Adoptive Transfer of Natural Killer (NK) Cells to Prevent Gvhd and Enhance GVT Effects After Allogeneic Hematopoietic Cell Transplantation (HCT): The Timing of Donor NK Cell Infusions Critically Impacts Transplant Outcome.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 786-786
Author(s):  
Andreas Lundqvist ◽  
Hisayuki Yokoyama ◽  
Richard W. Childs

Abstract Abstract 786 Murine models have shown infusions of donor NK cells from hematopoietic stem cell transplant donors can prevent GVHD while simultaneously mediating a graft-vs-tumor (GVT) effect. Reduction of GVHD by alloreactive NK cells can be mediated indirectly through the eradication of host antigen presenting cells (APC) or directly by NK cell killing of alloreactive T cells. To assess how the timing of NK cell administration impacts these effects, donor NK cells from B10.d2 (H-2d) mice (1-2×106 cells) were infused into MHC-matched BALB/c (H-2d) recipients following lethal irradiation (950cGy) at one of the following time-points: 1) two days prior to a T cell replete (TR) HCT to target host APC or 2) at the time of a TR-HCT or 3) five days following a TR-HCT to target both host APC and in vivo primed alloreactive donor T cells. We also evaluated whether donor NK cells given on the day of a T cell deplete (TD) HCT could be used to prevent GVHD in mice that subsequently received a delayed donor lymphocyte infusion (DLI) given four days following HCT. Administration of donor NK cells two days prior to allogeneic TR-HCT did not result in a reduction of GVHD (figure). In contrast, the administration of NK cells given either at the time of a TR-HCT or five days following a TR-HCT reduced the incidence of GVHD (GVHD incidence 70% (p=0.2) and 40% (p=0.01) respectively) compared to controls that did not receive NK cells following a TR-HCT (GVHD incidence 100%). Similarly, mice that received a TD-HCT followed by a DLI on day four had a significantly lower incidence of GVHD when NK cells were infused on the day of transplantation compared to controls that did not receive donor NK cells (GVHD incidence 40% vs 100% respectively, p=0.01). Using bioluminescence imaging, we next investigated the impact of the timing of NK cell infusions on GVT effects in tumor bearing mice. Luciferase transduced RENCA tumors were injected intravenously into BALB/c mice ten days prior to allogeneic HCT. Tumor progression was significantly delayed in recipients of a TR-HCT when NK cells were infused on day five compared to when NK cells were infused two days prior to or at the same day of a TR-HCT (tumor doubling times 22.9 days, 7.6 days and 8.5 days respectively; p=0.03) (figure). This delay in tumor progression correlated with a significant improvement in overall survival; recipients of a TR-HCT given NK cells on day five had significantly longer survival compared to recipients of TR-HCT that did not receive NK cells (median survival 54±14 days vs 44±9; p=0.008), whereas infusion of NK cells prior to or concomitant with a TR-HCT did not significantly prolong survival (median survival 49±4 days and 50±12 days respectively; p=0.23). A comparable delay in tumor progression and longer survival was observed in mice that received a TD-HCT followed by a DLI on day four when NK cells were infused at the time of transplant compared to controls not receiving NK cells (tumor doubling time 19.7 days vs 7.2 days and survival 62±16 days vs. 50±9 days respectively; p=0.07). In conclusion, these results show that the timing of adoptive donor NK cell transfer has a critical impact on the ability of NK cells to prevent GVHD and enhance GVT effects following both T-cell replete and T-cell depleted allogeneic HCT. Following a TR-HCT, a delayed add-back of NK cells maximizes GVHD reducing and anti-tumor effects of adoptively transferred donor NK cells. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 61-61 ◽  
Author(s):  
Janelle A Olson ◽  
Dennis B Leveson-Gower ◽  
Jeanette Baker ◽  
Andreas Beilhack ◽  
Robert Negrin

