Page 155 - Haematologica April 2020
P. 155

MSC-induced SP phenotype leads to chemoresistance
promoting effects on blasts, preliminary results on four patients suggest that cultivating MSC and blasts in autol- ogous conditions would bring a supplemental advantage on blast survival and chemoresistance (Boutin L et al., 2018, unpublished data). This suggests that an intimate crosstalk has taken place in vivo between MSC and blasts and has been conserved in vitro. Recently, Moschoi et al.31 proposed that mitochondrial transfers from BM stromal cells to AML blasts provided them with a protective effect following chemotherapy. Other communication modes such as exosome exchange, metabolite secretion, and nan- otube formation could also be suggested to participate in this process and must be identified to disrupt the stroma- induced chemoresistance of AML blasts.
Induction of SP/chemoresistance phenotype on AML blasts is of major interest to clinicians as ABC transporters implicated in drug/chemotherapy agent efflux could be targeted.32,33 In that context, we have shown that mitox- antrone efflux and blast survival are reversible in vitro by addition of verapamil. However, results from clinical trials using P-glycoprotein inhibitors in AML patients are dis- couraging.34,35 Our data showing that the functionality of the main ABC transporters implicated in MDR36 is pro- moted by blast-MSC interactions, and that this process is
patient-dependent, suggest that it is time to revisit the role of ABC transporters in therapeutic failure in the context of personalized medicine.37,38
In conclusion, our results show the critical role of stro- ma interactions in blast chemoresistance through SP phe- notype promotion and ABC transporter activation. Therefore, targeting these interactions in combination with the development of new improved inhibitors of ABC transporter function39 could be an attractive therapeutic alternative.
Acknowledgments
We thank the molecular core facility of Cochin Institute for transcriptomic analysis. We are very grateful to the polyclinic of Blois and particularly to Dr Denis Burgot as well as all the ortho- pedic surgery staff for providing human bone marrow samples from hip replacement. We thank Dr Cedric Thépenier for the sta- tistical analysis and sharpened advices. We also thank Pr Christophe Martinaud for blast analysis on blood smears.
Funding
This work was supported by grants from Association “Laurette Fugain” (ALF 2013/08) and Association “Vaincre le Cancer- Nouvelles Recherches Biomédicales”.
References
1. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neo- plasms and acute leukemia. Blood. 2016;127(20):2391–2405.
2. Taussig DC, Miraki-Moud F, Anjos-Afonso F, et al. Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initi- ating cells. Blood. 2008;112(3):568–575.
3. van Rhenen A, Feller N, Kelder A, et al. High stem cell frequency in acute myeloid leukemia at diagnosis predicts high mini- mal residual disease and poor survival. Clin Cancer Res. 2005;11(18):6520–6527.
4. Vergez F, Green AS, Tamburini J, et al. High levels of CD34+CD38low/−CD123+ blasts are predictive of an adverse outcome in acute myeloid leukemia: a Groupe Ouest- Est des Leucémies Aiguës et Maladies du Sang (GOELAMS) study. Haematologica. 2011;96(12):1792–1798.
5. Jordan CT, Upchurch D, Szilvassy SJ, et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myeloge- nous leukemia stem cells. Leukemia. 2000;14(10):1777–1784.
6. Sarry J-E, Murphy K, Perry R, et al. Human acute myelogenous leukemia stem cells are rare and heterogeneous when assayed in NOD/SCID/IL2Rγc-deficient mice. J Clin Invest. 2011;121(1):384–395.
7. Farge T, Saland E, de Toni F, et al. Chemotherapy-Resistant Human Acute Myeloid Leukemia Cells Are Not Enriched for Leukemic Stem Cells but Require Oxidative Metabolism. Cancer Discov. 2017;7(7):716–735.
8. Ye H, Adane B, Khan N, et al. Leukemic stem cells evade chemotherapy by meta- bolic adaptation to an adipose tissue niche. Cell Stem Cell. 2016;19(1):23–37.
9. Shafat MS, Gnaneswaran B, Bowles KM,
Rushworth SA. The bone marrow microenvironment - Home of the leukemic blasts. Blood Rev. 2017;31(5):277–286.
