Conditioning with fludarabine and targeted busulfan for transplantation of allogeneic hematopoietic stem cells

Blood ◽  
2003 ◽  
Vol 102 (3) ◽  
pp. 820-826 ◽  
Author(s):  
Martin Bornhäuser ◽  
Barry Storer ◽  
John T. Slattery ◽  
Frederick R. Appelbaum ◽  
H. Joachim Deeg ◽  
...  

Abstract A regimen of busulfan and cyclophosphamide is standard therapy before transplantation of allogeneic hematopoietic stem cells in patients with chronic myelogenous leukemia (CML) or myelodysplastic syndrome (MDS). The clinical trial reported here was undertaken to test the hypothesis that fludarabine can replace cyclophosphamide in this regimen and facilitate donor engraftment with reduced toxicity. The conditioning regimen consisted of 30 mg/m2 intravenous fludarabine daily from day -9 to day -6, and oral busulfan given at 1 mg/kg 4 times a day every 6 hours from day -5 to day -2, with doses adjusted to target plasma levels of 900 ± 100 ng/mL at steady state. Cyclosporine and methotrexate were used for prophylaxis for graft-versus-host disease. Enrolled were 42 patients with high-risk CML (n = 4) or MDS (n = 38). The median patient age was 52 years (range, 12-65 years). Mobilized blood stem cells were obtained from HLA-compatible siblings (n = 16) or unrelated donors (n = 26). Engraftment was achieved in all patients, and the day-100 regimen-related mortality was 7%. With a median follow-up of 18 months (range, 13-27 months), the probabilities of overall survival, disease-free survival, and nonrelapse mortality were 42.4%, 34.9%, and 24%, respectively. These data indicate that the combination of fludarabine and targeted busulfan is sufficiently immunosuppressive to facilitate engraftment of blood stem cells from HLA-matched siblings and unrelated donors. Based on these encouraging results, further studies of fludarabine and targeted busulfan are warranted in standard-risk patients.

Blood ◽  
2011 ◽  
Vol 118 (13) ◽  
pp. 3715-3720 ◽  
Author(s):  
Jennifer Cuellar-Rodriguez ◽  
Juan Gea-Banacloche ◽  
Alexandra F. Freeman ◽  
Amy P. Hsu ◽  
Christa S. Zerbe ◽  
...  

Abstract We performed nonmyeloablative HSCT in 6 patients with a newly described genetic immunodeficiency syndrome caused by mutations in GATA2—a disease characterized by nontuberculous mycobacterial infection, monocytopenia, B- and NK-cell deficiency, and the propensity to transform to myelodysplastic syndrome/acute myelogenous leukemia. Two patients received peripheral blood stem cells (PBSCs) from matched-related donors, 2 received PBSCs from matched-unrelated donors, and 2 received stem cells from umbilical cord blood (UCB) donors. Recipients of matched-related and -unrelated donors received fludarabine and 200 cGy of total body irradiation (TBI); UCB recipients received cyclophosphamide in addition to fludarabine and TBI as conditioning. All patients received tacrolimus and sirolimus posttransplantation. Five patients were alive at a median follow-up of 17.4 months (range, 10-25). All patients achieved high levels of donor engraftment in the hematopoietic compartments that were deficient pretransplantation. Adverse events consisted of delayed engraftment in the recipient of a single UCB, GVHD in 4 patients, and immune-mediated pancytopenia and nephrotic syndrome in the recipient of a double UCB transplantation. Nonmyeloablative HSCT in GATA2 deficiency results in reconstitution of the severely deficient monocyte, B-cell, and NK-cell populations and reversal of the clinical phenotype. Registered at www.clinicaltrials.gov as NCT00923364.


