Revisão Acesso aberto Revisado por pares

Optimizing autologous cell grafts to improve stem cell gene therapy

2016; Elsevier BV; Volume: 44; Issue: 7 Linguagem: Inglês

10.1016/j.exphem.2016.04.007

ISSN

1873-2399

Autores

Nikoletta Psatha, Garyfalia Karponi, Evangelia Yannaki,

Tópico(s)

Mesenchymal stem cell research

Resumo

•Challenges and interventions in optimizing cell grafts for gene therapy are discussed.•Large numbers of transduced hematopoietic stem cells (HSCs) with enhanced engraftment capacity are required for successful gene therapy.•Enrichment of HSC grafts may reduce target cell numbers and vector load.•Expansion of transduced HSCs without differentiation will potentially increase the effective dose of infused cells.•Priming of transduced HSCs with effector molecules will potentially enhance their repopulating capacity. Over the past decade, stem cell gene therapy has achieved unprecedented curative outcomes for several genetic disorders. Despite the unequivocal success, clinical gene therapy still faces challenges. Genetically engineered hematopoietic stem cells are particularly vulnerable to attenuation of their repopulating capacity once exposed to culture conditions, ultimately leading to low engraftment levels posttransplant. This becomes of particular importance when transduction rates are low or/and competitive transplant conditions are generated by reduced-intensity conditioning in the absence of a selective advantage of the transduced over the unmodified cells. These limitations could partially be overcome by introducing megadoses of genetically modified CD34+ cells into conditioned patients or by transplanting hematopoietic stem cells hematopoietic stem cells with high engrafting and repopulating potential. On the basis of the lessons gained from cord blood transplantation, we summarize the most promising approaches to date of increasing either the numbers of hematopoietic stem cells for transplantation or/and their engraftability, as a platform toward the optimization of engineered stem cell grafts. Over the past decade, stem cell gene therapy has achieved unprecedented curative outcomes for several genetic disorders. Despite the unequivocal success, clinical gene therapy still faces challenges. Genetically engineered hematopoietic stem cells are particularly vulnerable to attenuation of their repopulating capacity once exposed to culture conditions, ultimately leading to low engraftment levels posttransplant. This becomes of particular importance when transduction rates are low or/and competitive transplant conditions are generated by reduced-intensity conditioning in the absence of a selective advantage of the transduced over the unmodified cells. These limitations could partially be overcome by introducing megadoses of genetically modified CD34+ cells into conditioned patients or by transplanting hematopoietic stem cells hematopoietic stem cells with high engrafting and repopulating potential. On the basis of the lessons gained from cord blood transplantation, we summarize the most promising approaches to date of increasing either the numbers of hematopoietic stem cells for transplantation or/and their engraftability, as a platform toward the optimization of engineered stem cell grafts. Hematopoietic stem cell gene therapy (HSC-GT) represents an autologous therapeutic intervention by which a normal copy of a deficient gene is introduced into a patient's own HSCs to re-establish effective gene function. As such, HSC-GT bypasses the immunologic risks of allogeneic HSC transplantation and the immune suppression needed to prevent or control these risks. Today, HSC-GT offers a curative potential for diseases in which hematopoietic cell transplantation is suboptimal (i.e., metachromatic leukodystrophy) [1Peters C. Steward C.G. Hematopoietic cell transplantation for inherited metabolic diseases: An overview of outcomes and practice guidelines.Bone Marrow Transplant. 2003; 31: 229-239Crossref PubMed Scopus (0) Google Scholar] or the need for a well-matched donor precludes a significant number of patients from undergoing this therapeutic procedure (i.e., hemoglobinopathies) [2King A. Shenoy S. Evidence-based focused review of the status of hematopoietic stem cell transplantation as treatment of sickle cell disease and thalassemia.Blood. 