Page 29 - 2019_07 resto del Mondo-web
P. 29

CAR T-cells for AML
an immuno-suppressive microenvironment,50 failure of persistence of the CART population,51,52 or unacceptable or unexpected on-target off-tumor toxicity.53 As early results from CART trials for AML become available in the coming years, we will gain a better understanding of their relative role vis-à-vis that of other recently approved ther- apies for this disease targeted against specific genetic lesions (FLT3, IDH1/2) or survival pathways (BCL2). The T-cell manufacturing process is complicated and time- consuming, with the median time from enrollment to infusion in the ELIANA trial of CART-19 being 45 days (range 30-105 days),1 and so the availability of well-toler- ated, targeted, non-immunosuppressive therapies may improve the feasibility of bridging patients to potentially curative immunotherapy by providing disease control and clinical stability.
How close are we ... really?
Although the first CART trials for AML are now appear- ing in the clinical sphere, it is likely that other barriers will need to be overcome before this therapy becomes widely available. It remains to be seen if the ‘bridge to transplant’ approach is feasible, if it provides a sufficient duration of CART persistence, and how the on-target off-tumor toxic- ity of the various constructs, combinations and target anti-
gens will be tolerated. We suspect that CART therapy will not be suitable or efficacious for all patients with AML, for example, frail elderly patients who are at higher risk of tox- icity, or for those lacking access to expensive personalized therapies, by virtue of geographic and economic factors. However, select patients with AML are now being treated on CART trials and in the next 1-2 years data are likely to tell us more about the patient, disease and treatment char- acteristics that can predict success in this arena. We think of the future of CART cell therapy for AML as the next step in alloHSCT: a complex, resource-intensive but feasi- ble approach intended to provide curative therapy to selected patients. In addition, the lessons learned in treat- ing AML with CAR T cells may reveal other targetable pathways to be exploited in combination with immune- based or pharmacological therapies. We await the impact of CART therapy on AML with cautious optimism, noting the recent shower of drug approvals that followed a long dry spell in AML therapeutics. We are hopeful that the combination of alloHSCT and CAR T-cell therapy (the old master and new arrival in adoptive cellular therapy) may prove to be the key to unlocking relapsed-refractory AML. We, and many others, continue to create and develop new solutions to make CAR T cells for AML a safe, deliverable and effective reality.
References
1. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med. 2018;378(5):439-448.
2. June CH, O'Connor RS, Kawalekar OU, et al. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361-1365.
3. Ritchie DS, Neeson PJ, Khot A, et al. Persistence and efficacy of second genera- tion CAR-T cell against the LeY antigen in acute myeloid leukemia. Mol Ther. 2013;21(11):2122-2129.
4. Brenner MK. CAR T cells for acute myeloid leukemia: the LeY of the land. Mol Ther. 2013;21(11):1983-1984.
5. Ehninger A, Kramer M, Rollig C, et al. Distribution and levels of cell surface expres- sion of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J. 2014;4:e218.
6. Testa U, Pelosi E, Frankel A. CD 123 is a membrane biomarker and a therapeutic tar- get in hematologic malignancies. Biomark Res. 2014;2(4).
7. WangQ-s,WangY,LvH-y,etal.Treatment of CD33-directed Chimeric Antigen Receptor-modified T Cells in One Patient With Relapsed and Refractory Acute
Myeloid Leukemia. Mol Ther. 2015;23(1):184-191.
8. Liu F, Cao Y, Pinz K, et al. First-in-Human CLL1-CD33 Compound CAR T Cell Therapy Induces Complete Remission in Patients with Refractory Acute Myeloid Leukemia: Update on Phase 1 Clinical Trial. Blood. 2018;132(Suppl 1):901.
9. Munoz L, Nomdedeu JF, Lopez O, et al. Interleukin-3 receptor alpha chain (CD123) is widely expressed in hematologic malig- nancies. Haematologica. 2001;86(12):1261- 1269.
10. VergezF,GreenAS,TamburiniJ,etal.High
levels of CD34+CD38low/-CD123+ blasts
are predictive of an adverse outcome in
acute myeloid leukemia: a Groupe Ouest- 19. Est des Leucemies Aigues et Maladies du
Sang (GOELAMS) study. Haematologica. 2011;96(12):1792-1798. 20.
