Human regulatory T cells kill tumor cells through granzyme-dependent cytotoxicity upon retargeting with a bispecific antibody.
Journal: 2015/July - Cancer immunology research
ISSN: 2326-6074
Abstract:
A major mechanism by which human regulatory T cells (T(regs)) have been shown to suppress and kill autologous immune cells is through the granzyme-perforin pathway. However, it is unknown whether T(regs) also possess the capacity to kill tumor cells using similar mechanisms. Bispecific antibodies (bscAbs) have emerged as a promising class of therapeutics that activate T cells against tumor antigens without the need for classical MHC-restricted TCR recognition. Here, we show that a bscAb targeting the tumor-specific mutation of the epidermal growth factor receptor, EGFRvIII, redirects human CD4(+)CD25(+)FoxP3(+) T(regs) to kill glioblastoma (GBM) cells. This activity was significantly abrogated by inhibitors of the granzyme-perforin pathway. Notably, analyses of human primary GBM also displayed diffuse infiltration of granzyme-expressing FoxP3(+) T cells. Together, these data suggest that despite their known suppressive functions, tumor-infiltrating T(regs) possess potent cytotoxic mechanisms that can be co-opted for efficient tumor cell lysis.
Relations:
Content
Citations
(15)
References
(24)
Grants
(101)
Diseases
(1)
Chemicals
(2)
Organisms
(1)
Processes
(1)
Anatomy
(2)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Cancer Immunol Res 1(3): 163

Human regulatory T cells kill tumor cells through granzyme-dependent cytotoxicity upon retargeting with a bispecific antibody

Introduction

Despite maximal surgical resection, dose-intensive radiation therapy and multimodal chemotherapy, glioblastoma (GBM) remains uniformly lethal with a median survival of less than 15 months (1). Novel therapies are desperately needed to improve the prognosis of this disease, and immunologic targeting of tumor-specific mutations has emerged as a promising approach (2).

A major barrier that has impeded translation of efficacious immunotherapy is the inability to overcome profound immunosuppression associated with malignant disease (3). Regulatory T cells (Tregs) in particular are thought to play a central role in tumor escape from immune-mediated rejection. One mechanism by which Tregs are known to suppress and even kill autologous immune cells is through the granzyme-perforin pathway (4, 5). Despite this well-characterized cytotoxic capacity, whether Tregs can co-opt cytotoxic mechanisms to kill tumor cells has yet to be evaluated.

T-cell-activating bispecific antibodies (bscAbs)—particularly those of the bispecific T-cell engager (BiTE) subclass—represent a new therapeutic strategy that has the potential to treat even bulky, invasive disease (6, 7). BiTEs are tandem single-chain molecules that possess dual specificity for tumor-associated surface antigens and the CD3 complex on T cells, which allows them to divalently bind and afford potent, specific target cell lysis (8). Because CD3 is universally expressed among T cells, BiTEs have the theoretical capacity to redirect and activate even Tregs that are elevated and present in tumors of patients with cancer.

Among the few known tumor-specific antigens, perhaps the most widely-characterized is the truncated mutant epidermal growth factor receptor variant type III (EGFRvIII). EGFRvIII is a constitutively activated tyrosine kinase that is frequently expressed on the surface of GBM and other common neoplasms but is completely absent from healthy tissues (9).

In this study, we demonstrate that an EGFRvIII-specific BiTE, bscEGFRvIIIxCD3 (7), successfully redirects highly-purified Tregs and activates them in the presence of tumors expressing EGFRvIII. Despite their known suppressive properties, purified CD4CD25CD127 T cells efficiently lysed EGFRvIII-expressing GBM in vitro upon redirection and activation with bscEGFRvIIIxCD3. This activity was found to be dependent on the granzyme-perforin pathway. Immunohistochemistry (IHC) analysis from human primary GBMs also displayed diffuse infiltration of activated, granzyme-producing FoxP3 cells, demonstrating that Tregs with potent effector functions may already be present in tumors even under natural conditions.