2014; 123 (quiz 3210): 3089-3094Crossref PubMed Scopus (0) Google Scholar]. Over the last decade, the proof of principle that the genetic modification of autologous HSCs can provide durable cures in monogenic disorders has been reported for several diseases, including primary immunodeficiencies and lysosomal storage diseases [3Cavazzana-Calvo M. Hacein-Bev S. de Saint Basile G. et al.Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease.Science. 2000; 288: 669-672Crossref PubMed Scopus (1822) Google Scholar, 4Cartier N. Hacein-Bey-Abina S. Bartholomae C.C. et al.Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy.Science. 2009; 326: 818-823Crossref PubMed Scopus (760) Google Scholar, 5Aiuti A. Cattaneo F. Galimberti S. et al.Gene therapy for immunodeficiency due to adenosine deaminase deficiency.N Engl J Med. 2009; 360: 447-458Crossref PubMed Scopus (551) Google Scholar, 6Cavazzana-Calvo M. Payen E. Negre O. et al.Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia.Nature. 2010; 467: 318-322Crossref PubMed Scopus (540) Google Scholar, 7Biffi A. Montini E. Lorioli L. et al.Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy.Science. 2013; 341: 1233158Crossref PubMed Scopus (359) Google Scholar, 8Aiuti A. Biasco L. Scarmuzzo S. et al.Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott–Aldrich syndrome.Science. 2013; 341: 1233151Crossref PubMed Scopus (299) Google Scholar]. Despite the unequivocal success, depending on the underlying disease and transgene function, outcome may be suboptimal (chronic granulomatous disease [CGD], hemoglobinopathies), thus requiring improvements in culture conditions, vector design, numbers and quality of cells infused, conditioning, and so on. Although a single HSC is, theoretically, capable and sufficient to eventually repopulate the hematopoietic system in mice, the delayed reconstitution from a single cell or limited numbers of HSCs in humans is not compatible with life, and large numbers of infused cells are required for rapid engraftment and hematologic reconstitution after HSC transplantation [9Wagner J.E. Barker J.N. DeFor T.E. et al.Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: Influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival.Blood. 2002; 100: 1611-1618Crossref PubMed Scopus (33) Google Scholar, 10Gluckman E. Rocha V. Boyer-Chammard A. et al.Eurocord Transplant Group and the European Blood and Marrow Transplantation Group. Outcome of cord-blood transplantation from related and unrelated donors.N Engl J Med. 1997; 337: 373-381Crossref PubMed Scopus (0) Google Scholar]. In HSC-GT, in particular, where the ex vivo transduction process negatively affects the competitiveness and homing of gene-modified cells [11Dorrell C. Gan O.I. Pereira D.S. Hawley R.G. Dick J.E. Expansion of human cord blood CD34(+)CD38(−) cells in ex vivo culture during retroviral transduction without a corresponding increase in SCID repopulating cell (SRC) frequency: Dissociation of SRC phenotype and function.Blood. 2000; 95: 102-110PubMed Google Scholar], the need for large numbers of transduced HSCs with the capacity to robustly engraft long term is further magnified. Umbilical cord blood transplantation (UCBT) and HSC-GT face common challenges such as suboptimal HSC doses for infusion and impaired engraftment of transplanted cells. Toward overcoming several of the current shortcomings of UCBT and HSC-GT, investigators try to develop methods to expand ex vivo the HSCs or enhance their engraftment capacity. In this review, based mostly on lessons gained in the UCBT setting, we summarize current approaches and considerations toward this goal and deliberate on how these may be optimized for effective GT applications. Although the last decade granted clinical GT with sound achievements, successful implementation of GT still faces major constraints including, in certain cases, limited efficacy resulting from suboptimal transduction efficiency or engraftment incompetence of the gene-modified cells [6Cavazzana-Calvo M. Payen E. Negre O. et al.Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia.Nature. 