AMD3100 Combined with Standard Doses of G-Csf Leads to Rapid, Consistent Mobilization of Hematopoietic Progenitor Cells in Patients with Non-Hodgkin’s Lymphoma (NHL) and Multiple Myeloma.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2847-2847 ◽  
Author(s):  
Patrick Stiff ◽  
Ivana Micallef ◽  
Philip McCarthy ◽  
Margarida Magalhaes-Silverman ◽  
Neal Flomenberg ◽  
...  

Abstract CXCR4, the chemokine receptor for stromal derived factor-1 (SDF-1) plays an important role in the homing and migration of hematopoietic stem cells. Selective inhibition of the binding of the CXCR4 receptor by the molecule AMD3100 leads to the mobilization of hematopoietic progenitors, including CD34+ cells into the circulation, even in the steady state. Preliminary studies have indicated that AMD3100 given 10–11 hours before each apheresis along with standard doses of G-CSF mobilizes more CD34+ cells per kilogram than does G-CSF alone. A Phase II study of this combination in patients with NHL and myeloma is underway. G-CSF at 10μg/kg/day for a duration of up to 9 days is administered with AMD3100 at 240 μg/kg/day starting on the evening of day 4 of G-CSF therapy, (10–11 hours prior to first and subsequent daily aphereses) until 5 x 106 CD34/kg are collected, or for a maximum of 5 days of a standard 3 blood volume apheresis. Both agents are administered subcutaneously. To date 20 patients (myeloma-6, NHL-14) have been enrolled and analyzed. Of these 20, 14 are considered to have been ‘heavily pre-treated’ using standard definitions (≥ 10 cycles of chemotherapy, platinum based salvage chemotherapy and/or radiation therapy to bone marrow sites). Blood CD34 assays (cells/μl) were performed before and after each AMD 3100 dose and on each apheresis product. After the first dose of AMD3100 there was a 2.6 fold increase in CD34/μl in blood (23 → 60 CD34+ cells/μl). The median number of apheresis performed was 2 (range 1–5); the median total CD34 collected for all 20 patients was 5.7 x 106/kg (range 2.32–14.58 x 106/kg). All patients had collections of > 2.0 x106 CD34/kg, and in 12 of 20 the 5 x 106 CD34/kg cell dose goal was collected, including 8 of 14 in the heavily pre-treated group. In fact in 6, the 5 x 106 CD34/kg cell dose was collected in a single apheresis. The median CD34/kg cell dose collected for the 14 heavily pre-treated patients was 5.7 x 106/kg (2.32–6.48 x 106/kg). There were no serious AE’s related to the use of AMD3100. Transient GI toxicity (mostly diarrhea) occurred in 10 of 20 patients shortly after the injections of AMD3100, but only 3 required therapy. Engraftment data is available for the first 12 patients treated. Time to ANC >500/μl and platelets > 20,000/μl was 9 (range 8–10) and 12 (range 9–19) days respectively. This novel combination of AMD3100 and G-CSF appears to be effective in mobilizing large numbers of CD34+ cells, even in patients considered to be heavily pre-treated, and is an effective alternative to chemotherapy/cytokine mobilization. Compared to studies of G-CSF alone, this combination may reduce the number of apheresis procedures needed to collect an adequate graft for rapid hematopoietic engraftment. The study is ongoing with accrual to date of 40 patients.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2261-2261 ◽  
Author(s):  
Michael P. Rettig ◽  
Sandra Lopez ◽  
Kyle McFarland ◽  
John F. DiPersio

Abstract Abstract 2261 Background: Plerixafor (AMD3100) is a CXCR4 antagonist that was approved by the FDA for use in combination with G-CSF to mobilize hematopoietic stem and progenitor cells (HSPCs) in patients with non-Hodgkin's lymphoma and multiple myeloma. We have previously reported the use of plerixafor alone to mobilize HSPCs for HLA-matched allogeneic transplantation (Blood. 2008;112:990). In this trial, sibling donors were treated with plerixafor at a dose of 0.24 mg/kg by subcutaneous (SC) injection, followed 4 hours later by leukapheresis. To date, 15 of 45 donors (33%) required a second day of leukapheresis to collect the minimum cell dose of ≥ 2.0 × 106 CD34+ cells/kg actual recipient body weight that are required to proceed to transplantation. Based on preliminary data suggesting higher (2-fold) and earlier (1 hr vs. 3 hr) progenitor mobilization in mice after intravenous (IV) dosing of plerixafor, we amended our trial to test the safety and efficacy of IV plerixafor. Methods: In our Phase I trial, 21 healthy donors were initially mobilized with increasing doses of IV plerixafor (0.08, 0.16, 0.24, 0.32, 0.40 or 0.48 mg/kg). After 4 days