Abstract Natural Killer (NK) cells have the ability to suppress graft-versus-host disease (GVHD) while inducing a graft-versus-tumor response (GVT) following murine allogeneic bone marrow transplantation (BMT). Prior studies have shown that NK cells suppress GVHD by eliminating recipient dendritic cells. To assess additional potential mechanisms of GVHD suppression we evaluated the impact of donor NK cells on GVHD-inducing donor T cells. Interleukin-2 activated allogeneic NK cells isolated from C57Bl6 (H-2b) or FVB (H-2q) animals were transplanted along with T cell-depleted bone marrow (TCD-BM) into lethally irradiated BALB/c (H-2d) mice, followed 2 days later by luciferase-expressing CD4+ and CD8+ conventional T cells (Tcon) from the same donor strain (Tcon+NK group). Control mice received TCD-BM on day 0, and luciferase-expressing T cells on day 2 after transplant (Tcon group). Bioluminescence imaging of Tcon+NK mice revealed a significantly lower T cell bioluminescent signal compared to Tcon mice (p=0.01 on day 5 post T cell transplant). We assessed the impact of NK cells on donor T cell activation and proliferation. CFSE proliferation analysis of alloreactive CD4 and CD8 T cells reisolated on day 4 post transplant showed a decreased percentage of dividing donor T cells in the Tcon+NK group. A reduced percentage of T cells in the Tcon+NK group as compared to the Tcon group expressed the T cell activation marker CD25 (11% and 49%, respectively, among donor CD4) and a reduced percentage of T cells from the Tcon+NK group down-regulated CD62L. Reisolated donor T cell numbers were reduced in the Tcon+NK mice compared to Tcon control mice. The impact of donor NK cells on donor Tcon function was addressed by intracellular cytokine staining. Fewer donor T cells reisolated from the spleen and lymph nodes of Tcon+NK mice produced the proinflammatory cytokines IFN-γ and IL-2 on day 3 after transplant. These observations can be explained by an NK cell-mediated induction of apoptosis in the donor Tcon. T cells reisolated from the peripheral lymph nodes of Tcon+NK animals at day 4 post transplant stained higher for the TUNEL apoptosis marker than those from Tcon mice (p<0.0001 for CD4 and CD8). To determine if this increase in apoptosis was due to a direct interaction between the donor T cells and NK cells, donor Tcon were reisolated from transplanted mice and used as targets in a killing assay. We demonstrated direct, specific lysis of these reisolated T cells by activated NK cells, both of which are from the donor strain and thus syngeneic to each other. Donor T cells reisolated from the lymph nodes of transplanted mice upregulated the NKG2D ligand Rae1γ as compared to naïve T cells, as shown by FACS. Further, use of an NKG2D-blocking antibody decreased the specific lysis of donor Tcon reisolated from the lymph nodes by activated NK cells in the in vitro killing assay, compared to an isotype control antibody (p=0.004). These data indicate that NK cells are causing direct, NKG2D-dependent lysis of alloreactive donor T cells in vivo during GVHD induction. Recent data from our laboratory has shown a lack of NKG2D ligand expression on GVHD target tissues in irradiated recipient mice. The tissue-specific expression of NKG2D ligands may explain why allogeneic NK cells do not cause GVHD but do impact donor T cells. We further investigated the ability of T cells in this model to elicit a GVT effect in the presence or absence of NK cells. Using a luciferase-expressing A20 lymphoma cell line, we demonstrated tumor clearance in groups receiving A20+Tcon and A20+Tcon+NK, as measured by A20 bioluminescence signal. Animals in the A20+Tcon+NK group had a lower peak bioluminescent signal than animals in the A20+Tcon group (p=0.03 on day 4 post T cell transplant), indicating an additive GVT effect of the T cells and NK cells. Thus, the T cells in this model are capable of mounting an effective GVT response. In addition to the established mechanism of NK cell-mediated elimination of recipient dendritic cells, we have demonstrated a novel mechanism of NK cell action in murine models of GVHD, whereby the donor NK cells inhibit T cell proliferation and activation and cause direct, NKG2D-mediated lysis of alloreactive donor T cells.


Blood ◽  
2010 ◽  
Vol 115 (21) ◽  
pp. 4293-4301 ◽  
Author(s):  
Janelle A. Olson ◽  
Dennis B. Leveson-Gower ◽  
Saar Gill ◽  
Jeanette Baker ◽  
Andreas Beilhack ◽  
...  