2010; 467: 318-322Crossref PubMed Scopus (540) Google Scholar, 12Grez M. Reichenbach J. Schwäble J. Seger R. Dinauer M.C. Thrasher A.J. Gene therapy of chronic granulomatous disease: The engraftment dilemma.Mol Ther. 2011; 19: 28-35Abstract Full Text Full Text PDF PubMed Scopus (68) Google Scholar, 13Kang H.J. Bartholomae C.C. Paruzynski A. et al.Retroviral gene therapy for X-linked chronic granulomatous disease: Results from phase I/II trial.Mol Ther. 2011; 19: 2092-2101Abstract Full Text Full Text PDF PubMed Scopus (42) Google Scholar]. Despite highly successful transduction of HSCs in GT of immune deficiencies [8Aiuti A. Biasco L. Scarmuzzo S. et al.Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott–Aldrich syndrome.Science. 2013; 341: 1233151Crossref PubMed Scopus (299) Google Scholar] or lysosomal storage diseases [7Biffi A. Montini E. Lorioli L. et al.Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy.Science. 2013; 341: 1233158Crossref PubMed Scopus (359) Google Scholar], efficient gene transfer to HSCs with vectors bearing large and complex gene expression cassettes, such as globin vectors, remains challenging. The incorporation of elements of the human locus control region (LCR) into globin vectors improved gene transfer into HSCs, but at the expense of a severe compromise of vector titers because of the substantial length of the micro-LCR cassettes [14Hargrove P.W. Kepes S. Hanawa H. et al.Globin lentiviral vector insertions can perturb the expression of endogenous genes in beta-thalassemic hematopoietic cells.Mol Ther. 2008; 16: 525-533Abstract Full Text Full Text PDF PubMed Scopus (70) Google Scholar, 15Urbinati F. Arumugam P. Higashimoto T. et al.Mechanism of reduction in titers from lentivirus vectors carrying large inserts in the 3′LTR.Mol Ther. 2009; 17: 1527-1536Abstract Full Text Full Text PDF PubMed Scopus (39) Google Scholar]. The problem is further intensified when chromatin insulators are inserted into already large vector constructs, to protect the transgene expression from chromosomal position effects and/or shield the target genome from genotoxic events [15Urbinati F. Arumugam P. Higashimoto T. et al.Mechanism of reduction in titers from lentivirus vectors carrying large inserts in the 3′LTR.Mol Ther. 2009; 17: 1527-1536Abstract Full Text Full Text PDF PubMed Scopus (39) Google Scholar, 16Emery D.W. Yannaki E. Tubb J. Nishino T. Li Q. Stamatoyannopoulos G. Development of virus vectors for gene therapy of beta chain hemoglobinopathies: Flanking with a chromatin insulator reduces gamma-globin gene silencing in vivo.Blood. 2002; 100: 2012-2019Crossref PubMed Scopus (0) Google Scholar, 17Ramezani A. Hawley T.S. Hawley R.G. Performance- and safety-enhanced lentiviral vectors containing the human interferon-beta scaffold attachment region and the chicken beta-globin insulator.Blood. 2003; 101: 4717-4724Crossref PubMed Google Scholar]. Overall, in hemoglobinopathies, both gene transfer efficiency and titers remain suboptimal, and consequently, large vector production batches associated with high costs, as well as full myeloablation, are necessary to reach clinically relevant levels of engraftment [18Walters M.C. Rasko J. Hongeng S. et al.Update of results from the Northstar study (HGB-204): A phase 1/2 study of gene therapy for beta-thalassemia major via transplantation of autologous hematopoietic stem cells transduced ex-vivo with a lentiviral beta AT87Q-globin vector (LentiGlobin BB305).Blood. 2015; 126: 201Crossref Google Scholar]. Another obstacle that HSC-GT needs to circumvent is the significant loss of repopulating cells caused by culture conditions applied to facilitate successful gene transfer, which, in turn, hampers the long-term engraftment of gene-modified cells. Indeed, culture supplementation with cytokines induces changes in cell cycle, apoptosis, and adhesion molecules [19Szilvassy S.J. Meyerrose T.E. Ragland P.L. Grimes B. Homing and engraftment defects in ex vivo expanded murine hematopoietic cells are associated with downregulation of beta1 integrin.Exp Hematol. 2001; 29: 1494-1502Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar, 20Ahmed F. Ings S.J. Pizzey A.R. et al.Impaired bone marrow homing of cytokine-activated CD34+ cells in the NOD/SCID model.Blood. 