11. Gill S, Tasian SK, Ruella M, et al. Preclinical targeting of human acute myeloid leukemia
and myeloablation using chimeric antigen receptor–modified T cells. Blood. 2014;123 (15):2343-2354. 21.
12. Arcangeli S, Bardelli M, Rotiroti MC, et al. Balance of Anti-CD123 Chimeric Antigen Receptor (CAR) Binding Affinity and Density for the Treatment of Acute Myeloid Leukemia. Blood. 2016;128(22):2163. 22.
Paradoxical Role of NKG2D in Cancer Immunity. Front Immunol. 2018;9:1808. Champsaur M, Lanier LL. Effect of NKG2D ligand expression on host immune respons- es. Immunol Rev. 2010;235(1):267-285. Chauveau A, Tonnerre P, Pabois A, et al. Endothelial Cell Activation and Proliferation Modulate NKG2D Activity by Regulating MICA Expression and Shedding. J Innate Immun. 2014;6(1):89-104.
Baumeister SH, Murad J, Werner L, et al. Phase I Trial of Autologous CAR T Cells Targeting NKG2D Ligands in Patients with AML/MDS and Multiple Myeloma. Cancer Immunol Res. 2019;7(1):100-112.
Sallman DA, Brayer JB, Poire X, et al. Abstract CT129: The THINK clinical trial: Preliminary evidence of clinical activity of NKG2D chimeric antigen receptor T cell therapy (CYAD-01) in acute myeloid leukemia. Cancer Res. 2018;78(13
13. http://www.cellectis.com/en/press/cellectis- reports-clinical-hold-of-ucart123-studies.
Available from: http://www.cellectis.com/ en/press/cellectis-reports-clinical-hold-of-
ucart123-studies. [Last accessed 8 February
2019] Supplement):CT129.
14. Cummins KD, Frey N, Nelson AM, et al. 23. Treating Relapsed / Refractory (RR) AML
with Biodegradable AntiCD123 CAR Modified T Cells. Blood. 2017;130(Suppl 1):1359.
15. Budde LE, Schuster SJ, Del Real M et al. 24. CD123CAR displays clinical activity in relapsed/refractory acute myeloid leukemia (AML) and blastic plasmcytoid dendritic cell neoplasm (BPDCN): Safety and efficiacy results from a phase 1 study. AACR Tumor 25. Immunology and Immunotherapy. Miami,
FL, USA, 2018.
16. Spear P, Wu M-R, Sentman M-L, et al.
NKG2D ligands as therapeutic targets.
Cancer Immun. 2013;13:8. 26.
17. Spear P, Barber A, Rynda-Apple A, et al. NKG2D CAR T-cell therapy inhibits the growth of NKG2D ligand heterogeneous
tumors. Immunol Cell Biol. 2013;91(6):435- 27.
440.
18. Sheppard S, Ferry A, Guedes J, et al. The
Baragano Raneros A, Martin-Palanco V, Fernandez AF, et al. Methylation of NKG2D ligands contributes to immune system eva- sion in acute myeloid leukemia. Genes Immun. 2015;16(1):71-82.
Perna F, Berman SH, Soni RK, et al. Integrating Proteomics and Transcriptomics for Systematic Combinatorial Chimeric Antigen Receptor Therapy of AML. Cancer Cell. 2017;32(4):506-519.e5.
Tashiro H, Sauer T, Shum T, et al. Treatment of Acute Myeloid Leukemia with T Cells Expressing Chimeric Antigen Receptors Directed to C-type Lectin-like Molecule 1. Mol Ther. 2017;25(9):2202-2213.
Wang J, Chen S, Xiao W, et al. CAR-T cells targeting CLL-1 as an approach to treat acute myeloid leukemia. J Hematol Oncol. 2018;11(1):7.
Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remis- sions in leukemia. N Engl J Med. 2014;
haematologica | 2019; 104(7)
1307


































































































   27   28   29   30   31