Abstract Natural killer (NK) cells suppress graft-versus-host disease (GVHD) without causing GVHD themselves. Our previous studies demonstrated that allogeneic T cells and NK cells traffic similarly after allogeneic bone marrow transplantation (BMT). We therefore investigated the impact of donor NK cells on donor alloreactive T cells in GVHD induction. Animals receiving donor NK and T cells showed improved survival and decreased GVHD score compared with controls receiving donor T cells alone. Donor T cells exhibited less proliferation, lower CD25 expression, and decreased interferon-γ (IFN-γ) production in the presence of NK cells. In vivo, we observed perforin- and Fas ligand (FasL)–mediated reduction of donor T cell proliferation and increased T cell apoptosis in the presence of NK cells. Further, activated NK cells mediated direct lysis of reisolated GVHD-inducing T cells in vitro. The graft-versus-tumor (GVT) effect was retained in the presence of donor NK cells. We demonstrate a novel mechanism of NK cell–mediated GVHD reduction whereby donor NK cells inhibit and lyse autologous donor T cells activated during the initiation of GVHD.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2563-2563
Author(s):  
Hisayuki Yokoyama ◽  
Andreas Lundqvist ◽  
Maria Berg ◽  
Muthalagu Ramanathan ◽  
Rebecca Lopez ◽  
...  

Abstract Animal models show infusions of donor NK cells given after allogeneic HCT can prevent GVHD while simultaneously mediating a graft-vs-tumor effect. However, it is unclear whether adoptively infused NK cells can mediate these beneficial effects in the presence of CSA, which is commonly given after HCT to prevent GVHD. In this study, we analyzed the in vitro effects of pharmacological concentrations of CSA on NK cell phenotype, cell proliferation, and tumor cytotoxicity. We also evaluated in vivo whether CSA administration would reduce the anti-tumor effects of adoptively infused NK cells in tumor bearing mice. PBMCs collected from healthy donors were labeled with CFSE then were stimulated in vitro with IL-2 for 7 days in the presence or absence of CSA (1000ng/ml). CFSE proliferation assays on fresh PBMC showed CSA inhibited IL-2 stimulated CD3+ T-cell proliferation more than CD3−/CD56+ NK cell proliferation (mean percentage inhibition of proliferation 49.4% vs. 22.2% for T cells and NK cells respectively; p<0.05). CD3−/CD56+ NK cells were then isolated from PBMCs of healthy donors and expanded in vitro with irradiated EBV-LCL and IL-2 for 10 days. In contrast to T-cells, CSA only minimally inhibited IL-2 induced proliferation of expanded NK cells (mean 9.5% inhibition of proliferation by CFSE staining). T cells and NK cells were next isolated from PBMCs and stimulated with either OKT3, PMA-Ionomycin (PI), or IL-2. A [3H] TdR uptake assay showed T cell proliferation was inhibited at a substantially higher level by CSA (mean stimulation index for OKT3: 0.03, for PI: 0.35, for IL-2: 0.55) compared to that of expanded NK cells (mean stimulation index for IL-2: 0.82, p<0.05). Furthermore, an ELISA assay showed CSA treatment reduced IL-2 induced secretion of INF-g by T cells more than expanded NK cells (mean reduction in INF-g secretion in T cells of 94.7 % vs. 36.5 % in NK cells, p<0.05). Compared to controls, culturing in vitro expanded NK cells in CSA did not alter surface expression of the activating receptors NKp30, NKp42, and NKG2D but did reduce surface expression of NKp44 and TRAIL (mean reduction in surface expression 36% and 36.3% respectively). Cytotoxic granule release assessed by CD107a staining was inhibited by CSA in CD8+ melanoma specific T cells co-cultured with melanoma cells (mean 12.2 % inhibition) in contrast to NK cells co-cultured with K562 cells where CSA increased CD107a expression a mean 29.7% (p<0.05). Furthermore, at a 20:1 E:T ratio, 51Cr cytotoxity assays showed CSA did not reduce the cytotoxicity of in vitro expanded NK cells against renal cell carcinoma (RCC) cells (58% mean lysis) compared to NK cells cultured in control media (55% mean; p=n.s.). In contrast, melanoma specific T-cell killing of tumor targets was significantly lower in CTL cultures containing CSA compared to control media (38.0% vs. 50.2% respectively P<0.05). Next, we assessed the impact of CSA administration on the anti-tumor effects of adoptive NK cell infusions in tumor bearing animals where syngeneic NK cell infusions following bortezomib treatment have been shown to delay tumor progression and prolong survival. BALB/c mice injected with 100,000 luciferase transfected RENCA tumor cells i.v. received 3 weekly treatments with the combination of bortezomib (5ug/mouse i.v.) and 2×106 syngeneic NK cells i.v. with or without daily administration of CSA (15mg/kg sc). Bioluminescence imaging in controls that did not receive CSA showed tumor growth was slower and survival was prolonged in mice receiving adoptive NK cell infusions (median survival 53 days) compared to mice that did not receive NK cells (median survival 30.2 days; p<0.05). CSA administration did not impair the anti-tumor effects of adoptive NK cell infusions; mice receiving CSA and adoptive NK cell infusions had similar tumor growth and survival as recipients of NK cells without CSA (median survival 47 vs. 53 days; p=n.s.). These results show that CSA is significantly more immunosuppressive to T cells compared to NK cells and provide evidence that the anti-tumor effects of adoptively infused in vitro expanded NK cells are maintained even in the presence CSA.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3731-3731
Author(s):  
Sarah Cooley ◽  
John E. Wagner ◽  
Claudio Brunstein ◽  
Mie Hagiwara ◽  
Giordi Orreggio ◽  
...  

