CD8+ but Not CD4+ T Cells Require Cognate Interactions with Target Tissues To Mediate GVHD across Only Minor H Antigens but CD4+ and CD8+ T Cells Both Require Direct Leukemic Contact for GVL.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 580-580 ◽  
Author(s):  
Warren D. Shlomchik ◽  
Catherine Matte ◽  
Jinli Liu ◽  
Dhanpat Jain ◽  
Jennifer McNiff

Abstract There has been debate as to whether CD4+ or CD8+ T cells require direct cognate interactions with their tissue targets to mediate GVHD. In one report, GVHD mediated by CD4 or CD8 cells did not require target tissue MHCI or MHCII expression in MHC-disparate models (Teshima, et al 2002). However, GVHD lethality may have been due to cytokines produced by high-frequency alloreactive T cells primed by MHC-disparate host APCs. In contrast, MHC-matched, multiple minor H antigen (miHA) disparate GVHD mediated by CD4 or CD8 cells has been reported to be reduced or absent in host → donor chimeric recipients of a second transplant with donor BM and T cells. In this case, host antigen was restricted to hematopoietic cells and the authors concluded that tissue miHA expression must be required (Korngold and Sprent, 1982; Jones et al, 2003). However these experiments did not address whether T cells directly interacted with MHC on target tissues. Rather they demonstrated that a continuing source of host antigen was essential. To resolve this we created bone marrow chimeras in which hematopoiesis was wild type (wt) while the parenchyma was either MHC I- (B6→B6 beta-2-microglobulin−/− (β2M−/−)) or MHC II− (B6→B6 IAb−/−) and used these chimeras as recipients in GVHD-inducing second transplants. We found that B6→B6β2M−/− chimeras were completely resistant clinically and pathologically to CD8-mediated GVHD induced by a second transplant with C3H.SW (H-2b) BM and CD8+ T cells whereas control B6→B6 chimeras developed severe disease. Thus, CD8 cells require direct cognate interactions with target host tissues to induce MHC-matched, miHA disparate GVHD. In contrast, B6→B6 IAb−/− chimeras developed similar clinical and histologic GVHD (liver, ear, skin and bowel) as did control B6→B6 chimeras when retransplanted with 129/J (H-2b) bone marrow and purified CD4+ T cells. Notably we observed lymphocytic infiltrates in involved organs. Therefore, CD4 cells can mediate tissue damage without directly recognizing alloantigen presented by MHCII on target epithelial cells. This suggests an indirect mechanism, perhaps mediated by T cell release of factors after stimulation in tissues by donor-derived APCs presenting host antigens. Alternatively, donor CD4 cells may activate miHA-bearing macrophages to release inflammatory mediators. To investigate the requirement for cognate recognition in GVL, we created murine CML via retroviral-mediated bcr-abl (p210) transduction of bone marrow from wt B6, B6 IAb−/− and B6 β2M−/− mice. Using the C3H.SW→B6 and 129→B6 GVHD models we found that both CD8-and CD4-mediated GVL requires leukemic expression of MHCI and MHCII, respectively. Thus both CD8-mediated GVHD and GVL required cognate T cell:target interactions. However, for CD4 cells only GVL, but not GVHD required target cell MHCII expression. This indicates that CD4-mediated GVL and GVHD have distinct mechanisms of action. Further understanding of these may provide insight in how to deliver GVL with less GVHD.

Blood ◽  
2001 ◽  
Vol 98 (2) ◽  
pp. 467-474 ◽  
Author(s):  
Patricia A. Taylor ◽  
Christopher J. Lees ◽  
Herman Waldmann ◽  
Randolph J. Noelle ◽  
Bruce R. Blazar

