scholarly journals Memory T cells from minor histocompatibility antigen–vaccinated and virus-immune donors improve GVL and immune reconstitution

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 ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1332-1332
Author(s):  
Ning Li ◽  
Catherine Matte-Martone ◽  
Srividhya Venkatesan ◽  
Warren D. Shlomchik

Abstract Abstract 1332 Poster Board I-354 In allogeneic hematopoietic stem cell transplantation (alloSCT), donor T cells mediate graft-vs-leukemia (GVL) but also cause graft-vs.-host disease (GVHD). Our previous studies indicate that memory T cells (TM) induce less GVHD than do naïve T cells (TN). Therefore the selective infusion of memory T cells could improve immune reconstitution with less GVHD. However, human memory T cells have a more restricted TCR repertoire than do TN and therefore may be less effective in mediating GVL. One approach for addressing this concern would be to vaccinate donors against a single miHA expressed by host leukemic cells. Then the selective transfer of memory T cells could improve both GVL and immune reconstitution with less GVHD. Questions remain about how best to apply this strategy. For example, differences in target antigen distribution could affect the re-expansion of transferred memory cells and determine whether vaccination augments GVHD. Also, different types of memory cells could behave differently. To begin to optimize this approach in mouse models we chose as our target antigen the H-2Kb-restricted minor histocompatibility antigen (miHA) H60. We cloned the H60 peptide sequence (LTFNYRNL) into a genetic construct encoding the heavy chain of a monoclonal antibody against DEC205. Donor C3H.SW (H-2b, H60-) mice were vaccinated with a single injection of anti-DEC205-H60 plus an agonist antibody against CD40 (FGK45). By two months post vaccination, H60-reactive memory cells were a stable population that comprised approximately 4-8% of splenic and peripheral blood CD8 cells. Approximately 50% of H60-tetramer+ cells had central memory phenotype, which could be ideal as central memory T cells mount strong anti-tumor responses. Similar results were obtained by using a prime/boost approach with H60-pulsed DCs. To test their function, CD44+ memory CD8 cells from H60-vaccinated mice were sort-purified and transferred into recipient B6 mice congenic for H60 (B6.H60; expression hematopoietically restricted ). By day 7 post transplant, H60-specific CD8+ TM from vaccinated mice comprised 70-90% of total splenic and blood CD8 cells, as compared to 1-5% in recipients of TM from unvaccinated mice. In contrast to anti-H60 responses by naïve CD8 cells, expansion of H60-tetramer+ cells from H60-vaccinated mice did not require CD4 help. To test whether target antigen distribution affects the re-expansion of H60-reactive cells from H60-vaccinated mice, expansion was compared in B6.H60 and B6.H60<right arrow>B6.actH60 (ubiquitous expression of H60 driven by an actin promoter) bone marrow chimeras. Approximately 60% of splenic and lymph node CD8 cells in both recipient groups were H60-tetramer+, though there was a trend towards increased overall numbers of tetramer+ cells in B6.H60 recipients. We are currently testing the GVL potency of TM from H60-vaccinated against mouse models of chronic phase (CP-CML) and blast crisis chronic myelogenous leukemia (BC-CML). In an ongoing experiment, very low numbers of CD8+ TM from H60-vaccinated mice mediate potent GVL against CP-CML relative to CD8+ TM from unvaccinated donors. Small numbers of CD8+ TM cells from vaccinated mice also mediated GVL against BC-CML, which is typically GVL resistant. Future studies will better define the potency of TM from H60-vaccinated mice and test these cells against CP-CML and BC-CML that do or do not express H60 but are otherwise identical. GVHD effect will also be tested. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 191-191 ◽  
Author(s):  
Warren D. Shlomchik ◽  
Catherine C. Matte-Martone ◽  
D. Gary Gilliland ◽  
Lieping Chen