Abstract Abstract 3731 Both T cell and natural killer (NK) cell reconstitution have been shown to affect clinical outcomes after hematopoietic stem cell transplantation (HSCT). Killer immunoglobulin-like receptor (KIR) interactions between alloreactive NK cells and their targets can prevent relapse, but may be dysregulated, especially after T cell replete HSCT. T cell recovery is also affected by the stem cell source and T cell content of the graft. To better understand the effects of various NK and T cell subsets we evaluated lymphocyte recovery in 304 adult patients who received either UCB (n=116), Sib (n=84) or Auto (n=94) HSCT for hematologic malignancies between 2003 and 2010 at the University of Minnesota. Peripheral blood mononuclear cells obtained at 3 months after HSCT were stained with CD56, CD3, CD4, CD8, and a cocktail of anti-NK cell KIR antibodies to determine the relative percentage of lymphocyte subsets by flow cytometry. The absolute lymphocyte count (ALC) was measured and used to calculate the absolute (Abs) number of T and NK cells and their subsets. ALC recovery at 3 months was similar among groups (UCB: 901.9 ± 74.5, Sib 890.2 ± 73.0 and Auto 1076.7± 69.4 cells/ul). Abs NK cells were highest in the UCB cohort (375.4 ± 24.9) vs. Sib (183.8 ± 15.4; p<0.0001) or Auto (160.7 ± 11.0; p<0.0001), as were the CD56bright and KIR+ subsets (data not shown). In contrast, Abs T cell recovery was lowest in the UCB group (300.8 ± 39.6) vs. Sib (578.5 ± 57.9; p<0.0001) or Auto (737.3 ± 60.4; p<0.0001). Accordingly, the lowest Abs CD4 count was in the UCB group (158.8 ± 14.7) vs. Sib (272.5 ± 23.5; p<0.0001) or Auto (223.6 ± 20.2; p=0.01), with a similar pattern observed for Abs CD8 counts. We then examined the effect of lymphocyte recovery on clinical outcomes. Multivariate models were constructed for each transplant group with relevant covariates (risk status, conditioning, sex, age, number of UCB units, CMV status, HLA matching (4/6, 5/6, or 6/6), and ABO matching). The most significant effect of lymphocyte recovery on outcomes was observed specifically in the UCB group, where higher ALC was associated with improved OS with a hazard ratio (HR) of 0.86 (95% CI 0.78–0.95) for each unit increase in ALC of 100 cells/ul (p <0.01). A similar trend was observed in Sib recipients but not in the Auto group. Specifically, increases in Abs T cells (HR 0.75 [95% CI 0.58–0.98]; p=0.034), Abs CD4 count (HR 0.63 [95% CI 0.42–0.95]; p=0.03), Abs CD8 count (HR 0.31 [95% CI 0.13–0.73]; p=0.01) and to a lesser extent Abs NK cells (HR 0.85 [95% CI 0.71–1.02]; p=0.085) were associated with improved OS. In the Sib cohort, higher Abs CD4 count was associated with improved OS (HR 0.43 [95% CI 0.20–0.92]; p=0.03) and decreased relapse (HR 0.37 [95% CI 0.37–1.00]; p=0.02), with no other factor having a significant impact. In the Auto group, only Abs NK (HR 0.40 [95% CI 0.16–0.99]; p=0.05) and to a lesser extent Abs KIR+ NK cells (HR 0.17 [95% CI 0.02–1.36]; p=0.09) were associated with improved OS but no other outcomes. The effect of Abs CD4 count on OS in all groups is shown in Figure 1 with survival stratified by quartiles. Figure 1: Figure 1:. In summary, rapid recovery of T cells predicts significantly better survival in patients undergoing UCB and Sib HSCT, while the NK cell effects are less pronounced. In contrast, NK cell effects predominate after Auto HCT. This suggests that more rapid T cell recovery is critical for survival and that defects in NK cell education after allogeneic HSCT may affect their function such that just increasing numbers may not be sufficient for clinical benefit. Appropriate modifications to immune suppression or the use of agents that promote T cell (IL-7) and/or NK cell (IL-15) function and survival may positively influence survival outcomes. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 8 (11) ◽  
pp. 1904 ◽  
Author(s):  
Helena Stabile ◽  
Paolo Nisti ◽  
Cinzia Fionda ◽  
Daria Pagliara ◽  
Stefania Gaspari ◽  
...  

