flechtner H, Fleishman SB, Haes JCJM, Kaasa S, Klee M, Osoba D, Razavi D, Rofe PB, Schraub S, Sneeuw K, Sullivan M, Takeda F. The European Organisation for Research and Treatment of Cancer QLQ-C30: A quality-of-life instrument for use in international clinical trials in oncology. J Natl Cancer Inst. 1993;85(5):365–376. doi: 10.1093/jnci/85.5.365.
    1. Buysse DJ, Reynolds CF, Monk TH, et al. et al. Pittsburgh Sleep Quality Index. In: Rush AJ, et al., editors. APA task force. Handbook of psychiatric measures. Washington DC: APA; 2000. pp. 678–680.
    1. Patient helath questionnaire screeners. Instructions Instructions for Patient Health Questionnaire (PHQ) and GAD-7 retrived Dezember 2020 from .
    1. Ware J, Snoww KK, Kosinski MA, et al. SF36 Health Survey: Manual and Interpretation Guide. Lincoln, RI: Quality Metric, Inc.; 1993. p. 30.
    1. Brazier J, Roberts J, Deverill M. The estimation of a preference-based measure of health from the SF-36. J Health Econ. 2002;21(2):271–292. doi: 10.1016/s0167-6296(01)00130-8.
    1. van Buuren S. Multiple imputation of discrete and continuous data by fully conditional specification. Stat Methods Med Res. 2007;16(3):219–242. doi: 10.1177/0962280206074463.
    1. Kapp-Schwoerer S, Weber D, Corbacioglu A, Gaidzik VI, Paschka P, Krönke J, Theis F, Rücker FG, Teleanu MV, Panina E, Jahn N, Herzig J, Kubanek L, Schrade A, Göhring G, Fiedler W, Kindler T, Schroeder T, Mayer KT, Lübbert M, Wattad M, Götze KS, Horst HA, Koller E, Wulf G, Schleicher J, Bentz M, Krauter J, Bullinger L, Krzykalla J, Benner A, Schlenk RF, Thol F, Heuser M, Ganser A, Döhner H, Döhner K. Impact of gemtuzumab ozogamicin on MRD and relapse risk in NPM1 mutated AML patients: results from the AMLSG 09-09 Trial. Blood. 2020;136(26):3041–3050. doi: 10.1182/blood.2020005998.
    1. Kayser S, Walter RB, Stock W, Schlenk RF. Minimal residual disease in acute myeloid leukemia--current status and future perspectives. Curr Hematol Malig Rep. 2015;10(2):132–144. doi: 10.1007/s11899-015-0260-7.
    1. Grimwade D, Jovanovic JV, Hills RK, Nugent EA, Patel Y, Flora R, Diverio D, Jones K, Aslett H, Batson E, Rennie K, Angell R, Clark RE, Solomon E, Lo-Coco F, Wheatley K, Burnett AK. Prospective minimal residual disease monitoring to predict relapse of acute promyelocytic leukemia and to direct pre-emptive arsenic trioxide therapy. J Clin Oncol. 2009;27(22):3650–3658. doi: 10.1200/JCO.2008.20.1533.
    1. Ossenkoppele G, Schuurhuis GJ. MRD in AML: does it already guide therapy decision-making? Hematol Am Soc Hematol Educ Progr. 2016;1(1):356–365. doi: 10.1182/asheducation-2016.1.356.
    1. Ivey A, Hills RK, Simpson MA, Jovanovic JV, Gilkes A, Grech A, Patel Y, Bhudia N, Farah H, Mason J, Wall K, Akiki S, Griffiths M, Solomon E, McCaughan F, Linch DC, Gale RE, Vyas P, Freeman SD, Russell N, Burnett AK, Grimwade D. Assessment of Minimal Residual Disease in Standard-Risk AML. N Engl J Med. 2016;374(5):422–433. doi: 10.1056/NEJMoa1507471.
    1. Buckley SA, Wood BL, Othus M, Hourigan CS, Ustun C, Linden MA, DeFor TE, Malagola M, Anthias C, Valkova V, Kanakry CG, Gruhn B, Buccisano F, Devine B, Walter RB. Minimal residual disease prior to allogeneic hematopoietic cell transplantation in acute myeloid leukemia: A meta-analysis. Haematologica. 2017;102(5):865–873. doi: 10.3324/haematol.2016.159343.
    1. Inaba H, Coustan-Smith E, Cao X, Pounds SB, Shurtleff SA, Wang KY, Raimondi SC, Onciu M, Jacobsen J, Ribeiro RC, Dahl GV, Bowman WP, Taub JW, Degar B, Leung W, Downing JR, Pui CH, Rubnitz JE, Campana D. Comparative analysis of different approaches to measure treatment response in acute myeloid leukemia. J Clin Oncol. 2012;30(29):3625–3632. doi: 10.1200/JCO.2011.41.5323.
    1. FDA. FDA Approves Gemtuzumab Ozogamicin for CD33-positive AML, 2017. [Online]. Available:. [Accessed: 18-Jan-2020].
    1. Cheson BD, Bennett JM, Kopecky KJ, Büchner T, Willman CL, Estey EH, Schiffer CA, Doehner H, Tallman MS, Lister TA, Lo-Coco F, Willemze R, Biondi A, Hiddemann W, Larson RA, Löwenberg B, Sanz MA, Head DR, Ohno R, Bloomfield CD, International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia Revised recommendations of the international working group for diagnosis, standardization of response criteria treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia. J Clin Oncol. 2003;21(24):4642–4649. doi: 10.1200/JCO.2003.04.036.
    1. M. Buyse, et al. Leukemia-free survival as a surrogate end point for overall survival in the evaluation of maintenance therapy for patients with acute myeloid leukemia in complete remission Haematologica. 2011; 96: 1106-12. 10.3324/haematol.2010.039131, 8.
    1. FR Appelbaum, et al. End points to establish the efficacy of new agents in the treatment of acute leukemia. Blood. 2007 109: 1810-16. 10.1182/blood-2006-08-041152.

Source: PubMed

3
Subskrybuj