Limitations of G-CSF Mobilization in Splenectomized Patients with Beta-Thalassemia Major: Implications for Thalassemia Gene Therapy.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2150-2150
Author(s):  
Evangelia Yannaki ◽  
Thalia Papayannopoulou ◽  
Erica Jonlin ◽  
Ioannis Batsis ◽  
Pamela S. Becker ◽  
...  

Abstract Abstract 2150 Poster Board II-127 For gene therapy (GT) of thalassemia (TH), high numbers of genetically-modified hematopoietic stem cells (HSCs) are required to effectively compete for niche space in the hypercellular thalassemic bone marrow (bm). Mobilized peripheral blood is the preferable source of HSCs for thalassemia GT due to higher yields of CD34+cells compared to bm harvest. There is limited information on the mobilization efficacy of adult patients with major β-TH as well as on the safety of the procedure in a condition of splenomegaly and extramedullary hemopoiesis. Rare events of splenic rupture or thrombosis with G-CSF in normal donors raise safety concerns for its use in TH where chronic splenomegaly and hypercoagulability exist. Pretreatment of patients with hydroxyurea (HU) could reduce the risk of splenic rupture or thrombosis by decreasing the splenic hemopoiesis and thereby the spleen size in the non-splenectomized (non-SPL), and the circulating cells in the splenectomized (SPL) patients before G-CSF. In an on going mobilization study, we aim to assess the safety and efficacy of G-CSF mobilization with or without HU pretreatment in adult patients with β-TH major. Sufficient mobilization is considered to be the yield of ≥2×106CD34+cells/kg/2aphereses. Sixteen patients have been enrolled so far, 9 SPL and 7 non-SPL. One non-SPL patient withdrew during the study. Six SPL and 4 non-SPL patients received HU pretreatment (20mg/kg/d the non-SPL, 25-30mg/kg/d the SPL) for 1 month before G-CSF. There was a 1-2 weeks' interval between HU cessation and G-CSF initiation. No severe adverse events were observed. In non-SPL patients, HU decreased the spleen volume over baseline (306cm3 vs 536cm3, p=0,03) resulting in 9% max increase during mobilization compared to 45% size increase in patients w/o HU pretreatment. In non-SPL patients, HU negatively affected the CD34+yield when the ‘wash-out' period before G-CSF was 8 days (mean CD34+cells 1,86±0,76×106/kg/2aphereses, n=2). However, when the interval period from the HU stop to the G-CSF initiation increased up to 18 days allowing for bm recovery after the myelosuppressive effect of HU, mobilization was successful (P12:CD34+cells 6,5×106/kg/2aph). Non-SPL patients w/o HU pretreatment (n=3) yielded adequate numbers of HSCs (CD34+cells:5,8±3,89×106/kg/2aph). Surprisingly, CD34+cell yields were very low in the first 2 non-HU pretreated SPL patients (CD34+cells:0,98±0,14×106/kg/2aph). This was due to the development of early excessive leukocytosis (mean max WBCs 81×109/l, day 3) with the regular 10mcg/kg/d G-CSF dose, which necessitated dose hold or therapeutic leukapheresis and resulted in loss of the CD34+cell peak in blood. However, when mobilization started with lower (2,5mcg/kg/d) and adjusted to the WBCs doses of G-CSF (mean daily dose 3,21mcg/kg) and aphereses were initiated later (day6), CD34+cell yield markedly improved (P15:4,5×106/kg/2aph) without inducing early excessive leukocytosis (max WBCs 67×109/l, day 5). In SPL patients, HU was shown to decrease the high PLT and WBC numbers before G-CSF (PLTs:from 640±132×109/l at baseline to 240±53×109/l, p=0,0002 / WBCs:from 20,23±15,8×109/l at baseline to 11,47±6,11×109/l, p=0,23) potentially reducing the risk of thrombosis and partially preventing excessive leukocytosis during mobilization (max WBCs SPL-HU:51,68±29,37×109/l vs SPL-no HU:74,77±8,78×109/l, p=0,23). HU pretreatment negatively affected the yield in SPL patients when the ‘wash-out' period before G-CSF was 7-10 days (mean CD34+yield 0.62±0,41×106/kg/2aph, n=5). However, when the interval period from HU stop to G-CSF initiation increased up to 12 days, mobilization was successful (P16:CD34+cells 3,8×106/kg/2aphereses). G-CSF dose adjustment was also needed in HU-pretreated SPL patients with WBCs≥14,6×109/l before G-CSF (P9,P16). Overall, it seems that mobilization of SPL thalassemic patients is challenging. Mobilization is not inherently inefficient in SPL patients but it results from mandatory G-CSF-dose modifications to avoid hyperleukocytosis. Patient-tailored schemes of G-CSF mobilization or alternative ways of mobilization (ie AMD 3100) will be required in order to obtain high numbers of HSCs from SPL patients. HU seems to play a safety role as pretreatment before mobilization, especially in the SPL patients, however the time to G-CSF initiation after HU cessation is critical for a sufficient CD34+cell yield. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2901-2901
Author(s):  
Maria Rosa Lidonnici ◽  
Annamaria Aprile ◽  
Marta Claudia Frittoli ◽  
Giacomo Mandelli ◽  
Bernhard Gentner ◽  
...  