T-cell-depleted (TCD) human leukocyte antigen (HLA) haploidentical (haplo) hematopoietic stem cell transplantation (HSCT) (TCD-haplo-HSCT) has had a huge impact on the treatment of many haematological diseases. The adoptive transfer of a titrated number of T cells genetically modified with a gene suicide can improve immune reconstitution and represents an interesting strategy to enhance the success of haplo-HSCT. Natural killer (NK) cells are the first donor-derived lymphocyte population to reconstitute following transplantation, and play a pivotal role in mediating graft-versus-leukaemia (GvL). We recently described a CD56lowCD16low NK cell subset that mediates both cytotoxic activity and cytokine production. Given the multifunctional properties of this subset, we studied its functional recovery in a cohort of children given α/βT-cell-depleted haplo-HSCT followed by the infusion of a titrated number of iCasp-9-modified T cells (iCasp-9 HSCT). The data obtained indicate that multifunctional CD56lowCD16low NK cell frequency is similar to that of healthy donors (HD) at all time points analysed, showing enrichment in the bone marrow (BM). Interestingly, with regard to functional acquisition, we identified two groups of patients, namely those whose NK cells did (responder) or did not (non responder) degranulate or produce cytokines. Moreover, in patients analysed for both functions, we observed that the acquisition of degranulation capacity was not associated with the ability to produce interferon-gamma (IFN-γ Intriguingly, we found a higher BM and peripheral blood (PB) frequency of iCas9 donor T cells only in patients characterized by the ability of CD56lowCD16low NK cells to degranulate. Collectively, these findings suggest that donor iCasp9-T lymphocytes do not have a significant influence on NK cell reconstitution, even if they may positively affect the acquisition of target-induced degranulation of CD56lowCD16low NK cells in the T-cell-depleted haplo-HSC transplanted patients.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3906-3906
Author(s):  
Lixia Sheng ◽  
Huarui Fu ◽  
Yongxian Hu ◽  
Shan Fu ◽  
Yamin Tan ◽  
...  