2014 ◽  
Vol 6 (3) ◽  
pp. 115
Author(s):  
Anna Meiliana ◽  
Andi Wijaya

BACKGROUND: Since the first umbilical cord blood (UCB) transplant, performed 25 years ago, UCB banks have been established worldwide for the collection and cryopreservation of UCB for autologous and allogeneic transplants.CONTENT: Much has been learned in a relatively short time on the properties of UCB hematopoietic progenitors and their clinical application. More interestingly, non-hematopoietic stem cells have been isolated from UCB. These cells can be grown and differentiated into various tissues including bone, cartilage, liver, pancreas, nerve, muscle and so on. The non-hematopoietic stem cells have an advantage over other sources of stem cells, such as embryonic stem cells or induced pluripotent stem cells, because their supply is unlimited, they can be used in autologous or allogeneic situations, they need minimal manipulation and they raise no ethical concerns. Future studies will test the potential of UCB cells for the treatment of several diseases including, among other possibilities, diabetes, arthritis, burns, neurological disorder and myocardial infarction.SUMMARY: In addition to hematopoietic stem cells, UCB contain a large number of non-hematopoietic stem cells. In the absence of ethical concern, the unlimited supply of UCB cells explains the increasing interest of using UCB for developing regenerative medicine.KEYWORDS: UCB, transplantation, UCB bank, HSC, MSC, CD34, CD133, VSEL


1999 ◽  
Vol 189 (4) ◽  
pp. 693-700 ◽  
Author(s):  
Taila Mattern ◽  
Gundolf Girroleit ◽  
Hans-Dieter Flad ◽  
Ernst T. Rietschel ◽  
Artur J. Ulmer

CD34+ hematopoietic stem cells, which circulate in peripheral blood with very low frequency, exert essential accessory function during lipopolysaccharide (LPS)-induced human T lymphocyte activation, resulting in interferon γ production and proliferation. In contrast, stimulation of T cells by “conventional” recall antigens is not controlled by blood stem cells. These conclusions are based on the observation that depletion of CD34+ blood stem cells results in a loss of LPS-induced T cell stimulation as well as reduced expression of CD80 antigen on monocytes. The addition of CD34-enriched blood stem cells resulted in a recovery of reactivity of T cells and monocytes to LPS. Blood stem cells could be replaced by the hematopoietic stem cell line KG-1a. These findings may be of relevance for high risk patients treated with stem cells or stem cell recruiting compounds and for patients suffering from endotoxin-mediated diseases.


Blood ◽  
2000 ◽  
Vol 95 (12) ◽  
pp. 3945-3950 ◽  
Author(s):  
Yunfang Jiang ◽  
Hong Liang ◽  
Wei Guo ◽  
Lazar V. Kottickal ◽  
Lalitha Nagarajan

Abstract SMADs are evolutionarily conserved transducers of the differentiation and growth arrest signals from the transforming growth factor/BMP (TGF/BMP) family of ligands. Upon receptor activation, the ligand-restricted SMADs1–35 are phosphorylated in the C-terminal MH2 domain and recruit the common subunit SMAD4/DPC-4 gene to the nucleus to mediate target gene expression. Frequent inactivating mutations of SMAD4, or less common somatic mutations ofSMAD2 seen in solid tumors, suggest that these genes have a suppressor function. However, there have been no identified mutations of SMAD5, although the gene localizes to the critical region of loss in chromosome 5q31.1 (chromosome 5, long arm, region 3, band 1, subband 1) in myelodysplasia (MDS) and acute myelogenous leukemia (AML). A ubiquitously expressed novel isoform,SMAD5β, encodes a 351 amino acid protein with a truncated MH2 domain and a unique C-terminal tail of 18 amino acids, which may be the functional equivalent of inactivating mutations. The levels of SMAD5β transcripts are higher in the undifferentiated CD34+ hematopoietic stem cells than in the terminally differentiated peripheral blood leukocytes, thereby implicating the β form in stem cell homeostasis. Yeast 2-hybrid interaction assays reveal the lack of physical interactions between SMAD5β and SMAD5 or SMAD4. The expression ofSMAD5β may represent a novel mechanism to protect pluripotent stem cells and malignant cells from the growth inhibitory and differentiation signals of BMPs.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2204-2204
Author(s):  
Mary-Elizabeth A. Muchmore ◽  
Matthew J. Burge ◽  
Judith A. Shizuru