The promotion of alloengraftment in the absence of global immune suppression and multiorgan toxicity is a major goal of transplantation. It is demonstrated that the infusion of a single modest bone marrow dosage in 200 cGy-irradiated recipients treated with anti-CD154 (anti-CD40L) monoclonal antibody (mAb) resulted in chimerism levels of 48%. Reducing irradiation to 100 or 50 cGy permitted 24% and 10% chimerism, respectively. In contrast, pan–T-cell depletion resulted in only transient engraftment in 200 cGy-irradiated recipients. Host CD4+ cells were essential for alloengraftment as depletion of CD4+ cells abrogated engraftment in anti-CD154–treated recipients. Strikingly, the depletion of CD8+ cells did not further enhance engraftment in anti-CD154 mAb–treated recipients in a model in which rejection is mediated by both CD4+ and CD8+ T cells. However, anti-CD154 mAb did facilitate engraftment in a model in which only CD8+ T cells mediate rejection. Furthermore, CD154 deletional mice irradiated with 200 cGy irradiation were not tolerant of grafts, suggesting that engraftment promotion by anti-CD154 mAb may not simply be the result of CD154:CD40 blockade. Together, these data suggest that a CD4+regulatory T cell may be induced by anti-CD154 mAb. In contrast to anti-CD154 mAb, anti-B7 mAb did not promote donor engraftment. Additionally, the administration of either anti-CD28 mAb or anti-CD152 (anti–CTLA-4) mAb or the use of CD28 deletional recipients abrogated engraftment in anti-CD154 mAb–treated mice, suggesting that balanced CD28/CD152:B7 interactions are required for the engraftment-promoting capacity of anti-CD154 mAb. These data have important ramifications for the design of clinical nonmyeloablative regimens based on anti-CD154 mAb administration.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3605-3605 ◽  
Author(s):  
Haitham Abdelhakim ◽  
Luis M. Cortez ◽  
Meizhang Li ◽  
Mitchell Braun ◽  
Barry S. Skikne ◽  
...  