2004; 103: 2079-2087Crossref PubMed Scopus (0) Google Scholar, 21Mazurier F. Gan O.I. McKenzie J.L. Doedens M. Dick J.E. Lentivector-mediated clonal tracking reveals intrinsic heterogeneity in the human hematopoietic stem cell compartment and culture-induced stem cell impairment.Blood. 2004; 103: 545-552Crossref PubMed Scopus (103) Google Scholar, 22Yong K.L. Fahey A. Pizzey A. Linch D.C. Influence of cell cycling and cell division on transendothelial migration of CD34+ cells.Br J Haematol. 2002; 119: 500-509Crossref PubMed Scopus (0) Google Scholar, 23Liu B. Buckley S.M. Lewis I.D. Goldman A.I. Wagner J.E. van der Loo J.C. Homing defect of cultured human hematopoietic cells in the NOD/SCID mouse is mediated by Fas/CD95.Exp Hematol. 2003; 31: 824-832Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar], ultimately leading to differentiation and loss of the primitive phenotype [24Shpall E.J. Quinones R. Giller R. et al.Transplantation of ex vivo expanded cord blood.Biol Blood Marrow Transplant. 2002; 8: 368-376Abstract Full Text Full Text PDF PubMed Google Scholar, 25De Lima M. McMannis J. Gee A. et al.Transplantation of ex vivo expanded cord blood cells using the copper chelator tetraethylenepentamine: A phase I/II clinical trial.Bone Marrow Transplant. 2008; 41: 771-778Crossref PubMed Scopus (0) Google Scholar, 26Delaney C. Heimfeld S. Brashem-Stein C. Voorhies H. Manger R.L. Bernstein I.D. Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution.Nat Med. 2010; 16: 232-236Crossref PubMed Scopus (377) Google Scholar, 27Zhang C.C. Kaba M. Iizuka S. Huynh H. Harvey F. Angiopoietin-like 5 and IGFBP2 stimulate ex vivo expansion of human cord blood hematopoietic stem cells as assayed by NOD/SCID transplantation.Blood. 2009; 111: 3415-3423Crossref Scopus (0) Google Scholar] of the transduced HSCs. In addition, transduction per se compromises the homing and engraftment potential of HSCs through their exposure to vector particles and supernatant [28Hall K.M. Horvath T.L. Abonour R. Cornetta K. Srour E.F. Decreased homing of retrovirally transduced human bone marrow CD34+ cells in the NOD/SCID mouse model.Exp Hematol. 2006; 34: 433-442Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar]. To minimize the peritransplant toxicity of HSC-GT in myeloablated recipients, especially those with comorbidities, several groups have proposed testing a nonmyeloablative conditioning; in this case, successful HSC-GT becomes highly challenging, especially when the disease background (i.e., thalassemia, CGD) does not allow for a selective pressure at the gene-corrected stem or early progenitor cell level on engraftment [29Sadelain M. Riviere I. Wang X. et al.Strategy for a multicenter phase I clinical trial to evaluate globin gene transfer in beta-thalassemia.Ann NY Acad Sci. 2010; 1202: 52-58Crossref PubMed Scopus (0) Google Scholar, 30Yannaki E. Emery D.W. Stamatoyannopoulos G. Gene therapy for β-thalassaemia: The continuing challenge.Expert Rev Mol Med. 2010; 12: e31Crossref PubMed Scopus (0) Google Scholar]. Because of the increased competiveness from the endogenous unmodified HSCs in this case, very high doses of HSCs, transduced with robustly performing vectors, will be needed to establish clinically relevant long-term engraftment. For all the reasons mentioned above, various approaches have been widely sought to increase the numbers of infused cells, enrich the cell grafts in stem cells, expand the HSCs ex vivo, or enhance the engrafting capacity of infused cells. Various sources of HSCs are being used in HSC transplantation, including bone marrow (BM), mobilized peripheral blood (mPB), and UCB. Mobilization, the pharmacologic egress of HSCs from BM niches to the peripheral blood, yields by leukapheresis severalfold larger numbers of CD34+ cells than BM [31To L.B. Haylock D.N. Simmons P.J. Juttner C.A. The biology and clinical uses of blood stem cells.Blood. 1997; 89: 2233-2258Crossref PubMed Google Scholar, 32Champlin R.E. Schmitz N. Horowitz M.M. et al.Blood stem cells compared with bone marrow as a source of hematopoietic cells for allogeneic transplantation.Blood. 