scholarly journals 196: Ability of anti c-Kit targeting monoclonal antibody ACK-2 to target hematopoietic stem cells, and facilitate engraftment of human CD34+ engraftment and hematolymphoid development in immunodeficient mice

2007 ◽  
Vol 13 (2) ◽  
pp. 72
Author(s):  
D.L. Kraft ◽  
A. Czechowicz ◽  
D. Bhattacharya ◽  
I.L. Weissman
PLoS ONE ◽  
2010 ◽  
Vol 5 (10) ◽  
pp. e13109 ◽  
Author(s):  
Yoshinori Sato ◽  
Hiroshi Takata ◽  
Naoki Kobayashi ◽  
Sayaka Nagata ◽  
Naomi Nakagata ◽  
...  

Stem Cells ◽  
1997 ◽  
Vol 15 (S2) ◽  
pp. 13-19 ◽  
Author(s):  
Vladimir Bazil ◽  
John E. Brandt ◽  
Ronald Hoffman

1997 ◽  
Vol 15 (4) ◽  
pp. 1518-1528 ◽  
Author(s):  
M Tempero ◽  
P Leichner ◽  
G Dalrymple ◽  
K Harrison ◽  
S Augustine ◽  
...  

PURPOSE A phase I trial that evaluated for extrahematopoietic toxicity was conducted with iodine-131 (131I) labeled monoclonal antibody (MAb) CC49. Correlative studies included pharmacokinetic and biodistribution analyses, estimates of absorbed radiation dose, and measurement of human antimonoclonal antibodies (HAMA). PATIENTS AND METHODS After collection and cryopreservation of hematopoietic stem cells, 15 patients with gastrointestinal cancers were administered a tracer dose of 131I-MAb CC49. Within 5 to 6 days, 14 patients (two to three per activity level) underwent a single treatment with 131I-MAb CC49 (50, 100, 150, 200, 250, and 300 mCi/m2). Biodistribution was determined using planar and single photon emission computer tomographic (SPECT) imaging. Pharmacokinetic studies were performed by measuring radioactivity in serial blood samples. In some patients, biopsies of metastases and related normal tissues were obtained for radioactivity measurements. Radiation dosimetry estimates were calculated using available biodistribution, pharmacokinetic, and tissue biopsy data. Toxicity was evaluated using the National Cancer Institute (NCI) Common Toxicity Criteria. RESULTS No dose-limiting extrahematopoietic toxicity was identified. Twelve patients experienced grade IV myelosuppression and met criteria for infusion of hematopoietic stem cells. Radioimmunolocalization was excellent. The T1/2 for 131I-MAb CC49 after diagnostic and therapeutic administration was 39.7 +/- 10.4 and 46.1 +/- 10.6 hours, respectively. The percent injected dose per killigram of tumor ranged from 0.2 to 2.1. Absorbed radiation dose in metastatic tumor sites ranged from 630 to 3300 cGy. CONCLUSION Although extrahematopoietic dose-limiting toxicity was neither observed or predicted, suboptimal absorbed dose estimates suggested that further escalation of 131I-MAb CC49 would not be useful. Future studies should focus on the use of radionuclides with high energy beta emissions, such as yttrium 90, and on strategies to optimize access of antibody to target antigens.


Blood ◽  
1994 ◽  
Vol 83 (10) ◽  
pp. 3041-3051 ◽  
Author(s):  
JA Nolta ◽  
MB Hanley ◽  
DB Kohn

Abstract We have developed a novel cotransplantation system in which gene- transduced human CD34+ progenitor cells are transplanted into immunodeficient (bnx) mice together with primary human bone marrow (BM) stromal cells engineered to produce human interleukin-3 (IL-3). The IL- 3-secreting stroma produced sustained circulating levels of human IL-3 for at least 4 months in the mice. The IL-3-secreting stroma, but not control stroma, supported human hematopoiesis from the cotransplanted human BM CD34+ progenitors for up to 9 months, such that an average of 6% of the hematopoietic cells removed from the mice were of human origin (human CD45+). Human multilineage progenitors were readily detected as colony-forming units from the mouse marrow over this time period. Retroviral-mediated transfer of the neomycin phosphotransferase gene or a human glucocerebrosidase cDNA into the human CD34+ progenitor cells was performed in vitro before cotransplantation. Human multilineage progenitors were recovered from the marrow of the mice 4 to 9 months later and were shown to contain the transduced genes. Mature human blood cells marked by vector DNA circulated in the murine peripheral blood throughout this time period. This xenograft system will be useful in the study of gene transduction of human hematopoietic stem cells, by tracing the development of individually marked BM stem cells into mature blood cells of different lineages.


Blood ◽  
2010 ◽  
Vol 115 (18) ◽  
pp. 3704-3707 ◽  
Author(s):  
Faiyaz Notta ◽  
Sergei Doulatov ◽  
John E. Dick

Abstract Repopulation of immunodeficient mice remains the primary method to assay human hematopoietic stem cells (HSCs). Here we report that female NOD/SCID/IL-2Rgc-null mice are far superior in detecting human HSCs (Lin−CD34+CD38−CD90+CD45RA−) compared with male recipients. When multiple HSCs were transplanted, female recipients displayed a trend (1.4-fold) toward higher levels of human chimerism (female vs male: injected femur, 44.4 ± 9.3 vs 32.2 ± 6.2; n = 12 females, n = 24 males; P = .1). Strikingly, this effect was dramatically amplified at limiting cell doses where female recipients had an approximately 11-fold higher chimerism from single HSCs (female vs male: injected femur, 8.1 ± 2.7 vs 0.7 ± 0.7; n = 28 females, n = 20 males; P < .001). Secondary transplantations from primary recipients indicate that females more efficiently support the self-renewal of human HSCs. Therefore, sex-associated factors play a pivotal role in the survival, proliferation, and self-renewal of human HSCs in the xenograft model, and recipient sex must be carefully monitored in the future design of experiments requiring human HSC assays.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3178-3178
Author(s):  
Toshinao Kawai ◽  
Uimook Choi ◽  
Po-Ching Liu ◽  
Harry L. Malech