Background: Acute Myeloid Leukemia (AML) is an aggressive hematologic malignancy with known immune dyregulation. In addition to their capacity to rapidly divide, AML cells directly inhibit the activation and proliferation of immune cells in culture. Immunosuppressive features observed in the bone marrow of AML patients include upregulation of Tregs and production of immunosuppressive cytokines (e.g., TGFβ). Irradiating AML cells diminishes their immunosuppressive capacity while maintaining antigen presentation, leading to increased activation of T cells in co-culture. We subsequently identified the immune checkpoint LAG3 as an important mediator of AML-induced immunosuppression and LAG3 modulation as potential treatment strategy. Methods: Normal PBMC were isolated from healthy donors. PBMC were co-cultured with non-irradiated and irradiated (40 Gy) human AML cell lines (Kasumi1 (K1), THP1) separately at a 1:2 ratio. On day 3 of co-culture, immunophenotypic characterization of T cells was performed on a flow cytometer using the following surface markers: CD3, CD4, CD8, CD25, CD137, CD154, PD-1, TIM3, TIGIT, and LAG3 and intracellular IFNg and FOXP3. Supernatant from co-culture media were analyzed for cytokine (IL-2, IL-6, IL-10 & TGFβ) secretion by ELISA. CFSE-labeled AML cells were incubated with healthy donor PBMCs in the presence or absence of LAG3, then viability was measured by 7-ADD on flow cytometry. PBMCs were also isolated from AML patients' peripheral blood and mononuclear cells were isolated from their respective bone marrow samples. Primary AML cultures were established in RPMI complete media with 20% FBS. CFSE-7-ADD killing assay was conducted after incubation of AML cells with autologous PBMCs. Results: Healthy donor PBMC co-cultured with irradiated K1 AML cells showed higher intracellular IFNg expression (11.8% ± 3.1 v. 7% ± 3.3; n=7, P=0.012) and higher CD137 expression (9.3% ± 1.21 v. 5.7% ± 3.4; n=7, P<0.001) on CD8+ T cells, and higher CD154 expression on CD4+ cells (44.7% ± 20.3 v. 26.3% ± 14.2; n=5, P=0.002) when compared to the non-irradiated K1-PBMC co-cultures. There were fewer Tregs (CD4+ CD25+ FOXP3+) in the PBMC co-cultured with irradiated K1 cells (1.96% ± 0.37 v. 3.39% ± 0.58; n=4, P=0.03) compared to the non-irradiated K1-PBMC co-cultures. LAG3 expression on CD8+ T cells co-cultured with irradiated K1 was decreased (11.8% ± 2.4 v. 17.5% ± 2.5; n=4, P=0.002) compared to the PBMC co-cultured with non-irradiated K1 cells. No other changes in checkpoint expression on CD8+ T cells were observed. No changes were observed in MHCI or PDL1 expression on non-irradiated K1 AML cells before or after co-culture with PBMC. We observed similar findings with healthy donor PBMC co-cultured with a different AML cell line, THP1; CD137 expression was higher on CD8+ T cells (17.6% v. 6.5%; P=0.02, n=3). ELISA of the supernatant of culture media showed higher mean OD for secreted TGFβ in the non-irradiated AML co-cultures compared to the irradiated AML co-cultures at 6 hours (2.5 v. 2.0, P=0.03, n=3) and 72 hours (7.9 v. 5.3, P=0.04, n=3). Adding anti-LAG3 antibody (3DS223H; 100 µg/ml) to PBMC co-cultured with non-irradiated AML cells resulted in higher IFNg (16.3% v. 6.6%, P=0.01, n=4) and CD137 expression (6.5% v. 4.1%, p=.007, n=4) on CD8+ cells and fewer Tregs (1.7% v. 3.8%, P=.04, n=4) compared to no antibody added. Healthy donor PBMC (n=3) were incubated with CFSE labeled AML cells (K1 and THP1) separately at an effector:target ratio of 5:1. The addition of anti-LAG3 antibody lead to increased killing of both K1 and THP1 AML cells at 4 and 24 hours (Figure 1A). To eliminate the HLA mismatch effect, we incubated PBMC from AML patients with autologous AML cells in the presence or absence of anti-LAG3 (Figure 1B). MHC-I blocking (W6/32, 30 µg/ml) lead to inhibition of cell mediated killing in the presence of anti-LAG3 (Figure 1B). Conclusion: In this in vitro model, AML cells showed immunosuppressive features with decreased activation of CD8+ T cells, upregulation of Tregs, increased secretion of TGFβ and higher expression of LAG3 on CD8+ T cells. Antibody blocking of LAG3 mitigated this effect, resulting in increased activation of T cells, fewer T regs and improved MHC-I-mediated killing against AML cells. These results demonstrate that the immunosuppressive effects of AML cells can be modulated through inhibition of LAG-3, suggesting a potential strategy for future combination therapy in AML. Disclosures Lin: Jazz Pharmaceuticals: Honoraria; Pfizer: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2004-2004
Author(s):  
Sarah Morin-Zorman ◽  
Christian Wysocki ◽  
Edwina Kisanga ◽  
David Gonzalez ◽  
David Rothstein ◽  
...  

Abstract Graft-versus-host disease (GVHD) limits the broader application of allogeneic hematopoietic stem cell transplantation. In prior studies we defined roles for both host and donor-derived antigen presenting cells (APCs) in the activation of alloreactive donor T cells and promotion of GVHD. While initial T cell activation in GVHD occurs predominantly in secondary lymphoid organs (SLO), we have consistently observed MHCII+ donor-derived tissue APCs (t-APCs), including tissue dendritic cells (t-DCs) in histopathologic GVHD lesions, frequently adjacent to infiltrating T cells. We hypothesize that productive interactions occur between donor APCs and T cells in situ in GVHD target tissues, which propagate disease locally. We could not use knockout or APC depletion approaches to study T cell: t-APCs interactions as they impact APCs systemically and might therefore affect T cell stimulation in SLO. We therefore utilized two-photon intravital microscopy to analyze interactions between fluorescent donor CD4+ T cells and t-DCs in skin. 129 (H-2b) hosts were irradiated and reconstituted with B6 (H-2b) CD11c-YFP transgenic (Tg) Bone Marrow (BM) with B6 RFP Tg CD4 cells and nonfluorescent B6 CD8 cells. We imaged ear skin in GVHD mice 4 weeks later. In general CD4 cells co-localized with DCs. We observed CD4+ T cells that were highly motile and only in transient contact with DCs and others that made stable contact with DCs. To determine how much TCR: MHCII interactions drive sustained CD4+ T cell: DC interactions and arrest CD4+ T cell motility, mice were imaged and then injected with an MHCII blocking antibody (Ab; Y3P) with continued imaging of the same regions. After injection, T cell mean speed significantly increased and the proportion of T cells in stable contact with DCs decreased, indicating that transient disruption of TCR: MHCII is sufficient to restore motility to some T cells. In a second approach to assess the specificity of CD4+ T cell: t-DC interactions we transplanted 129 mice with B6 RFP+ CD4 cells, nonfluorescent CD8 cells and a mix of CD11c-YFP MHCII-/- and RFP wt BM or a mix of CD11c-YFP wt and RFP Tg MHCII-/- BM. We are currently comparing the motility of CD4 cells that make contact with MHCII+ as compared to MHCII- DCs, with the prediction that contact times will be shorter with the latter. Our data suggest that CD4+ T cells make cognate interactions with t-DCs in skin and we hypothesize that these interactions promote GVHD locally. Because the graft-versus-leukemia effect occurs primarily in bone marrow and secondary lymphoid tissues, targeting of tissue-infiltrating APCs could represent a unique strategy to ameliorate GVHD while preserving GVL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3551-3551
Author(s):  
Huihui Ma ◽  
Caisheng Lu ◽  
Judith Ziegler ◽  
Suzanne Lentzsch ◽  
Markus Y. Mapara