2000; 95: 3702-3709PubMed Google Scholar, 33Bensinger W.I. Martin P.J. Storer B. et al.Transplantation of bone marrow as compared with peripheral-blood cells from HLA-identical relatives in patients with hematologic cancers.N Engl J Med. 2001; 344: 175-181Crossref PubMed Scopus (736) Google Scholar], providing also earlier hematopoietic recovery on transplantation. Therefore, not only are the majority of autologous and allogeneic HSC transplantations performed with mPBHSCs [34Gratwohl A. Baldomero H. Frauendorfer K. et al.The EBMT activity survey 2006 on hematopoietic stem cell transplantation: Focus on the use of cord blood products.Bone Marrow Transplant. 2008; 41: 687-705Crossref PubMed Scopus (0) Google Scholar], but also GT approaches use, in most cases, the granulocyte colony-stimulating factor (G-CSF)-mPB as HSC source for genetic modification. Purified CD34+ cell populations, obtained from mPB, remain markedly heterogeneous and contain, apart from HSCs with long-term repopulating capacity, a pool of multilineage progenitors with short-term engraftment potential [35Guenechea G. Gan O.I. Dorrell C. Dick J.E. Distinct classes of human stem cells that differ in proliferative and self-renewal potential.Nat Immunol. 2001; 2: 75-82Crossref PubMed Scopus (236) Google Scholar, 36McKenzie J.L. Gan O.I. Doedens M. Wang J.C.Y. Dick J.E. Individual stem cells with highly variable proliferation and self-renewal properties comprise the human hematopoietic stem cell compartment.Nat Immunol. 2006; 7: 1225-1233Crossref PubMed Scopus (99) Google Scholar]. To increase the chance that HSCs endowed with high repopulating capacity are targeted by a therapeutic vector while reducing the number of target cells that need to be modified ex vivo and, thereby, the large vector load and relevant cost, further purification of hematopoietic stem and progenitor cells beyond the standard CD34+ cell fraction may be needed. Currently, there is no unique immunophenotypic profile that safely and clearly demarcates primitive HSCs from transiently engrafting multipotent progenitors (MMPs). The similarity in immunophenotypic profile between long-term engrafting HSCs and MMPs [37Majeti R. Park C.Y. Weissman I.L. Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood.Cell Stem Cell. 2007; 1: 635-645Abstract Full Text Full Text PDF PubMed Scopus (199) Google Scholar] complicates the identification of a functional measure of stem and progenitor cell content. Short-term (colony-forming units [CFUs]) as well as long-term culture-initiating cell (LTC-IC) culture-based assays, together with xenogeneic transplantation models, are frequently used to assess the potency of human HSCs to sustain functional hematopoiesis and characterize “true” HSCs [38Doulatov S. Notta F. Laurenti E. Dick J.E. Hematopoiesis: A human perspective.Cell Stem Cell. 2012; 10: 120-136Abstract Full Text Full Text PDF PubMed Scopus (212) Google Scholar]. On the basis of such studies, populations of CD34+/CD90+/CD133+ cells [38Doulatov S. Notta F. Laurenti E. Dick J.E. Hematopoiesis: A human perspective.Cell Stem Cell. 2012; 10: 120-136Abstract Full Text Full Text PDF PubMed Scopus (212) Google Scholar], CD34+/CD38−/CD133+ and CD34+/CD38−/CD45RA−/CD90+ cells [37Majeti R. Park C.Y. Weissman I.L. Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood.Cell Stem Cell. 2007; 1: 635-645Abstract Full Text Full Text PDF PubMed Scopus (199) Google Scholar], or CD34+/CD38−/CD45RA−/CD90+/CD49f+ cells [39Notta F. Doulatov S. Laurenti E. Poeppl A. Jurisica I. Dick J.E. Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment.Science. 2011; 333: 218-221Crossref PubMed Scopus (258) Google Scholar] are considered to be enriched in the most primitive human HSCs. Very recently, using multistep screening instead of multiparametric flow cytometry analysis, Chen et al. [40Chen J.Y. Miyanishi M. Wang S.K. et al.Hoxb5 marks long-term haematopoietic stem cells and reveals a homogenous perivascular niche.Nature. 2016; 530: 223-227Crossref PubMed Scopus (0) Google Scholar] identified HoxB5, a gene whose expression is limited to the BM, as a marking gene of functional long-term HSCs in the mouse. By this approach, they demonstrated that only 7% to 35% of cells identified as HSCs with the previous solely immunophenotype-based approaches were LT-HSCs [40Chen J.Y. Miyanishi M. Wang S.K. et al.Hoxb5 marks long-term haematopoietic stem cells and reveals a homogenous perivascular niche.Nature. 