Protection Against Graft-Versus-Host Disease and Retention of Graft-Versus-Tumor Effect after Allogeneic BMT in Mice Conditioned with TLI and Anti-Thymocyte Serum.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3049-3049
Author(s):  
Asha Pillai ◽  
Pearline Teo ◽  
Samuel Strober

Abstract In a completely MHC-mismatched model of bone marrow transplantation [C57BL/6 (H-2b) donors into BALB/c (H-2d) hosts], we have developed a technique of non-myeloablative host conditioning using fractionated total lymphoid irradiation (TLI) and and anti-thymocyte serum (ATS) that prevents lethal graft-versus-host disease (GVHD). We have previously reported that this GVHD prevention is dependent on the secretion of IL-4 and on host regulatory NKT cells. In the current study, we assessed the graft-versus-tumor (GVT) effect of BMT to determine whether the GVT effect remains intact in this non-myeloablative conditioning model. Male BALB/c mice were given fractionated TLI (17 doses of 240 cGy each), 3 doses of ATS, and subsequently received intravenous infusion of 50 x 106 bone marrow and 60x 106 splenocytes from C57BL/6 donors, with and without the BCL1 B-cell lymphoma. Animals were observed for minimum of 100 days, and underwent autopsy at death to assess for sub-clinical evidence of GVHD or tumor infiltration. TLI/ATS-conditioned mice achieved a high percentage of donor chimerism, in the range of 50–100% in all lineages. TLI/ATS-conditioned hosts uniformly survived without signs of GVHD beyond day +100. By contrast, hosts conditioned with a single dose of 800cGy total body irradiation (TBI) died of acute GVHD (severe diarrhea, hunched back, weight loss) by day +21. When TBI/ATS or TBI-treated mice receive bone marrow, splenocytes, and BCL1 lymphoma, all hosts died with signs of acute GVHD by day +28. TLI/ATS-conditioned hosts receiving marrow, splenocytes and tumor cells showed no evidence of disease progression by day +100 and either cleared tumor idiotype completely or had persistence of low-intensity staining for tumor idiotype (tumor dormancy). TLI/ATS-conditioned hosts given BCL1 tumor cells without allogeneic BMT all succumbed to tumor. TLI/ATS hosts receiving bone marrow plus BCL1 without splenocytes all died by day +108 with high circulating BCL1 tumor burden and no clinical evidence of GVHD. The data indicate that peripheral donor T cells are necessary to maintain a robust graft-versus-tumor effect after TLI/ATS conditioning, and that complete donor chimerism is not a requirement for tumor eradication. In conclusion, using the TLI/ATS non-myeloablative conditioning regimen, it is possible to maintain a clinically significant graft-versus-tumor effect without inducing GVHD despite a high dose of infused donor peripheral T cells.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 5167-5167
Author(s):  
Yihuan Chai ◽  
Huiying Qiu ◽  
Hui Lv

Abstract One of the main goals in allogeneic bone marrow(BM) transplantation is the abrogation of graft-versus-host disease (GVHD) with the preservation of antileukemia and antiviral activity. The Study present a selective T cell depletion strategy based on the physical separation of the alloreactive T cells, which were identified by expression of two activation-induced antigens (CD25 and CD69). T cells from C57BL/6(H-2b) mice were first activated with BALB/c (H-2d) recipient spleen cells in a 2-day mixed-lymphocyte-culture (MLC). Following this activation, this compound is selectively depleted based on expression of two activation-induced antigens CD25 and CD69 using magnetic cell sorting. The depleted cells or the untreated cells were then rechallenged respectively in a secondary MLC, with the same stimulator cells or a third-party (DBAH-2k) or tumor- specific (SP2/0, BALB/c-origin myeloma) cells. Cells proliferation were assayed at the indicated time points(1, 2, 3, 4, 5 days). These treated cells or control-cultured cells (2.0×106) mixed with 5.0×106 BM cells from C57BL/6 were transfused respectively by the trail vain into the lethally irradiated BALB/c to observe the survival time, GVHD incidence and pathological analysis. MLC assays demonstrated that this technique led to a significant decrease in alloreactivity of donor cells(29.02~64.17%), which at the same time preserved reactivity against third party cells(49.61~75.69%)and anti-tumor cells(61.14~68.62%). The mice in the group of control-coclutured were died of acute GVHD within 24days. The 7 recipient mice in the treated group were free of acute GVHD, and 3 mice were died of acute GVHD (aGVHD) within 23 days. MACS-based ex-vivo depletion of alloreactive donor T cells based on expression of two activation-induced antigens (CD25 and CD69) could inhibit anti-host responses, by contrast, anti-SP2/O and anti-third-party responses were preserved. Cotransplantation of these selected depleted cells and BM cells could reduce aGVHD.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1316-1316
Author(s):  
William J. Murphy ◽  
Olga Frolova ◽  
Marina Konopleva ◽  
Michael Andreeff ◽  
Weihong Ma ◽  
...  