Abstract Donor T cells mediate a graft-versus-leukemia effect that is responsible for much of the efficacy of allogeneic hematopoietic stem cell transplantation (alloSCT) in treatment of hematologic malignancies. Chronic phase chronic myeloid leukemia (CP-CML) is the most GVL-sensitive neoplasm. Unfortunately, most other malignancies are relatively GVL-resistant. A striking example is blast crisis CML (BC-CML) which, although sharing its genetic etiology with CP-CML, is nearly refractory to alloimmune T cells. A detailed understanding of GVL-resistance has been hindered by the absence of GVL-sensitive and GVL-resistant murine leukemias that are similar to their human counterparts and are inducible on different mouse strains. In particular, generating gene-deficient leukemias is important for mechanistic experiments. To address these limitations, we have adopted murine models of CP-CML (mCP-CML) and BC-CML (mBC-CML) that share pathology and genetic etiology with their human counterparts. mCP-CML is generated by retroviral transduction of murine bone marrow (BM) with the bcr-abl fusion cDNA (p210), the defining genetic abnormality in human CP-CML. As is the case with human CP-CML, mCP-CML is extremely GVL-sensitive at least in part due to the redundant immune mechanisms sufficient for GVL (Matte et al, Blood 2004). mBC-CML is induced by the retroviral transduction of BM with both p210 and the fusion cDNA NUP98/HOXA9 (Dash, PNAS, 2002), a translocation found in human BC-CML and AML. Relative to mCP-CML, mBC-CML is GVL-resistant. In the MHC-matched C3H.SW→B6 (H-2b) strain pairing, 30–40% of recipients of 4–6 million donor CD4 or CD8 cells die from mBC-CML. This dose is nearly 10-fold higher than required for a similar survival from mCP-CML, even though recipients of mBC-CML and no donor T cells die nearly a week later than recipients of only mCP-CML. Having established that mBC-CML is GVL-resistant, we investigated mechanisms of T cell killing and the roles of donor and recipient antigen presenting cells (APCs). Direct T cell:mBC-CML cognate interactions were required as MHCI− and MHCII− mBC-CML cells (generated in β2microglobulin (β2M) or IAb β chain knockout (KO) BM) were completely insensitive to CD8 and CD4-mediated GVL, respectively. In contrast, neither CD8 nor CD4-mediated GVL was impaired against mBC-CML generated from TNF-receptor1/2 double KO or Faslpr BM. These are the same basic mechanisms of cytotoxicity we observed in GVL against mCP-CML. CD8-mediated GVL against mBC-CML required functional recipient APCs as we observed no GVL when recipients were MHCI− β2M KOs. As was the case with GVL against mCP-CML (Matte, N.Med. 2004), donor APCs were not required as GVL was equivalent in recipients of wild type and β2M KO C3H.SW donor BM. We observed no GVL in MHCII− recipients demonstrating that CD4-mediated GVL also requires functional recipient APCs. In sum, the basic rules of immunogenicity for GVL against mCP-CML and mBC-CML are similar, suggesting that other pathways are responsible for GVL-resistance. One possibility is differential sensitivity to TRAIL-mediated killing and we are currently generating TRAILR-deficient mBC-CML. Another candidate is PD-L1, a B7 family member that can suppress T cell responses. PD-L1 is highly expressed on mBC-CML relative to mCP-CML. We have already generated PD-L1-deficient mBC-CML and GVL experiments with it are underway.


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.


2006 ◽  
Vol 6 ◽  
pp. 246-253 ◽  
Author(s):  
Elizabeth Hexner

Much attention has focused on the immune recovery of donor T cells following hematopoietic stem cell transplantation (HSCT). Termed immune reconstitution, a better understanding of the dynamics of the functional recovery of immune cells following HSCT has important implications both for fighting infections and, in the allogeneic setting, for providing antitumor activity while controlling graft-vs.-host disease (GVHD). The immune cells involved in immune reconstitution include antigen-presenting cells, B lymphocytes, natural killer cells, and, in particular, T lymphocytes, the immune cell that will be the subject of this review. In addition, T cells can play an important role in the process of engraftment of hematopoietic stem cells. The evidence for a T cell tropic effect on hematopoietic engraftment is both direct and indirect, and comes from the clinic as well as the research lab. Animal models have provided useful clues, but the molecular mechanisms that govern the interaction between donor stem cells, donor T cells, the host immune system, and the stem cell niche remain obscure. This review will describe the current published clinical and basic evidence related to T cells and stem cell engraftment, and will identify future directions for translational research in this area.


Blood ◽  
2010 ◽  
Vol 116 (22) ◽  
pp. 4501-4511 ◽  
Author(s):  
Willemijn Hobo ◽  
Frans Maas ◽  
Niken Adisty ◽  
Theo de Witte ◽  
Nicolaas Schaap ◽  
...  

