Page 198 - Haematologica April 2020
P. 198

A. Vidal-Crespo et al.
MCL) and other where exhibits superiority (transformed CD20dim FL). Moreover, daratumumab potentiates the antitumor activity of CHOP and R-CHOP in all three models.
In addition, as demonstrated in MM, it is likely that daratumumab may modulate the activity and frequency of CD38 expressing immune suppressive cell populations present in NHL, an effect not exerted by rituximab, and therefore may offer a superior overall antitumor effect.29 These may be of special relevance in FL and DLBCL where infiltration of diverse T and myeloid subpopulations is specially prominent.48 The immune-compromised mouse models used in this study preclude the analysis of this immune-modulating activity of daratumumab.
In conclusion, our findings warrant further clinical development of daratumumab in NHL providing a strong rationale for examining its clinical efficacy in different sce- narios, including maintenance therapy after induction therapy, cases with anti-CD20 resistance, FL histologic transformation, and as frontline in combination with the standard of care (CHOP/ R-CHOP).
Acknowledgments
We thank Dr. Adrian Wiestner for his support with this study and his critical revision of the manuscript. Jocabed Roldan, Laura Jiménez, Sandra Cabezas and Ariadna Giro for their technical assistance. The authors would like to thank Dr J Rouquette and Ms L Teyssedre (Imaging facility, ITAV, Toulouse) for performing Daratumumab imaging within the 3D lymphoma cultures by SPIM, and Dr JJ Fournié (CRCT, Toulouse) for article comments.
Funding
This work was carried out at the Esther Koplowitz Center, Barcelona. Genmab and Janssen pharmaceuticals funded this research. Additional grants that contributed to this work included: Spanish Ministry of Economy and Competitiveness & European Regional Development Fund (ERDF) “Una manera de hacer Europa” for SAF2011/29326 and SAF2014/57708R to PP-G, SAF2015/31242R and SAF2015-31242-R to DC, CIBERONC (CB16/12/00334 and CB16/12/00225), the Integrated Excellence Grant from the Instituto de Salud Carlos III (ISCIII) PIE1313/00033 to EC and PP-G, and finally Generalitat de Catalunya support for AGAUR 2017SGR1009 to DC.
References
1. Chihara D, Nastoupil LJ, Williams JN, Lee P, Koff JL and Fowers CR. New insights into the epidemiology of non-Hodgkin lymphoma and implications for therapy. Expert Rev Anticancer Ther. 2015; 15(5):531-544.
2. Swerdlow SH, Campo E, Harris NL, et al. WHO Classification of tumours of haematopoietic and lymphoid tissues. 2008.
3. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization (WHO) classification of lym- phoid neoplasms. Blood. 2016; 127(20):2375-2390.
4. LossosIS,GascoyneRD.Transformationof follicular lymphoma. Best Pr Res Clin Haematol. 2011;24(2):147-163.
5. HiddemannW,KnebaM,DreylingM,etal. Frontline therapy with rituximab added to the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) significantly improves the out- come for patients with advanced-stage fol- licular lymphoma compared with therapy with CHOP alone. Blood. 2005; 106(12):3725-3732.
6. Federico M, Luminari S, Dondi A, et al. R- CVP versus R-CHOP versus R-FM for the initial treatment of patients with advanced- stage follicular lymphoma: Results of the FOLL05 trial conducted by the Fondazione Italiana Linfomi. J Clin Oncol. 2013; 31(12):1506-1513.
7. Kridel R, Sehn LH, Gascoyne RD. Pathogenesis of follicular lymphoma. J Clin Invest. 2012;122(10):3424-3431.
8. Alizadeh AA, Eisen MB, Davis RE, et al. Distinct types of diffuse large B-cell lym- phoma identified by gene expression profil- ing. Nature. 2000;403(6769):503-511.
9. Tilly H, Dreyling M. Diffuse large B-cell non-Hodgkin’s lymphoma: ESMO Clinical Recommendations for diagnosis, treatment and follow-up. Ann Oncol. 2009;20 Suppl 4:110-112.