Abstract Transplantation of purified allogeneic hematopoietic stem cells (HSC) has the potential to be a curative treatment for autoimmune diseases. Before it becomes a viable therapy, however, the treatment-related mortality and difficulty of achieving engraftment must be addressed. Our research has focused on developing non-myeloablative regimens that lead to donor-derived engraftment of purified HSC in a murine model. Total lymphoid irradiation (TLI) consists of low-dose fractionated irradiation targeted to the thymus, abdomen, and peripheral nodes, while the skull, lungs, and long bones remain shielded. The non-myeloablative conditioning regimen of TLI and anti-thymocyte globulin (ATG) was followed by HSC transplantation. HSCs were isolated by the composite phenotype of Thy1.1+, c-kit+, Sca-1+, and lineage- (KTLS) or, in strains lacking the Thy1.1 marker, c-kit+, Sca-1+, and lineage- (KSL). We tested HSC transplantations across three major histocompatiblity complex (MHC)-matched strain combinations known through previous studies in our group to have significantly different barriers to engraftment. In all three strain combinations we observed stable mixed chimerism (approximately 50% donor-derived cells) when high doses of HSC (10,000/mouse) were administered. Chimerism was measured at thirty-day intervals, and initially sharply increased and then stabilized around day ninety post-transplantation. In prior studies from our laboratory in a spontaneously arising autoimmune diabetes model, we demonstrated that mixed allogeneic chimerism alone following low dose total body irradiation (TBI) and HSC transplantation was sufficient to block the autoimmune pathogenesis. In order to establish a second clinically relevant conditioning regimen, we attempted here to lower the dose of TBI by using cyclophosphamide and ATG in addition to low dose TBI. However, less robust engraftment was observed as compared to the TLI/ATG approach. To study how TLI/ATG allows engraftment, we have examined the marrow of TLI/ATG conditioned, untransplanted animals. Though TUNEL and Caspase-3 assays did not show a significant increase in apoptosis compared to controls, a 71% decrease in the quantitative number of HSCs isolated from these animals was observed. This depletion of HSCs in the marrow could provide a niche for donor HSCs to inhabit. Further histologic studies on lymphoid organs exposed to radiation through TLI, including the thymus and spleen, are ongoing and may further elucidate the mechanisms by which TLI reconditions the host immune system. The durable mixed chimerism observed following TLI/ATG conditioning and HCT will be applied to mice affected with the rodent form of multiple sclerosis (experimental autoimmune encephalomyelitis) and to tolerance induction of solid-organ grafts. SUMMARY: The combination of TLI/ATG non-myeloablative conditioning and transplantation of allogeneic HSC leads to a durable mixed chimeric state between donor and host and will next be applied to the induction of tolerance to autoantigens and alloantigens.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. LB2-LB2
Author(s):  
Agnieszka Czechowicz ◽  
Daniel L. Kraft ◽  
Deepta Bhattacharya ◽  
Irving L. Weissman