Abstract CD26/DPP-IV (CD26) is a membrane-anchored ectoenzyme with N terminus exopeptidase activity that cleaves X-Pro-dipeptides. Stromal cell-derived factor 1 (SDF-1) and its receptor CXCR4 play a central role in trafficking of hematopoietic stem cells in the bone marrow. SDF-1 has a proline second from the N-terminus and is cleaved by CD26. Mouse hematopoietic progenitors express CD26 and a brief treatment of these cells with Diprotin A (Ile-Pro-Ile), a specific inhibitor of CD26, enhances engraftment. We examined the effect of Diprotin A treatment of peripheral blood human CD34+ stem cells (PBSC) with respect to subsequent responses to SDF-1 and with respect to engraftment in the NOD/SCID mouse xenograft model. We found that human CD34+ PBSC with colony forming potential are unlike mouse hematopoietic stem cells in that they lack the equivalent of CD26; and also are unlike mouse cells in that their response to SDF-1 and their engraftment in the NOD/SCID xenograft model are not affected by pre-treatment with Diprotin A. However, administration of Diprotin A intravenously to the NOD/SCID mouse at the time of transplant of human PBSC greatly enhances engraftment of the human PBSC, suggesting an effect primarily on the mouse stroma. Previous reports suggested that 70% of mouse lineage negative/sca-1 positive hematopoietic stem cells express CD26. However, freshly selected mobilized human CD34+ PBSC do not express detectable CD26, though after 4 days and 7 days of ex vivo culture in growth factors (SCF, flt3-ligand, TPO, IL3) 8.9% and 26.6% of cells express CD26, indicating that CD26 may only appear in later progenitors. At day 4 the cultured human PBSC were sorted by flow cytometry into CD26 positive and negative fractions. Only the CD26 negative fraction contained colony forming cells. 4 day-cultured human PBSC were exposed to Diprotin A 5mM for 15 minutes, washed and used in a filter transwell migration assay in response to SDF-1 at concentrations from 0.5 to 10 nM. There was no statistical difference between migration of Diprotin A treated and control PBSC, even in experiments with longer treatment with Diprotin A. When these human PBSC were transplanted into NOD/SCID mice there was no difference of engraftment between the Diprotin A treated group and control group. However, when 1x106 of 4 day-cultured PBSC were injected into NOD/SCID mice without or together with 2μmol of Diprotin A, there was a profound enhancement on subsequent engraftment in the group of mice injected with Diprotin A at time of transplant. At 6 weeks after transplantation the CD45+ human cell engraftment of the Diprotin A group was 6-fold increased compared to control group (49.6±8.2% vs. 8.1 ± 3.4%, p<.0001). Taken together with the colony assay, the in vitro migration studies, and lack of effect on engraftment when only the human PBSC are treated with Diprotin A, this result suggests that the enhanced engraftment of human PBSC in NOD/SCID mice is due to an action of Diprotin A on endogenous mouse CD26/DPP-IV (where the target is unknown, but possibly stromal cells). Although, further work is required to determine levels of expression of CD26/DPP-IV in human marrow stromal cells, it is possible to speculate that inhibitors of CD26/DPP-IV activity may provide a novel approach to improve stem cell engraftment in humans.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4622-4622
Author(s):  
Nadja Grund ◽  
Patrick Maier ◽  
Uwe Appelt ◽  
Heike Allgayer ◽  
Frederik Wenz ◽  
...  

Abstract Hematologic side effects of cancer chemotherapy like myelosuppression are frequently dose-limiting. Lentiviral gene therapy with cytostatic drug resistance gene transfer to human hematopoietic stem cells (CD34+) is a promising approach to overcome this problem. In this context it is of interest if chemotherapy mediated selection has an impact on lentiviral integration site patterns of transduced hematopoietic stem cells (CD34+). Concerning this issue, human CD34+ cells transduced with a lentiviral self-inactivating (SIN) vector encoding MGMTP140K (the O6-BG resistant mutant of O6-methylguanine- DNA methyltransferase) were in vitro treated with the alkylating agent BCNU. For integration site analysis LM-PCR was performed and integration patterns of the treated and untreated CD34+ cells were analyzed and compared with an in silico set of 106 random integrations. We found different integration preferences of the lentiviral vector between either the treated (82 integrations) or the untreated (30 integrations) CD34+ cells and the in silico set: both groups showed chromosomal preferences, a significant bias for integrations in genes (74,4% in the treated, respectively 70% in the untreated to 40% in the in silico group), especially by favouring introns, a random integration distribution regarding transcription start sites (TSS), and most importantly no significant differences concerning the number of integrations in or near cancer genes. Concerning all integration characteristics we could not find significant differences when comparing the untreated with the treated group. In conclusion, the general distribution of lentiviral integrations in either untreated or treated human CD34+ cells showed no distinct differences between both groups but significant differences compared to the in silico integration set. These results suggest that chemoselection of cells lentivirally overexpressing a specific chemoresistence gene might not influence the integration pattern. Therefore chemotherapy pressure seems not to hamper the safety of lentiviral vectors in gene transfer studies.


2017 ◽  
Vol 53 ◽  
pp. S124
Author(s):  
Dariusz Krenz ◽  
Faidra Aivazidou ◽  
Andreas Bosio ◽  
Sebastian Knöbel ◽  
Ute Bissels

Sign in / Sign up

Export Citation Format

Share Document