Abstract In murine models, donor natural killer cells(NK) exhibit immunoregulatory functions to alloreactive T cells during the initiation of acute graft versus host disease(aGVHD). The immunoregulatory role of NK cells in human aGVHD remains unclear. Here we compared the regulation of alloreactive donor T cell response by donor CD56+NK cells in 63 patients receiving allogeneic hematopoietic stem cell transplantation(allo-HSCT) and their donors. We found that NK cells from donors effectively suppressed T cell proliferation in response to Allo-DCs, showing cytotoxicity against activated proliferating T cells but not resting T cells. Subgroup of NK cells influenced the cytotoxicity against allo-reactive T cells, NKG2A-CD57+ NK cells degranulated to activated auto-T cells more potently than NKG2A+CD57- subgroup, suggesting NKG2A and CD57 expression patterns influenced NK cytotoxicity against activated T cells. When we analyzed the alteration in potential ligands for NK activating receptors on CD3+T cells during stimulated by allo-antigens, we found that activated T cells expressed higher levels of NKG2D-L(MICA/B,ULBP-1/ 2/ 4), DNAM1-L(PVR), and LFA-L(ICAM-1 and ICAM-2). Using neutralizing antibodies to block the interaction between NK receptors and correspondence ligands, we found that both activating receptor(LFA-1,NKG2D and DNAM-1) and inhibited receptor(NKG2A and TIM-3) participated this process. In the first 3 months post HSCT, reconstituted NK cells were mainly CD56bright and NKG2A+ CD57- subgroup, and percent of CD11b+CD27+ subgroup was significantly higher than in health donors, indicating relative immature subgroup predominated the early reconstituted NK cells after transplantation. By evaluating the dynamic restitution regularity of NK cell receptoires after Allo-HSCT, we found that the early reconstituted NK cells had a notably decreased surface expression of DNAM-1 and NKG2D compared with their corresponding donors. Furthermore, we compared the expression of receptors on CD56+NK cells from patients who developed aGVHD (group GVHD) with those without aGVHD (group non-GVHD) at 4 weeks after transplantation. Interestingly, we found that decreased expression of DNAM-1 and NKG2D and enhanced NKG2A expression are associated with aGVHD. When we assessed the expression of ligands for activating NK-cell receptors on activated T cells in aGVHD and non-aGVHD patients, we found that T cells in aGVHD patients expressed higher level of PVR(ligand for DNAM-1) and MICA/B(ligand for NKG2D) when compared with no-aGVHD patients or donors. To explore whether the subgroup alteration and reduced activating receptors expression on NK cells in aGVHD patients affected their capacity of GVHD regulation, we next examined NK-cell degranulation and cytotoxicity to allogeneic antigen activated T cells. The results demonstrated that the ability of donor NK cells to inhibit and lyse autologous activated T cells is impaired during human GVHD. Of clinical relevance, the tyrosine kinase inhibitor(TKI) dasatinib enhanced NK cytotoxicity towards activated T cells by up-regulating the expression of CD226 and NKG2D and enhancing the proportion of CD57+NKG2A- subgroup. This study demonstrates for the first time that the ability of donor NK cells to inhibit alloreactive T cells response is impaired during human GVHD and dasatinib may reinforced the GVHD-regulation function of NK cells, which potentially may provide an opportunity for therapeutic treatment of GVHD. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 ◽  
Author(s):  
Estéfani García-Ríos ◽  
Marcos Nuévalos ◽  
Francisco J. Mancebo ◽  
Pilar Pérez-Romero

During the last decade, many studies have demonstrated the role of CMV specific T-cell immune response on controlling CMV replication and dissemination. In fact, it is well established that transplanted patients lacking CMV-specific T-cell immunity have an increased occurrence of CMV replication episodes and CMV-related complications. In this context, the use of adoptive transfer of CMV-specific T-cells has been widely investigated and applied to Hematopoietic Stem Cell Transplant patients and may be useful as a therapeutic alternative, to reconstitute the CMV specific T-cell response and to control CMV viremia in patients receiving a transplantation. However, only few authors have explored the use of T-cell adoptive transfer in SOT recipients. We propose a novel review in which we provide an overview of the impact of using CMV-specific T-cell adoptive transfer on the control of CMV infection in SOT recipients, the different approaches to stimulate, isolate and expand CMV-specific T-cells developed over the years and a discussion of the possible use of CMV adoptive cellular therapy in this SOT population. Given the timeliness and importance of this topic, we believe that such an analysis will provide important insights into CMV infection and its treatment/prevention.


Author(s):  
Shannon L. McArdel ◽  
Anne-Sophie Dugast ◽  
Maegan E. Hoover ◽  
Arjun Bollampalli ◽  
Enping Hong ◽  
...  