of drug clearance, the same donors were then mobilized with a single SC dose of 0.24 mg/kg plerixafor followed 4 hours later by leukapheresis. In our Phase II study, 28 sibling donors were treated with plerixafor at a dose of 0.32 mg/kg by IV injection, followed 4 hours later by leukapheresis. Successful mobilization was defined as a minimum leukapheresis yield of ≥ 2.0 × 106/kg CD34+ cell/kg actual recipient body weight. Results: Peak CD34+ cells/mm3 were observed 4 hours after IV dosing (vs. 6–9 hours after SC dosing) and donors given 0.24 mg/kg IV plerixafor, had significantly higher peak levels of CD34+ cells/mm3 compared to the same donors who received 0.24 mg/kg SC plerixafor. There was a clear dose-response relationship of IV plerixafor on mobilization of CD34+ HSPCs, with the 0.32 mg/kg dose yielding a maximum increase in circulating CD34+ cells of 27 CD34+ cells/mm3 at 4 hours after injection, representing a median eight-fold increase from baseline. Interestingly, the median CD34+ cells/mm3 in donors receiving 0.40 mg/kg (N=3) and 0.48 mg/kg (N=3) was 17 and 24 CD34+ cells/mm3, respectively, not significantly better than the 320 mcg/kg dose at the four hour time-point. We also noted that IV dosing (especially doses >0.24 mg/kg) resulted in prolonged mobilization of CD34+ cells such that levels approached 20 CD34+ cells/mm3 at 24 hours after IV dosing. Pharmacokinetic studies demonstrated that the Cmax of plerixafor following the 0.32 mg/kg IV dose remained below 1.0 μg/mL whereas the 0.40 and 0.48 mg/kg doses resulted in Cmax levels of 1.8–2.2 μg/mL. A total of 28 HLA-identical sibling donor/recipient pairs have been enrolled in the Phase II trial, with all donors mobilized using IV plerixafor at a dose 0.32 mg/kg. At 4 hours after IV plerixafor, the CD34+ cell count rose to a median of 16 CD34+ cells/mm3 (range, 4–46), representing a 6.5-fold increase. The efficacy of CD34+ cell collection following 0.32 mg/kg IV plerixafor was not evaluable in five donors due to incomplete apheresis collections or the extremely large size of one recipient (recipient >200 kg). Six of the remaining 23 donors (26%) did not achieve the minimum cell dose of 2 × 106 CD34+ cells/kg in a single 20 L leukapheresis procedure. This mobilization failure rate of 26% with 0.32 mg/kg IV plerixafor is similar to the failure rate of 33% we observed following administration of 0.24 mg/kg SC plerixafor to healthy donors. Four of the six patients who failed to collect ≥ 2.0 × 106 CD34+ cells/kg after the first mobilization and apheresis procedure reached goal following a second mobilization and collection procedure. No adverse events or acute grade 3 or 4 toxicities have been observed in any of the donors given IV plerixafor doses up to 0.48 mg/kg. We have transplanted 24 of the 28 patients entered in the phase II trial. All but one patient engrafted neutrophils and platelets promptly and only 2 of the 24 (8.3%) transplant recipients have developed clinical evidence of acute GVHD (minimum follow-up of 45 days); 1 had grade II and the other grade III. Summary: IV plerixafor increases the magnitude and duration of CD34+ mobilization compared to SC plerixafor in healthy donors. These observations suggest that IV plerixafor may be a more effective mobilization agent with a low side effect profile for allogeneic transplantation. Disclosures: Rettig: Genzyme Corp.: Consultancy, Honoraria. DiPersio: Genzyme: Honoraria.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 884-884 ◽  
Author(s):  
Xinxin Huang ◽  
Bin Guo ◽  
Hal E. Broxmeyer

Abstract Stromal cell derived factor-1α (SDF-1α)/chemokine C-X-C receptor 4 (CXCR4) interactions play a crucial role in hematopoietic stem cells (HSC) trafficking and homing to the bone marrow (BM) environment. To identify new epigenetic regulators of CXCR4 receptor, we screened a chemical compound library of epigenetic enzyme inhibitors to evaluate their effects on membrane CXCR4 expression in CB CD34+ cells. We found that treatment with a couple of histone deacetylase (HDAC) inhibitors, including M344, strongly upregulated membrane CXCR4 expression. We also investigated the effect of M344 on membranal CXCR4 expression in a rigorously defined more primitive HSC cell population (CD34+CD38-CD45RA-CD49f+CD90+) and found that there was a 2.5 fold increase in the M344 treated group compared with vehicle control treated cells. Quantitative RTPCR also showed increased CXCR4 mRNA levels in M344-treated CD34+ cells compared with vehicle control (3.1 fold), indicating that this