First in Human Engraftment of Anti-HIV Lentiviral Vector Gene Modified CD34+ Peripheral Blood Progenitor Cells in the Treatment of AIDS Related Lymphoma (ARL).

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2348-2348 ◽  
Author(s):  
Amrita Krishnan ◽  
John A. Zaia ◽  
John Rossi ◽  
David DiGiusto ◽  
Michael Kalos ◽  
...  

Abstract Background: Autologous stem cell transplantation (ASCT) has become an accepted treatment option for high risk or relapsed ARL. Treatment related mortality is similar to the HIV negative setting. However, ultimately further improvement in ASCT will depend on both effective anti lymphoma therapy and better control of the HIV infection. Highly active antiretroviral therapy (HAART) can lower HIV viral loads to undetectable levels in the peripheral blood, but reservoirs of HIV are still present in the tissues and acquired resistance to HAART also remains a problem. A treatment strategy that would confer intrinsic resistance to HIV could circumvent theses issues. Herein we report on one such strategy using multiplexed RNA based anti-HIV gene transfer strategies to render autologous peripheral blood progenitor cells resistant to HIV. Patients with high risk ARL deemed candidates for ASCT were eligible. Seven subjects with NHL have been enrolled. (4PR, 2 REL, 1CR2), of whom 2 failed screening phase, 1 failed product release test, 2 are pending transplant, and 2 patients have undergone successful transplantation. Median age was 43 yrs at enrollment. Four pts to date were mobilized with chemotherapy plus GCSF and cells were collected for the clinical product (Fx1) and for CD34-selection (CliniMACSª, Miltenyi) and research treatment (Fx2). (see table ) UPN # Fx1 (CD34+/kg) Fx2 (CD34+/kg) Post Selection and transductionCD34+/kg 301 2.8X106 3.5X106 .26 X106 not infused 304 3.9X106 3.6X106 1.2 X106 305 3.4X106 3.8X106 1.4X106 306 5.6X106 8.8X106 pending Three days prior to the completion of CBV (cyclophosphamide 100mg/kg, BCNU 450mg/ m2, VP16 60mg/kg) conditioning, the Fx2 cells were thawed and transduced with a lentivirus vector (LV,rHIV7-ShI-TAR-CCR5Z) encoding 3 RNA elements including short hairpin RNA (shRNA) targeted to HIV tat/rev, a nucleolar localizing TAR decoy sequence, and a ribozyme targeted to CCR5. Cell viability post transduction ranged between 52–64% in three pts. On day 0 Fx2 is given and Fx1 is given 24hrs later (day+1). UPN301 did not receive the transduced Fx2 cells due to a low cell dose. For UPN304 and 305 who received the gene modified Fx2 cells, WBC engraftment occurred at day +11, platelet engraftment at day+16, and there have been no serious adverse events. Results to date at 30 and 60 days post ASCT reveal peripheral blood marking consistent with the ratio of gene modified to unmodified cells infused. Q-PCR analysis demonstrated distribution of genetically modified cells in myeloid and lymphoid lineages, and RT-PCR evidence of shRNA in progeny cells provided further evidence of successful transduction and engraftment of progenitor cells. Follow-up data for these and subsequent patients will be presented at the meeting. Conclusion: Lentiviral vector transduction of autologous peripheral blood progenitor cells with multiplexed RNA is feasible, well tolerated, and led to successful engraftment following high dose chemotherapy for ARL.

Blood ◽  
1994 ◽  
Vol 84 (9) ◽  
pp. 2898-2903 ◽  
Author(s):  
R Henschler ◽  
W Brugger ◽  
T Luft ◽  
T Frey ◽  
R Mertelsmann ◽  
...  