Abstract The use of hematopoietic stem cell transplantation (HSCT) in cancer treatment is seriously hampered by the occurrence of graft-versus-host disease (GVHD) and cancer relapse. During acute GVHD, inflammatory cytokines play a pivotal role in the amplification of GVHD. Therefore, assessment of agents with known anti-neoplastic activity that also reduce cytokine production may be useful in both the prevention of GVHD and cancer relapse. The synthetic triterpenoid, CDDO (2-cyano-3, 12-dioxooleana-1, 9-dien-28-oic acid) is multifunctional molecule which has shown potent anti-cancer activities both in vitro and in vivo through the induction of apoptosis. We first examined the effects of CDDO on both human and murine T cell mitogen responses in vitro. CDDO significantly inhibited mitogen responsiveness of both human and murine T cells in vitro with evidence of cell cycle arrest of the human T cells. We then proceeded to examine the effects of CDDO on acute GVHD induction and progression. In these studies, lethally irradiated C57BL/6 mice received 10 million bone marrow cells (BMC) and 40 million spleen cells from fully MHC-mismatched BALB/c donors. All of the control mice succumbed rapidly due to acute GVHD. In contrast, the mice that received CDDO (120 ug/BID) given from days 0-3 following BMT exhibited significant improvement in survival (P < 0.001). Body weights from the treated mice also were significantly increased compared to untreated controls. We found that the timing of CDDO administration was a critical factor for protection from GVHD as protection only occurred when CDDO was administered early after BMT. Importantly, donor myeloid reconstitution was not adversely affected by CDDO treatment as determined by peripheral blood cell count and donor chimerism assessment on day +14 post-transplant. No adverse toxicities or effects on reconstitution were observed in the mice receiving BMC alone with CDDO being administered continuously. Given the reported direct anti-tumor effects of CDDO, it will be of particular interest in examining the effects of CDDO and allogeneic BMT in tumor-bearing recipients. Our findings suggest that CDDO can enhance the efficacy of allogeneic BMT by decreasing acute GVHD in mice.


Blood ◽  
1998 ◽  
Vol 91 (9) ◽  
pp. 3315-3322 ◽  
Author(s):  
Bimalangshu R. Dey ◽  
Yong-Guang Yang ◽  
Gregory L. Szot ◽  
Denise A. Pearson ◽  
Megan Sykes