Previous efforts to enhance antitumor immunity via Treg depletion have been limited, in part due to an inability to efficiently eliminate suppressive cells that infiltrate tumor tissue (10). Alternatively, our data suggest that BiTEs can activate even suppressive Tregs to lyse tumor cells by redirecting their natural granzyme-mediated cytotoxic potential. These findings not only highlight a new mechanism by which BiTEs may circumvent certain aspects of Treg-mediated suppression, but also have broader implications with regard to the natural functional role of activated, tumor-infiltrating Tregs that express granzyme and perforin in the tumor microenvironment.

Materials and Methods

Tumor cell lines and reagents

The human glioma cell line U87MG and its subline U87MG.ΔEGFR, which expresses EGFRvIII, are described elsewhere and were kind gifts from Dr. Webster Cavenee (11). EGFRvIII expression was verified by flow cytometry using the L8A4 antibody (12). These cell lines were regularly confirmed to be Mycoplasma-free by nucleic acid hybridization assay (GEN-PROBE MTC-NI). Characterization, production and purification of bscEGFRvIIIxCD3 as well as control antibody constructs were performed as previously described by us (7). Antibodies to CD4 (RPA-T4), CD25 (M-A251), CD69 (L78), CD152 (BNI3), Granzyme A (CB9), Granzyme B (GB11) and Perforin (δG9) were purchased from BD Biosciences. Anti-FoxP3 antibody (PCH101) and the FoxP3 Staining Buffer Set were purchased from eBioscience and intracellular staining was performed according to manufacturer instructions. Antibodies against human FoxP3 (259D, BioLegend) and granzyme B (Cat No. ab4059) were used for immunohistochemical staining.

Treg isolation and preparation

All human samples were obtained at Duke University Medical Center from individuals who had given written, informed consent. Human PBMCs were prepared by density gradient centrifugation from buffy coats of healthy donor leukaphereses. Highly purified regulatory T cells were isolated from PBMCs by magnetic separation using the CD4CD25CD127 Regulatory T Cell Isolation Kit II and autoMACS Separator (Miltenyi Biotec) according to manufacturer instructions. Following isolation, purity was confirmed by flow cytometric analysis to be greater than 95% (CD25FoxP3). Where noted, purified Tregs were prepared with the Treg Expansion Kit (Miltenyi Biotec) at a MACSiBead Particle-to-Treg ratio of 4:1 according to manufacturer instructions, with recombinant interleukin 2 (rIL-2) at 500 U/mL, without rapamycin. Cells were verified for their suppressive ability in vitro prior to all cytotoxicity assays.

In vitro activation and functional assays

Activation, proliferation, cytokine secretion and measures of specific lysis were performed as previously described by us (7). In assays assessing activation and proliferation, freshly thawed and sorted lymphocytes were incubated in 96-well round-bottom plates with 1 × 10 target tumor cells and EGFRvIII BiTE (E:T ratio, 20:1; incubation time, 48 h at 37 °C; [bscEGFRvIIIxCD3], 10 μg/mL) in a total volume of 200 μL. Supernatant was removed after incubation and analyzed by cytometric bead array analysis according to manufacturer instructions (BD Biosciences). Cells were also harvested and assessed for surface and activation markers as above. In experiments assessing proliferation 1 μCi H-thymidine was added to each well of a 96-well round-bottom plate for an additional 24 h and cells were collected by a cell harvester. Counts were performed using a Wallac 1450 Microbeta Trilux Liquid Scintillation/Luminescence Counter (Perkin-Elmer). Similarly, cytotoxicity assays were performed by a standard chromium release assay as previously described by us (7). In brief, target cells were labeled with Cr and incubated with EGFRvIII BiTE and effector cells (E:T ratio, 20:1; incubation time, 18 h at 37 °C; [bscEGFRvIIIxCD3], 10 μg/mL) in a total volume of 200 μL. Following incubation, supernatants were removed and measured by gamma counter. Where noted, for inhibition of the granzyme-perforin axis, concanamycin A (CMA, 100 nM; Sigma), Granzyme B Inhibitor I (Z-AAD-CMK, 50 μM; Calbiochem), or ethylene glycol tetraacetic acid (EGTA, 4 mM; Calbiochem) was added to each well. Inhibitor concentrations were chosen based on previously published studies to establish dependence of cell-mediated cytotoxicity on perforin-granzyme activity (4, 13, 14). Prior to use, each inhibitor was found to have insignificant effects on the viability of Tregs following 18 h incubation at 37 °C as assessed by LIVE/DEAD Fixable Violet Dead Cell Stain Kit (Invitrogen). Blockade of FasL- and TRAIL-mediated apoptosis was carried out with antibody clones NOK-1 (25 μg/mL; BD Biosciences) and RIK-2 (25 μg/mL; BD Biosciences), respectively.