Abstract CD34(+)-selected hematopoietic progenitor cells are being increasingly used for autotransplantation, and recent evidence indicates that these cells can be expanded ex vivo. Of 15 patients with solid tumors undergoing a phase I/II clinical trial using CD34(+)-selected peripheral blood progenitor cells (PBPCs) after high-dose chemotherapy, we analyzed the frequency of long-term culture-initiating cells (LTCIC) as a measure of transplantation potential before and after ex vivo expansion of CD34+ cells. PBPCs were mobilized by combination chemotherapy and granulocyte colony-stimulating factor (G-CSF). The original unseparated leukapheresis preparations, the CD34(+)-enriched transplants, as well as nonabsorbed fractions eluting from the CD34 immunoaffinity columns (Ceprate; CellPro, Bothell, WA) were monitored for their capacity to repopulate irradiated allogeneic stroma in human long-term bone marrow cultures. We found preservation of more than three quarters of fully functional LTCIC in the CD34(+)-selected fractions. Quantitation of LTCIC by limiting dilution analysis showed a 53-fold enrichment of LTCIC from 1/9,075 in the unseparated cells to an incidence of 1/169 in the CD34+ fractions. Thus, in a single apheresis, it was possible to harvest a median of 1.65 x 10(4) LTCIC per kg body weight (range, 0.71 to 3.72). In addition, in six patients, large-scale ex vivo expansions were performed using a five-factor cytokine combination consisting of stem cell factor (SCF), interleukin-1 (IL-1), IL-3, IL-6, and erythropoietin (EPO), previously shown to expand committed progenitor cells. LTCIC were preserved, but not expanded during the culture period. Optimization of ex vivo expansion growth factor requirements using limiting dilution assays for LTCIC estimation indicated that the five-factor combination using SCF, IL-1, IL-3, IL-6, and EPO together with autologous plasma was the most reliable combination securing both high progenitor yield and, at the same time, optimal preservation of LTCIC. Our data suggest that ex vivo-expanded CD34+ PBPCs might be able to allow long-term reconstitution of hematopoiesis.


Blood ◽  
1993 ◽  
Vol 81 (8) ◽  
pp. 2031-2035 ◽  
Author(s):  
NJ Chao ◽  
JR Schriber ◽  
K Grimes ◽  
GD Long ◽  
RS Negrin ◽  
...  

Abstract Hematopoietic growth factors have been used to accelerate engraftment after bone marrow transplantation and to “mobilize” peripheral blood progenitor cells (PBPC). We report on the data in 85 consecutive patients with Hodgkin's disease who were treated in a single institution using different methods to obtain PB progenitor cells. Use of granulocyte colony-stimulating factor for mobilization resulted in a significantly accelerated time to recovery of granulocytes (10 days v 12 days, P < .01) when compared with “nonmobilized” PBPC recipients. Similarly, use of mobilized PBPC resulted in a significantly accelerated time to platelet engraftment (13 days v 30 days, P < .001) when compared with “nonmobilized” recipients. Moreover, there was a statistically significant difference in total costs in favor of the group receiving “mobilized” PBPC.


1998 ◽  
Vol 21 (6_suppl) ◽  
pp. 1-10
Author(s):  
C. Carlo-Stella ◽  
V. Rizzoli

Mobilized peripheral blood progenitor cells (PBPC) are increasingly used to reconstitute hematopoiesis in patients undergoing high-dose chemoradiotherapy. PBPC collections comprise a heterogeneous population containing both committed progenitors and pluripotent stem cells and can be harvested (i) in steady state, (ii) after chemotherapeutic conditioning, (iii) growth factor priming, or (iv) both. The use of PBPC has opened new therapeutic perspectives mainly related to the availability of large amounts of mobilized hematopoietic stem and progenitor cells. Extensive manipulation of the grafts, including the possibility of exploiting these cells as vehicles for gene therapy strategies, are now possible and will be reviewed.


Blood ◽  
2000 ◽  
Vol 96 (9) ◽  
pp. 3001-3007 ◽  
Author(s):  
Ian McNiece ◽  
Roy Jones ◽  
Scott I. Bearman ◽  
Pablo Cagnoni ◽  
Yago Nieto ◽  
...  

