Page 13 - 2019_04-Haematologica-web
P. 13

Editorials
hydroxycarbamide. A trend was also observed towards reduction in megakaryocyte cluster formation and decrease in numbers of large megakaryocytes with staghorn-like morphology, both cardinal features of MPN. Possible confounding factors are that the participants had established MPN with a median time between MPN diag- nosis and trial registration of 3.6 years (range 1.6-8.6 years) and received a relatively short duration of therapy. It is possible that longer exposure to mirabegron, which was well tolerated, and considering its use earlier in the disease course may have demonstrated different results since potentially the chronicity and degree of increase in nestin+ MSC may be paramount in ultimately modulating the clin- ical phenotype. Mirabegron itself, as noted above, is a less potent agent and may have additional effects when com- pared to BRL37344, which was used in the murine work.
+ Moreover, although significant increases in Nestin MSC
were seen, this was limited to those patients not receiving hydroxycarbamide, an agent that most likely affected MSC senescence.
As we begin to explore the complex BM HSC-neuro- stromal interaction through niche targeting therapies, such as that described in the novel study described above, sig- nificant thought needs to be given to the timing of such therapies within the disease course and that logical sequencing / dual agent approaches are considered when targeting of this axis is contemplated. For example, the JAK inhibitor ruxolitinib has been shown to abrogate MSC- growth and an ability to secrete MCP and IL-6 which could potentially be beneficial through downregulation of pro- inflammatory MSC, yet it is unknown what effect prior or concurrent JAK inhibitor therapy would have on β3 adren- ergic agonist mediated-increases in Nestin+ MSC.24 It is increasingly evident, however, that strategies with the abil- ity to break through the self-perpetuating inflammatory and pro-oncogenic MPN microenvironment are required. Potential combination approaches with drugs acting on this axis to explore include the human pentraxin-2 protein analog PRM-151 (Promedia Pharmaceuticals), which may potentially synergistically reduce fibrosis, or with interfer- on, which may ‘switch on’ mutant-HSC-directed immune responses. Both of which are being assessed as single agents and in combination with ruxolitinib (reviewed by Harrison and McLornan25). Furthermore, an additional ben- efit of focusing on the stem cell niche could generate a sur- rogate marker of disease response that would facilitate more rapid evaluation of niche-targeting therapies in the clinical arena. Surrogate markers of disease response mov- ing beyond the blood count, symptoms and spleen are urgently required in this field. An enhanced understanding of the role of the neural network within the MPN niche and how this can be successfully modulated will accelerate the design of such synergistic therapeutic approaches and help the field to move forward. This work from Méndez- Ferrer and Skoda is also an inspiration in following the aca- demic trail from bench to bedside.13,14,17,23
References
1. Schmitt-Graeff AH, Nitschke R, Zeiser R. The Hematopoietic Niche in Myeloproliferative Neoplasms. Mediators Inflamm. 2015;2015:347270.
2. Schepers K, Campbell T, Passegué E. Normal and leukemic stem cell niches: insights and therapeutic opportunities. Cell Stem Cell.
2015;16(3):254-267.
3. GaoX,XuC,AsadaN,FrenettePS.Thehematopoieticstemcellniche:
from embryo to adult. Development. 2018;145(2).
4. Mead AJ, Mullally A. Myeloproliferative neoplasm stem cells. Blood.
2017;129(12):1607-1616.
5. ZhanH,LinCHS,SegalY,KaushanskyK.TheJAK2V617F-bearingvas-
cular niche promotes clonal expansion in myeloproliferative neo-
plasms. Leukaemia. 2018;32(2):462-469.
6. SchepersK,PietrasEM,ReynaudD,etal.Myeloproliferativeneoplasia
remodels the endosteal bone marrow niche into a self-reinforcing
leukemic niche. Cell Stem Cell. 2013;13(3):285-299.
7. Kim YW, Koo BK, Jeong HW, et al. Defective Notch activation in
microenvironment leads to myeloproliferative disease. Blood.
2008;112(12):4628-4638.
