Spontaneous Memory CD4+ T Cells Preserve Graft-Versus-Leukemia without Causing Graft-Versus-Host Disease.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 597-597
Author(s):  
Hong Zheng ◽  
Catherine C. Matte ◽  
Britt E. Anderson ◽  
Mark J. Shlomchik ◽  
Warren D. Shlomchik

Abstract We have recently shown in a major histocompatibility complex (MHC) compatible, multiple minor histocompatibility antigen mismatched CD4-dependent murine model of allogeneic stem cell transplantation (alloSCT) that donor spontaneous effector memory (EM) CD4 cells depleted of regulatory (Treg) CD25+ cells (CD4+CD44+CD62L−CD25−) do not cause graft-vs.-host disease (GVHD), but engraft and mount recall responses to a model antigen (Anderson et al., JCI, 2003). Thus EM cells might allow immune reconstitution with less GVHD. However, as alloSCT is most frequently used to treat malignant diseases, it is important to determine whether spontaneous EM cells can mediate graft-vs.-leukemia (GVL). Chen et al (Blood, 2004) found that memory T cells from donors immunized to host antigens can mediate GVL in an MHC disparate model; however Tregs were not depleted and whether spontaneous memory T cells can mediate GVL, which would be a likely clinical scenario, was not addressed. Here, we report that donor CD25− EM CD4 cells do not cause GVHD in the MHCII-disparate B6bm12→B6 model, but mediate GVL against a murine model of chronic phase CML (mCP-CML) induced by retroviral insertion into bone marrow (BM) cells of the bcr-abl (p210) fusion cDNA, the defining genetic abnormality in human CML (Matte et al., Blood, 2004). mCP-CML is manifest by a high WBC count and splenomegaly dominated by maturing myeloid cells. The retroviral construct also expresses the human nerve growth factor receptor (NGFR) which allows the tracking of transduced cells by FACS. We first addressed GVHD. Lethally irradiated B6 mice were reconstituted with T cell depleted B6bm12 BM with no T cells or with 5x105 CD4+CD25−CD62L+CD44− naïve (N) or CD4+CD25−CD62L−CD44+ EM B6bm12 T cells. In 2 independent experiments EM cells induced neither clinical nor histologic GVHD. In contrast, N CD4 cells induced GVHD manifest by weight loss (P<0.007; EM vs. N), hunched posture and ruffled fur. GVHD was confirmed histologically in the liver, intestine and skin (P<0.0002 for liver and bowel, P=0.0553 for skin; EM vs. N). To determine whether EM CD4 cells mediate GVL, lethally irradiated B6 mice were reconstituted with p210-infected B6 BM, donor B6bm12 BM, with or without a source of donor T cells (2.5x105 or 5x105 N CD4 cells or 106 EM CD4 cells). 5/5 mice that did not receive donor T cells died from mCP-CML between days 18-21 post BMT. N CD4 recipients (n=5 for both 2.5 and 5x105groups) cleared blood NGFR+ cells by day 22, but suffered from GVHD as expected. Strikingly, EM CD4 cells also mediated GVL as measured by improved survival and a 10-fold reduction in NGFR+ WBCs as compared to mice that received no T cells. Only 1/6 recipients died of mCP-CML; 5/6 recipients were still alive with a low WBC count at day 28 (P<0.0001; BM vs. BM/EM), although some NGFR+ cells were still present. Importantly, none of the EM recipients developed GVHD. EM cells were 99% pure and it is unlikely that less that 104 contaminating N cells were responsible for the GVL we observed. Taken together, our prior published work and this new data suggest that donor EM cells can contribute to both immune reconstitution and GVL and thus selective infusion of EM cells may be a successful strategy in clinical alloSCT.

Blood ◽  
2011 ◽  
Vol 118 (22) ◽  
pp. 5965-5976 ◽  
Author(s):  
Ning Li ◽  
Catherine Matte-Martone ◽  
Hong Zheng ◽  
Weiguo Cui ◽  
Srividhya Venkatesan ◽  
...  