10. Tabe Y, Konopleva M. Leukemia Stem Cells Microenvironment. Adv Exp Med Biol. 2017;1041:19–32.
11. Cukierman E, Bassi DE. The mesenchymal tumor microenvironment. Cell Adhes Migr. 2012;6(3):285–296.
12. Goodell MA, Brose K, Paradis g, Conner AS, Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med. 1996;183(4):1797–1806.
13. Malfuson J-V, Boutin L, Clay D, et al. SP/drug efflux functionality of hematopoi- etic progenitors is controlled by mesenchy- mal niche through VLA-4/CD44 axis. Leukemia. 2014;28(4):853–864.
14. Pierre-Louis O, Clay D, Brunet de la Grange P, et al. Dual SP/ALDH functionali- ties refine the human hematopoietic Lin- CD34+CD38- stem/progenitor cell com- partment. Stem Cells. 2009;27(10):2552– 2562.
15. Saland E, Boutzen H, Castellano R, et al. A robust and rapid xenograft model to assess efficacy of chemotherapeutic agents for human acute myeloid leukemia. Blood Cancer J. 2015;5(3):e297.
16. Challen GA, Little MH. A side order of stem cells: the SP phenotype. Stem Cells. 2006;24(1):3–12.
17. Svirnovski AI, Shman TV, Serhiyenka TF, Savitski VP, Smolnikova VV, Fedasenka UU. ABCB1 and ABCG2 proteins, their functional activity and gene expression in concert with drug sensitivity of leukemia cells. Hematol Amst Neth. 2009;14(4):204– 212.
18. Imrichova D, Messingerova L, Seres M, et al. Selection of resistant acute myeloid leukemia SKM-1 and MOLM-13 cells by vincristine-, mitoxantrone- and lenalido- mide-induced upregulation of P-glycopro-
tein activity and downregulation of CD33 cell surface exposure. Eur J Pharm Sci. 2015;77:29–39.
19. Nieth C, Lage H. Induction of the ABC- transporters Mdr1/P-gp (Abcb1), mrpl (Abcc1), and bcrp (Abcg2) during establish- ment of multidrug resistance following exposure to mitoxantrone. J Chemother. 2005;17(2):215–223.
20. Eppert K, Takenaka K, Lechman ER, et al. Stem cell gene expression programs influ- ence clinical outcome in human leukemia. Nat Med. 2011;17(9):1086–1093.
21. Ng SWK, Mitchell A, Kennedy JA, et al. A 17-gene stemness score for rapid determi- nation of risk in acute leukaemia. Nature. 2016;540(7633):433–437.
22. Birbrair A, Frenette PS. Niche heterogene- ity in the bone marrow. Ann N Y Acad Sci. 2016;1370(1):82–96.
23. Crane GM, Jeffery E, Morrison SJ. Adult haematopoietic stem cell niches. Nat Rev Immunol. 2017;17(9):573–590.
24. Wei Q, Frenette PS. Niches for Hematopoietic Stem Cells and Their Progeny. Immunity. 2018;48(4):632–648.
25. Arai F, Hirao A, Suda T. Regulation of hematopoietic stem cells by the niche. Trends Cardiovasc Med. 2005;15(2):75–79.
26. Gillet J-P, Gottesman MM. Overcoming multidrug resistance in cancer: 35 years after the discovery of ABCB1. Drug Resist Updat. 2012;15(1–2):2–4.
27. Balzano M, De Grandis M, Vu Manh T-P, et al. Nidogen-1 Contributes to the Interaction Network Involved in Pro-B Cell Retention in the Peri-sinusoidal Hematopoietic Stem Cell Niche. Cell Rep. 2019;26(12):3257-3271.e8.
28. De Grandis M, Mancini SJ, Aurrand-Lions M. In quest for leukemia initiating cells in AML. Oncoscience. 2018;5(1–2):9–10.
29. Hosokawa K, Arai F, Yoshihara H, et al. Function of oxidative stress in the regula- tion of hematopoietic stem cell-niche inter-
haematologica | 2020; 105(4)
997


































































































   153   154   155   156   157