Tumor relapse after human leukocyte antigen–matched allogeneic stem cell transplantation (SCT) remains a serious problem, despite the long-term presence of minor histocompatibility antigen (MiHA)–specific memory T cells. Dendritic cell (DC)–based vaccination boosting MiHA-specific T-cell immunity is an appealing strategy to prevent or counteract tumor recurrence, but improvement is necessary to increase the clinical benefit. Here, we investigated whether knockdown of programmed death ligand 1 (PD-L1) and PD-L2 on monocyte-derived DCs results in improved T-cell activation. Electroporation of single siRNA sequences into immature DCs resulted in efficient, specific, and long-lasting knockdown of PD-L1 and PD-L2 expression. PD-L knockdown DCs strongly augmented interferon-γ and interleukin-2 production by stimulated T cells in an allogeneic mixed lymphocyte reaction, whereas no effect was observed on T-cell proliferation. Moreover, we demonstrated that PD-L gene silencing, especially combined PD-L1 and PD-L2 knockdown, resulted in improved proliferation and cytokine production of keyhole limpet hemocyanin–specific CD4+ T cells. Most importantly, PD-L knockdown DCs showed superior potential to expand MiHA-specific CD8+ effector and memory T cells from leukemia patients early after donor lymphocyte infusion and later during relapse. These data demonstrate that PD-L siRNA electroporated DCs are highly effective in enhancing T-cell proliferation and cytokine production, and are therefore attractive cells for improving the efficacy of DC vaccines in cancer patients.


2006 ◽  
Vol 135 (3) ◽  
pp. 413-414 ◽  
Author(s):  
Yoshiki Akatsuka ◽  
Hiroki Torikai ◽  
Yoshihiro Inamoto ◽  
Kunio Tsujimura ◽  
Yasuo Morishima ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3521-3521
Author(s):  
Mohammad Hossain ◽  
Andrew T Gewirtz ◽  
John D Roback ◽  
Edmund K. Waller

Abstract Bacground: Graft-vs-host disease (GvHD) is a major complication in allogeneic Hematopoietic Stem Cell Transplant (HSCT) recipients. Flagellin is a bacterial protein and a TLR5 agonist that showed diverse immunological responses in both human and animal including both activation of dendritic cells and immuno-suppression. We recently observed that prophylactic use of flagellin protected allogeneic HSCT recipient from GvHD without affecting host immune reconstitution. Acute GvHD has been reported to be mediated by allo-reactive CD62L+ T cells, and over 80% of murine naïve splenic CD4+ and CD8+ T cells express CD62L. In order to test the effect of flagellin on GvHD mediated by the CD62L+ CD4+ and CD62L+CD8+ donor T cells, we investigated clinical manifestation of GvHD as well as the in vivo expression of CD62L on donor T cells in flagellin treated versus control treated allogeneic HSCT recipients. Methods: We established a parent →F1 MHC major mismatched model (C57BL/6 → C57BL/6 × BALB/c) for allogeneic HSCT for which GvHD is the major complication. Recipient mice received 5 × 10^6 T cell depleted (TCD) bone marrow cells and 5×10^6 or 10×10^6 CFSE labeled donor splenocytes from naïve C57Bl/6 congenic donors. 50 μg flagellin per recipient was administered intraperitoneally 3 hours before irradiation and 24 hours after allogeneic HSCT (treated). CB6F1 recipients that received no flagellin (untreated) and recipients of syngeneic HSCT were used as control. Recipients were sacrificed on day 66+ transplant and the numbers of CD62L+ T cells and foxp3+CD4+CD25+ T cells were determined by FACS. Recipients of CFSE treated donor splenocytes were sacrificed on day 4 post HSCT, splenocytes were harvested and analyzed for CD62L expression on T cell subsets undergone in vivo cell division by Flow cytometry. 5 mice were used per group. Results: Flagellin treated recipients did not have GvHD and had no mortality. Untreated control recipients had 87% survival at 30 days post transplant and had signs of chronic GvHD. While total cell number and also donor spleen- and BM-derived CD4+ and CD8+ T cells per spleen in untreated recipients were significantly lower compared to flagellin treated recipients (p=0.0006) on day 66 post transplant, persistent of donor spleen-derived CD62L+CD4+ T cells and CD62L+CD8+ T cells per spleen were not significantly different (p=0.13 and p=0.07, respectively). Moreover, higher number of foxp3+CD25+CD4+ regulatory T cells were found in the spleen and thymus in treated recipients compared to untreated recipients. Within day 4 post transplant, the number of CD4+ T cells per spleen of treated and untreated recipients increased significantly compared to syngeneic recipients (p=0.001 and p=0.03, respectively). Although equivalent numbers of CD62L+CD4+ T cells were observed in both treated and untreated recipients (p=0.3), significantly increased numbers of CD62L+CD8+ T cells was found in treated recipients compare to untreated recipients (p=0.02). Moreover, significantly higher numbers of divided (far left CFSE staining population) CD62L+CD4+ and CD62L+CD8+ T cells were found in recipients of treated splenocytes within day 4 post transplant followed by down regulation of CD62L surface marker compared to untreated recipients (p=0.02 and p=0.01, respectively). Conclusion: Flagellin treated recipients had limited GvHD and had rapid increased divided CD4+CD62L+ T cells followed by CD62L-ve activated CD4+ T cells per spleen in treated recipients compared to untreated recipients may be one of the major affect mediated by flagellin. Flagellin-TLR5 receptor agonistic effect may reduce production of biological factor(s) essential to generate allo-reactive T cells or directly stimulate CD62L+CD4+ and CD62L+CD8+ T cells in different activation status other than allo-reactive T cells; maintain a balanced immune reconstitution in lymphoid organs by producing regulatory T cells through their thymus. Therefore, use of flagellin may be a novel therapeutic approach to treat blood cancer patients with allogeneic HSCT without GvHD and toxicity.