2016; 530: 223-227Crossref PubMed Scopus (0) Google Scholar]. A number of groups used various CD34+-enriched cell populations to exploit the higher repopulating capacity of specific, competitive, CD34+ cell subpopulations or to indirectly improve the suboptimal transduction efficiencies or even reduce the high vector production costs by targeting lower cell numbers. Baldwin et al. [41Baldwin K. Urbinati F. Romero Z. et al.Enrichment of human hematopoietic stem/progenitor cells facilitates transduction for stem cell gene therapy.Stem Cells. 2015; 33: 1532-1542Crossref PubMed Scopus (4) Google Scholar] reported increased susceptibility to transduction with markedly smaller vector amounts by further purification of HSCs beyond the standard CD34+ cell-enriched fraction, through the isolation and transduction with lentiviral vectors of CD34+/CD38− cells as compared with the unfractionated CD34+ cells [41Baldwin K. Urbinati F. Romero Z. et al.Enrichment of human hematopoietic stem/progenitor cells facilitates transduction for stem cell gene therapy.Stem Cells. 2015; 33: 1532-1542Crossref PubMed Scopus (4) Google Scholar]. These sorted and gene-modified CD34+/CD38− cells were approximately one-hundredfold more competitive for engraftment than their CD34+/CD38+ counterparts when xenotransplanted into NSG mice. These data imply that in a clinical setting, transduction of CD34+/CD38− cells would allow the use of limited amounts of viral vector to achieve high rates of transduced HSCs with good engraftment capacity, thus generating treatment opportunities for more patients. In another approach to target HSCs with a more primitive phenotype, Brendel et al. [42Brendel C. Goebel B. Daniela A. et al.CD133-targeted gene transfer into long-term repopulating hematopoietic stem cells.Mol Ther. 2015; 23: 63-70Abstract Full Text Full Text PDF PubMed Scopus (10) Google Scholar] has recently reported that direct gene transfer into unstimulated CD34+ cells by a receptor-targeting CD133-LV lentiviral vector, which uses the CD133 surface marker of primitive HSCs as entry receptor. This allows for sustained long-term engraftment of gene-corrected cells in immunodeficient mice, even after secondary transplantation [42Brendel C. Goebel B. Daniela A. et al.CD133-targeted gene transfer into long-term repopulating hematopoietic stem cells.Mol Ther. 2015; 23: 63-70Abstract Full Text Full Text PDF PubMed Scopus (10) Google Scholar], an effect that was attributed to preferential gene transfer into cells with higher engrafting and repopulating capacity. Novel mobilization approaches combining plerixafor with G-CSF have resulted in increased mobilization of the CD34+/CD38− or CD34+/CD133+/CD38− cell subsets, thus providing pharmacologic enrichment in HSCs of mobilized cell grafts [43Fruehauf S. Veldwijk M.R. Seeger T. et al.A combination of granulocyte-colony-stimulating factor (G-CSF) and plerixafor mobilizes more primitive peripheral blood progenitor cells than G-CSF alone: Results of a European phase II study.Cytotherapy. 2009; 11: 992-1001Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar, 44Varmavuo V. Mäntymaa P. Silvennoinen R. Nousiainen T. Kuittinen T. Jantunen E. CD34+ cell subclasses and lymphocyte subsets in blood grafts collected after various mobilization methods in myeloma patients.Transfusion. 2013; 53: 1024-1032Crossref PubMed Scopus (0) Google Scholar, 45Varmavuo V. Mäntymaa P. Nousiainen T. Valonen P. Kuittinen T. Jantunen E. Blood graft composition after plerixafor injection in patients with NHL.Eur J Haematol. 2012; 89: 128-135Crossref PubMed Scopus (0) Google Scholar]. Extensive studies from our group, using thalassemia as a model, have defined plerixafor + G-CSF-mobilized cells as an optimal graft source for thalassemia GT, but also for HSC-GT applications in general; they not only contain very large numbers of CD34+ cells obtained through single collections [46Yannaki E. Karponi G. Zerrvou F. et al.Hematopoietic stem cell mobilization for gene therapy: Superior mobilization by the combination of granulocyte-colony stimulating factor plus plerixafor in patients with β-thalassemia major.Hum Gene Ther. 2013; 24: 852-860Crossref PubMed Google Scholar], but importantly also exhibit superior competitive long-term engraftment in a thalassemic mouse model [47Psatha N. Sgouramali E. Gkountis A. et al.Superior long-term repopulating capacity of G-CSF + plerixafor-mobilized blood: implications for stem cell gene therapy by studies in the Hbb(th-3) mouse model.Hum Gene Ther Methods. 