Page 46 - 2019_05-HaematologicaMondo-web
P. 46

E. González-Romero et al.
and to ensure that epigenetic modification is heritable upon cell division.
Last but not least, the contribution of CRISPR/Cas9 to multiplex editing in CAR-T cells to create safer and more effective treatments cannot be underestimated, and it is highly likely that this technique will move forward the field of cancer immunotherapy for personalized T-cell- based therapies. The engineering of novel CAR-T cells by pharmaceutical industries, resulting in costly and unaf- fordable treatments for the general population, should be accompanied by their production by academia institu- tions, which could make it easier to tailor CAR-T cells for each patient. Thus, drug regulatory authorities should facilitate their academic production and provide resources for CAR-T- cell manufacturing processes, so that these can be simplified and automated to enable scaling up of these cell products.
Conclusions
CRISPR/Cas9 technology is a revolutionary approach for genome editing with wide applications in molecular biology, genetics, and medicine. It has great potential for dissecting gene function, modeling diseases and editing human genes for curative treatment. The number of pub- lications in this field has doubled every year since its intro- duction, and the CRISPR/Cas9 system is now more wide- ly used in biotechnology and research laboratories than
other, more time-consuming and expensive approaches such as zinc-finger nucleases or TALEN. In hematology, CRISPR/Cas9 can be used to model diseases using cul- tured cells or model organisms, but perhaps more impor- tantly, it can be a valuable approach to correct ex vivo mutations and chromosomal aberrations in cells from patients with blood disorders for autologous HSC trans- plantation. However, many pitfalls and challenges need to be overcome for the translation of CRISPR/Cas9 gene editing to the clinic. For example, we do not know the minimum number of edited cells needed to functionally correct a genetic defect or if gene editing can be applied to treat multigenic diseases. Further research is necessary to implement CRISPR/Cas9 in the clinical context, so that genome editing-based treatments are available to patients. In conclusion, the CRISPR/Cas9 revolution brings us a specific, efficient and versatile tool for editing genes. Nowadays, technology is no longer a limitation and scien- tists can probably do any genetic manipulation they can dream of.
Acknowledgments
This work was supported by Ayudas FEDER CIBERONC [CB16/12/00284], Instituto de Salud Carlos III [PI16/01113, PI17/00011, CP16/00011], Conselleria de Educación, Investigación, Cultura y Deporte [PROMETEOII/2015/008, ACIF/2018/255], MINECO [RYC-2015-17534, SAF2017- 82171-R], and Beca Leonardo a Investigadores y Creadores Culturales de la Fundación BBVA.
References
1. Mortensen R. Overview of gene targeting by homologous recombination. Curr Protoc Neurosci. 2007;Chapter 4:Unit 4.29.
2. Porteus MH, Baltimore D. Chimeric nucle- ases stimulate gene targeting in human cells. Science. 2003;300(5620):763.
3. Smith J, Grizot S, Arnould S, et al. A combi- natorial approach to create artificial homing endonucleases cleaving chosen sequences. Nucleic Acids Res. 2006;34(22):e149.
4. Urnov FD, Miller JC, Lee YL, et al. Highly efficient endogenous human gene correc- tion using designed zinc-finger nucleases. Nature. 2005;435(7042):646-651.
5. Miller JC, Tan S, Qiao G, et al. A TALE nuclease architecture for efficient genome editing. Nat Biotechnol. 2011;29(2):143-148.
6. Ishino Y, Shinagawa H, Makino K, Amemura M, Nakata A. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. J Bacteriol. 1987;169(12):5429-5433.
7. Mojica FJ, Ferrer C, Juez G, Rodriguez- Valera F. Long stretches of short tandem repeats are present in the largest replicons of the Archaea Haloferax mediterranei and Haloferax volcanii and could be involved in replicon partitioning. Mol Microbiol. 1995;17(1):85-93.
8. Mojica FJ, Diez-Villasenor C, Garcia- Martinez J, Soria E. Intervening sequences of regularly spaced prokaryotic repeats derive from foreign genetic elements. J Mol Evol. 2005;60(2):174-182.
9. Mojica FJ, Diez-Villasenor C, Garcia- Martinez J, Almendros C. Short motif sequences determine the targets of the prokaryotic CRISPR defence system. Microbiology. 2009;155(Pt3):733-740.
10. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816-821.
11. Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014;346 (6213):1258096.
12. Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas sys- tems. Science. 2013;339(6121):819-823.
13. Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science. 2013;339(6121):823-826.
14. Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J. RNA-programmed genome edit- ing in human cells. Elife. 2013;2:e00471.
15. Bibikova M, Golic M, Golic KG, Carroll D. Targeted chromosomal cleavage and muta- genesis in Drosophila using zinc-finger nucleases. Genetics. 2002;161(3):1169-1175.
16. Capecchi MR. Altering the genome by homologous recombination. Science. 1989;244(4910):1288-1292.
17. Wu WY, Lebbink JHG, Kanaar R, Geijsen N, van der Oost J. Genome editing by natural and engineered CRISPR-associated nucleas- es. Nat Chem Biol. 2018;14(7):642-651.
18. Lino CA, Harper JC, Carney JP, Timlin JA. Delivering CRISPR: a review of the chal- lenges and approaches. Drug Deliv. 2018;25(1):1234-1257.
19. Shi X, Kitano A, Jiang Y, Luu V, Hoegenauer KA, Nakada D. et al. Clonal expansion and myeloid leukemia progression modeled by multiplex gene editing of murine hematopoietic progenitor cells. Exp Hematol. 2018;64:33-44.e5.
20. Wu ZJ, Zhao X, Banaszak LG, et al. CRISPR/Cas9-mediated ASXL1 mutations in U937 cells disrupt myeloid differentia- tion. Int J Oncol. 2018;52(4):1209-1223.
21. Hannon GJ, Rossi JJ. Unlocking the potential of the human genome with RNA interfer- ence. Nature. 2004;431(7006):371-378.
22. Liu ET, Bolcun-Filas E, Grass DS, et al. Of mice and CRISPR: the post-CRISPR future of the mouse as a model system for the human condition. EMBO Rep. 2017;18(2): 187-193.
23. Zhang J, Lieu YK, Ali AM, et al. Disease- associated mutation in SRSF2 misregulates splicing by altering RNA-binding affinities. Proc Natl Acad Sci U S A. 2015;112(34): E4726-4734.
24. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209-2221.
25. Brabetz O, Alla V, Angenendt L, et al. RNA- guided CRISPR-Cas9 system-mediated engineering of acute myeloid leukemia mutations. Mol Ther Nucleic Acids. 2017;6:243-248.
26. Valletta S, Dolatshad H, Bartenstein M, et al. ASXL1 mutation correction by CRISPR/Cas9 restores gene function in leukemia cells and increases survival in mouse xenografts. Oncotarget. 2015;6(42): 44061-44071.
892
haematologica | 2019; 104(5)


































































































   44   45   46   47   48