Page 127 - 2019_12-Haematologica-web
P. 127

MDM2 inhibition in chronic lymphocytic leukemia
incidence of clonal evolution leading to ibrutinib resist- ance due to mutations in BTK and PLCG2 have been reported in patients progressing on treatment.52
Continued preclinical studies to develop innovative therapeutic strategies for CLL therefore remain a high pri- ority. In particular, new agents promoting CLL cell apop- tosis with limited toxicity to normal cells represent an attractive therapeutic strategy for CLL, which is a disease of elderly patients who would benefit from the use of compounds with a therapeutic window associated with minimal effects on normal cells. Moreover, given the clin- ical heterogeneity of CLL, there is a constant need to iden- tify treatment strategies that can be effective also in the most aggressive subtypes of this disease. In our cohort, RG7388 significantly decreased the viability of CLL cells isolated from patients in different poor prognosis sub- groups, including cases with advanced-stage disease, cases with unmutated IGHV genes and cases with 11q deletion and trisomy 12, which are usually more prone to progres- sion. This indicates that inhibiting the p53-MDM2 inter- action is a promising treatment strategy to explore for high-risk CLL patients with functional p53.
Taken together, our data demonstrate that MDM2
inhibitors induce a pro-apoptotic response in cells from patients with both low- and high-risk subtypes of CLL, at doses which show a lesser effect on normal blood cells and hematopoietic stem cells. This therapeutic window supports the clinical evaluation of new-generation, non- genotoxic MDM2 inhibitors, used in combined treatment strategies with other targeted therapies for the treatment of CLL.
Acknowledgments
This study was supported by Bloodwise (grant # 13034), the JGW Patterson Foundation (grant # BH152495) and the Newcastle Healthcare Charity (grant # BH152694).
The authors gratefully acknowledge Newcastle University/Astex Pharmaceuticals Alliance and CRUK which funded the Drug Discovery Programme at the Northern Institute for Cancer Research, Newcastle University for their support and encouragement.
The authors would also like to thank Jane Cole for recruiting patients and providing clinical information, Dr Kenneth Rankin for providing bone marrow samples, Dr Sally Hall for providing blood samples from healthy donors and all the CLL patients for generously donating samples.
References
1. Chiorazzi N, Rai KR, Ferrarini M. Chronic lymphocytic leukemia. N Engl J Med. 2005;352(8):804-815.
2. Müller-Hermelink HK, Montserrat E, Catovsky D, Campo E, Harris NL, Stein H. Chronic lymphocytic leukemia/small lym- phocytic lymphoma. In: Swerdlow SH, Campo E, Harris NL, et al. (Eds.), World Health Organization Classification of Tumours, Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues, IARC, Lyon, France: 2008:180–182.
3. Zenz T, Mertens D, Küppers R, Döhner H, Stilgenbauer S. From pathogenesis to treat- ment of chronic lymphocytic leukaemia. Nat Rev Cancer. 2010;10(1):37-50.
4. Herishanu Y, Perez-Galan P, Liu D, et al. The lymph node microenvironment promotes B- cell receptor signaling, NF-kappaB activa- tion, and tumor proliferation in chronic lym- phocytic leukemia. Blood. 2011;117(2):563- 574.
5. Kay NE, Wasil T. Survival of chronic lym- phocytic leukemia cells: CD40L and the vas- cular endothelial growth factor (VEGF) con- nection. Leukemia. 2005;19(4):531-532.
6. Munk Pedersen I, Reed J. Micro- environmental interactions and survival of CLL B-cells. Leuk Lymphoma. 2004;45 (12):2365-2372.
7. Herishanu Y, Katz BZ, Lipsky A, Wiestner A. Biology of chronic lymphocytic leukemia in different microenvironments: clinical and therapeutic implications. Hematol Oncol Clin North Am. 2013;27(2):173-206.
8. Badoux XC, Keating MJ, Wang X, et al. Fludarabine, cyclophosphamide, and ritux- imab chemoimmunotherapy is highly effec- tive treatment for relapsed patients with CLL. Blood. 2011;117(11):3016-3024.
9. Burger JA, Tedeschi A, Barr PM, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015;373(25):2425-2437.
10. Billard C. Apoptosis inducers in chronic lymphocytic leukemia. Oncotarget. 2014;5(2):309–325.
11. Besbes S, Mirshahi M, Pocard M, Billard C. Strategies targeting apoptosis proteins to improve therapy of chronic lymphocytic leukemia. Blood Rev. 2015;29(5):345-350.