Abstract Ex vivo expanded peripheral blood progenitor cells (PBPCs) have been proposed as a source of hematopoietic support to decrease or eliminate the period of neutropenia after high-dose chemotherapy. CD34 cells were selected from rhG-CSF mobilized PBPCs from patients with breast cancer and were cultured for 10 days in defined media containing 100 ng/mL each of rhSCF, rhG-CSF, and PEG-rhMGDF in 1 L Teflon bags at 20 000 cells/mL. After culture the cells were washed and reinfused on day 0 of transplantation. On day +1, cohort 1 patients (n = 10) also received an unexpanded CD34-selected PBPC product. These patients engrafted neutrophils (absolute neutrophil count, &gt;500/μL) in a median of 6 (range, 5-14) days. Cohort 2 patients (n = 11), who received expanded PBPCs only, engrafted neutrophils in a median of 8 (range, 4-16) days. In comparison, the median time to neutrophil engraftment in a historical control group of patients (n = 100) was 9 days (range, 7-30 days). All surviving patients are now past the 15-month posttransplantation stage with no evidence of late graft failure. The total number of nucleated cells harvested after expansion culture was shown to be the best predictor of time to neutrophil engraftment, with all patients receiving more than 4 × 107 cells/kg, engrafting neutrophils by day 8. No significant effect on platelet recovery was observed in any patient. These data demonstrate that PBPCs expanded under the conditions defined can shorten the time to engraftment of neutrophils compared with historical controls and that the rate of engraftment is related to the dose of expanded cells transplanted.


1993 ◽  
Vol 11 (8) ◽  
pp. 1583-1591 ◽  
Author(s):  
I Tepler ◽  
S A Cannistra ◽  
E Frei ◽  
R Gonin ◽  
K C Anderson ◽  
...  

PURPOSE Attempts to increase dose-intensity in clinical practice have been limited by cumulative hematologic toxicity despite the use of hematopoietic growth factors. To address this problem, we designed a study to determine whether four cycles of dose-intensive chemotherapy with carboplatin could be administered in the outpatient setting using granulocyte-macrophage colony-stimulating factor (GM-CSF) and peripheral-blood progenitor cells (PBPCs) that had been harvested before initiation of treatment. PATIENTS AND METHODS An initial cycle (cycle no. 0) of cyclophosphamide 4 g/m2 followed by GM-CSF was used to mobilize PBPCs harvested by leukapheresis for 6 consecutive days. Cycles no. 1 through 4 consisted of outpatient carboplatin 600 mg/m2 and cyclophosphamide 600 mg/m2 followed by GM-CSF 5 micrograms/kg subcutaneously (SC) twice per day every 28 days. In cycle no. 1, PBPC were not reinfused to assess the effects of GM-CSF alone. In cycles no. 2 through 4, PBPCs were reinfused on day 3 in an outpatient setting. RESULTS In eight assessable patients, the addition of PBPCs in cycle no. 2 resulted in a significant reduction in the median duration of thrombocytopenia less than 20,000/microL (6.5 v 1 day; P = .016), days to platelets more than 50,000/microL (20.5 v 15 days; P = .020), number of platelet transfusions (five v 1.5; P = .016), and duration of neutropenia (absolute neutrophil count [ANC] < 1,000/microL (7 v 2.5 days; P = .008) when compared with cycle no. 1. Dose-limiting hematologic toxicity, defined as more than 7 days of platelets less than 20,000/microL or ANC less than 500/microL, was observed in four of eight patients during cycle no. 1, but not during cycles no. 2, 3, and 4 of chemotherapy supported by PBPCs (a total of 19 cycles in eight patients). Five of eight patients completed all four cycles of high-dose therapy. Three patients did not complete four cycles due to late thrombocytopenia (n = 2) or tumor progression (n = 1). CONCLUSION These results indicate a benefit of PBPCs in addition to GM-CSF in alleviating myelosuppression of dose-intensive chemotherapy. Initial collection of PBPCs may allow administration of repetitive cycles of high-dose chemotherapy with acceptable toxicity to outpatients at disease onset.


Sign in / Sign up

Export Citation Format

Share Document