regulation occurs at the transcriptional level. We next evaluated the effect of M344 treatment on HSC chemotaxis in in vitro transwell migration assays. Both vehicle and M344 treated CB CD34+ cells showed significant migration to 50ng/mL SDF-1α, however, chemotaxis was 2.1 fold higher in M344 treated group. Enhanced migration to SDF-1α by M344 was also observed in the more primitive HSC population. Chemotaxis of CB CD34+ cells to SDF-1α was blocked by CXCR4 antagonist AMD3100, suggesting that the effect was mediated through the CXCR4 receptor. To directly evaluate in vivo homing, vehicle and M344 treated CB CD34+ cells were injected into sublethally irradiated NSG mice, and human cells homing to mouse BM were analyzed 24 hours after transplantation. Consistently, M344 treatment enhanced CB CD34+ cells homing by 2.3 fold in NSG mice. Next, we performed a limiting dilution assay to compare the frequency of SCID-repopulating cells (SRCs) in vehicle and M344 treated CB CD34+ cells. Poisson distribution analysis revealed an SRC frequency of 1/3216 in vehicle control treated group and 1/746 in M344 treatment. We calculated the respective presence of 310.9 SRCs and 1340.5 SRCs in 1×106 cells from vehicle control and M344-treated cultures, so M344 treatment resulted in a 4.3 fold increase in the number of functionally detectable SRCs compared with vehicle control. Eighteen HDACs have been identified in humans and they are divided into four classes. The mechanisms regarding HDAC regulation of HSC homing and engraftment are largely unknown. Using shRNA to knockdown expression of individual HDACs in CB CD34+ cells, we surprisingly found that HDAC5 shRNA transfection resulted in upregulation of membrane CXCR4 expression. LMK235, a selective inhibitor of HDAC5, also increased membrane CXCR4 expression in CB CD34+ cells. In contrast, inhibition of other HDACs did not show any effect on membrane CXCR4 expression. Similar to M344, LMK235 treatment resulted in significantly higher CXCR4 mRNA, membrane CXCR4 expression in CB HSCs, enhanced migration to SDF-1α in chemotaxis assay, and higher number of cells homed to the BM in NSG mice. We next performed chromatin immunoprecipitation (ChIP) assays to examine the chromatin status at the CXCR4 promoter region. H3K9 acetylation levels were significant higher in LMK235-treated CB CD34+ cells compared with vehicle control, suggesting increased H3K9 levels at the CXCR4 promoter region contributes to increased CXCR4 transcription. To further explore the mechanisms underlying HDAC5 regulation of HSC homing, we tested a couple of signaling pathways and found that inhibition of the NF-κB signaling pathway by Andrographolide and Pyrrolidinedithiocarbamate Ammonium, suppressed M344 and LMK235-mediated CXCR4 upregulation on CB CD34+ cells. It has been reported that acetylation of NF-κB p65 subunit enhances its transcriptional activity. We examined the acetylation levels of p65 and found that LMK235 treatment resulted in increased levels of p65 acetylation in CB CD34+ cells, indicating p65 could be a downstream target of HDAC5. Consistently, using ChIP assay we detected increased levels of acetylated p65 binding to the CXCR4 promoter region in the LMK235-treated group. Taken together, our results reveal previously unknown specific epigenetic regulation of HSC homing and engraftment by HDAC5, which suggests a new translational strategy to enhance HSC transplantation. Disclosures Broxmeyer: CordUse: Other: SAB Member.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2889-2889 ◽  
Author(s):  
Neeta Shirvaikar ◽  
Jencet Montano ◽  
Andrew R. Turner ◽  
Mariusz Z. Ratajczak ◽  
Anna Janowska-Wieczorek

Abstract Hyaluronic acid (HA), the bone marrow (BM) extracellular matrix microenvironment (ECM) component, not only supports cell adhesion but also promotes migration and homing of hematopoietic stem/progenitor cells (HSPC) by interacting with its cell surface receptor CD44. CD44 has been shown to co-localize with matrix metalloproteinases (MMPs), particularly membrane-type (MT)-1 MMP and MMP-9, at the leading edge of migrating tumor cells, and the cleavage of CD44 by MT1-MMP is critical for tumor cell migration and invasion. MT1-MMP has strong pericellular proteolytic activity and also activates latent forms of MMP-2 and MMP-9. In this study we examined the effect of HA on MT1-MMP expression and migration of BM, peripheral blood and cord blood CD34+ cells. We found that HA upregulates mRNA for MT1-MMP and MMP-9, increases MT1-MMP protein (as evaluated by Western