Abstract Successful gene therapy of inherited blood diseases relies on transplantation and engraftment of autologous genetically engineered hematopoietic stem/progenitor cells (HSPCs) in myeloablated patients. Hematopoietic reconstitution and clinical benefit are related to cell dose, although single disease features might play a role favoring selection of relevant progenitor populations. Gene therapy trials in young pediatric patients are performed isolating CD34+ cells from bone marrow (BM), while in adults mobilized peripheral blood stem cells (PBSC) should represent the favorite target. In the context of gene therapy for thalassemia, the choice of HSPC source is crucial since intrinsic characteristics of patients (splenomegaly and thrombophilia) dictate caution in the use of G-CSF as mobilization agent and prompt investigation of new agents. Moreover, adult thalassemic patients may possibly have a decreased BM stem cell reservoir, due to the BM suppression in response to multiple transfusions. A phase II clinical protocol exploring the use of Plerixafor as a single mobilizing agent in adult patients affected by transfusion dependent beta-thalassemia (EudraCT 2011-000973-30) started in 2012 at our hospital. Plerixafor selectively and reversibly antagonizes the binding of SDF-1 to its receptor CXCR4 with subsequent egress of HSCs to the peripheral blood. The availability of a new source of HSPCs, potentially superior in terms of CD34+ cell yield, transduction efficiency and biological features to steady-state BM, would have a significant impact on the feasibility and efficacy of gene therapy. Four subjects were enrolled and treated by subcutaneously administration of Plerixafor at the single dose of 0.24 mg/kg followed by leukoapheresis. Mobilization of CD34+ cells occurred very rapidly with a peak between 7 to 9 hrs. Three out of four patients achieved the minimal target cell dose (2 x 106 cells/kg) and no severe adverse event occurred. To the aim of engineering Plerixafor-mobilized CD34+ cells for gene therapy, we performed a comprehensive characterization of their biological, molecular and functional properties. In vivo reconstitution potential and lympho-myeloid differentiation were tested following transplantation in NSG mice and compared to those of PBSCs mobilized by G-CSF. Percentages of engrafted human cells in NSG mice transplanted with Plerixafor -PBSCs were about 2- to 5-fold higher than those found in mice transplanted with G-CSF PBSCs. On the same line, the SRC frequency, obtained by pooled engraftment data, was significantly higher (1 SRC out of 47.875 CD34+ cells vs.1 SRC out of 141.203 CD34+ cells). The phenotypic analysis of the frequency of primitive hematopoietic sub-populations revealed that Plerixafor mobilizes preferentially HSPCs and LT-HSPCs, with a percentage of CD34+ CD38-/low CD90+ CD45RA- CD49f+ cells higher than that found in G-CSF PBSCs. This result mirrors the enhanced number of SRCs found in the CD34+ cell population mobilized by Plerixafor. In order to further define the molecular features of HSPCs from different sources, we are studying signalling networks in response to specific cytokines by phospho-proteins analysis and gene expression by microarrays analysis. Our studies are focused on self-renewal, homing, engraftment and multilineage differentiation processes and bioinformatic analysis will reveal the molecular machinery underlying 'stemness' properties of Plerixafor mobilized cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. SCI-16-SCI-16
Author(s):  
Mitchell J Weiss

