Post-Transplant Cyclophosphamide and Sirolimus Are Synergistic in Preventing Rejection and Inducing Stable Mixed Chimerism Independently of Regulatory T Cells.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3540-3540 ◽  
Author(s):  
Courtney Fitzhugh ◽  
Matthew M. Hsieh ◽  
Oswald Phang ◽  
Camille Madison ◽  
Leo Luznik ◽  
...  

Abstract Abstract 3540 Poster Board III-477 A strategy that can induce stable mixed chimerism across human leukocyte antigen (HLA) barriers would be beneficial in extending the application of hematopoietic stem cell transplantation (HSCT) to patients with severe sickle cell disease (SCD) who are in need of this potentially curative procedure. Indeed, we have recently demonstrated the feasibility of an HLA-matched sibling protocol employing low dose total body irradiation (TBI, 300cGy), the lymphocyte depleting agent alemtuzumab, and sirolimus to reverse the phenotype with minimal side effects. Due to the lack of HLA-matched siblings in the majority of patients, our goal is to develop a safe haploidentical regimen. In this work, we focused on determining optimal postgrafting immunosuppression and examined sirolimus and post-transplant cyclophosphamide (PT-cy), agents known to induce transplantation tolerance. To determine the optimal sequence for combining these drugs and whether this combination is synergistic in promoting stable donor chimerism despite the antiproliferative effects of sirolimus, we used a mismatched murine model with BalbC donors and C57Bl6 recipients. Twenty-five to 40 recipient mice received 200cGy TBI and PT-cy (200mg/kg intraperitoneally (IP) 2 days post transplant) with or without sirolimus (3mg/kg IP) for 14 to 30 days starting 1 day before or 4, 6, or 10 days post transplant. We found that in contrast to sirolimus or PT-cy alone, the combination of PT-cy and a limited course of sirolimus resulted in stable mixed chimerism: all mice that received PT-cy and sirolimus starting between 1 day before and 6 days after transplant attained donor chimerism levels ranging from 15-35%. Further, a 14 day course of sirolimus was sufficient to maintain stable mixed chimerism in our model (See Figures 1 and 2). To examine whether this synergistic effect is mediated by regulatory T cells, we administered anti-CD25 monoclonal antibody (CD25 mAb), an agent known to transiently deplete these cells in vivo. Fifteen mice received 200cGy TBI, sirolimus, PT-Cy, and either no CD25 mab, CD25 mab (1mg IP) on 7 and 3 days before and 1 day after transplant, or CD25 mab starting 14 days after transplant. CD25 mab was given biweekly for 5 weeks to mice in both groups. Donor engraftment levels did not differ in the three groups, with donor chimerism levels ranging from 30-40%. Our data show that the anti-proliferative effects of sirolimus do not inhibit the efficacy of the cytotoxic agent cyclophosphamide. Rather, our data demonstrate that the combination of PT-cy and a limited course of sirolimus synergistically promote mixed bone marrow chimerism in a complete mismatched setting. Further, the synergistic effect of this drug combination appears to be mediated independently from CD25+ regulatory T cell expression. In light of our previous success using sirolimus in an HLA-matched HSCT protocol, these findings lay the groundwork for developing PT-cy and sirolimus as a novel, safe, and effective means of promoting stable mixed chimerism in the haploidentical setting and thus greatly enhancing our ability to successfully apply this approach to patients with severe SCD. Figure 1 PT-cy and sirolimus are synergistic. 25 to 40 C57BI6 mice received 200cGy TBI, 22-25 × 106 bone marrow cells from BalbC mice, and PT-cy (Cy) 200mg/kg IP 2 days post transplant with or without sirolimus (Sir) 3mg/kg IP for 30 days starting from 1 day before to 4 days after transplant. Figure 1. PT-cy and sirolimus are synergistic. 25 to 40 C57BI6 mice received 200cGy TBI, 22-25 × 106 bone marrow cells from BalbC mice, and PT-cy (Cy) 200mg/kg IP 2 days post transplant with or without sirolimus (Sir) 3mg/kg IP for 30 days starting from 1 day before to 4 days after transplant. Figure 2 Fourteen days of sirolimus is sufficient to maintain stable mixed chimerism. 25 to 40 C57BI6 mice received 200cGy TBI, 22-25 × 106 bone marrow cells from BalbC mice, and PT-cy (Cy) 200mg/kg IP 2 days post transplant with or without sirolimus (Sir) 3mg/kg IP for 14 days starting from 1 day before to 10 days after transplant. Figure 2. Fourteen days of sirolimus is sufficient to maintain stable mixed chimerism. 25 to 40 C57BI6 mice received 200cGy TBI, 22-25 × 106 bone marrow cells from BalbC mice, and PT-cy (Cy) 200mg/kg IP 2 days post transplant with or without sirolimus (Sir) 3mg/kg IP for 14 days starting from 1 day before to 10 days after transplant. Disclosures: No relevant conflicts of interest to declare.