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 ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 45-45 ◽  
Author(s):  
Rosa Bacchetta ◽  
Claudia Sartirana ◽  
Barbarella Lucarelli ◽  
Patrick Miqueu ◽  
Maria Teresa Lupo Stanghellini ◽  
...  

Abstract Abstract 45 Background: Adoptive transfer of regulatory T cells is a potentially attractive alternative to conventional immunosuppressive therapy in allogeneic hematopoietic stem cell transplantation (HSCT) (Roncarolo MG., Nat Rev Immunol 2007). Among CD4+ T cells, the subset known as Type 1 regulatory T (Tr1) cells are induced in an antigen specific manner by interleukin-10 (IL-10) and suppress via production of high levels of IL-10 (Groux H., Nature 1997). The aim of this phase I/II study was to establish the safety and efficacy of a new cellular therapy with Tr1 cells in a non-randomized study. Patients and Methods: A cellular therapy protocol for the adoptive transfer of IL-10 induced alloantigen specific donor-derived Tr1 cells in patients transplanted with CD34+ selected cells from haploidentical donor, has been applied to patients with high risk hematopoietic malignancies (www.risetfp6.org). Donor T cells, anergized ex vivo toward host alloantigens, presented by monocytes (original protocol) or tolerogenic dendritic cells (modified protocol) as host antigen presenting cells, in the presence of IL-10, are infused post-transplant into the host (IL-10 DLI). The infusion is made in the absence of immunosuppression for graft-versus-host-disease (GVHD) prophylaxis, with the ultimate goal to provide immune reconstitution without severe GVHD. The infused donor T cells, at the dose of 105 CD3+ cells/kg or 3 × 105 CD3+ cells/kg, are anergic towards host-HLA antigens and contain precursors of host-specific Tr1 cells but, at the same time, comprise memory T cells able to respond to nominal and viral antigens. Results: Eighteen patients have been enrolled, sixteen received CD34+ selected cells from haploidentical donor after myeloablative conditioning. Twelve patients have been treated with IL-10 anergized cell therapy at day +30 post transplant, at the dose of 105 CD3+ cells/kg with the exception of two patients who received 3 × 105 CD3+ cells/kg. No severe immediate reactions post infusion were registered. Five patients died from infections by day +30 after Tregs cell infusion and two patients dropped out for graft rejection. Five patients achieved immune reconstitution at a median of 30,5 days (range 15–46 days) after IL-10 DLI, followed by progressive normalization of TCR repertoire, memory/naïve phenotype and T cell functions in vitro and in vivo. Acute GVHD grade III was observed in one patient who received 3 × 105 CD3+ cells/kg; GVHD grade II was observed in 4 patients who received 105 CD3+ cells/kg and were successfully immune reconstituted. The median follow-up of the IL-10 DLI treated patients is 980 days (range 291–1624); 4 patients are alive and disease free and they do not require immunosuppressive treatment. Conclusions: Cellular therapy with IL-10 anergized donor T cells has proven to be safe and feasible, and to sustain immune reconstitution associated with a reduced severity of GVHD and no occurrence of relapse. This trial represents the first step towards an extended use of Tr1 cells as adjuvant treatment in allogeneic HSCT. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document