Abstract Hematopoietic stem cells (HSCs) are used therapeutically in bone marrow/hematopoietic stem cell transplantation (BMT/HSCT) to correct hematolymphoid abnormalities. Upon intravenous transplantation, HSCs can home to specialized bone marrow niches, self-renew and differentiate and thus generate a new, complete hematolymphoid system. Unfortunately BMT has had limited applications, due to the risks associated with the toxic conditioning regimens, such as irradiation and chemotherapy, that are deemed necessary for HSC engraftment. Elimination of these toxic conditioning regimens could expand the potential applications of BMT to include many non-malignant hematologic disorders, a wide variety of autoimmune disorders such as diabetes and multiple sclerosis, as well as in the facilitation of organ transplantation. The exact function of these traditional myeloablative conditioning regimens is not clear. To elucidate the barriers of HSC engraftment, we transplanted 50–1000 purified HSCs (Ckit+Lin−Sca1+CD34+CD150−) into immunodeficient, Rag2−/− or Rag2−/−gc−/− recipient mice and show that HSC engraftment levels rarely exceed 0.5% following transplantation without toxic conditioning, indicating that the immune system is not the only barrier to engraftment. Additionally, we did not observe a significant increase in HSC engraftment when HSC doses of >250 cells were transplanted. Even when up to 18000 HSC were transplanted, we did not see a linear increase in HSC engraftment, indicating that the increased doses of HSCs transplant inefficiently. We believe this is due to the naturally low frequency of available HSC niches, which we postulate may result from the physiologic migration of HSCs into circulation. Conversely, separation of the graft into small fractions and the subsequent time-delayed transplantation of these doses did result in increased engraftment due to the natural physiologic creation of new available HSC niches. When 1800 HSC were transplanted daily for seven days, the engraftment was 6.1-fold higher than transplantation of 12800 HSC in a single bolus. Here, we provide evidence that, aside from immune barriers, donor HSC engraftment is restricted by occupancy of appropriate niches by host HSCs. Through elimination of host HSCs we are able to increase available HSC niches for engraftment. We have developed a novel system where HSCs can be eliminated by targeting C-kit, a cell surface antigen that is highly expressed on the surface of HSCs. Cultivation of HSCs with ACK2, a depleting antibody specific for c-kit, prevented stem-cell factor (SCF) dependent HSC proliferation in vitro and resulted in cell death. Administration of ACK2 to mice led to the rapid and transient removal of >98% of endogenous HSCs in vivo thus resulting in equal numbers of available niches for engraftment. Following ACK2 clearance from serum, transplantation of these animals with donor HSCs led to chimerism levels of up to 90%, representing a 180-fold increase as compared to unconditioned animals. This non-myeloablative conditioning regimen had few side effects, other than temporary loss of coat color. The HSCs in even untransplanted animals rapidly recovered and animals remained healthy and fertile. This work redefines the way we approach BMT/HSCT, and places great emphasis on the necessity to create available HSC niches prior to transplantation. Extrapolation of these methods to humans may enable efficient yet mild conditioning regimens for transplantation, thus expanding the potential applications of BMT/HSCT.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3384-3384
Author(s):  
Andrew Daly ◽  
Douglas A. Stewart ◽  
Ahsan Chaudhry ◽  
Nizar J Bahlis ◽  
Christopher Brown ◽  
...  

Abstract Abstract 3384 Poster Board III-272 Purpose: Hematopoietic stem cell transplantation (HSCT) is routinely offered to suitable candidates with high-risk or advanced acute lymphoblastic leukemia. In this report we describe our experience with a novel conditioning regimen, previously reported to confer low TRM and high OS in AML, in this population. Patients and Methods: Between 05/2000 and 06/2008 44 patients with high-risk (either adverse cytogenetics, age > 35 or high WBC at diagnosis) or advanced (>CR1) ALL received HSCT after myeloablative conditioning using Fludarabine 50 mg/m2 days -6 to -2, Busulfan 3.2 mg/kg days -5 to -2, and TBI 2 Gy x 2 doses. GVHD prophylaxis was with rabbit ATG 0.5 mg/kg day -2, then 2 mg/kg days -1 and 0, CyA (tapered on day +56) and short-course methotrexate. All patients were in remission at the time of transplant. Imatinib mesylate (Gleevec) was not used routinely before or after transplant for patients with BCR-Abl+ ALL. Median (range) follow-up of surviving patients is 4.3 (1.0 – 9.0) years. All patients were followed for at least 1 year after BCT. Results: The cohort consists of 32 patients with high-risk (median age 40 (19-64) years) and 12 patients with advanced (median age 25 (19-65) years) disease who received bone marrow (n=5), G-CSF mobilized blood stem cells (n=38) or umbilical cord blood stem cells (n=1) from 25 related (21 fully-matched) or 19 unrelated (16 fully-matched) donors. Median times to neutrophil and platelet engraftment were 14 (11-28) days and 18 (9-105) days, respectively. Five patients did not require platelet transfusion. Cumulative incidences of grade II-IV and grade III-IV acute GVHD were 53.2% (95% CI 36.3%-67.5%) and 20.6% (95% CI 3.5%-47.6%), respectively. Chronic GVHD complicated 55% (95% CI 38.4%-68.8%) of transplants. Six patients (13.6%) died in remission before day +100. Event-free and overall survivals at 5 years were 56.7% (95% CI 39.1%-71.0%) and 66.0% (95% CI 48.8%-78.6%), respectively. Nine patients (20%) died in remission, 6 (14%) died after relapse and two patients remain alive following second transplants for relapsed disease. Five of 11 patients age > 50, 8/12 patients with advanced disease and 13/23 patients with adverse-risk cytogenetics remain alive. Conclusion: We found encouraging results with FluBup-ATG-TBI in a cohort of patients with advanced or high-risk ALL. These results warrant comparison with other conditioning regimens in a randomized, multi-center study. Disclosures: Daly: Hoffmann-Laroche: Advisory Board, Honoraria. Stewart:Hoffmann La Roche: Advisory Board, Honoraria, Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 873-873
Author(s):  
Andrea Toma ◽  
Marie-Lorraine Balère-Appert ◽  
Jean-Michel Boiron ◽  
Pierre Bordigoni ◽  
Gerard Socie ◽  
...  