Genetic forms of anemia caused by HBB gene mutations that impair beta globin production are extremely common worldwide. The resultant disorders, mainly sickle cell disease (SCD) and beta-thalassemia, cause substantial morbidity and early mortality. Treatments for these diseases include medical therapies and bone marrow transplantation (BMT), which can be curative. However, medical therapies are suboptimal and BMT is associated with serious toxicities, particularly because HLA-matched allogeneic sibling donors are not available for most patients. Thus, new therapies are urgently needed for millions of affected individuals. Gene therapy offers great promise to cure SCD and beta thalassemia and emerging genome editing technologies represent a new form of gene therapy. Approaches to cure SCD and beta-thalassemia via genome editing include: 1) Correction of HBB mutations by homology directed repair (HDR); 2) use of non-homologous end joining (NHEJ) to activate gamma globin production and raise fetal hemoglobin (HbF) levels; 3) NHEJ to disrupt alpha-globin genes (HBA1 or HBA2) and thereby alleviate globin chain imbalance in intermediately severe forms of beta thalassemia. Challenges for these approaches include selection of the most effective genome editing tools, optimizing their delivery to hematopoietic stem cells (HSCs), improving specificity and better understanding potential off target effects, particularly those that are biologically relevant. Technologies for genome editing are advancing rapidly and being tested in preclinical models for HBB-mutated disorders. Ultimately, however, the best strategies can only be identified in clinical trials. This will require close collaborations between basic/translational researchers who study genome editing, clinical hematologists and collaboration between experts in academia and the bio-pharmaceutical industry. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3378-3378
Author(s):  
Jianyun Wen ◽  
Yuelin He ◽  
Libai Chen ◽  
Jing Du ◽  
Zhiyong Peng ◽  
...  

Abstract Background: Peripheral blood stem cells (PBSC) are increasingly used as a source of stem cells for either autologous or allogeneic hematopoietic transplantation in children.Although technically similar to adult procedures, PBSC harvest may be difficult in young children, especially in the very small children. Aim: In this study, we aimed to evaluatethe safety and efficacy of harvesting peripheral blood hematopoietic stem cells in very small children,and to provide a guideline. Methods: Between Jan 2013 to Mar 2016, we evaluated 118 children weighing 20 kg or less, with the smallest patient weighing 11 kg. The patients had a median age of 59 months and included 72 children with thalassemia major and 46 young donors. The granulocyte-colony stimulating factor (G-CSF) analogs were used at a dose of 10 mg/kg/day administered subcutaneously once a day and receiving oral calcium for five days before harvesting. Blood was withdrawn at a mean rate of 30-40 ml/min through a temporaryfemoral vein catheter (12 or 14 guage) to ensure adequate blood flow and returned through a larger catheter in a peripheral vein.Total nucleated cells(TNC) and CD34+ cells were estimated in the peripheral blood before collection of the apheresis product. Results: We collected sufficient products from all the children with one to three apheresis procedures. No serious complication was detected in all children and all aphereses were completed within 4 hours.The volume of blood per kilogram processed for each apheresis ranged from 55 to 160ml (median, 85ml). The median number of TNC and CD34+ cells collected were 12×108/kg and 15×106/kg per apheresis, respectively. Conclusions:We conclude that collection of PBSC is a safe and practical procedure in children, even in very small children. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. SCI-23-SCI-23
Author(s):  
Giuliana Ferrari

Beta-thalassemia and sickle cell disease (SCD) are congenital anemias caused by mutations in the beta-globin gene, resulting in either reduced/absent production of globin chains or abnormal hemoglobin structure. At present, the definitive cure is represented by allogeneic hematopoietic stem cell transplantation, with a probability to find a well-matched donor of <25%. Experimental gene therapy for hemoglobinopathies is based on transplantation of autologous hematopoietic stem cells genetically modified to express therapeutic hemoglobin levels. Approaches to genetically modify HSCs for treatment of hemoglobinopathies include: 1) the addition of globin genes by lentiviral vectors and 2) gene editing by nucleases to reactivate fetal hemoglobin either through inhibition of repressors or by reproducing mutations associated with high fetal hemoglobin levels. The outcomes of early clinical trials are showing the safety and potential efficacy, as well as the hurdles still limiting a general application.Current challenges and improved strategies will be presented and discussed. Disclosures No relevant conflicts of interest to declare. OffLabel Disclosure: Plerixafor