blotting) and stimulates MMP-9 and MMP-2 activity (as determined by zymography) in CD34+ cells. In chemotaxis assays HA alone did not show any chemotactic activity but primed the chemotaxis of CD34+ cells to a low SDF-1 gradient (10 ng/mL) and their trans-Matrigel chemoinvasion to a low SDF-1 gradient. Similarly, SDF-1 besides stimulating MMP-2 and MMP-9 (as we previously described in Exp Hematol2000; 28:1274), also upregulated MT1-MMP in CD34+ cells. In addition, we found that the HA enhanced activation of latent MMP-2 in co-cultures of CD34+ cells with HUVEC. In conclusion, we demonstrate for the first time that (i) HA primes CD34+ cell chemotaxis and chemoinvasion to a low SDF-1 gradient; (ii) that both HA and SDF-1 stimulate MT1-MMP, MMP-2 and MMP-9 in CD34+ cells, and (iii) HA enhances activation of proMMP-2 in the ECM. Hence the interaction of HA and SDF-1 with MT1-MMP could play an important role in HSPC migration and homing and we postulate that pretreatment of HSPC with HA before transplantation could enhance their homing by inducing expression of MMPs that is optimal for stem cell engraftment.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1962-1962 ◽  
Author(s):  
David A. Hess ◽  
Louisa Wirthlin ◽  
Timothy P. Craft ◽  
Jesper Bonde ◽  
Ryan W. Lahey ◽  
...  

Abstract Interactions between stromal derived factor-1 (SDF-1 or CXCL12), and its receptor CXCR4 regulate hematopoietic stem and progenitor cell retention in the bone marrow. AMD3100, a bicyclam molecule that selectively blocks the interaction between CXCL12 and CXCR4, has recently been used in clinical trials to rapidly mobilize hematopoietic progenitor cells. However, the functional properties of human stem and progenitor cells mobilized with this agent are not well characterized. Here, we directly compared the NOD/SCID repopulating function of CD34+ cells rapidly mobilized (4 hours) by AMD3100 versus CD34+ cells mobilized after 5 days of G-CSF treatment. A total of 7 HLA-matched sibling donors were leukapheresed after a single injection of 240ug/kg AMD3100. After 1 week of drug clearance, the same donor was mobilized with G-CSF, allowing a paired comparison of the repopulating function of cells mobilized by the two agents. Total CD34+ cells mobilized by AMD3100 treatment averaged 1.2±0.4x106 CD34+ cells/kg (range 0.4–2.1x106 CD34+ cells/kg), as compared to G-CSF treatment at 3.2±0.9x106 CD34+ cells/kg (range 1.7–5.7 x106 CD34+ cells/kg). Leukapheresis total mononuclear cell (MNC) fraction or purified CD34+ cells (>90% purity), were isolated and transplanted into sublethally irradiated NOD/SCID mice at varying doses. BM, spleen, and peripheral blood of mice were harvested 7–8 weeks post-transplantation and analyzed by flow cytometry for the presence or absence of engrafting human cells. Low frequency human engraftment events (<0.2% human cells) were confirmed by PCR for P17H8 alpha-satellite human DNA sequences. Injection of 1–40x106 MNC or 0.5–5x105 CD34+ cells produced consistent human engraftment and allowed limiting dilution analysis using Poisson statistics to be performed on paired samples of AMD3100 and G-CSF leukapheresis products from 3 individual patients. The calculated frequencies of NOD/SCID repopulating cells (SRC) were 1 SRC in 11.5x106 AMD3100-mobilized MNC (n=50) compared to 1 SRC in 44.8x106 G-CSF-mobilized MNC (n=55). For purified CD34+ populations, the overall frequency of repopulating cells was 1 SRC in 1.0x105 AMD3100-mobilized CDC34+ cells (n=53) compared to 1 SRC in 3.1x105 G-CSF-mobilized CD34+ cells (n=45). These data correspond to a 3–4-fold increase in overall repopulating function demonstrated by AMD3100 mobilized cells. Multilineage hematopoietic differentiation of transplanted CD34+ cells was similar for AMD3100 and G-CSF-mobilized CD34+ cells, with equivalent production of myelo-monocytic cells (CD33+CD14+), immature B-lymphoid cells (CD19+CD20+), and primitive repopulating (CD34+CD133+CD38−) cells 7–8 weeks post-transplantation. These studies indicate that human AMD3100-mobilized MNC and purified CD34+ cells possess enhanced repopulating capacity, as compared to G-CSF mobilized counterparts from the same donor. Thus, AMD3100 mobilized peripheral blood represents a rapidly obtained and highly functional source of repopulating hematopoietic stem cells for clinical transplantation procedures.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1817-1817
Author(s):  
Yves Chalandon ◽  
Christoph Schmid ◽  
Kimmo Porkka ◽  
Alvaro Urbano-Ispizua ◽  
Bernd Hertenstein ◽  
...  