We have recently made the paradoxical observation that a single injection of recombinant murine interleukin-12 (IL-12) on the day of bone marrow transplantation (BMT) inhibits graft-versus-host disease (GVHD) in lethally irradiated mice receiving fully major histocompatability complex (MHC)-mismatched bone marrow and spleen cells. We have now examined the mechanism of this effect of IL-12 on acute GVHD. By day 4 post-BMT, IL-12–treated mice showed marked reductions in splenic donor CD4+ and CD8+ T cells compared with GVHD controls. Expression of the early activation markers IL-2R alpha chain (CD25) and CD69 on splenic donor CD4+ cells was considerably higher at early time points (36 and 72 hours post-BMT) in IL-12–treated mice compared with GVHD controls. However, the later, GVHD-associated increase in CD25 and very late antigen-4 (VLA-4) expression on donor T cells was greatly depressed in IL-12–protected mice compared with GVHD controls. The marked GVHD-associated expansion of host-reactive T helper cells by day 4 was also completely inhibited in the IL-12–treated group. Expression of Fas was increased on donor CD4 cells of IL-12–treated mice compared with those of controls on days 3 through 7 post-BMT. Furthermore, the ability of IL-12 to protect against GVHD was at least partially dependent on the ability of donor cells to express functional Fas molecules. We conclude that IL-12 treatment at the time of BMT markedly perturbs the activation of alloreactive donor CD4+ T cells that play a critical role in the pathogenesis of acute GVHD. We hypothesize that these perturbations culminate in Fas-dependent apoptosis of donor T cells, thus impeding their expansion and their GVHD-promoting activity.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3245-3245
Author(s):  
Jun-ichi Nishimura ◽  
Divino DeOliveira ◽  
Benny J. Chen ◽  
Yuzuru Kanakura ◽  
Russell P. Rother ◽  
...  

Abstract Graft versus-host disease (GVHD) is a major cause of morbidity and mortality after bone marrow transplantation (BMT). An allogeneic GVH reaction is a response of donor lymphoid cells to host minor or major histocompatibility antigens. Donor T cells can be activated through the innate and the adaptive immune mechanisms. Donor B cells produce antibodies directed to host cells. These mechanisms may activate complement pathways. Thus, complement may have a crucial role in inflammation during a GVH reaction, but direct evidence for this has not been shown. In this study, we investigated the possibility of complement inhibitor, anti-mouse C5 antibody (BB5.1), to ameliorate the symptoms of GVHD using an acute GVHD mouse model: C57BL/6 (H2b) →BALB/c (H2d). One million T cells were injected together with 1 x 107 T-cell-depleted bone marrow (TCD BM) cells via tail vein into lethally irradiated BALB/c (8.5 Gy) recipients. Anti-mouse C5 antibody or its isotype matched control was administered intraperitoneally at a dose of 1 mg/mouse, 3 doses/week, for 4 weeks. Recipients were weighed weekly, and their survival was monitored daily. Average body weight of C5 antibody treated mice was 15.8 g at day 84 (19.2 g at day 0, N=12), whereas average weight of control mice was 13.3 g (19.2 g at day 0, N=12) (P=0.05, Student’s t-test). Kaplan-Meier survival curves were also compared as shown in the Figure. Eight of 12 mice were alive at day 84 in the treated group, as compared to only 2 of 12 in the control group (P=0.03, Logrank test). A second experiment showed similar data. We, thus, observed the effect of anti-mouse C5 antibody to reduce the symptoms of GVHD using an acute GVHD mouse model. These results might open a new window for the prevention of acute GVHD. Further experiments are currently ongoing to clarify the exact mechanism between complement and GVHD. Figure Figure


Blood ◽  
1998 ◽  
Vol 91 (9) ◽  
pp. 3315-3322 ◽  
Author(s):  
Bimalangshu R. Dey ◽  
Yong-Guang Yang ◽  
Gregory L. Szot ◽  
Denise A. Pearson ◽  
Megan Sykes

Abstract We have recently made the paradoxical observation that a single injection of recombinant murine interleukin-12 (IL-12) on the day of bone marrow transplantation (BMT) inhibits graft-versus-host disease (GVHD) in lethally irradiated mice receiving fully major histocompatability complex (MHC)-mismatched bone marrow and spleen cells. We have now examined the mechanism of this effect of IL-12 on acute GVHD. By day 4 post-BMT, IL-12–treated mice showed marked reductions in splenic donor CD4+ and CD8+ T cells compared with GVHD controls. Expression of the early activation markers IL-2R alpha chain (CD25) and CD69 on splenic donor CD4+ cells was considerably higher at early time points (36 and 72 hours post-BMT) in IL-12–treated mice compared with GVHD controls. However, the later, GVHD-associated increase in CD25 and very late antigen-4 (VLA-4) expression on donor T cells was greatly depressed in IL-12–protected mice compared with GVHD controls. The marked GVHD-associated expansion of host-reactive T helper cells by day 4 was also completely inhibited in the IL-12–treated group. Expression of Fas was increased on donor CD4 cells of IL-12–treated mice compared with those of controls on days 3 through 7 post-BMT. Furthermore, the ability of IL-12 to protect against GVHD was at least partially dependent on the ability of donor cells to express functional Fas molecules. We conclude that IL-12 treatment at the time of BMT markedly perturbs the activation of alloreactive donor CD4+ T cells that play a critical role in the pathogenesis of acute GVHD. We hypothesize that these perturbations culminate in Fas-dependent apoptosis of donor T cells, thus impeding their expansion and their GVHD-promoting activity.