8. Walkley CR, Olsen GH, Dworkin S, et al. A microenvironment-
induced myeloproliferative syndrome caused by retinoic acid receptor
gamma deficiency. Cell. 2007;129(6):1097-1110
9. Wang L, Zhang H, Rodriguez S, et al. Notch-dependent repression of
κB-dependent manner. Cell Stem Cell. 2014;15(1):51-65.
10. Avanzini MA, Bernardo ME, Novara F, et al. Functional and genetic aberrations of in vitro-cultured marrow-derived mesenchymal stromal cells of patients with classical Philadelphia-negative myeloproliferative
miR-155 in the bone marrow niche regulates hematopoiesis in an NF-
neoplasms. Leukemia. 2014;28(8):1742-1745.
11. Martinaud C, Desterke C, Konopacki J, et al. Osteogenic Potential of
Mesenchymal Stromal Cells Contributes to Primary Myelofibrosis.
Cancer Res. 2015;75(22):4753-4765.
12. Ramos TL, Sánchez-Abarca LI, Rosón-Burgo B, et al. Mesenchymal
stromal cells (MSC) from JAK2+ myeloproliferative neoplasms differ from normal MSC and contribute to the maintenance of neoplastic hematopoiesis. PLoS One. 2017;12(8):e0182470.
13. Méndez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452(7186):442-447.
14. Méndez-Ferrer S, Michurina TV, Ferraro F, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010; 466(7308):829-834.
15. KatayamaY,BattistaM,KaoWM,etal.Signalsfromthesympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124(2):407-421.
16. MaryanovichM,ZahalkaAH,PierceH,etal.Adrenergicnervedegen- eration in bone marrow drives aging of the hematopoietic stem cell niche. Nat Med. 2018;24(6):782-791.
17. Arranz L, Sánchez-Aguilera A, Martín-Pérez D, et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neo- plasms. Nature. 2014;512(7512):78-81.
18. Zahalka AH, Arnal-Estapé A, Maryanovich M, et al. Adrenergic nerves activate an angio-metabolic switch in prostate cancer. Science. 2017;358(6361):321-326.
19. WolterJK,WolterNE,BlanchA,etal.Anti-tumoractivityofthebeta- adrenergic receptor antagonist propranolol in neuroblastoma. Oncotarget. 2014;5(1):161-172.
20. Magnon C, Hall SJ, Lin J, et al. Autonomic nerve development con- tributes to prostate cancer progression. Science. 2013; 341(6142):1236361.
21. GrytliHH,FagerlandMW,FossåSD,etal.Associationbetweenuseof β-bockers and prostate cancer-specific survival: A cohort study of 3561 prostate cancer patients with high-risk or metastatic disease. Eur Urol. 2014; 65(3):635-641.
22. Qiao G, Chen M, Bucsek MJ, et al. Adrenergic Signaling: A Targetable Checkpoint Limiting Development of the Antitumor Immune Response. Front Immunol. 2018;9:164.
23. Drexler B, Passweg JR, Tzankov A, et al. The sympathomimetic ago- nist mirabegron did not lower JAK2-V617F allele burden, but restored nestin-positive cells and reduced reticulin fibrosis in patients with myeloproliferative neoplasms: results of phase 2 study SAKK 33/14. Haematologica. 2019;104(4):710-716.
24. ZacharakiD,GhazanfariR,LiH,etal.EffectsofJAK1/2inhibitionon bone marrow stromal cells of myeloproliferative neoplasm (MPN) patients and healthy individuals. Eur J Haematol. 2018;101(1):57-67.
25. HarrisonCN,McLornanDP.Currenttreatmentalgorithmfortheman- agement of patients with myelofibrosis, JAK inhibitors, and beyond. Hematology Am Soc Hematol Educ Program. 2017;2017(1):489-497.
haematologica | 2019; 104(4)
641


































































































   11   12   13   14   15