10. Lim SH, Vaughan AT, Ashton-Key M, et al. Fc gamma receptor IIb on target B cells pro- motes rituximab internalization and reduces clinical efficacy. Blood. 2011; 118(9):2530-2540.
11. Edelmann J, Gribben JG. Obinutuzumab for the treatment of indolent lymphoma. Futur Med. 2016;12(15):1769-1781.
12. Mantei K, Wood BL. Flow cytometric eval- uation of CD38 expression assists in distin- guishing follicular hyperplasia from follicu- lar lymphoma. Cytom Part B Clin Cytom. 2009;76(5):315-320.
13. Pérez-Galán P, Dreyling M, Wiestner A. Mantle cell lymphoma: biology, pathogen- esis, and the molecular basis of treatment in the genomic era. Blood. 2011;117(1):26- 38.
21. Pérez-Galán P, Mora-Jensen H, Weniger MA, et al. Bortezomib resistance in mantle cell lymphoma is associated with plasma- cytic differentiation. Blood. 2011; 117(2):542-553.
22. Deaglio S, Vaisitti T, Aydin S, Ferrero E, Malavasi F. In-tandem insight from basic science combined with clinical research: CD38 as both marker and key component of the pathogenetic network underlying chronic lymphocytic leukemia. Blood. 2006;108(4):1135-1144.
23. McKeage K. Daratumumab: First Global Approval. Drugs. 2016;76(2):275-281.
24. https://www.accessdata.fda.gov/drugsatf- da_docs/label/2017/761036s005lbl.pdf. Janssen Biotech Inc. DarzalexTM (daratu- mumab): prescribing information 2017;1-
14. Campo E and Rule S. Mantle cell lym- 29.
phoma: evolving management strategies.
Blood. 2015;125(1):48-55.
15. Dreyling M, Campo E, Hermine O, et al.
Newly diagnosed and relapsed mantle cell lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and fol- low-up. Ann Oncol. 2017;28(suppl-4):iv62- iv71.
16. Cavalli F. Bortezomib-based therapy for mantle-cell lymphoma. N Engl J Med. 2015;372(23):2271.
17. Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013;369(6):507-516.
18. Martin P, Maddocks K, Leonard JP, et al. Postibrutinib outcomes in patients with mantle cell lymphoma. Blood. 2016; 127(12):1559-1663.
19. Orchard J, Garand R, Davis Z, et al. A sub- set of t(11; 14) lymphoma with mantle cell features displays mutated IgV H genes and includes patients with good prognosis, nonnodal disease. Blood. 2003; 101(12):4975-4981.
20. Camacho FI, Algara P, Rodríguez A, et al. Molecular heterogeneity in MCL defined by the use of specific V H genes and the fre- quency of somatic mutations. Blood. 2003; 101(10):4042-4046.
25. de Weers M, Tai Y-T, van der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol. 2011;186(3):1840-1848.
26. Overdijk MB, Verploegen S, Bögels M, et al. Antibody-mediated phagocytosis con- tributes to the anti-tumor activity of the therapeutic antibody daratumumab in lym- phoma and multiple myeloma. MAbs. 2015;7(2):311-320.
27. Overdijk MB, Jansen JHM, Nederend M, et al. The therapeutic CD38 monoclonal anti- body daratumumab induces programmed cell death via Fc Receptor-mediated cross- linking. J Immunol. 2016;197(3):807-813.
28. Lammerts van Bueren J, Jakobs D, Kaldenhoven N, et al. Direct in vitro com- parison of daratumumab with Surrogate analogs of CD38 antibodies MOR03087, SAR650984 and Ab79 [abstract]. Blood. 2014;124(21):Abstract 3474.
29. Krejcik J, Casneuf T, Nijhof IS, et al. Daratumumab depletes CD38+ immune- regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood. 2016;128(3):384-394.
30. Matas-Céspedes A, Vidal-Crespo A, Rodriguez V, et al. The human CD38 mon-
1040
haematologica | 2020; 105(4)


































































































   196   197   198   199   200