2019 ◽  
Vol 20 (16) ◽  
pp. 1603-1623 ◽  
Author(s):  
Eman M. Hamed ◽  
Mohamed Hussein Meabed ◽  
Usama Farghaly Aly ◽  
Raghda R.S. Hussein

Beta-thalassemia is a genetic disorder characterized by the impaired synthesis of the betaglobin chain of adult hemoglobin. The disorder has a complex pathophysiology that affects multiple organ systems. The main complications of beta thalassemia are ineffective erythropoiesis, chronic hemolytic anemia and hemosiderosis-induced organ dysfunction. Regular blood transfusions are the main therapy for beta thalassemia major; however, this treatment can cause cardiac and hepatic hemosiderosis – the most common cause of death in these patients. This review focuses on unique future therapeutic interventions for thalassemia that reverse splenomegaly, reduce transfusion frequency, decrease iron toxicity in organs, and correct chronic anemia. The targeted effective protocols include hemoglobin fetal inducers, ineffective erythropoiesis correctors, antioxidants, vitamins, and natural products. Resveratrol is a new herbal therapeutic approach which serves as fetal Hb inducer in beta thalassemia. Hematopoietic stem cell transplantation (HSCT) is the only curative therapy for beta thalassemia major and is preferred over iron chelation and blood transfusion for ensuring long life in these patients. Meanwhile, several molecular therapies, such as ActRIIB/IgG1 Fc recombinant protein, have emerged to address complications of beta thalassemia or the adverse effects of current drugs. Regarding gene correction strategies, a phase III trial called HGB-207 (Northstar-2; NCT02906202) is evaluating the efficacy and safety of autologous cell transplantation with LentiGlobin. Advanced gene-editing approaches aim to cut DNA at a targeted site and convert HbF to HbA during infancy, such as the suppression of BCL11A (B cell lymphoma 11A), HPFH (hereditary persistence of fetal hemoglobin) and zinc-finger nucleases. Gene therapy is progressing rapidly, with multiple clinical trials being conducted in many countries and the promise of commercial products to be available in the near future.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 5473-5473
Author(s):  
Giuliana Ferrari ◽  
Emanuela Roselli ◽  
Francesca Tiboni ◽  
Erica Biral ◽  
Sarah Marktel ◽  
...  

Abstract Gene therapy for beta-thalassemia is based on the transplantation of genetically-modified autologous hematopoietic stem cells (HSC) into patients affected by the severe form of disease. The genetic treatment of the hemoglobinopathies poses the general challenge of efficient level of gene transfer into HSC and high and persistent transgene expression, in the differentiated progeny of a genetically modified stem cell. The validation of a gene therapy approach to thalassemia requires to obtain results of gene correction in a broad number of patients’ cells, since different molecular defects in the beta-globin gene lead to the clinical phenotype. The heterogeneity in the molecular defects and in the proportion of alpha and non-alpha (beta, gamma and delta) chains will represent a key element to set a threshold in the amount of vector-derived beta-chain required to correct a thalassemic phenotype. Additionally, the impact of some biological parameters, such as the degree of BM erythroid hyperplasia, the BM subpopulations proportion and the apoptotic index, on the successful correction of thalassemic phenotype needs to be studied in the perspective of clinical translation. In order to address these issues, we collected samples from BM aspirates and isolated CD34+ cells from 25 beta+ and beta0 thalassemic patients, characterized by different genotypes and biochemical profiles of globin chains synthesis. A novel, erythroid specific LV expressing human beta-globin from a minimal promoter enhanced by only 2 LCR elements (HS2 and HS3) was used to transduce BM derived CD34+ cells at high efficiency (>80%). The efficacy of the beta-globin LV in correcting the human thalassemic phenotype was tested in an in vitro model of erythropoiesis and in the human-mouse hematological chimera. Upon transduction, normal level of HbA expression was achieved in erythroblastic cultures and BFU-E, associated with a progression towards erythroid maturation, which was impaired in mock-transduced thalassemic cells. Molecular analysis showed proviral integrity, with no detectable rearrangements and an average proviral copy number of 2.4. Analysis of specific globin chains proportion and contribution to phenotype correction in the context of different genotypes is under evaluation.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 549-549 ◽  
Author(s):  
Alexis A. Thompson ◽  
John E Rasko ◽  
Suradej Hongeng ◽  
Janet L. Kwiatkowski ◽  
Gary Schiller ◽  
...  