Immunohistochemistry

Tissues for immunohistochemical analysis were derived from human brain tumor biopsy material. Paraffin-embedded tissues were fixed, cut into 5 μm sections, mounted on glass slides and subjected to primary and secondary staining using the MACH 2 Double Stain 1 polymer detection kit (Biocare Medical) according to manufacturer instructions. Diaminobenzidine (DAB) and AminoEthyl Carbazole (AEC) were used to detect granzyme B and FoxP3, respectively.

Statistical analysis

Groups were compared using a two-sample two-tailed t-test and statistical significance was determined at a value of P < 0.05.

Tumor cell lines and reagents

The human glioma cell line U87MG and its subline U87MG.ΔEGFR, which expresses EGFRvIII, are described elsewhere and were kind gifts from Dr. Webster Cavenee (11). EGFRvIII expression was verified by flow cytometry using the L8A4 antibody (12). These cell lines were regularly confirmed to be Mycoplasma-free by nucleic acid hybridization assay (GEN-PROBE MTC-NI). Characterization, production and purification of bscEGFRvIIIxCD3 as well as control antibody constructs were performed as previously described by us (7). Antibodies to CD4 (RPA-T4), CD25 (M-A251), CD69 (L78), CD152 (BNI3), Granzyme A (CB9), Granzyme B (GB11) and Perforin (δG9) were purchased from BD Biosciences. Anti-FoxP3 antibody (PCH101) and the FoxP3 Staining Buffer Set were purchased from eBioscience and intracellular staining was performed according to manufacturer instructions. Antibodies against human FoxP3 (259D, BioLegend) and granzyme B (Cat No. ab4059) were used for immunohistochemical staining.

Treg isolation and preparation

All human samples were obtained at Duke University Medical Center from individuals who had given written, informed consent. Human PBMCs were prepared by density gradient centrifugation from buffy coats of healthy donor leukaphereses. Highly purified regulatory T cells were isolated from PBMCs by magnetic separation using the CD4CD25CD127 Regulatory T Cell Isolation Kit II and autoMACS Separator (Miltenyi Biotec) according to manufacturer instructions. Following isolation, purity was confirmed by flow cytometric analysis to be greater than 95% (CD25FoxP3). Where noted, purified Tregs were prepared with the Treg Expansion Kit (Miltenyi Biotec) at a MACSiBead Particle-to-Treg ratio of 4:1 according to manufacturer instructions, with recombinant interleukin 2 (rIL-2) at 500 U/mL, without rapamycin. Cells were verified for their suppressive ability in vitro prior to all cytotoxicity assays.