Abstract Abstract 3551 Poster Board III-488 We have previously demonstrated that activation of STAT1 and STAT3 in target tissue and secondary lymphoid organs belong to the earliest events during induction of GVHD. Using STAT1-gene-deficient (STAT1KO) mice we tested the role of donor STAT1 in fully MHC-mismatched (129Sv[H2b] to BALB/c [H2d]) and MHC-matched minor histocompatibility antigen (mHA)-mismatched strain combinations (129Sv[H2b] to B6[H2b]). GVHD was induced lethal irradiation and transplantation of allogeneic donor bone marrow cells and whole spleen cells. GVHD in the MHC-mismatched model is primarily CD4 dependent. Induction of GVHD was associated with activation of STAT1 and significant expansion of activated STAT1 expressing CD4+ and CD8+ T cells as assessed by analysis of STAT1 Tyr701 phosphorylation using phosphoflow staining. Using STAT1KO whole splenocytes we were able to show that lack of STAT1 significantly inhibited development of GVHD in both major and mHA mismatched recipients with significantly extended median survival times (MST) and lower GVHD morbidity. Protection against GVHD in recipients of STAT1KO splenocytes was associated with significant contraction of CD8+ T cells, but expansion of CD4 T cells on days +3 and +6 post-BMT in the MHC-mismatched setting. Most importantly, we observed a significant expansion of CD4+CD25+ FOXP3+ Treg cells in recipients of STAT1KO splenocytes. Lack of STAT1 in donor splenocytes resulted in a significantly attenuated and skewed systemic inflammatory response on day +6 post-BMT as demonstrated by significantly reduced IFN-g levels 508pg/ml vs 84.pg/ml (p<0.05), but significantly increased IL-4 (p=0.003), IL-5 (p=0.007) and IL-17 (p=0.03) levels. IL-6 levels were also increased with a trend towards statistical significance (p=0.08). In vitro studies demonstrated that STAT1KO CD8+ T cells produced much less IFN-g upon combined engagement of TCR and costimulation, but that this decrease in IFN-g secretion could be rescued if cells were simultaneously cultured under Th1 conditions (ie in the presence of IL-12 and anti-IL4 antibody). In contrast, lack of STAT1 completely inhibited the differentiation of naïve CD4+ T cells to IFN-g -producing cells upon TCR commitment and this capacity was also severely impaired under Th1 conditions. Furthermore, we observed a significantly reduced number of CXCR3−expressing CD4+ T cells in recipients of STAT1 KO splenocytes. In parallel to the afore-mentioned observations, tissue samples from BMT mice on day +3 and day +6 showed significantly less inflammation in liver and gut in recipients of STAT1 KO splenocytes compared to wild type cells. These data indicate that donor STAT1 is important for the induction of acute GVHD and that attenuation of GVHD in the absence of STAT1 involves expansion of Treg cells, perturbation of T cell polarization and subsequent reduced expression of the chemokine receptor CXCR3 on donor T cells leading to impaired target organ infiltration. Disclosures: Lentzsch: Celgene: Consultancy, Research Funding; cephalon: Consultancy, Research Funding. Mapara:Genzyme: Membership on an entity's Board of Directors or advisory committees; Resolvyx: Consultancy, Honoraria, Research Funding; Gentium: Stock Ownership.