Abstract Using data submitted to the EBMT registry, we analyzed outcome on 344 patients (pts) who had received donor lymphocyte infusions (DLI) for relapse after allogeneic hematopoietic stem cell transplantation (HSCT) for chronic myeloid leukemia (CML) in 31 centers. 113/344 pts (33%) developed acute graft-versus-host disease (aGVHD) a median of 50 days post DLI (max grade: I=42, II=30, III=31, IV=6)(60% grade II–IV). Organs involved (%): skin (88), liver (42), gut (30). Median age was 38 (4–59), 58% pts were male, 62 transplants were HLA-identical sibling and 51 unrelated. 74 were T-cell depleted, 92 transplanted in CP1, 21 beyond CP1. Relapse was molecular in 19 pts, cytogenetic in 31, hematological in 49, accelerated or blastic in 12. Median initial cell dose was 107CD3+ cells/kg (0.01–32), median number of DLI was 1 (1–10). aGvHD was treated with prednisone in 92% of pts, CSA in 52 %, ATG and monoclonal antibodies in 2% and other in 19%. aGVHD resolved in 53% of the pts within a median of 63 d (7–546). 82/344 pts (24%) had chronic GVHD (cGVHD)(30 limited, 50 extensive, 2 not specified), of those 46 (56%) following aGVHD post DLI. Organs involved (%): skin (75), liver (35), lungs (13), mouth (43), eyes (22) and gut (5). Median age was 35 (6–58), 51% were male, stem cell source was PB in 15% and marrow in 85%, 43 underwent HLA-identical sibling HSCT and 39 unrelated donor HSCT. Forty-three were T-cell depleted, 66 transplanted in CP1, 16 beyond CP1. Relapse was molecular in 21 pts, cytogenetic in 29, hematological in 22, accelerated or blastic in 7. Median initial cell dose was 107 CD3+ cells/kg (0.05–40), median number of DLI was 1 (1–7). 61 pts are alive with a median follow-up of 50 mth. Treatment was with steroids in 83% of pts, CSA in 58 %, MMF in 20%, thalidomide in 15%, photopheresis in 15%, PUVA in 10% and other in 17%. cGVHD resolved in 39% of the pts within a median of 354 d (44–1588). The estimated 5-y OS post-DLI was significantly lower in pts who developed aGVHD post-DLI, 61 ± 10% vs 74 ± 7% in the one that did not, p=0.007 and also a tendency to have a lower 5-y EFS, 58 ± 10% vs 65 ± 7%, p=0.19. Median duration of response to DLI in aGVHD pts was 4 y. aGVHD post-DLI did not influence the relapse rate (5 ± 5% vs 6 ± 5% in the absence of aGVHD). 5-y DLI related mortality was significantly higher in aGVHD pts, 31 ± 8% vs 4 ± 4%, p<0.00001. On the other hand, pts that developed cGVHD post-DLI had a tendency to have a better 5-y OS and EFS, 74 ± 11% and 71 ± 11% respectively vs 69 ± 6% and 62 ± 7% in those that did not, p=0.32 and 0.09. This was related to a tendency to lower incidence of relapse, 2 ± 3% in pts with cGVHD vs 9 ± 6% without, p=0.2. DLI related mortality was not different, 11 ± 8% vs 10 ± 5%, p=0.77. aGVHD post-DLI for CML relapse is mainly of advanced stage and negatively influence OS and EFS with a higher DLI related mortality. cGVHD post-DLI is mainly extensive, but pts with cGHVD tend to have better outcome with better 5-y OS, EFS and less relapse than those without, although this was not statistically significant.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4208-4208
Author(s):  
Hiroto Araki ◽  
Nadim Mahmud ◽  
Mohammed Milhem ◽  
Mingjiang Xu ◽  
Ronald Hoffman

Abstract The fixed number of hematopoietic stem cells (HSCs) within a single cord blood (CB) unit has limited the use of CB grafts for allogeneic transplantation in adults. Efforts to promote self-renewal and expansion of HSCs have been met with limited success. Using presently available ex-vivo culture techniques HSCs lose their functional properties in proportion to the number of cellular divisions they have undergone. We hypothesized that chromatin modifying agents, 5-aza-2′-deoxycytidine (5azaD) and histone deacetylase inhibitor, trichostatin A (TSA) could reactivate pivotal genes required for retaining the functional properties of dividing HSC. We have demonstrated previously that the fate of human bone marrow CD34+ cells could be altered by the addition of 5azaD/TSA (Milhem et al. Blood.2004;103:4102). In our current studies we hypothesized that in vitro exposure of CB CD34+ cells to chromatin modifying agents might lead to optimal HSC expansion to permit transplantation of adults. A 12.5-fold expansion was observed in the 5azaD/TSA treated CD34+CD90+ cell cultures containing SCF, thrombopoietin and FLT3 ligand (cytokines) in comparison to the input cell number. Despite 9 days of culture, 35.4% ± 5.8% (n = 10) of the total cells in the cultures exposed to chromatin modifying agents were CD34+CD90+ as compared to 1.40 % ± 0.32% in the culture containing cytokines alone. The 12.5-fold expansion of CD34+CD90+ cells was associated with a 9.8-fold increase in the numbers of CFU-mix and 11.5-fold expansion of cobblestone area-forming cells (CAFC). The frequency of SCID repopulating cells (SRC) was 1 in 26,537 in primary CB CD34+CD90+ cells but was increased to 1 in 2,745 CD34+CD90+ cells following 9 days of culture in the presence of 5azaD/TSA resulting in a 9.6-fold expansion of the absolute number of SRC. In contrast, the cultures lacking 5azaD/TSA had a net loss of both CFC/CAFC as well as SRC. The expansion of cells maintaining CD34+CD90+ phenotype was not due to the retention of a quiescent population of cells since all of the CD34+CD90+ cells in the culture had undergone cellular division as demonstrated by labeling with a cytoplasmic dye. CD34+CD90+ cells that had undergone 5–10 cellular divisions in the presence of 5azaD/TSA but not in the absence still retained the ability to repopulate NOD/SCID mice. 5azaD/TSA treated CD34+CD90+ cells, but not CD34+CD90- cells were responsible for in vivo hematopoietic repopulation of NOD/SCID assay, suggesting a strong association between CD34+CD90+ phenotype and their ability to repopulate NOD/SCID mice. We next assessed the effect of 5azaD/TSA treatment on the expression of HOXB4, a transcription factor which has been implicated in HSC self-renewal. A significantly higher level of HOXB4 protein was detected by western blot analysis after 9 days of culture in the cells treated with 5azaD/TSA as compared to cells exposed to cytokines alone. The almost 10-fold increase in SRC achieved using the chromatin modifying agents should be sufficient to increase the numbers of engraftable HSC within a single human CB unit so as to permit these expanded grafts to be routinely used for transplanting adult recipients.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4040-4040
Author(s):  
Szabolcs Fatrai ◽  
Simon M.G.J. Daenen ◽  
Edo Vellenga ◽  
Jan J. Schuringa

Abstract Mucin1 (Muc1) is a membrane glycoprotein which is expressed on most of the normal secretory epithelial cells as well as on hematopoietic cells. It is involved in migration, adhesion and intracellular signalling. Muc1 can be cleaved close to the membrane-proximal region, resulting in an intracellular Muc1 that can associate with or activate various signalling pathway components such as b-catenin, p53 and HIF1a. Based on these properties, Muc1 expression was analysed in human hematopoietic stem/progenitor cells. Muc1 mRNA expression was highest in the immature CD34+/CD38− cells and was reduced upon maturation towards the progenitor stage. Cord blood (CB) CD34+ cells were sorted into Muc1+ and Muc1− populations followed by CFC and LTC-IC assays and these experiments revealed that the stem and progenitor cells reside predominantly in the CD34+/Muc1+ fraction. Importantly, we observed strongly increased Muc1 expression in the CD34+ subfraction of AML mononuclear cells. These results tempted us to further study the role of Muc1 overexpression in human CD34+ stem/progenitor cells. Full-length Muc1 (Muc1F) and a Muc1 isoform with a deleted extracellular domain (DTR) were stably expressed in CB CD34+ cells using a retroviral approach. Upon coculture with MS5 bone marrow stromal cells, a two-fold increase in expansion of suspension cells was observed in both Muc1F and DTR cultures. In line with these results, we observed an increase in progenitor counts in the Muc1F and DTR group as determined by CFC assays in methylcellulose. Upon replating of CFC cultures, Muc1F and DTR were giving rise to secondary colonies in contrast to empty vector control groups, indicating that self-renewal was imposed on progenitors by expression of Muc1. A 3-fold and 2-fold increase in stem cell frequencies was observed in the DTR and Muc1F groups, respectively, as determined by LTC-IC assays. To determine whether the above mentioned phenotypes in MS5 co-cultures were stroma-dependent, we expanded Muc1F and DTR-transduced cells in cytokine-driven liquid cultures. However, no proliferative advantage or increase in CFC frequencies was observed suggesting that Muc1 requires bone marrow stromal cells. In conclusion, our data indicate that HSCs as well as AML cells are enriched for Muc1 expression, and that overexpression of Muc1 in CB cells is sufficient to increase both progenitor and stem cell frequencies.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2393-2393
Author(s):  
Stefanie Geyh ◽  
Ron Patrick Cadeddu ◽  
Julia Fröbel ◽  
Ingmar Bruns ◽  
Fabian Zohren ◽  
...  