In vitro activation and functional assays

Activation, proliferation, cytokine secretion and measures of specific lysis were performed as previously described by us (7). In assays assessing activation and proliferation, freshly thawed and sorted lymphocytes were incubated in 96-well round-bottom plates with 1 × 10 target tumor cells and EGFRvIII BiTE (E:T ratio, 20:1; incubation time, 48 h at 37 °C; [bscEGFRvIIIxCD3], 10 μg/mL) in a total volume of 200 μL. Supernatant was removed after incubation and analyzed by cytometric bead array analysis according to manufacturer instructions (BD Biosciences). Cells were also harvested and assessed for surface and activation markers as above. In experiments assessing proliferation 1 μCi H-thymidine was added to each well of a 96-well round-bottom plate for an additional 24 h and cells were collected by a cell harvester. Counts were performed using a Wallac 1450 Microbeta Trilux Liquid Scintillation/Luminescence Counter (Perkin-Elmer). Similarly, cytotoxicity assays were performed by a standard chromium release assay as previously described by us (7). In brief, target cells were labeled with Cr and incubated with EGFRvIII BiTE and effector cells (E:T ratio, 20:1; incubation time, 18 h at 37 °C; [bscEGFRvIIIxCD3], 10 μg/mL) in a total volume of 200 μL. Following incubation, supernatants were removed and measured by gamma counter. Where noted, for inhibition of the granzyme-perforin axis, concanamycin A (CMA, 100 nM; Sigma), Granzyme B Inhibitor I (Z-AAD-CMK, 50 μM; Calbiochem), or ethylene glycol tetraacetic acid (EGTA, 4 mM; Calbiochem) was added to each well. Inhibitor concentrations were chosen based on previously published studies to establish dependence of cell-mediated cytotoxicity on perforin-granzyme activity (4, 13, 14). Prior to use, each inhibitor was found to have insignificant effects on the viability of Tregs following 18 h incubation at 37 °C as assessed by LIVE/DEAD Fixable Violet Dead Cell Stain Kit (Invitrogen). Blockade of FasL- and TRAIL-mediated apoptosis was carried out with antibody clones NOK-1 (25 μg/mL; BD Biosciences) and RIK-2 (25 μg/mL; BD Biosciences), respectively.

Immunohistochemistry

Tissues for immunohistochemical analysis were derived from human brain tumor biopsy material. Paraffin-embedded tissues were fixed, cut into 5 μm sections, mounted on glass slides and subjected to primary and secondary staining using the MACH 2 Double Stain 1 polymer detection kit (Biocare Medical) according to manufacturer instructions. Diaminobenzidine (DAB) and AminoEthyl Carbazole (AEC) were used to detect granzyme B and FoxP3, respectively.

Statistical analysis

Groups were compared using a two-sample two-tailed t-test and statistical significance was determined at a value of P < 0.05.

Results and Discussion

The EGFRvIII BiTE, bscEGFRvIIIxCD3, has previously been shown to redirect polyclonal T-cell populations against EGFRvIII-expressing gliomas (7); however, whether this BiTE also signals activation of FoxP3 Tregs—which are not only elevated among tumor-infiltrating lymphocytes (15) but also known to possess potent suppressive properties in the setting of GBM (16)—is currently unknown. To evaluate the impact of the EGFRvIII BiTE on human Tregs, we first isolated naturally occuring human Tregs according to phenotypic CD4CD25CD127 expression (17) and cultured these cells in the presence of EGFRvIII BiTE and glioma cells expressing EGFRvIII (U87MG.ΔEGFR). A control BiTE directed against a nonspecific antigen was also tested in order to exclude the possibility that the CD3-binding portion of bscEGFRvIIIxCD3 alone was sufficient for activation. After 24 hours, T cells were harvested and examined for surface and intracellular activation markers.

In line with previous literature, isolation of CD4CD25CD127 cells yielded a highly pure population of T cells expressing elevated levels of transcription factor FoxP3, consistent with the phenotypic definition of the Treg subset (18) (Figure 1A). Following coculture with target cells expressing EGFRvIII, flow cytometric analysis of sorted Tregs demonstrated that, in the presence of control BiTE, surface activation markers CD69 and CD25 remained unchanged. However, in wells cultured with EGFRvIII BiTE, Tregs exhibited significant upregulation of both early-activation associated glycoprotein, CD69, as well as IL-2 receptor alpha chain, CD25 (Figure 1B). These results were also consistent on repeat using sorted Tregs isolated from three separate healthy donors (P < 0.05, Figure 1C). Based on this evidence of activation, we also sought to determine functional characteristics of Tregs stimulated by the EGFRvIII BiTE in the presence of tumor. Importantly, we found that following BiTE-mediated activation, Tregs remained refractory both in terms of their ability to produce inflammatory cytokines (Figure 1D) and proliferate when compared to CD4CD25 helper T cells (Th) (Figure 1E). Together, these results indicate that although the EGFRvIII BiTE clearly has the capacity to activate Tregs, the functional outcome of this activation may be similar to what would otherwise be expected through standard TCR-restricted engagement of Tregsin vitro.