Blood ◽  
1991 ◽  
Vol 78 (8) ◽  
pp. 2139-2149 ◽  
Author(s):  
JH Antin ◽  
BE Bierer ◽  
BR Smith ◽  
J Ferrara ◽  
EC Guinan ◽  
...  

Seventy-one patients with hematologic malignancies received bone marrow from a histocompatible sibling (n = 48) or a partially matched relative (n = 23) that had been depleted of CD5+ T cells with either an anti-CD5 mooclonal antibody (MoAb) plus complement (anti-Leu1 + C) or an anti- CD5 MoAb conjugated to ricin A chain (ST1 immunotoxin [ST1-IT]). These patients received intensive chemoradiotherapy consisting of cytosine arabinoside, cyclophosphamide, and fractionated total body irradiation. Both anti-Leu1 + C and ST1-IT ex vivo treatments effectively depleted bone marrow of T cells (97% and 95%, respectively). Overall, primary and late graft failure each occurred in 4% of evaluable patients. The diagnosis of myelodysplasia was a significant risk factor for graft failure (P less than .001), and if myelodysplastic patients were excluded, there were no graft failures in major histocompatibility complex (MHC)-matched patients and 2 of 23 (8.7%) in MHC-mismatched patients. The actuarial risk of grade 2 to 4 acute graft-versus-host disease (GVHD) was 23% in MHC-matched patients and 50% in MHC- mismatched patients. In MHC-matched patients, acute GVHD tended to be mild and treatable with corticosteroids. Chronic GVHD was observed in 6 of 36 (17%) MHC-matched patients and none of 11 MHC-mismatched patients. There were no deaths attributable to GVHD in the MHC-matched group. Epstein-Barr virus-associated lymphoproliferative disorders were observed in 3 of 23 MHC-mismatched patients. The actuarial event-free survival was 38% in the MHC-matched patients versus 21% in the MHC- mismatched patients. However, if outcome is analyzed by risk of relapse, low-risk patients had a 62% actuarial survival compared with 11% in high-risk patients. These data indicate that the use of anti-CD5 MoAbs can effectively control GVHD in histocompatible patients, and that additional strategies are required in MHC-mismatched and high-risk patients.


Blood ◽  
2020 ◽  
Author(s):  
Cheng Yin Yuan ◽  
Vivian Zhou ◽  
Garrett Sauber ◽  
Todd M Stollenwerk ◽  
Richard Komorowski ◽  
...  

Graft versus host disease (GVHD) pathophysiology is a complex interplay between cells that comprise the adaptive and innate arms of the immune system. Effective prophylactic strategies are therefore contingent upon approaches that address contributions from both immune cell compartments. In the current study, we examined the role of the type 2 cannabinoid receptor (CB2R) which is expressed on nearly all immune cells and demonstrated that absence of the CB2R on donor CD4+ or CD8+ T cells, or administration of a selective CB2R pharmacological antagonist, exacerbated acute GVHD lethality. This was accompanied primarily by the expansion of proinflammatory CD8+ T cells indicating that constitutive CB2R expression on T cells preferentially regulated CD8+ T cell alloreactivity. Using a novel CB2R-EGFP reporter mouse, we observed significant loss of CB2R expression on T cells, but not macrophages, during acute GVHD, indicative of differential alterations in receptor expression under inflammatory conditions. Therapeutic targeting of the CB2R with the agonists, tetrahydrocannabinol (THC) and JWH-133, revealed that only THC mitigated lethal T cell-mediated acute GVHD. Conversely, only JWH-133 was effective in a sclerodermatous chronic GVHD model where macrophages contribute to disease biology. In vitro, both THC and JWH-133 induced arrestin recruitment and ERK phosphorylation via CB2R, but THC had no effect on CB2R-mediated inhibition of adenylyl cyclase. These studies demonstrate that the CB2R plays a critical role in the regulation of GVHD and suggest that effective therapeutic targeting is dependent upon agonist signaling characteristics and receptor selectivity in conjunction with the composition of pathogenic immune effector cells.