Abstract Abstract 873 The use of peripheral blood stem cells (PBSC) for hematopoietic stem cell transplantation (HSCT) is associated with a higher risk of chronic graft versus host disease (GvHD) but its impact on survival is not clear since it may favor a greater graft versus leukemia (GvL) effect. However, in the context of HSCT from unrelated donors (UD), the balance between GvH and GvL may differ from the context of sibling donors and thus the use of PBSC may be deleterious. In this retrospective study, we analyzed 103 patients from the french registry who received a graft from an UD after a reduced intensity conditioning regimen (RIC) to evaluate the role of various parameters including the source of stem cells on the outcome. Seventy-one D/R pairs (69%) were 10/10 HLA match at the allelic level. Mismatches concerned 5, 6, 15, 2 and 7 D/R pairs for HLA-A, -B, -C, -DRB1 and -DQB1, respectively. The median age was 46 years (18-67). All patients had hematologic malignancies: AL (n=35), MM (n=18), CLL (n=5), NHL (n=11), HD (n=9), CML (n=12), MDS (n=9), and MPS (n=4). 39% of the patients were in an advanced phase of the disease at time of HSCT. The conditioning regimen was Fluda/TBI 2Gys for 26 patients, Bu/Fluda/ATG for 24 patients, Fluda/Melph for 16 patients and others for 37 patients. Overall, anti-thymocytes globulins (ATG) were part of the conditioning regimen for 77% of patients. The source of stem cells was PBSC for 65 patients and bone marrow (BM) for 38 patients. The median follow up of the cohort is 61,3 months (1,2-113,7). The results showed that 95% of patients engrafted. Five patients did not engraft (4 in the BM group and 1 in the PBSC group). Acute GvHD grade II to IV and grade III/IV occurred in 47% and 19% of patients, respectively. The risk of developing chronic GvHD was 49% at 2 years. Overall survival (OS) was 36% at five years. The median disease free survival (DFS) was 55 months among the 36 patients alive. We performed univariate and multivariate analysis of factors susceptible to impact on GvHD and survival. The multivariate analysis included the impact of HLA mismatch, disease status, diagnosis, source of stem cells, patient's and donor's ages. This multivariate analysis performed on the global population shows a trend towards an improved OS with the use of BM instead of PBSC. However, when focusing the multivariate analysis on the 71 patients transplanted with a 10/10 match donor, the most potent factor influencing the outcome is the use of BM which is associated with an improved OS (p=0.03) and DFS (p=0.02), less acute GvHD grade II-IV (p=0.05), or grade III/IV (p=0.05) and less chronic GvHD (p=0.05). These results suggest that the use of BM as the source of stem cells should be reconsidered in the context of matched UD after RIC transplantation. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document