AbstractDonor T cells contribute to the success of allogeneic hematopoietic stem cell transplantation (alloSCT). Alloreactive donor T cells attack leukemia cells, mediating the GVL effect. Donor T cells, including the memory T cells (TM) that are generated after infection, also promote immune reconstitution. Nonetheless, leukemia relapse and infection are major sources of treatment failure. Efforts to augment GVL and immune reconstitution have been limited by GVHD, the attack by donor T cells on host tissues. One approach to augmenting GVL has been to infuse ex vivo–generated T cells with defined specificities; however, this requires expertise that is not widely available. In the present study, we tested an alternative approach, adoptive immunotherapy with CD8+ TM from donors vaccinated against a single minor histocompatibility antigen (miHA) expressed by leukemia cells. Vaccination against the miHA H60 greatly augmented TM-mediated GVL against mouse chronic-phase (CP-CML) and blast crisis chronic myeloid leukemia (BC-CML). TM-mediated GVL was antigen specific and was optimal when H60 expression was hematopoietically restricted. Even when H60 was ubiquitous, donor H60 vaccination had a minimal impact on GVHD. TM from lymphocytic choriomeningitis virus (LCMV)–immune and H60-vaccinated donors augmented GVL and protected recipients from LCMV. These data establish a strategy for augmenting GVL and immune reconstitution without elaborate T-cell manipulation.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 579-579 ◽  
Author(s):  
Hong Zheng ◽  
Catherine C. Matte ◽  
Srividhya Venkatesan ◽  
Britt E. Anderson ◽  
Mark J. Shlomchik ◽  
...  

Abstract One of the major challenges in allogeneic stem cell transplantation (alloSCT) is to separate graft-versus-host-disease (GVHD) from graft-versus-leukemia (GVL). We and others have previously demonstrated, in both major histocompatibility complex (MHC)-compatible/multiple minor histocompatibility antigen-mismatched and MHC-mismatched murine models of alloSCT, that spontaneous effector memory (EM) CD4+T cells depleted of regulatory CD25+ cells (CD4+CD44+CD62L-CD25-) do not cause GVHD. We have also shown that these EM CD4+ T cells can mediate GVL against a model of murine chronic phase of CML (mCP-CML) induced via retroviral transduction of BM cells with the bcr-abl fusion cDNA without causing GVHD (Zheng, et al ASH meeting 2004). In the present study we analyzed the effector mechanisms of these EM CD4+ cells in the B6bm12 → B6 MHCII disparate bone marrow transplantation (BMT) model. First, we demonstrated that the GVL activity of both EM and naïve CD4+ T cells required cognate interactions with CML targets as GVL was ineffective against mCP-CML induced in bone marrow from B6.I-Ab−/− (MHCII−) mice. Recipients of MHCII− mCP-CML died from mCP-CML between day 15-20 post BMT, regardless of whether they received EM or naïve CD4+ cells or no T cells at all. In light of data in the same model that parenchymal MHCII expression is not required for GVHD (Teshima et al, 2002), these data demonstrate distinct mechanisms for the cytotoxicity by CD4+ cells in GVL and GVHD—direct in the former and indirect in the latter. To further investigate the specific mechanisms of T cell killing, we tested the effectiveness of EM CD4+ cells in eradicating mCP-CML induced in bone marrow cells from Fas−/− and TNFR1/R2−/− mice. Both EM and naïve CD4+ cells mediated GVL against these gene deficient leukemias that was similar to that against wild type mCP-CML. In summary, these results suggest that EM and naive CD4+ cells mediate GVL via direct cognate engagement with targets. Their killing, however, does not depend on either FasL or TNF-α which suggests a dominant role for perforin, TRAIL, or both. Interestingly, although the mechanisms of recognition and killing of mCP-CML by either naïve or EM CD4+ T cells are so far indistinguishable, whereas only the naïve cells cause GVHD. Whereas a number of investigators have been able to separate mechanisms of killing in GVHD vs. GVL, this is to our knowledge the first clear demonstration of a difference in the mechanism of recognition between GVHD and GVL.