1989 ◽  
Vol 169 (1) ◽  
pp. 213-238 ◽  
Author(s):  
H Mayumi ◽  
R A Good

A new method of cyclophosphamide (CP)-induced skin allograft tolerance in mice that can regularly overcome fully allogeneic (major H-2 plus non-H-2) antigen barriers in mice has been established. The components of the method are intravenous or intraperitoneal administration of 50-100 micrograms of anti-Thy-1.2 mAb on day -1, intravenous injection of 90 x 10(6) allogeneic spleen cells mixed with 30 x 10(6) allogeneic bone marrow cells from the same donor on day 0, and intraperitoneal injection of 200 mg/kg CP on day 2. In each of four fully allogeneic donor----recipient combinations, including C3H/HeJ (C3H; H-2k)----C57BL/6J(B6; H-2b), B6----C3H, BALB/cByJ (BALB; H-2d)----B6, and BALB----C3H, long-lasting survival of skin allografts was induced in most of the recipient mice. The specific tolerant state induced was dependent on the doses of the antibody and bone marrow cells used. The optimal timing of CP treatment to induce tolerance was found to be 1-3 d after the stimulating cell injection. Treatment with the anti-Thy-1.2 antibody together with CP on day 2 after the cell injection on day 0 also induced profound tolerance. In the B6 mice made tolerant of C3H with antibody, C3H spleen cells plus C3H bone marrow cells, and then CP, a minimal degree of stable mixed chimerism was established and the antitolerogen (C3H) immune responses examined here, including delayed footpad reaction (DFR), CTL activity, and capacity for antibody production against donor-strain antigens were abrogated in a tolerogen-specific manner. From cell transfer experiments, the mechanism of tolerance could be largely attributed to reduction of effector T cells reactive against the tolerogen, and strong suppressive influences that might prolong skin allograft survival directly were not detected in the tolerant mice. Moreover, pretreatment with anti-Thy-1.2 antibody or anti-L3T4 (CD4) antibody was more effective than pretreatment with anti-Lyt-1 (CD5) antibody or anti-Lyt-2 (CD8) antibody as an initial step in tolerance induction. These results suggest that permanent tolerance to fully allogeneic skin grafts may be induced because antibody given before the stimulating cell injection reduces the number of reactive T cells in the recipient mice. This antibody treatment may facilitate an antigen-stimulated destruction of responding and thus proliferating cells with CP by preventing a possibly less proliferative, more rapid maturation of reactive T cells or by destroying residual effector T cells.(ABSTRACT TRUNCATED AT 400 WORDS)


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5341-5341
Author(s):  
Reza Tabrizi ◽  
Francis Belloc ◽  
Xavier Lafarge ◽  
Virginie Perreau ◽  
Krimo Bouabdallah ◽  
...  