Blood ◽  
2003 ◽  
Vol 102 (1) ◽  
pp. 180-183 ◽  
Author(s):  
Carl E. Mackewicz ◽  
Baikun Wang ◽  
Sunil Metkar ◽  
Matthew Richey ◽  
Christopher J. Froelich ◽  
...  

Abstract In HIV infection, CD8+ cells show cytotoxic and noncytotoxic anti-HIV activity. The latter function is mediated, at least in part, by a secreted antiviral protein, the CD8+ cell antiviral factor (CAF). Because antiviral effector molecules, such as perforin and granzymes, reside in the exocytic granules of CD8+ T cells, we examined the possibility that granules contain CAF-like activity. CD8+ cells from HIV-infected individuals showing strong CAF-mediated antiviral activity were induced to release their granule constituents into culture media. Within 1 hour of stimulation, high levels of granzyme B (a primary granule constituent) were found in the culture fluids of previously activated CD8+ cells. The same culture fluids contained no or very low amounts of CAF activity, as measured with HIV-infected CD4+ cells. Maximal levels of CAF activity were not observed until 5 or 7 days after stimulation, consistent with typical CAF production kinetics. In addition, extracts of granules purified from antiviral CD8+ cells did not show any CAF activity, whereas the cytoplasmic fraction of these cells showed substantial levels of antiviral activity. These findings suggest that CAF does not reside at appreciable levels in the exocytic granules of antiviral CD8+ T cells. (Blood. 2003;102: 180-183)


Blood ◽  
1989 ◽  
Vol 74 (6) ◽  
pp. 2270-2277 ◽  
Author(s):  
S Cayeux ◽  
S Meuer ◽  
A Pezzutto ◽  
M Korbling ◽  
R Haas ◽  
...  

Abstract T cells generated during a second round of ontogeny after autologous bone marrow transplantation (ABMT) represent a unique model of early T- cell ontogeny in an autologous situation. Since grafted bone marrows were pretreated in vitro with the cyclophosphamide derivative ASTA Z 7557, circulating T cells had to be regenerated from reinfused hematopoietic progenitor cells. The T-cell population derived from 25 patients post-ABMT was phenotypically characterized: an increase in CD8+ cells, a low percentage of CD4+ cells, and a median of 12% CD56+ (NKH1+) cells were found. When the T cells were stimulated with phytohemagglutinin (PHA) and phorbol myristate acetate (PMA), defective interleukin-2 (IL-2) secretion was observed. In addition, proliferative responses of the T cells after activation through the antigen-receptor- dependent CD3 pathway, through the CD2 dependent alternative T-cell pathway, and by the lectin PHA were investigated. Despite the presence of CD2, CD3, alpha/beta chains of the T-cell receptor, and CD25+ IL-2 surface receptors, abnormal proliferative responses were obtained even in the presence of exogeneous IL-2. In experiments where the T-cell population was separated into CD4+ cells and CD8+ cells, both the CD4- and CD8+ subsets were unable to respond to activating and proliferating signals. Thus, T cells at early stages of ontogeny not only possess an intrinsic defect in IL-2 synthesis but, in addition, were unable to express functional IL-2 receptors in response to mitogenic stimuli.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 5262-5262
Author(s):  
Emma Morris ◽  
Aristotle Tsallios ◽  
Gavin Bendle ◽  
Shao-an Xue ◽  
Hans Stauss