Blood ◽  
1999 ◽  
Vol 94 (2) ◽  
pp. 390-400 ◽  
Author(s):  
Sylvie Brochu ◽  
Benjamin Rioux-Massé ◽  
Jean Roy ◽  
Denis-Claude Roy ◽  
Claude Perreault

After hematopoietic stem cell transplantation, the persistence and expansion of grafted mature postthymic T cells allow both transfer of donor immunologic memory and generation of a diverse T repertoire. This thymic-independent process, which is particularly important in humans, because most transplant recipients present severe thymus atrophy, is impaired by graft-versus-host disease (GVHD). The goal of this study was to decipher how GVHD influences the fate of grafted postthymic T cells. Two major findings emerged. First, we found that, after a brisk proliferation phase, alloreactive antihost T cells underwent a massive activation-induced cell death (AICD). For both CD4+ and CD8+ T cells, the Fas pathway was found to play a major role in this AICD: alloreactive T cells upregulated Fas and FasL, and AICD of antihost T cells was much decreased in the case of lpr (Fas-deficient) donors. Second, whereas non–host-reactive donor T cells neither upregulated Fas nor suffered apoptosis when transplanted alone, they showed increased membrane Fas expression and apoptosis when coinjected with host-reactive T cells. We conclude that GVHD-associated AICD of antihost T cells coupled with bystander lysis of grafted non–host-reactive T cells abrogate immune reconstitution by donor-derived postthymic T lymphocytes. Furthermore, we speculate that massive lymphoid apoptosis observed in the acute phase of GVHD might be responsible for the occurrence of autoimmunity in the chronic phase of GVHD.


Blood ◽  
2011 ◽  
Vol 118 (23) ◽  
pp. 6209-6219 ◽  
Author(s):  
Kathryn W. Juchem ◽  
Britt E. Anderson ◽  
Cuiling Zhang ◽  
Jennifer M. McNiff ◽  
Anthony J. Demetris ◽  
...  

Abstract Effector memory T cells (TEM) do not cause graft-versus-host disease (GVHD), though why this is has not been elucidated. To compare the fates of alloreactive naive (TN) or memory (TM) T cells, we developed a model of GVHD in which donor T cells express a transgene-encoded TCR specific for an antigenic peptide that is ubiquitously expressed in the recipient. Small numbers of naive TCR transgenic (Tg) T cells induced a robust syndrome of GVHD in transplanted recipients. We then used an established method to convert TCR Tg cells to TM and tested these for GVHD induction. This allowed us to control for the potentially different frequencies of alloreactive T cells among TN and TM, and to track fates of alloreactive T cells after transplantation. TEM caused minimal, transient GVHD whereas central memory T cells (TCM) caused potent GVHD. Surprisingly, TEM were not inert: they, engrafted, homed to target tissues, and proliferated extensively, but they produced less IFN-γ and their expansion in target tissues was limited at later time points, and local proliferation was reduced. Thus, cell-intrinsic properties independent of repertoire explain the impairment of TEM, which can initiate but cannot sustain expansion and tissue damage.


2015 ◽  
Vol 21 (7) ◽  
pp. 1215-1222 ◽  
Author(s):  
Pooja Khandelwal ◽  
Adam Lane ◽  
Vijaya Chaturvedi ◽  
Erika Owsley ◽  
Stella M. Davies ◽  
...  

2012 ◽  
Vol 18 (10) ◽  
pp. 1488-1499 ◽  
Author(s):  
Ping Zhang ◽  
Jieying Wu ◽  
Divino Deoliveira ◽  
Nelson J. Chao ◽  
Benny J. Chen

Blood ◽  
2006 ◽  
Vol 109 (7) ◽  
pp. 3115-3123 ◽  
Author(s):  
Benny J. Chen ◽  
Divino Deoliveira ◽  
Xiuyu Cui ◽  
Ngocdiep T. Le ◽  
Jessica Son ◽  
...  