Abstract The circulating dendritic cells (DC) are known to have an immunoregulatory role after allogeneic HSC transplantation, and recipient DC have been shown to be important in the development of GVHD in animal model. We studied the DC chimerism of 21 patients (pts) transplanted with reduced intensity conditioning regimen between January 2004 and August 2005. The blood was sampled at days -1, 15, 28 and 56 after transplantation. A series of 17 control normal bloods were also analyzed. DC were identified as ILT3-expressing cells negative for CD14. These cells were sorted by flow cytometry and chimerism was analyzed by PCR of Short Tandem Repeat motifs. Preliminary experiments showed that at least 500 sorted cells were necessary to perform chimerism analysis. Eight females and 13 males (median of age: 54 yrs; 25–61) were enrolled in the study. Diagnoses were 6 AML, 2 sAML, 1 MDS, 3 ALL, 6 MM, 2 NHL and 1 CML. Fifteen pts had high-risk disease. As conditioning regimen, all but 3 pts received cumulative dose of ATG (Thymoglobulin, Genzyme, Lyon, France) (2.5 mg/kg for sibling and 7.5 mg/kg for MUD), in addition to Busulfan 8 mg/kg and Fludarabine 150mg/m2. Eight pts received stem cells from a 10/10 MUD, 2 pts from 9/10 MUD, and 11 pts from sibling donor. For all but one patient, the stem cell source was blood. CsA alone was used for 11 pts, CsA with methotrexate for 8 pts and CsA with MMF for 2 pts. In the absence of aGVHD, the immunosuppressive therapy was tapered within 4 weeks (after day 28 in sibling donor and after day 90 for MUD). The kinetics of the absolute number of DC showed significantly lower count of circulating DC than in control samples at day -1, and a rapid increase, reaching normal values at day 15 post-transplant while the other leukocytes remained at a low value. To determine the origin of post-transplant blood DC, chimerism was analyzed on sorted DC. From 20 pts DC chimerism at day 15 was of full donor origin for 8 pts, mixed in 10 pts. Two pts had no detectable DC. At day 28 from 18 pts, only 4 pts had mixed chimerism. Of these 4 pts, 3 presented at day 56 a full donor chimerism and one patient died from relapse. For T cells at day 15, only one/17 pt had full donor chimerism, and one had no detectable circulating T cells. At day 28, 7/20 pts had full donor chimerism and one without detectable T cells. Only 2/17 had still mixed chimerism at 3 months. Six out of 21 pts relapsed and 3 died from relapse. Among these 6 pts, all but one reached full donor T cells, 3 had a full donor DC at day 28. Six pts from 21 had grade ≥ 2 aGVHD and 3 died from aGVHD. 7/17 evaluable pts had cGVHD. We didn’t found any correlation between DC chimerism and engraftment or relapse. At day 15, the median percentage of recipient DC was lower in pts who developed cGVHD (P<0.017) while it was higher in those with aGVHD (but p not significant). In conclusion, this study demonstrates that the circulating DC pool is rapidly reconstituted from both donor and recipient origins. Thereafter at day 28, donor engraftment of DC became predominant. The median of recipient DC was significantly higher in pts without cGVHD. An analysis on a larger series would be useful to determine if the chimerism in DC could be predictive for cGVHD.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 431-431
Author(s):  
Hidekazu Nishikii ◽  
Antonio Pierini ◽  
Yasuhisa Yokoyama ◽  
Takaharu Kimura ◽  
Hye-Sook Kwon ◽  
...  

Abstract Background: Foxp3+regulatory T cells (Treg) are a subpopulation of T cells, which regulate the immune system, maintain self-tolerance and enhance immune tolerance after transplantation. It was also reported that recipient derived Treg could provide immune privilege niche to allogeneic hematopoietic stem cells (HSC) after transplantation. However, the precise role of Treg in hematopoiesis has not been fully elucidated. Methods: We used Foxp3-DTR mice (B6, CD45.2) for in vivo depletion of Treg through diphtheria toxin (DT) injection and investigated whether Treg depletion would affect hematopoiesis derived from HSC. To investigate whether Treg depletion affects the function of the bone marrow microenvironment, we transplanted wild type bone marrow cells into lethally irradiated Foxp3-DTR mice after Treg depletion. Results: We found 1) a significant defect on B cell progenitors including mature B cells (IgM+B220+, P<0.001), pre-B cells (IgM-B220+CD19+cKit-, P<0.001) and pro-B cells (IgM-B220+CD19+cKit+, P<0.05), 2) LT-HSC population (CD34-/lowFlit3-cKit+Sca1+Lin-) was significantly expanded (p<0.01) and entered into cell cycle, 3) the residual Foxp3-CD4+ or CD8+ T cells in the bone marrow had an activated immune phenotype and clustered at sinusoids when bone marrow cells from Treg depleted mice were analyzed. Expanded LT-HSC from Treg depleted mice had reduced long-term reconstitution capacity when we performed competitive repopulation experiments using purified LT-HSC from Foxp3-DTR mice with or without Treg depletion (100 cells/mice, CD45.2), total bone marrow cells (2x10e5/mice, B6-F1, CD45.1/CD45.2) and congenic recipient mice (lethally irradiated B6, CD45.1). B cell reconstitution was also severely abrogated following transplantation using Treg depleted mice as recipients (p<0.01). In those mice, we observed a significant reduction of IL-7 production (p<0.01). Interestingly, we found that a subpopulation of CD45-TER119-CD31- ICAM1+ perivascular stromal cells are a major source of IL-7 in the bone marrow. ICAM1+ perivascular stromal cells also secrete SCF and CXCL12, which is crucial for the maintenance of LT-HSC. In Treg depleted BM cells, a significant reduction in IL-7 secretion from ICAM1+ perivascular stromal cells was observed, suggesting that this population is the target of activated T cells after Treg depletion. Conclusions: These data demonstrate that Treg play a key role in B cell differentiation from HSCs by maintaining the immunological homeostasis in the bone marrow microenvironment. These data provide new insights into Treg biology and function in normal and stress hematopoiesis. Disclosures Negrin: Stanford University: Patents & Royalties.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3464-3464
Author(s):  
Ellen Meijer ◽  
Lucia Duinhouwer ◽  
Eric Braakman ◽  
Joke Boonstra ◽  
Inge de Greef ◽  
...  