2014; 25: 317-327Crossref Scopus (0) Google Scholar] or enhanced early human chimerism after transduction/transplantation under nonmyeloablative conditioning in xenografts [48Karponi G. Psatha N. Lederer C.W. et al.Plerixafor + G-CSF–mobilized CD34+ cells represent an optimal graft source for thalassemia gene therapy.Blood. 2015; 126: 616-619Crossref Scopus (0) Google Scholar]. Even though the transduction rates with a lentiviral globin vector were lower for plerixafor + G-CSF-mobilized cells than for cells mobilized with either agent alone, there was increased β-globin expression per vector copy, implying that a given required level of expression might be achieved at a lower vector copy number, thus providing higher biosafety in GT applications. Because cells bearing a more primitive phenotype may be less permissive of gene transfer, optimization of the culture process with preservation of their primitive cell nature may be needed, as was recently suggested [49Genovese P. Schiroli G. Escobar G. et al.Targeted genome editing in human repopulating haematopoietic stem cells.Nature. 2014; 510: 235-240Crossref PubMed Scopus (134) Google Scholar]. The need for large numbers of transplantable HSCs becomes particularly challenging in UCBT because of the size of suitable grafts in blood banks and the small number of recovered stem cells, as well as in HSC-GT approaches, in which the culture conditions, suboptimal transduction rates, or nonmyeloablative conditioning may lead to impaired engraftment. Ex vivo expansion of HSCs would be a most satisfactory resolution in a GT setting by providing large numbers of gene-corrected cells, should the expanded cells retain their competitiveness and repopulating capacity in vivo. However, ex vivo culture of HSCs is neither easy nor always successful, as a plethora of intrinsic and extrinsic mechanisms are responsible for the fate and renewal of HSCs. Initial attempts to expand HSCs using various combinations of cytokines generated significant expansions in myeloid progenitors but, at best, modest increases in long-term repopulating cells [50Bhatia M. Bonnet D. Kapp U. Wang J.C. Murdoch B. Dick J.E. Quantitative analysis reveals expansion of human hematopoietic repopulating cells after short-term ex vivo culture.J Exp Med. 1997; 186: 619-624Crossref PubMed Scopus (313) Google Scholar, 51Ueda T. Tsuji K. Yoshino H. et al.Expansion of human NOD/SCID-repopulating cells by stem cell factor, Flk2/Flt3 ligand, thrombopoietin, IL-6, and soluble IL-6 receptor.J Clin Invest. 2000; 105: 1013-1021Crossref PubMed Google Scholar]. Early clinical trials of UCBT, conducted with the expectation that the increased committed progenitors would at least enhance the delayed neutrophil count recovery, failed to achieve, in most cases, an improvement in neutrophil or platelet recovery [24Shpall E.J. Quinones R. Giller R. et al.Transplantation of ex vivo expanded cord blood.Biol Blood Marrow Transplant. 2002; 8: 368-376Abstract Full Text Full Text PDF PubMed Google Scholar, 52Jaroscak J. Goltry K. Smith A. et al.Augmentation of umbilical cord blood (UCB) transplantation with ex vivo-expanded UCB cells: Results of a phase 1 trial using the AastromReplicell System.Blood. 2003; 101: 5061-5067Crossref PubMed Google Scholar]. Therefore, current approaches to HSC expansion target molecular pathways governing HSC self-renewal or cell fate decisions. In an attempt to simulate the HSC niche and BM microenvironment that allows self-renewal and proliferation of the more primitive hematopoietic cells, as well as to recapitulate the molecular signals and interactions directing these properties, co-cultures of HSCs with feeder, nonhematopoietic cells have been assayed by numerous groups to ex vivo expand HSCs. Hematopoietic stem and progenitor cell (HSPC) expansion was achieved by culturing hematopoietic cells on feeder cells derived from various sources including

Referência(s)