Abstract CD4 helper T cells play a critical role in the anti-tumour immune response. Cytokines secreted by CD4 T cells can have a direct effect on tumour cells and provide help for CTL priming and effector function. In this study we tested if it was possible to generate MHC class I-restricted helper T cells by retroviral TCR gene transfer into CD4 lymphocytes. Methods: We used a TCR (utilising V11) that recognises the influenza virus A nucleoprotein (NP366–379) peptide in the context of murine Db MHC class I. Murine splenocytes were isolated from C57BL/6 mice (H2b) and activated with conconavalin A and IL-7, and after 48 hours transduced with the pMX-TCR-IRES-TCR retroviral vector. The transduced splenocytes were then cultured in the presence of IL2 for a further 48 hours before staining with anti-murine CD4, CD8 and V11 antibodies and sorting into CD4+ V11+ and CD8+ V11+ populations. Sorted cells were expanded for a further 48–72 hours prior to functional assays. Functional Assays: Purified TCR-transduced (TCR-Td) CD8+ cells and purified TCR-Td CD4+ cells were tested for IFN secretion in response to dendritic cells (DCs) pulsed with NP peptide, an irrelevant peptide (pMDM100) or no peptide. Further experiments examined IFN secretion in response to peptide-loaded tumour cells (EL4 murine lymphoma cells) or transfected tumour cells expressing NP endogenously. Secretion of IFN was measured by ELISA. Results: (1) Antigen-specific IFN secretion was observed by both CD8+ (100% purity) and CD4+ cells (99.93% purity) expressing the class I-restricted TCR when incubated with peptide-loaded DCs. When tested with no peptide or irrelevant peptide, no IFN secretion was observed. The CD8+ cells were more sensitive, recognizing lower concentrations of peptide (10pM) than CD4+ cells (100pM). With peptide-coated EL4 tumour cells as stimulator cells, CD8+ cells showed a peptide-specific response. In contrast, the TCR-Td CD4+ cells were only able to elicit a weak peptide-specific response. Similarly, TCR-Td CD8+ cells were able to recognise NP transfected EL4 tumour cells (EL4NP68), whereas the CD4+ cells were unable to. However, the addition of syngeneic DCs restored the CD4+ cell response to NP transfected EL4 tumour cells to one equivalent to that seen with the TCR-Td CD8+ populations (Table 1). Summary: We have demonstrated that it is feasible to generate MHC class I-restricted CD4+ helper T cells, that are specific for peptide epitopes presented in the context of MHC class I. The CD4+ T cells can recognise antigen-expressing tumour cells in the presence of professional APC, such as DCs. The mechanism by which APC restore tumour recognition may involve trans-costimulation or cross presentation. The data suggest that class I-restricted CD4+ T cells may be able to contribute to enhanced anti-tumour immunity. αββββγγγγγβ γIFN Secretion (ng/ml) After Stimulation with DCs or Tumour Cells T Cell (Responder Cell) Stimulator Cell/s No Peptide NP (100nM) pMDM100 (100nM) Abbreviations: ND not done; DC, EL4 and EL4NP68 as indicated in text. TCR-Td CD8+ DCs 0.1 163.2 0.7 TCR-Td CD8+ EL4 0.1 19.9 0.2 TCR-Td CD8+ EL4NP68 16.6 ND ND TCR-Td CD8+ EL4NP68 + DCs 31.2 ND ND TCR-Td CD4+ DCs 0.1 163.9 0.2 TCR-Td CD4+ EL4 0.1 0.8 0.0 TCR-Td CD4+ EL4NP68 0.2 ND ND TCR-Td CD4+ EL4NP68 + DCs 25.3 ND ND


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 ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 454-454
Author(s):  
Jakub Tolar ◽  
Angela Panoskaltsis-Mortari ◽  
Anton K. Yerich ◽  
Ron T. McElmurry ◽  
Georg A. Hollander ◽  
...  