Abstract Abstract 3464 Introduction: Monitoring hematopoietic chimerism following allogeneic hematopoietic stem cell transplantation (alloSCT) after non-myeloablative (NMA) conditioning is commonly used to document engraftment. While mixed chimerism (MC) is frequently observed after NMA alloSCT, it occurs only rarely after myeloablative (MA) alloSCT. Persistent mixed chimerism is generally considered a risk factor for both relapse and rejection of the donor graft, irrespective of the type of conditioning regimen. However, it is still unknown to what extent mixed chimerism quantitatively predicts for relapse and whether patients at high risk for relapse can accurately be identified by assaying chimerism in either blood, marrow, and/or T cells. Therefore, we prospectively evaluated the establishment of complete and mixed chimerism in blood, marrow, and CD3+ selected T cells at 3, 6 and 12 months following alloSCT to investigate the predictive value of mixed chimerism in either subset for relapse. Methods: The study cohort included 152 recipients of an alloSCT, performed between October 2005 and December 2009 because of hematological malignancies (AML: n=61, ALL: n=25, NHL: n=18, Myeloma: n=15, CML: n=5, CLL: n=18, MDS: n=6, MPN: n=4). Median age was 50 years (range 17–67). Seventy-eight patients received a sibling donor transplant, 74 a transplant from a matched related donor. MA and NMA conditioning consisted of Cyclophosphamide/TBI 1200 cGy (n=45) and Fludarabine/TBI 200cGy (n=107), respectively. Chimerism tests were routinely performed in bone marrow (BM), peripheral blood (PB) and CD3 selected samples at 3, 6 and 12 months post transplant, using PCR and electrophoresis of variable number of tandem repeats or fluorescent in-situ hybridization by sex-chromosome specific probes. Complete (donor) chimerism (CC) was defined as >95% donor hematopoiesis, MC as ≤95% donor hematopoiesis. The cumulative incidence of disease recurrence/progression and progression free survival by chimerism status was evaluated as from 3, 6 and 12 months post transplant and adjusted for type of conditioning, donor type, patient/donor sexe and age. Results: MC appeared very rare after MA conditioning, but was more frequently observed after NMA alloSCT with incidences of BM-MC of 28, 22 and 9% at the 3, 6 and 12 month timepoints, respectively. MC in BM as well as PB samples at 6 and 12 months post transplant was highly predictive for disease recurrence/progression, both after univariate and multivariate analyses (BM: hazard ratio (HR) at 6 months: 3.52 (1.30-9.53 95% confidence interval (CI)), p-value 0.013; at 12 months: 5.42 (1.17-25.13 95%CI), p-value 0.031). The probability to develop a relapse increased to 40% in time if MC was detected at 6 months following transplantation, as compared to 15% in the CC group. MC detected at 12 months resulted in a relapse incidence of ≥50% (Figure 1). Moreover, MC at these timepoints also predicted for decreased progression free survival (BM: HR at 6 months: 2.35 (1.04-5.27 95%CI), p-value 0.039; at 12 months: 9.36 (2.94-29.92 95%CI), p-value 0.000). Chimerism results in CD3+ selected T cell fractions did not show a significant association with relapse/progression or progression free survival. Conclusion: These results show that patients with MC at 6 and 12 months post transplant in either BM or PB are at a 3 to 9 fold higher risk of disease recurrence/progression. T cell chimerism appeared not associated with relapse, which may be explained by a significant different and protracted pattern of kinetics after transplantation as compared to kinetics of PB and BM chimerism, thereby identifying different subgroups of patients. Collectively, this study highlights the need to rapidly establish complete donor chimerism both after MA and NMA alloSCT. The preferred medical intervention in patients with MC after NMA conditioning, however, remains to be established in a prospective study. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 87 (10) ◽  
pp. 4136-4142 ◽  
Author(s):  
I Kawashima ◽  
ED Zanjani ◽  
G Almaida-Porada ◽  
AW Flake ◽  
H Zeng ◽  
...  