AbstractRecombinant agonists that activate co-stimulatory and cytokine receptors have shown limited clinical anticancer utility, potentially due to narrow therapeutic windows, the need for coordinated activation of co-stimulatory and cytokine pathways and the failure of agonistic antibodies to recapitulate signaling by endogenous ligands. RTX-240 is a genetically engineered red blood cell expressing 4-1BBL and IL-15/IL-15Rα fusion (IL-15TP). RTX-240 is designed to potently and simultaneously stimulate the 4-1BB and IL-15 pathways, thereby activating and expanding T cells and NK cells, while potentially offering an improved safety profile through restricted biodistribution. We assessed the ability of RTX-240 to expand and activate T cells and NK cells and evaluated the in vivo efficacy, pharmacodynamics and tolerability using murine models. Treatment of PBMCs with RTX-240 induced T cell and NK cell activation and proliferation. In vivo studies using mRBC-240, a mouse surrogate for RTX-240, revealed biodistribution predominantly to the red pulp of the spleen, leading to CD8 + T cell and NK cell expansion. mRBC-240 was efficacious in a B16-F10 melanoma model and led to increased NK cell infiltration into the lungs. mRBC-240 significantly inhibited CT26 tumor growth, in association with an increase in tumor-infiltrating proliferating and cytotoxic CD8 + T cells. mRBC-240 was tolerated and showed no evidence of hepatic injury at the highest feasible dose, compared with a 4-1BB agonistic antibody. RTX-240 promotes T cell and NK cell activity in preclinical models and shows efficacy and an improved safety profile. Based on these data, RTX-240 is now being evaluated in a clinical trial.


2008 ◽  
Vol 205 (13) ◽  
pp. 2965-2973 ◽  
Author(s):  
Susan Gilfillan ◽  
Christopher J. Chan ◽  
Marina Cella ◽  
Nicole M. Haynes ◽  
Aaron S. Rapaport ◽  
...  

Natural killer (NK) cells and CD8 T cells require adhesion molecules for migration, activation, expansion, differentiation, and effector functions. DNAX accessory molecule 1 (DNAM-1), an adhesion molecule belonging to the immunoglobulin superfamily, promotes many of these functions in vitro. However, because NK cells and CD8 T cells express multiple adhesion molecules, it is unclear whether DNAM-1 has a unique function or is effectively redundant in vivo. To address this question, we generated mice lacking DNAM-1 and evaluated DNAM-1–deficient CD8 T cell and NK cell function in vitro and in vivo. Our results demonstrate that CD8 T cells require DNAM-1 for co-stimulation when recognizing antigen presented by nonprofessional antigen-presenting cells; in contrast, DNAM-1 is dispensable when dendritic cells present the antigen. Similarly, NK cells require DNAM-1 for the elimination of tumor cells that are comparatively resistant to NK cell–mediated cytotoxicity caused by the paucity of other NK cell–activating ligands. We conclude that DNAM-1 serves to extend the range of target cells that can activate CD8 T cell and NK cells and, hence, may be essential for immunosurveillance against tumors and/or viruses that evade recognition by other activating or accessory molecules.


2018 ◽  
Vol 93 (3) ◽  
Author(s):  
Abena K. R. Kwaa ◽  
Chloe A. G. Talana ◽  
Joel N. Blankson

ABSTRACTCurrent shock-and-kill strategies for the eradication of the HIV-1 reservoir have resulted in blips of viremia but not in a decrease in the size of the latent reservoir in patients on suppressive antiretroviral therapy (ART). This discrepancy could potentially be explained by an inability of the immune system to kill HIV-1-infected cells following the reversal of latency. Furthermore, some studies have suggested that certain latency-reversing agents (LRAs) may inhibit CD8+T cell and natural killer (NK) cell responses. In this study, we tested the hypothesis that alpha interferon (IFN-α) could improve the function of NK cells from chronic progressors (CP) on ART. We show here that IFN-α treatment enhanced cytokine secretion, polyfunctionality, degranulation, and the cytotoxic potential of NK cells from healthy donors (HD) and CP. We also show that this cytokine enhanced the viral suppressive capacity of NK cells from HD and elite controllers or suppressors. Furthermore, IFN-α enhanced global CP CD8+T cell cytokine responses and the suppressive capacity of ES CD8+T cells. Our data suggest that IFN-α treatment may potentially be used as an immunomodulatory agent in HIV-1 cure strategies.IMPORTANCEData suggest that HIV+individuals unable to control infection fail to do so due to impaired cytokine production and/cytotoxic effector cell function. Consequently, the success of cure agendas such as the shock-and-kill strategy will probably depend on enhancing patient effector cell function. In this regard, NK cells are of particular interest since they complement the function of CD8+T cells. Here, we demonstrate the ability of short-course alpha interferon (IFN-α) treatments to effectively enhance such effector functions in chronic progressor NK cells without inhibiting their general CD8+T cell function. These results point to the possibility of exploring such short-course IFN-α treatments for the enhancement of effector cell function in HIV+patients in future cure strategies.


Sign in / Sign up

Export Citation Format

Share Document