Abstract Gene correction is an attractive strategy for gene therapy since it allows the corrected gene to remain regulated within its native genome location. We have explored gene correction of murine severe combined immunodeficiency (SCID) with single-stranded DNA oligonucleotides (SSO). Murine SCID is characterized by severe T- and B-cell lymphopenia and is caused by a point mutation in the DNA protein kinase subunit (DNA-PK). To correct the mutant missense sequence (T to A substitution), a silent mutation was introduced by synthesizing the SSO non-transcribed sequence (45 bp) surrounding the site of the SCID mutation and replacing the T nucleotide with a C nucleotide to permit production of wild-type (wt) DNA-PK protein. Since the fetus is potentially an ideal permissive environment for gene correction due to the high proliferative rate of its tissues, SSO were injected in utero either directly into the liver of the fetus or transplacentally (via hydrodynamic infusion to the pregnant dam). E15/16 BALB/c-SCID recipients (N = 78) were injected with SSO (20 mcg/fetus). Twenty nine mice survived to term and, when evaluated by peripheral blood (PB) FACS at 15–30 weeks of life, 11 had significant phenotypic evidence of immune restoration defined as ≥ 2% CD4+ or CD8+ T cells: 6 had both CD4+ and CD8+ T cells, 2 had CD4+ cells only and 3 had CD8+ T cells only. The highest level of CD4+ cells seen was 9%, the highest level of CD8+ cells was 2% and both had TCR rearrangement and 27% and 15% genotypic correction of the mutated bp by quantitative pyrosequencing (PSQ) of DNA isolated from whole blood. Since placental membranes are permeable to some molecules, SSO were hydrodynamically delivered to pregnant BALB/c-SCID dams (100 mcg). Two of 8 evaluable mice injected on day E5/6 had significant numbers of T cells, one of which had 20% CD8+ with 3% CD4+ cells at 13 weeks of life, and PSQ showed a 13% correction rate. Sixteen offspring injected at E13/14 were analyzed: 2 had 4% and 5% CD4+ cells and the latter also had 6% of CD8+ cells with PSQ correction rates of 22% and 11%, respectively. Of 40 mice evaluated after transplacental injections at age E15/16, 9 had &gt;2% CD4+ or CD8+cells. The four with the highest T cell count had a genotypic correction of 12–25% of wt levels. Notably, littermates with no phenotypic correction had no evidence of gene correction at the DNA-PK mutation site. However, in all immune-restored animals that were analyzed for gene correction, (2/78 after in utero; 7/64 after transplacental delivery) an A to T rather than the anticipated A to C correction occurred. This is consistent with the hypothesis that SSO stimulated homologous recombination with a preferred utilization of the endogenous T rather than the exogenous C due to preferential pairing of two pyrimidines (A with T) than pyrimidine with purine (A with C). In summary, we show that SSO therapy for correction of DNA-PK mutation is possible when SSO are injected in utero at late gestation or are hydrodynamically delivered to the pregnant dam. These findings also suggest that while DNA homology around the mutation site is necessary for correction, the wt nucleotide is favored by the endogenous DNA repair pathway.


2018 ◽  
Author(s):  
Tyler R. McCaw ◽  
Mei Li ◽  
Dmytro Starenki ◽  
Sara J. Cooper ◽  
Selene Meza-Perez ◽  
...  

AbstractThe expression of major histocompatibility complex II (MHCII) on tumor cells correlates with survival and responsiveness to immunotherapy. However, the mechanisms underlying these observations are poorly defined. Using a murine breast tumor line, we tested how MHCII expression affected anti-tumor immunity. We found that MHCII-expressing tumors grew more slowly than controls and recruited more functional CD4+ and CD8+ T cells. Additionally, MHCII-expressing tumors contained more TCR clonotypes expanded to a larger degree than control tumors. Functional CD8+ T cells in tumors depended on CD4+ T cells. However, both CD4+ and CD8+ T cells eventually became exhausted, even in MHCII-expressing tumors. PD1 blockade had no impact on tumor growth, potentially because tumor cells poorly expressed PD-L1. These results suggest tumor cell expression of MHCII facilitates the local activation of CD4+ T cells and indirectly helps the activation and expansion of CD8+ T cells, but by itself, cannot prevent T cell exhaustion.PrécisThe expression of MHCII on tumor cells augments CD4 and CD8 T cell responses, expands the TCR repertoire and delays exhaustion. Hence, strategies to induce MHCII expression may be a powerful adjuvant to immunotherapeutic regimens of solid tumors.


Sign in / Sign up

Export Citation Format

Share Document