Using in utero transplantation into fetal sheep, we examined the capability of human bone marrow CD34+ cells fractionated based on Kit protein expression to provide long-term in vivo engraftment. Twelve hundred to 5,000 CD34+ Kit-, CD34+ Kit(low), and CD34+ Kit(high) cells were injected into a total of 14 preimmune fetal sheep recipients using the amniotic bubble technique. Six fetuses were killed in utero 1.5 months after bone marrow cell transplantation. Two fetuses receiving CD34+ Kit(low) cells showed signs of engraftment according to analysis of CD45+ cells in their bone marrow cells and karyotype studies of the colonies grown in methylcellulose culture. In contrast, two fetuses receiving CD34+ Kit(high) cells and two fetuses receiving CD34+ Kit- cells failed to show evidence of significant engraftment. Two fetuses were absorbed. A total of six fetuses receiving different cell populations were allowed to proceed to term, and the newborn sheep were serially examined for the presence of chimerism. Again, only the two sheep receiving CD34+ Kit(low) cells exhibited signs of engraftment upon serial examination. Earlier in studies of murine hematopoiesis, we have shown stage-specific changes in Kit expression by the progenitors. The studies of human cells reported here are in agreement with observations in mice, and indicate that human hematopoietic stem cells are enriched in the Kit(low) population.


Blood ◽  
1986 ◽  
Vol 67 (2) ◽  
pp. 479-483
Author(s):  
T Nagasawa ◽  
T Sakurai ◽  
H Kashiwagi ◽  
T Abe

We studied a patient with a rare complication of amegakaryocytic thrombocytopenia (AMT) associated with systemic lupus erythematosus (SLE). To investigate the underlying pathogenesis of AMT, the effects of peripheral blood T cells and serum on human megakaryocyte progenitor cells were studied using in vitro coculture techniques. Mononuclear bone marrow cells (2 X 10(5) from normal donors produced 33.6 +/- 8.8 (n = 10) colony-forming unit-megakaryocytes (CFU-M) in our plasma clot system. When 2 X 10(5) of the patient's T cells were added to the culture system, the number of CFU-M decreased to only 3.5 +/- 0.6/2 X 10(5) bone marrow cells. No evidence of inhibitory effects was found by the addition of the patient's serum and complement to the culture system. The T cells stored at -80 degrees C on admission were also capable of suppressing autologous CFU-M after recovery from AMT. These results indicate that in vitro suppression of CFU-M from allogenic and autologous bone marrow cells by this patient's T cells provides an explanation for the pathogenesis of AMT associated with SLE.


Blood ◽  
2000 ◽  
Vol 95 (2) ◽  
pp. 700-704 ◽  
Author(s):  
Kimberly A. Gush ◽  
Kai-Ling Fu ◽  
Markus Grompe ◽  
Christopher E. Walsh

Fanconi anemia (FA) is a genetic disorder characterized by bone marrow failure, congenital anomalies, and a predisposition to malignancy. FA cells demonstrate hypersensitivity to DNA cross-linking agents, such as mitomycin C (MMC). Mice with a targeted disruption of the FANCC gene (fancc −/− nullizygous mice) exhibit many of the characteristic features of FA and provide a valuable tool for testing novel therapeutic strategies. We have exploited the inherent hypersensitivity offancc −/− hematopoietic cells to assay for phenotypic correction following transfer of the FANCC complementary DNA (cDNA) into bone marrow cells. Murine fancc −/− bone marrow cells were transduced with the use of retrovirus carrying the humanfancc cDNA and injected into lethally irradiated recipients. Mitomycin C (MMC) dosing, known to induce pancytopenia, was used to challenge the transplanted animals. Phenotypic correction was determined by assessment of peripheral blood counts. Mice that received cells transduced with virus carrying the wild-type gene maintained normal blood counts following MMC administration. All nullizygous control animals receiving MMC exhibited pancytopenia shortly before death. Clonogenic assay and polymerase chain reaction analysis confirmed gene transfer of progenitor cells. These results indicate that selective pressure promotes in vivo enrichment offancc-transduced hematopoietic stem/progenitor cells. In addition, MMC resistance coupled with detection of the transgene in secondary recipients suggests transduction and phenotypic correction of long-term repopulating stem cells.


