Page 135 - 2020_07-Haematologica-web
P. 135

Danger signaling to NK cells in AML
Table 2. Univariate Cox proportional hazard analysis. RFS
Table 3. Multivariate Cox proportional hazard analysis. RFS
Variable
Age
Sex
Peripheral blast counts HSCT
Ecto-CRT+ blasts (%) KLRK1 expression NKG2D+ NK cells (%)
HR (95% CI)
1.02 (0.99-1.04)
0.79 (0.44-1.43) 1.00 (0.99-1.01) 0.76 (0.40-1.44) 0.99 (0.98-1.00) 0.80 (0.53-1.19) 0.96 (0.92-1.01)
P
0.19
0.45 0.26 0.40 0.22 0.27 0.13
Variable
Age
Sex
Peripheral blast counts HSCT
Ecto-CRT+ blasts (%) KLRK1 expression NKG2D+ NK cells (%)
HR (95% CI)
1.07 (1.01-1.13)
1.34 (0.50-3.60) 1.00 (0.98-1.02) 0.62 (0.21-1.83) 0.98 (0.96-1.00) 0.69 (0.44-1.07) 0.95 (0.90-1.01)
P
0.007*
0.55 0.71 0.39 0.10 0.10 0.11
CI, 95% confidence interval; HR, hazard ratio; *p< 0.05; RFS, relapse-free survival.
this possibility. Indeed, in vivo application of C1498 AML cells engineered to constitutively expose CRT on their sur- face elicited an accumulation of highly functional NK cells and CD4+ and CD8+ T cells in mouse tumors and/or spleen (Figure 4B-D).
Finally, both CRT exposure on malignant blasts and NK cell-related marker NKG2D were associated with improved RFS amongst AML patients (Figure 5A-C), corroborating previously published data.10,34 Combinatorial assessment of the prognostic value of these parameters identified signifi- cantly prolonged RFS in KLRK1HiCRTHi and NKG2DHiCRTHi subgroup of patients (Figure 5D-E). However, these findings could not be confirmed using univariate and multivariate Cox proportional hazard analysis, potentially reflecting a limited follow-up period, the small size of the patient cohort, disease subset and/or inter-patient heterogeneity. Thus, the precise prognostic value of CRT exposure on AML blasts and NKGD2 levels on NK cells remains to be validated in independent patient series.
Taken together, our results support the association of CRT with enhanced activation of the innate and adap- tive anticancer immunity. Parallel assessment of CRT exposure on malignant blasts and immune cell parame- ters, such as NK-cell markers, may provide prognostic
CI, 95% confidence interval; HR, hazard ratio; *p< 0.05; RFS, relapse-free survival.
information and have therapeutic relevance for AML patients in the future.
Funding
This study was exclusively sponsored by Sotio, Prague, Czech Republic.
Acknowledgments
The authors thank Anna Fialova for her valuable help with statistical analysis and to Jana Bieblova for help with FACS and ELISA analysis. LG is supported by a Breakthrough Level 2 grant from the US Department of Defense (DoD), Breast Cancer Research Program (BRCP) (#BC180476P1), by the 2019 Laura Ziskin Prize in Translational Research (#ZP-6177, PI: Formenti) from the Stand Up to Cancer (SU2C), by a Mantle Cell Lymphoma Research Initiative (MCL-RI, PI: Chen-Kiang) grant from the Leukemia and Lymphoma Society (LLS), by a startup grant from the Dept. of Radiation Oncology at Weill Cornell Medicine (New York, NY, USA), by a Rapid Response Grant from the Functional Genomics Initiative (York, NY, USA), by industrial collaborations with Lytix (Oslo, Norway) and Phosplatin (York, NY, USA), and by donations from Phosplatin (York, NY, USA), the Luke Heller TECPR2 Foundation (Boston, USA) and Sotio a.s. (Prague, Czech Republic).
References
1. Fucikova J, Moserova I, Truxova I, et al. High hydrostatic pressure induces immunogenic cell death in human tumor cells. Int J Cancer. 2014;135(5):1165-1177.
2. Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in can- cer and infectious disease. Nat Rev Immunol. 2017;17(2):97-111.
3. Spisek R, Charalambous A, Mazumder A, Vesole DH, Jagannath S, Dhodapkar MV. Bortezomib enhances dendritic cell (DC)- mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: ther- apeutic implications. Blood. 2007;109(11): 4839-4845.
4. Vanpouille-Box C, Demaria S, Formenti SC, Galluzzi L. Cytosolic DNA sensing in organismal tumor control. Cancer Cell. 2018;34(3):361-378.
5. Galluzzi L, Chan TA, Kroemer G, Wolchok
JD, Lopez-Soto A. The hallmarks of suc- cessful anticancer immunotherapy. Sci Transl Med. 2018;10(459).
6. Garg AD, Vandenberk L, Fang S, et al. Pathogen response-like recruitment and activation of neutrophils by sterile immunogenic dying cells drives neutrophil- mediated residual cell killing. Cell Death Differ. 2017;24(5):832-843.
7. Mehta MM, Weinberg SE, Chandel NS. Mitochondrial control of immunity: beyond ATP. Nat Rev Immunol. 2017; 17(10):608-620.
8. Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012; 12(12):860-875.
9. Fucikova J, Becht E, Iribarren K, et al. Calreticulin Expression in human non-small cell lung cancers correlates with Increased accumulation of antitumor immune cells and favorable prognosis. Cancer Res. 2016; 76(7):1746-1756.
10. Fucikova J, Truxova I, Hensler M, et al. Calreticulin exposure by malignant blasts correlates with robust anticancer immunity and improved clinical outcome in AML patients. Blood. 2016;128(26):3113-3124.
11. Peng RQ, Chen YB, Ding Y, et al. Expression of calreticulin is associated with infiltration of T-cells in stage IIIB colon can- cer. World J Gastroenterol. 2010;16(19): 2428-2434.
12. Wemeau M, Kepp O, Tesniere A, et al. Calreticulin exposure on malignant blasts predicts a cellular anticancer immune response in patients with acute myeloid leukemia. Cell Death Dis. 2010;1:e104.
13. Fucikova J, Moserova I, Urbanova L, et al. Prognostic and predictive value of DAMPs and DAMP-associated processes in cancer. Front Immunol. 2015;6:402.
14. Lopez-Soto A, Gonzalez S, Smyth MJ, Galluzzi L. Control of metastasis by NK cells. Cancer Cell. 2017;32(2):135-154.
15. Gehrmann M, Schonberger J, Zilch T, et al. Retinoid- and sodium-butyrate-induced
haematologica | 2020; 105(7)
1877


































































































   133   134   135   136   137