Abstract Background: Hematopoietic stem cell (HSC) gene therapy has the potential to induce globin production and mitigate the need for blood transfusions in β-thalassemia major. Promising early results for 2 subjects with β0/βE -thalassemia major in the ongoing HGB-205 study suggested that transplantation with autologous CD34+ cells transduced with a replication-defective, self-inactivating LentiGlobin BB305 lentiviral vector containing an engineered β-globin gene (βA-T87Q) can be safe and yield robust production of βA-T87Qglobin resulting in rapid transfusion independence. The Northstar study (HGB-204), which uses the same lentivirus vector and analogous study design as study HGB-205, is multi-center and multi-national, and centralizes drug product manufacturing. Herein, we provide the initial data on subjects enrolled and treated in this study. Subjects and Methods: Transfusion-dependent subjects with β-thalassemia major undergo HSC collection via mobilized peripheral blood apheresis and CD34+ cells are selected. Estimation of the mean ex-vivo vector copy number (VCN) is obtained by quantitative PCR performed on pooled colony-forming progenitors. Subjects undergo myeloablation with intravenous busulfan, followed by infusion of transduced CD34+ cells. Subjects are monitored for hematologic engraftment, βA-T87Q -globin expression (by high performance liquid chromatography) and transfusion requirements. Integration site analysis (ISA, by linear amplification-mediated PCR and high-throughput sequencing on nucleated cells) and replication-competent lentivirus (RCL) assays are performed for safety monitoring. Results: As of 31 July 2014, 3 subjects have undergone HSC collection and ex-vivo LentiGlobin BB305 gene transfer. One subject (Subject 1102) has undergone myeloablation and drug product infusion. Outcomes data are shown in Table 1. The initial safety profile is consistent with myeloablation, without serious adverse events or gene therapy-related adverse events. This subject has increasing production of βA-T87Q-globin: the proportion of βA-T87Qglobin was 1.5%, 10.9% and 19.5% of total Hb at 1, 2 and 3 months post-infusion, respectively. This subject received pRBCs on Day +14 following drug product infusion and required no further transfusions until a single unit of pRBC was transfused on Day +96 for a Hb of 8.6 g/dL and fatigue. Two additional subjects have undergone drug product manufacture and are awaiting transplantation. Safety data related to ISA and RCL assays are pending. Abstract 549. Table 1 Preliminary results of dosing parameters and transplantation outcomes Subject Age (years) and Gender Genotype BB305 Drug Product Day of Neutrophil Engraftment Drug Product- related Adverse Events βA-T87Q-Hb at last follow-up visit /Total Hb (g/dL) VCN CD34+ cell dose (x106 per kg) 1102 18 F β0/βE 1.0/1.1a 6.5 Day +17 None 1.77/8.6 1104 21 F β0/βE 0.7/0.7a 5.4 P P P 1106 20 F β0/β0 1.5 12.3 P P P As of 31 July 2014; P, pending a If more than one drug product were manufactured, the VCN of each drug product lot is presented. Conclusion: The first subject treated on the Northstar study has safely undergone drug product infusion with autologous HSCs transduced with LentiGlobin BB305 lentiviral vector and is producing steadily increasing amounts of βA-T87Q-globin. Additional follow-up of this subject plus data on additional subjects who undergo drug product infusion will be presented at the meeting. Ex-vivo gene transfer of βA-T87Q-globin to autologous HSCs is a promising approach for the treatment of patients with β-thalassemia major. Disclosures Thompson: ApoPharma: Consultancy; Novartis: Consultancy, Research Funding; Amgen: Research Funding; Glaxo Smith Kline: Research Funding; Mast: Research Funding; Eli Lilly: Research Funding. Kwiatkowski:Shire Pharmaceuticals and Sideris Pharmaceuticals: Consultancy. Schiller:Sunesis, Amgen, Pfizer, Bristol Myers Squibb: Research Funding. Leboulch:bluebird bio: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding. Petrusich:bluebird bio, Inc.: Employment, Equity Ownership. Soni:bluebird bio, Inc.: Employment. Walters:Via Cord and AllCells, Inc.: Medical Director Other.