An external file that holds a picture, illustration, etc.
Object name is nihms525715f1.jpg

BscEGFRvIIIxCD3 activates highly-purified Tregs but does not reverse defects in cytokine secretion and proliferation in response to stimulation. (A) Representative flow cytometric analysis of purified CD4CD25CD127 Tregs demonstrates greater than 97% purity as determined by FoxP3 and CD25 phenotypic markers. (B) Purified Tregs express elevated levels of activation markers CD69 and CD25 in response to bscEGFRvIIIxCD3 specifically in the presence of EGFRvIII-expressing tumors when compared to cells incubated with non-specific, control BiTE. These activity was consistent and statistically significant among lymphocyte donors from three separate individuals (C). (D) Supernatants from wells containing U87MG.ΔEGFR, bscEGFRvIIIxCD3 and Tregs contained significantly lower levels inflammatory cytokines compared to wells in which responder cells consisted of purified CD4CD25 helper T cells (Th). (E) Proliferation of Tregs and Th in response to bscEGFRvIIIxCD3 and solid phase EGFRvIII as measured by H-thymidine incorporation demonstrates that proliferative defects in the Treg compartment persist following activation with bscEGFRvIIIxCD3. Statistical analysis in (D) and (E) were performed in triplicate wells with lymphocytes from a single donor and all experiments were repeated twice. Horizontal bars represent a statistical significance of P < 0.05.

Despite their lack of classical proinflammatory mechaisms, it is well-known that even naturally activated Tregs possess sigificant cytotoxic capacity, largely through the production of high levels of granzyme B expression (19). In fact, the perforin-granzyme axis is a major mechanism by which Tregs are known to suppress immune responses, through the direct killing of effector T cells (4, 13). Given this well-characterized cytotoxic function, we hypothesized that the EGFRvIII BiTE might actually have the ability to coopt the perforin-granzyme axis in Tregs to kill cells other than lymphocytes, resulting in the redirected lysis of even tumor cells expressing a target antigen of interest, in this case EGFRvIII.

To test this hypothesis, we first sought to evaluate the impact of bscEGFRvIIIxCD3-mediated activation on the expression of perforin and granzymes among Tregs in the presence of target tumor cells expressing EGFRvIII. Using flow cytometric analysis of CD4CD25FoxP3 cells, we found that Tregs cultured with control BiTE and target tumor cells expressed only low levels of perforin, granzyme A (GrA) and granzyme B (GrB). However, Tregs activated in the presence of bscEGFRvIIIxCD3 exhibited significant upregulation of these cytotoxic molecules (Figure 2A). These results were repeated with consistent and significant upregulation of GrA, GrB and perforin in samples isolated from three separate healthy donors (P < 0.05, Figure 2B). Moreover, when analyzed for surface markers, back-gating anlysis of GrB-positive cells (dots) displayed a distinct population with dual positivity for Treg phenotypic markers, FoxP3 and CD25 (Figure 2C). The level of FoxP3 expression did not appear to change significantly when gating for Tregs with either high or low levels of GrB (data not shown). Although the impact of BiTEs on Treg expression of GrB has not been previously described, these results are consistent with previous studies which have demonstrated substantial GrB upregulation in Tregs on stimulation with anti-CD3 antibody (5), which in theory provides the same signal delivered through the T-cell engaging arm of bscEGFRvIIIxCD3.

An external file that holds a picture, illustration, etc.
Object name is nihms525715f2.jpg

BscEGFRvIIIxCD3 activates Tregs to elevate expression of granzymes and perforin specifically in the presence of tumor cells expressing EGFRvIII. (A) Human PBMCs were incubated in the presence of U87MG.ΔEGFR and control BiTE (left) or EGFRvIII BiTE (right), harvested and stained for flow cytometric analysis. CD4 cells were isolated and gated for high CD25 and FoxP3 expression and then analyzed for Perf, GrA and GrB expression with positivity determined by isotype control. Plots are representative of at least three repeated experiments. (B) Percent positive cells expressing Perf, GrA or GrB were determined to be significantly elevated in the presence of EGFRvIII BiTE over non-specific control BiTE across lymphocytes from three separate healthy donors. (C) Flow cytometric plot of backgated GrB-positive cells from Figure 2A corresponds to a discrete population (dots) with dual positivity for CD25 and FoxP3. All experiments were repeated twice. Horizontal bars represent a statistical significance of P < 0.05 between groups of three donors each defined by the presence of either EGFRvIII-specific or control BiTE.