Abstract Abstract 2393 Background: Myelodysplastic syndromes (MDS) represent a heterogeneous group of hematopoietic stem cell disorders and research in this field has mainly focused on hematopoietic stem and progenitor cells (HSPC). Still, recent data from mouse models indicate that the bone marrow (BM) microenvironment might be involved in the pathogenesis MDS (Raaijmakers et al., 2010). The role of mesenchymal stromal cells (MSC) in particular as a key component of the BM microenvironment remains elusive in human MDS and data so far are controversial. Design/Methods: We therefore investigated MSC and immunomagnetically enriched CD34+ HSPC from BM of 42 untreated patients (pts) with MDS (12 RCMD, 12 RAEB, 12 sAML, 3 del5q, 1 CMML-1, 1 MDS hypocellular, 1 MDS unclassifiable according to WHO) and age-matched healthy controls (HC, n=13). MSC were examined with regard to growth kinetics, morphology and differential potential after isolation and expansion according standard procedures in line with the international consensus criteria (Dominici et al., 2006). Furthermore corresponding receptor-ligand pairs on MSC and CD34+ cells (Kitlg/c-kit; CXCL12/CXCR4; Jagged1/Notch1; Angpt1-1/Tie-2; ICAM1/LFA-1) were investigated by RT-PCR. Results: In MDS, the colony forming activity (CFU-F) of MSC was significantly reduced in comparison to HC (median number of colonies per 1×107MNC in MDS: 8, range 2–74 vs. HC: 175, 10–646, p=0.003) and this was also true when looking at the different subtypes (RCMD median: 16, p=0.04; RAEB median: 8, p=0.31; sAML median: 26, p=0.02). According to this, MSC from pts with RCMD and del5q could only be maintained in culture for a lower number of passages (median number of passages: MDS 3 passages, range 1–15; HD 14 passages, range 8–15, p=0.01), had a lower number of cumulative population doublings (CPD) and needed a longer timer to reach equivalent CPD (MDS median: 18,16 CPD, HD median: 33,68 CPD, p=0,0059). All types of MDS-MSC showed an abnormal morphology, while an impaired osteogenic differentiation potential was exclusively observed in pts with RCMD. These findings of an altered morphology together with a diminished growth and differentiation potential prompted us to test, whether the interaction between MSC and CD34+ HSPC in BM of pts with MDS was also disturbed. On the MDS-MSC, we found a significant lower expression of Angpt1 in pts with RAEB (3.5-fold, p=0.01) and del5q (4.9-fold, p=0.009) compared to HD. The expression of CXCL12 (2.5-fold, p=0.057) and jagged1 was reduced in trend in MSC from pts with MDS, while no differences were observed with regard to the expression of kitlg and ICAM1. When looking on CD34+ cells, we found a significantly reduced expression of CXCR4 (RCMD 2.5-fold, p=0.02; RAEB 2.46-fold, p=0.02), notch1 (RCMD 6-fold, p=0.04) and Tie-2 (RAEB 5.91-fold, p=0.02) in pts with MDS, while LFA-1 was overexpressed in pts with RAEB (2.6-fold, p=0.036). Conclusion: Taken together, our data indicate that MSC from pts with MDS are structurally altered and that the crosstalk between CD34+ HSPC and MSC in the BM microenvironment of pts with MDS might be deregulated as a result of an abnormal expression of relevant receptor-ligand pairs. Ongoing research is required to corroborate these findings and to definitely address their functional relevance for the pathogenesis of MDS. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5542-5542 ◽  
Author(s):  
Shane A Gangatharan ◽  
John Kuruvilla ◽  
Vishal Kukreti ◽  
Armand Keating ◽  
Manjula Maganti ◽  
...  