Blood ◽  
2000 ◽  
Vol 95 (5) ◽  
pp. 1572-1579 ◽  
Author(s):  
Franco Locatelli ◽  
Marco Zecca ◽  
Roberto Rondelli ◽  
Federico Bonetti ◽  
Giorgio Dini ◽  
...  

Leukemia relapse is a major cause of treatment failure for patients with acute leukemia given allogeneic bone marrow transplantation (BMT). This study evaluated whether a reduction of the dosage of cyclosporine-A (Cs-A) used for graft versus host disease (GVHD) prophylaxis could reduce relapse rate (RR) in children with acute leukemia given BMT. Fifty-nine children who had transplantation from HLA-identical siblings were randomized to receive Cs-A intravenously at a dosage of 1 mg/kg/d (Cs-A1) or of 3 mg/kg/d (Cs-A3) until patients were able to tolerate oral intake. Subsequently, both groups received Cs-A orally at a dosage of 6 mg/kg/d, with discontinuation 5 months after BMT. The probability of developing grade II-IV acute GVHD was 57% for the Cs-A1 group versus 38% for the Cs-A3 group (P = .06); the probability of developing chronic GVHD was 30% for the Cs-A1 group and 26% for the Cs-A3 group (P = NS). Three patients died of grade IV acute GVHD: 2 were in the Cs-A1 and the third in the Cs-A3 group. The RR was 15% for the Cs-A1 group and 41% for the Cs-A3 group (P = .034); 1-year transplant-related mortality estimates were 17% and 7%, respectively (P = NS). With a median observation time of 44 months from BMT, the 5-year event-free survival for children belonging to Cs-A1 and Cs-A3 groups was 70% and 51%, respectively (P = .15). Our data demonstrate that the use of low Cs-A doses is associated with a statistically significant reduction of leukemia relapse, probably due to an increased graft versus leukemia effect.


Blood ◽  
2006 ◽  
Vol 109 (4) ◽  
pp. 1756-1764 ◽  
Author(s):  
Yukimi Sakoda ◽  
Daigo Hashimoto ◽  
Shoji Asakura ◽  
Kengo Takeuchi ◽  
Mine Harada ◽  
...  

Abstract Chronic graft-versus-host disease (GVHD) is the most common cause of poor long-term outcomes after allogeneic bone marrow transplantation (BMT), but the pathophysiology of chronic GVHD still remains poorly understood. We tested the hypothesis that the impaired thymic negative selection of the recipients will permit the emergence of pathogenic T cells that cause chronic GVHD. Lethally irradiated C3H/HeN (H-2k) recipients were reconstituted with T-cell–depleted bone marrow cells from major histocompatibility complex [MHC] class II–deficient (H2-Ab1−/−) B6 (H-2b) mice. These mice developed diseases that showed all of the clinical and histopathological features of human chronic GVHD. Thymectomy prevented chronic GVHD, thus confirming the causal association of the thymus. CD4+ T cells isolated from chronic GVHD mice were primarily donor reactive, and adoptive transfer of CD4+ T cells generated in these mice caused chronic GVHD in C3H/HeN mice in the presence of B6-derived antigen-presenting cells. Our results demonstrate for the first time that T cells that escape from negative thymic selection could cause chronic GVHD after allogeneic BMT. These results also suggest that self-reactivity of donor T cells plays a role in this chronic GVHD, and improvement in the thymic function may have a potential to decrease chronic GVHD.


Sign in / Sign up

Export Citation Format

Share Document