Due to the significant upregulation of the perforin-granzyme pathway in BiTE-activated Tregs, we next decided to test whether the EGFRvIII BiTE could redirect the cytotoxic payload expressed in Tregs to lyse target tumor cells expressing EGFRvIII. CD4CD25CD127 T cells were expanded between 4–7 days to provide sufficent numbers for in vitro cytotoxicity, a period of time known to preseve both suppressive capacity and FoxP3 expression (20). Under these conditions, sorted Tregs efficiently lysed target cells expressing EGFRvIII when redirected by the EGFRvIII-specific BiTE. Importantly, this effect was not detected in the presence of a nonspecific control bscAb or Tregs alone (Figure 3A). Moreover, redirected Treg-mediated lysis was shown to be actually dependent on the perforin-granzyme pathway, since cytotoxicity was not impacted by TRAIL or FasL blockade, but significantly abrogated in the presence of previously-characterized inhibitors of perforin- and granzyme-mediated cytotoxicity (Figure 3B). Adding clinical relevance to these findings, infiltrating FoxP3 Tregs in human glioma samples were actually found to coexpress detectable levels of granzyme B by immunohistochemical analysis (Figure 3C). Consistent with what has been reported elsewhere, granzyme B expression appeared to be largely cytoplasmic while subcellular localization of FoxP3 was exhibited prominently in the nucleus (21). Overall, these data suggest that activated Tregs with potent effector functions may be present within GBMs, and could potentially serve as effector cells upon infusion with T-cell-engaging bscAb therapies in vivo.

An external file that holds a picture, illustration, etc.
Object name is nihms525715f3.jpg

Tregs expressing GrB are present in human glioma and possess cytotoxic activity against EGFRvIII-expressing tumor in the presence of EGFRvIII BiTE. (A) Upon redirection with bscEGFRvIIIxCD3, but not a non-specific control bscAb (Control), activated Tregs demonstrate enhanced lysis against EGFRvIII-expressing tumor (Treg:target, 20:1; incubation time 18 hours; [BiTE] 10 μg/mL). (B) Specific lysis against target tumor cells expressing EGFRvIII is not significantly inhibited by blockade of Fas ligand- and TRAIL-mediated apoptosis but is significantly abrogated by partial inhibitors of the granzyme-perforin pathway, Z-AAD-CMK, EGTA and CMA. Pairwise comparisons with respect to Treg, BiTE and inhibitors of the granzyme-perforin pathway were made. All tests were performed in triplicate wells and independently repeated. Horizontal bars represent a statistical significance between compared groups of P < 0.05. (C) IHC analysis of human GBM shows diffuse infiltration of FoxP3 Tregs (AEC) expressing detectable levels of granzyme B (DAB).

In light of recent findings demonstrating that Tregs may actually be required for the priming of high-avidity CD8 T-cell responses (22), innovative methods to reappropriate the Treg compartment without ablating these cells completely could provide an attractive alternative to currently available depletive strategies. As such, we have provided evidence and a mechanism by which Tregs might be redirected to kill tumor cells through engagement with a bscAb. One previous study has explored the impact of bscAbs on Tregs, and demonstrated that bscAb-mediated activation of Tregs suppresses effector cell proliferation and abrogates antitumor efficacy (23). However, the direct effects of bscAb-redirected Tregs on target tumor cells were not discussed. Furthermore, because their work employed the use of rapamycin to expand Tregsex vivo—an additive which is known to completely suppress GrB expression in Tregs (19)—it may ultimately not be feasible to directly compare the results of our studies. One limitation of our work is the use of a GBM cell line that may ellicit low-level allogeneic responses and thus a permissive effect on the mechanisms of Treg activation explored here. Further studies will be necessary to explore the degree to which this is the case, likely through the use of syngeneic mouse models or autologous human tumors.