Blood ◽  
1994 ◽  
Vol 83 (4) ◽  
pp. 939-948 ◽  
Author(s):  
Y Tomita ◽  
DH Sachs ◽  
M Sykes

Abstract We have investigated the requirement for whole body irradiation (WBI) to achieve engraftment of syngeneic pluripotent hematopoietic stem cells (HSCs). Recipient B6 (H-2b; Ly-5.2) mice received various doses of WBI (0 to 3.0 Gy) and were reconstituted with 1.5 x 10(7) T-cell-depleted (TCD) bone marrow cells (BMCs) from congenic Ly-5.1 donors. Using anti-Ly-5.1 and anti-Ly-5.2 monoclonal antibodies and flow cytometry, the origins of lymphoid and myeloid cells reconstituting the animals were observed over time. Chimerism was at least initially detectable in all groups. However, between 1.5 and 3 Gy WBI was the minimum irradiation dose required to permit induction of long-term (at least 30 weeks), multilineage mixed chimerism in 100% of recipient mice. In these mice, stable reconstitution with approximately 70% to 90% donor-type lymphocytes, granulocytes, and monocytes was observed, suggesting that pluripotent HSC engraftment was achieved. About 50% of animals conditioned with 1.5 Gy WBI showed evidence for donor pluripotent HSC engraftment. Although low levels of chimerism were detected in untreated and 0.5-Gy-irradiated recipients in the early post-BM transplantation (BMT) period, donor cells disappeared completely by 12 to 20 weeks post-BMT. BM colony assays and adoptive transfers into secondary lethally irradiated recipients confirmed the absence of donor progenitors and HSCs, respectively, in the marrow of animals originally conditioned with only 0.5 Gy WBI. These results suggest that syngeneic pluripotent HSCs cannot readily engraft unless host HSCs sustain a significant level of injury, as is induced by 1.5 to 3.0 Gy WBI. We also attempted to determine the duration of the permissive period for syngeneic marrow engraftment in animals conditioned with 3 Gy WBI. Stable multilineage chimerism was uniformly established in 3-Gy-irradiated Ly-5.2 mice only when Ly-5.1 BMC were injected within 7 days of irradiation, suggesting that repair of damaged host stem cells or loss of factors stimulating engraftment may prevent syngeneic marrow engraftment after day 7.


Blood ◽  
1987 ◽  
Vol 69 (5) ◽  
pp. 1363-1367 ◽  
Author(s):  
RJ Berenson ◽  
WI Bensinger ◽  
D Kalamasz ◽  
F Schuening ◽  
HJ Deeg ◽  
...  

Abstract Previous work has shown failure of engraftment in lethally irradiated dogs when autologous marrow was depleted of Ia-positive cells with an anti-Ia antibody and complement before infusion. In the current study, we have utilized an avidin-biotin immunoadsorption procedure to obtain a population of highly enriched Ia-positive cells for autologous bone marrow transplantation in dogs given lethal irradiation. Dog marrow cells (2.4 to 7.0 X 10(9) cells) that contained 8.6% to 19.9% Ia- positive cells were treated successively with monoclonal antibody 7.2, which reacts with a framework determinant of Ia-antigen, and biotin- conjugated goat antimouse immunoglobulin. These treated cells were passed over a column of avidin-Biogel (polyacrylamide) and the adherent cells removed by mechanical agitation. Seven lethally irradiated dogs were transplanted with 5.9 to 33.4 X 10(6) recovered adherent cells per kilogram of which 69.0% to 88.0% were Ia-positive. All dogs had hematologic recovery; six are alive and well with durable engraftment and one died on day 15 posttransplant. They are immunologically normal as determined by lymph node and bone marrow biopsies, lymphocyte function, and immunophenotyping of peripheral blood and bone marrow cells. These data provide further evidence that canine hematopoietic stem cells express Ia-like antigens and that these cells are capable of complete hematopoietic and immunologic reconstitution in an autologous model.


Sign in / Sign up

Export Citation Format

Share Document