Abstract Several groups, including our own, have independently demonstrated that effector memory T cells from non–alloantigen-primed donors do not cause graft-versus-host disease (GVHD). In the current study, we further investigated whether this approach could be extended to all memory T cells, and we studied the underlying mechanisms. Neither total memory T cells nor purified central memory T cells were able to induce GVHD. Memory T cells were at least 3-log less potent than bulk T cells in mediating GVHD. As expected, memory T cells failed to elicit cytotoxicity and proliferated poorly against alloantigens in standard 5-day mixed-lymphocyte cultures. However, the proliferative responses of memory T cells were more comparable with those of bulk and naive T cells when the culture time was shortened. Moreover, the frequencies of IL-2–secreting cells measured by 42-hour enzyme-linked immunosorbent spot (ELISPOT) assay were similar among naive, memory, and bulk T cells. These data indicated that memory T cells are able to respond to alloantigens initially but fail to develop to full potential. The abortive immune response, which was mediated by non–alloantigen-specific memory T cells in response to alloantigens, may explain why memory T cells from unprimed and non–alloantigen-primed donors could not induce GVHD.


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 ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 78-78
Author(s):  
Jifeng Zhang ◽  
Divino DeOliveira ◽  
Jessica Son ◽  
Xiuyu Cui ◽  
Nelson J. Chao ◽  
...  

Abstract Abstract 78 Allogeneic effector memory T Cells (TEM) are able to kill host radioresistant T cells and host-type tumor cells without causing GVHD, suggesting that allogeneic TEM may facilitate hematopoietic engraftment. We investigated the effects of allogeneic TEM on hematopoietic engraftment and immune reconstitution using a rejection model. TEM were prepared after negative selection of T cells followed by depletion of CD62L+ cells using magnetic beads. Lethally irradiated (8.5Gy) BALB/c mice were transplanted with minimum number (5×105) of C57BL/6 T cell depleted bone marrow (TCD BM) cells. Only 40% of TCD BM recipients survived more than 100 days after transplantation. In contrast, addition of donor TEM in the stem cell graft rescued 90% of the recipients (P<0.01, compared with TCD BM alone group). While all the recipients receiving TCD BM alone were mixed chimera (37.6±14.6% donor), all the recipients of additional donor TEM became full chimera (99.7±0.07% donor). These data suggest that facilitation of engraftment is likely due to donor TEM activation. Upon transfer into BALB/c recipients, donor TEM were activated, secreted multiple inflammatory cytokines, and expressed all known cytotoxic molecules. Facilitation of engraftment may also lead to enhanced immune reconstitution. Faster stem-cell-derived T cell generation was observed in allogeneic TEM recipients compared with the recipients of stem cells (c-Kit+ Thy1.1low Lin− Sca-1+) alone (P<0.01). Enhancement of stem-cell-derived T cell reconstitution was dramatically decreased in SCID recipients (lacking T and B cells) and could not be observed in NOG recipients (lacking T, B, and NK cells), further indicating that allogeneic TEM enhance immune reconstitution through depletion of host radioresistant immune cells. To confirm that allogeneic TEM enhance functional immune reconstitution, we injected BCL1 cells to the recipients at day +7 after transplantation. While 6 out of 8 C57BL/6 TCD BM alone recipients died of tumor, all allogeneic TEM recipients were tumor-free and survived more than 100 days after transplantation (P<0.001). We hypothesize that donor TEM can react to alloantigens initially but the alloresponses can not be sustained upon transfer to GVHD recipients, thus explaining the dichotomy of lack of GVHD with preservation of GVL. To test this hypothesis, we injected BCL1 cells into lethally irradiated BALB/c recipients of Rag-2−/−gC−/− BM and TEM from C57BL/6 mice at different time points (day +7, +14, +21, +28) after transplantation. When injected at day +7, allogeneic TEM recipients survived significantly longer than Rag-2−/−gC−/− BM only controls (medium survival time: 66 days vs. 26 days). When injected after day +14, the development of tumor and survival were comparable between Rag-2−/−gC−/− BM only and allogeneic TEM groups. These data demonstrate that allogeneic TEM enhance hematopoietic engraftment and immune reconstitution by reducing host resistance. The activation of allogeneic TEM does not lead to GVHD because the alloresponse induced by TEM can not be sustained in allogeneic stem cell recipients. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document