In the current study, redirected cytotoxicity was dependent on the presence of bscEGFRvIIIxCD3; though, it is reasonable to suspect that similar results might be observed upon even endogenous TCR-specific engagement of granzyme-producing Tregs present in the tumor microenvironment. Supporting this, previous studies have shown that, in general, BiTE-mediated antitumor activity is indeed contact-mediated, and has the ability to induce cytolytic synapses that are indistinguishable from those naturally occurring between effector TCR and MHC (24). Thus, while our findings here were demonstrated using a single test molecule against EGFRvIII, we expect that BiTEs targeting other tumor antigens may likewise mediate similar effects. Overall, this work advances our understanding of potential functions for suppressive immune cells in the tumor microenvironment; however, the true functional relevance of Treg-mediated tumor killing remains unknown. The implications of these findings will need to be explored in future studies across broader areas of autoimmunity and immunotherapy where Treg biology is relevant.

Acknowledgments

Financial Support

This work was supported by grants from the National Institutes of Health 5R01-CA135272-04 (J.H. Sampson), 5P50-NS020023-29 (D.D. Bigner and J.H. Sampson), 3R25-NS065731-03S1 (J.H. Sampson), 1F30CA177152-01 (B.D. Choi) as well as grants from the Pediatric Brain Tumor Foundation (D.D. Bigner and J.H. Sampson), Ben and Catherine Ivy Foundation (J.H. Sampson), Duke Cancer Institute (J.H. Sampson and B.D. Choi), Cancer Research Institute (B.D. Choi), and Miami Brain Tumor Coalition (B.D. Choi).

Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC 27710
Department of Pathology, Duke University Medical Center, Durham, NC 27710
Department of Biomedical Engineering, Duke University, Durham, NC 27708
Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710
The Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, NC 27710
Corresponding author: John H. Sampson; Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery; Duke University Medical Center; Box 3050; Durham, NC 27710. Telephone: (919) 684-9043; Fax: 919-419-1741; ude.ekud@nospmas.nhoj

Abstract

A major mechanism by which human regulatory T cells (Tregs) have been shown to suppress and kill autologous immune cells is through the granzyme-perforin pathway. However, it is unknown whether Tregs also possess the capacity to kill tumor cells using similar mechanisms. Bispecific antibodies (bscAbs) have emerged as a promising class of therapeutics that activate T cells against tumor antigens without the need for classical MHC-restricted TCR recognition. Here, we show that a bscAb targeting the tumor-specific mutation of the epidermal growth factor receptor, EGFRvIII, redirects human CD4CD25FoxP3 Tregs to kill glioblastoma (GBM) cells. This activity was significantly abrogated by inhibitors of the granzyme-perforin pathway. Notably, analyses of human primary GBM also displayed diffuse infiltration of granzyme-expressing FoxP3 T cells. Together, these data suggest that despite their known suppressive functions, tumor-infiltrating Tregs possess potent cytotoxic mechanisms that can be co-opted for efficient tumor cell lysis.

Keywords: Regulatory T Cells, Granzymes, Bispecific Antibodies, Glioblastoma, Immunomodulation
Abstract

Footnotes

Conflict of Interest Statement: The authors B.D.C., D.D.B. and J.H.S. have a patent pending for EGFRvIII as a tumor-specific target for bispecific antibody therapy.