Hematology ◽  
2012 ◽  
Vol 2012 (1) ◽  
pp. 276-283 ◽  
Author(s):  
Emmanuel Payen ◽  
Philippe Leboulch

Abstract High-level production of β-globin, γ-globin, or therapeutic mutant globins in the RBC lineage by hematopoietic stem cell gene therapy ameliorates or cures the hemoglobinopathies sickle cell disease and beta thalassemia, which are major causes of morbidity and mortality worldwide. Considerable efforts have been made in the last 2 decades in devising suitable gene-transfer vectors and protocols to achieve this goal. Five years ago, the first βE/β0-thalassemia major (transfusion-dependent) patient was treated by globin lentiviral gene therapy without injection of backup cells. This patient has become completely transfusion independent for the past 4 years and has global amelioration of the thalassemic phenotype. Partial clonal dominance for an intragenic site (HMGA2) of chromosomal integration of the vector was observed in this patient without a loss of hematopoietic homeostasis. Other patients are now receiving transplantations while researchers are carefully weighing the benefit/risk ratio and continuing the development of further modified vectors and protocols to improve outcomes further with respect to safety and efficacy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3759-3759
Author(s):  
Cecilia Barese ◽  
Connor King ◽  
Stephanie Sellers ◽  
Allen E Krouse ◽  
Mark E Metzger ◽  
...  

Abstract Abstract 3759 For genetic blood diseases, such as primary immunodeficiencies, gene therapy targeted to hematopoietic stem cells (HSCs) is a feasible and now proven effective therapeutic option for patients who lack a histocompatible HSC. However, the risk of adverse outcomes resulting from insertional oncogenesis is a major concern. We are investigating whether inclusion of the herpes simplex virus thymidine kinase (HSVtk) gene into integrating vectors into rhesus macaque HSCs confers ganciclovir (GCV) sensitivity allowing ablation of vector-containing cells from the blood and other hematopoietic compartments, as an approach to increasing safety of gene therapy procedures. HSVtk suicide genes have been studied in detail in transduced mature T cells, but never in stem and progenitor cells. We infused autologous CD34+ cells transduced ex vivo with gammaretrovirus vectors encoding the HSVtk as suicide gene along with marker genes into 4 rhesus macaques, following myeloablative irradiation. In the first animal, a vector consisting of the MND backbone driving the sr39 high affinity tk mutant, and IRES and a truncated NGFR marker gene was used. A stable marking level of 5% NGFR+ circulating cells was observed for 6 months following transplantation, confirmed by q-PCR. The drug GCV was infused at 5 mg/Kg BID for 21 days. This animal had complete elimination of vector-containing cells in all peripheral blood lineages as assessed by flow cytometry and qPCR, and remains negative now 4 months after GCV discontinuation. Three additional animals were transplanted with autologous CD34+ cells transduced with a vector containing a standard HSVtk gene and GFP as a marker. These animals had lower stable marking levels of approximately 1% at 4 months post-transplant, and after 21 days of GCV, had a clear decrease in the level of GFP+ cells, but not complete ablation, likely due to lower drug-sensitivity of the tk protein expressed by this vector. Cells with a lower level of GFP expression were not eliminated, supporting this hypothesis. Additional animals receiving cells transduced with the sr39 tk retroviral vector and with a lentiviral vector containing a codon-optimized HSVtk are in progress. These data suggest that inclusion of a suicide gene in integrating vectors may be an effective way to address genotoxicity concerns, should clonal outgrowth occur, and increase safety of HSC-targeted gene therapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 166-166
Author(s):  
Evangelia Yannaki ◽  
Thalia Papayannopoulou ◽  
Erica Jonlin ◽  
Fani Zervou ◽  
Garyfalia Karponi ◽  
...  