12. Faderl S, Keating MJ, Do KA, et al. Expression profile of 11 proteins and their prognostic significance in patients with chronic lymphocytic leukemia (CLL). Leukemia. 2002;16(6):1045-1052.
13. Robertson LE, Plunkett W, McConnell K, Keating MJ, McDonnell TJ. Bcl-2 expression in chronic lymphocytic leukemia and its cor- relation with the induction of apoptosis and clinical outcome. Leukemia. 1996;10(3):456- 459.
14. Molica S, Dattilo A, Giulino C, Levato D, Levato L. Increased Bcl-2/ bax ratio in B-cell chronic lymphocytic leukemia is associated with a progressive pattern of disease. Haematologica. 1998;83(12):1122-1124.
15. Zenz T, Eichhorst B, Busch R, et al. TP53 mutation and survival in chronic lympho- cytic leukemia. J Clin Oncol. 2010;28(29): 4473-4479.
16. Dicker F, Herholz H, Schnittger S, et al. The detection of TP53 mutations in chronic lym- phocytic leukemia independently predicts rapid disease progression and is highly cor- related with a complex aberrant karyotype. Leukemia. 2009;23(1):117-124.
17. Puente XS, Pinyol M, Quesada V, et al. Whole-genome sequencing identifies recur- rent mutations in chronic lymphocytic leukaemia. Nature. 2011;475(7354):101-105.
18. Quesada V, Conde L, Villamor N, et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat Genet. 2011;44(1):47-52.
19. Rossi D, Rasi S, Fabbri G, et al. Mutations of NOTCH1 are an independent predictor of survival in chronic lymphocytic leukemia. Blood. 2012;119(2):521-529.
20. Chène P. Inhibiting the p53–MDM2 interac-
tion: an important target for cancer therapy.
Nat Rev Cancer. 2003;3(2):102-109.
21. Brown CJ, Lain S, Verma CS, Fersht AR, Lane DP. Awakening guardian angels: drug- ging the p53 pathway. Nat Rev Cancer.
2009;9(12):862-873.
22. Haupt Y, Maya R, Kazaz A, Oren M. Mdm2
promotes the rapid degradation of p53.
Nature. 1997;387(6630):296-299.
23. Moll UM, Petrenko O. The MDM2-p53 interaction. Mol Cancer Res. 2003;1(14):
1001-1008.
24. Vu B, Wovkulich P, Pizzolato G, et al.
Discovery of RG7112: a small-molecule MDM2 inhibitor in clinical development. ACS Med Chem Lett. 2013;4(5):466-469.
25. Ding Q, Zhang Z, Liu J-J, et al. Discovery of RG7388, a potent and selective p53-MDM2 inhibitor in clinical development. J Med Chem. 2013;56(14):5979-5983.
26. Endo S, Yamato K, Hirai S, et al. Potent in vitro and in vivo antitumor effects of MDM2 inhibitor nutlin-3 in gastric cancer cells. Cancer Sci. 2011;102(3):605-613.
27. Drakos E, Singh RR, Rassidakis GZ, et al. Activation of the p53 pathway by the MDM2 inhibitor nutlin-3a overcomes BCL2 overexpression in a preclinical model of dif- fuse large B-cell lymphoma associated with t(14;18)(q32;q21). Leukemia. 2011;25(5): 856-867.
28. Ray-Coquard I, Blay J-Y, Italiano A, et al. Effect of the MDM2 antagonist RG7112 on the P53 pathway in patients with MDM2- amplified, well-differentiated or dedifferen- tiated liposarcoma: an exploratory proof-of- mechanism study. Lancet Oncol. 2012;13 (11):1133-1140.
29. Andreeff M, Kelly KR, Yee K, et al. Results of the phase I trial of RG7112, a small-molecule MDM2 antagonist in leukemia. Clin Cancer Res. 2016;22(4):868-876.
30. Verreault M, Schmitt C, Goldwirt L, et al. Preclinical efficacy of the MDM2 inhibitor RG7112 in MDM2-amplified and TP53 wild-type glioblastomas. Clin Cancer Res. 2016;22(5):1185-1196.
haematologica | 2019; 104(12)
2441


































































































   125   126   127   128   129