Footnotes

References

  • 1. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, et al Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10:459–66.[PubMed][Google Scholar]
  • 2. Mellman I, Coukos G, Dranoff GCancer immunotherapy comes of age. Nature. 2011;480:480–9.[Google Scholar]
  • 3. Zou WImmunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. 2005;5:263–74.[PubMed][Google Scholar]
  • 4. Grossman WJ, Verbsky JW, Barchet W, Colonna M, Atkinson JP, Ley TJHuman T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity. 2004;21:589–601.[PubMed][Google Scholar]
  • 5. Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJCutting edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J Immunol. 2005;174:1783–6.[PubMed][Google Scholar]
  • 6. Bargou R, Leo E, Zugmaier G, Klinger M, Goebeler M, Knop S, et al Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 2008;321:974–7.[PubMed][Google Scholar]
  • 7. Choi BD, Kuan CT, Cai M, Archer GE, Mitchell DA, Gedeon PC, et al Systemic administration of a bispecific antibody targeting EGFRvIII successfully treats intracerebral glioma. Proc Natl Acad Sci U S A. 2013;110:270–5.[Google Scholar]
  • 8. Choi BD, Cai M, Bigner DD, Mehta AI, Kuan CT, Sampson JHBispecific antibodies engage T cells for antitumor immunotherapy. Expert Opin Biol Ther. 2011;11:843–53.[PubMed][Google Scholar]
  • 9. Choi BD, Archer GE, Mitchell DA, Heimberger AB, McLendon RE, Bigner DD, et al EGFRvIII-targeted vaccination therapy of malignant glioma. Brain Pathol. 2009;19:713–23.[Google Scholar]
  • 10. El Andaloussi A, Han Y, Lesniak MSProlongation of survival following depletion of CD4+CD25+ regulatory T cells in mice with experimental brain tumors. J Neurosurg. 2006;105:430–7.[PubMed][Google Scholar]
  • 11. Nishikawa R, Ji XD, Harmon RC, Lazar CS, Gill GN, Cavenee WK, et al A mutant epidermal growth factor receptor common in human glioma confers enhanced tumorigenicity. Proc Natl Acad Sci U S A. 1994;91:7727–31.[Google Scholar]
  • 12. Wikstrand CJ, Hale LP, Batra SK, Hill ML, Humphrey PA, Kurpad SN, et al Monoclonal antibodies against EGFRvIII are tumor specific and react with breast and lung carcinomas and malignant gliomas. Cancer Res. 1995;55:3140–8.[PubMed][Google Scholar]
  • 13. Grossman WJ, Verbsky JW, Tollefsen BL, Kemper C, Atkinson JP, Ley TJDifferential expression of granzymes A and B in human cytotoxic lymphocyte subsets and T regulatory cells. Blood. 2004;104:2840–8.[PubMed][Google Scholar]
  • 14. Quezada SA, Simpson TR, Peggs KS, Merghoub T, Vider J, Fan X, et al Tumor-reactive CD4(+) T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J Exp Med. 2010;207:637–50.[Google Scholar]
  • 15. El Andaloussi A, Lesniak MSAn increase in CD4+CD25+FOXP3+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neuro Oncol. 2006;8:234–43.[Google Scholar]
  • 16. Fecci PE, Mitchell DA, Whitesides JF, Xie W, Friedman AH, Archer GE, et al Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res. 2006;66:3294–302.[PubMed][Google Scholar]
  • 17. Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, et al CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. 2006;203:1701–11.[Google Scholar]
  • 18. Sakaguchi S, Miyara M, Costantino CM, Hafler DAFOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500.[PubMed][Google Scholar]
  • 19. Efimova OV, Kelley TWInduction of granzyme B expression in T-cell receptor/CD28-stimulated human regulatory T cells is suppressed by inhibitors of the PI3K-mTOR pathway. BMC Immunol. 2009;10:59.[Google Scholar]
  • 20. Marek N, Bieniaszewska M, Krzystyniak A, Juscinska J, Mysliwska J, Witkowski P, et al The time is crucial for ex vivo expansion of T regulatory cells for therapy. Cell Transplant. 2011;20:1747–58.[PubMed][Google Scholar]
  • 21. Magg T, Mannert J, Ellwart JW, Schmid I, Albert MHSubcellular localization of FOXP3 in human regulatory and nonregulatory T cells. Eur J Immunol. 2012;42:1627–38.[PubMed][Google Scholar]
  • 22. Pace L, Tempez A, Arnold-Schrauf C, Lemaitre F, Bousso P, Fetler L, et al Regulatory T cells increase the avidity of primary CD8+ T cell responses and promote memory. Science. 2012;338:532–6.[PubMed][Google Scholar]
  • 23. Koristka S, Cartellieri M, Theil A, Feldmann A, Arndt C, Stamova S, et al Retargeting of human regulatory T cells by single-chain bispecific antibodies. J Immunol. 2012;188:1551–8.[PubMed][Google Scholar]
  • 24. Offner S, Hofmeister R, Romaniuk A, Kufer P, Baeuerle PAInduction of regular cytolytic T cell synapses by bispecific single-chain antibody constructs on MHC class I-negative tumor cells. Mol Immunol. 2006;43:763–71.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.