Abstract Abstract 166FN2 In preparation for a trial for gene therapy of thalassemia, we investigated the safety and efficacy of stem cell mobilization in adult patients with severe thalassemia using either GCSF or Plerixafor. We first assessed mobilization with G-CSF or G-CSF following pretreatment with Hydroxyurea (HU). HU was used in non splenectomized patients in order to reduce extramedullary hemopoiesis in spleen and the splenic size and in splenectomized patients in order to decrease the high number of platelets and mainly WBCs before G-CSF, so as to reduce the chance of complicated leukocytosis during mobilization. We present data on 23 patients from the G-CSF study (EudraCT number 2005-000315-10, NCT00336362) and on 14 patients from the Plerixafor study (EudraCT number 2009–014136-37, NCT01206075). The bulk of the CD34+cell enriched leukaphereses product in either study was cryopreserved for the future gene therapy trial. Mobilization with G-CSF was safe and effective in non-splenectomized patients (CD34+cellsX106/kg/2aphereses: 6.67±2.87, n=6). HU+G-CSF-treated subjects (n=4), mobilized successfully only when an optimal wash out period of approximately two weeks was maintained before G-CSF administration (CD34+cellsX106/kg/2aphereses: 7.34±1.19 vs 1.86±0.76, respectively, p=0.03). HU reduced the spleen size before G-CSF (376±96cm3 vs 556±218cm3) and resulted in less splenic enlargement during mobilization as compared to non-HU pretreated subjects (26.8% vs 60%, p=0.1), although this difference did not reach statistical significance. Splenectomized patients responded excessively to G-CSF by developing early hyperleukocytosis (day 3 mean WBCs: 80.0±7.5×103/μl) without a corresponding rise in blood CD34+cells. This necessitated a significant G-CSF dose reduction or hold resulting in poor yields in the majority of cases (CD34+cells/kg/2aphereses: 2.0±1.7, n=4). One-month HU-pretreatment (n=9), prevented the G-CSF-associated hyperleukocytosis during mobilization (mean WBCs: 60.4±20.8×103/μl) allowing for safe and successful CD34+cell collections (CD34+cells/kg/2aphereses: 6.6±2.3, p=0.02 vs G-CSF-alone), but again, only when an optimal 2-week wash out period was maintained. Despite the safe and effective mobilization with the optimal HU+G-CSF combination of splenectomized patients, HU pretreatment significantly prolonged the mobilization procedure. Plerixafor induced a rapid and effective mobilization in both the splenectomized and non-splenectomized patients (CD34+celsX106/kg/1 or 2 aphereses: 7.29±1.82, n=8 and 5.38±1.99, n=6, respectively) and it was very well tolerated. In SPL subjects, the mean yield per apheresis with plerixafor was higher, with a trend to significance, over its G-CSF counterpart and similar to the mean yield obtained by the optimally HU+G-CSF-treated patients in the G-CSF study (CD34+cells/kgX106/apheresis: 4.19±3.13 vs 1.01±0.85 vs 3.32±1.16, p=0.06 and p=0.6 respectively). One patient who was remobilized with the combination of G-CSF+plerixafor due to previous failure to collect 32×106/kg/2aphereses with G-CSF-alone, yielded 6.5χ10^6 CD34+cells/kg by one apheresis. Importantly also for the non-SPL patients treated with plerixafor, a mean increase of splenic volume of 10.9±14.3% was encountered which was significantly lower than the 60% mean spleen volume increase during G-CSF mobilization (p=0.03). There was no significant difference in the clonogenic capacity (CFU-GM, BFU-E) of CD34+cells mobilized by G-CSF, HU+G-CSF or Plerixafor. However, there was a trend for Plerixafor to mobilize more primitive HSC subpopulations (CD34+/CD38−, CD34+/CD38−/HLADR−) as compared to HU+G-CSF- or G-CSF-alone-mobilized cells. These results suggest that either G-CSF or Plerixafor could be used for mobilizing non-splenectomized patients. Plerixafor seems to represent the agent of choice for mobilization of splenectomized patients with thalassemia. Disclosures: Off Label Use: Plerixafor is used off-label for mobilization of thalassemic subjects.


Sign in / Sign up

Export Citation Format

Share Document