Abstract Introduction Autologous stem cell transplantation (ASCT) is the standard of care for relapsed aggressive lymphomas. Time to neutrophil and platelet engraftment is strongly correlated with CD34+ cell number infused but data are conflicting as to whether patients who receive greater numbers of CD34+ stem cells have improved outcomes. We sought to determine whether short term engraftment predicts progression-free survival (PFS) independent of other disease-specific prognostic factors. Methods From the Princess Margaret Cancer Centre transplant database, we identified patients undergoing ASCT for relapsed aggressive lymphoma between 2007-2011. Data were extracted on prognostic features at relapse/progression, stem cell collection, engraftment, and time to progression and death. All patients received platinum-based salvage chemotherapy and those with chemosensitivity were mobilised with cyclophosphamide, etoposide and filgrastim (minimum threshold 2x106 CD34 cells/kg) and proceeded to ASCT. Patients who failed initial mobilisation were remobilised using plerixafor. High-dose therapy consisted of etoposide 60mg/kg Day -4 and melphalan 180mg/m2 Day -3 with stem cells infused on Day 0. Filgrastim 300µg daily was started from Day +7 until neutrophil recovery to >1.0 x106/uL. Platelet engraftment was defined as an unsupported platelet count >20 x 109/L. Results 97 patients with DLBCL (n=66), transformed (n=24) and T-cell lymphoma (n=7) were reviewed. Median age was 54 (range 20-67), 61% were male and median IPI score on relapse was 2. Fifty one percent relapsed within 12 months of last therapy, and of the patients with B-cell lymphoma, 81% received rituximab prior to salvage chemotherapy. Median stem cell dose was 5.7x106 CD34+ cells/kg (range 1.69-17.82) with a median number of apheresis sessions to achieve this of 2 (range 1-4). Median time to neutrophils >0.5x106/uL was 11 days (range 9-14) and platelets >20x106/uL was 14 days (range 10-23). The Spearman correlation test confirmed a higher stem cell dose was significantly associated with shorter time to neutrophil (p=0.0014) and platelet engraftment (p=0.0003). From date of ASCT, median follow-up was 3.1 years in progression-free patients. For the entire cohort, PFS was 50% and overall survival (OS) was 74% at 3 years. On univariable analysis, patients with B-cell lymphoma with IPI score of 0-2 had a 3-year PFS of 59%, v 28% for those with IPI of 3-4 (p=0.03) (n=90). Patients with early relapse within 12 months of last therapy had inferior 3-year PFS, 42% v 59% for those with initial PFS > 1 year (p=0.08). Patients with B-cell lymphoma who received rituximab with primary chemotherapy had worse 3-year PFS: 47% v 69% (p=0.11). There were no associations of PFS with lymphoma subtype, dose of stem cells infused, number of apheresis sessions and neutrophil or platelet engraftment. Similarly, on univariable analysis of OS for the entire cohort, only secondary IPI (0-2 v 3-4) was significant, with 3-year OS 82% v 48%, respectively (p=0.01). Multivariable Cox regression analysis of outcomes for patients with B-cell lymphoma in a model including IPI score, time to relapse, prior rituximab, CD34+ cell dose and neutrophil and platelet engraftment times confirmed IPI was the only significant variable predicting PFS (HR 1.99, p=0.03) and OS (HR 3.2, p=0.006). Conclusions In this cohort of patients with aggressive lymphomas, CD34+ cell dose was correlated with time to neutrophil and platelet engraftment but was not predictive of PFS or OS. Secondary IPI score, relapse within 12 months, and for B-cell lymphomas previous use of rituximab were predictive of outcomes post ASCT. Disclosures: Kuruvilla: Roche: Honoraria. Kukreti:Millennium Pharmaceuticals: Research Funding; Onyx: Research Funding. Keating:Roche: Honoraria. Crump:Roche: Honoraria; Jansen-Ortho: Honoraria; Celgene: Honoraria; Lundbeck: Honoraria; Novartis: Research Funding; Seattle Genetics: Honoraria.


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.


Sign in / Sign up

Export Citation Format

Share Document