Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Immunology

Antitumor Activity Mediated by Double-Negative T Cells

Kevin J. Young, Lyndsey S. Kay, M. James Phillips and Li Zhang
Kevin J. Young
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lyndsey S. Kay
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
M. James Phillips
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Li Zhang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI:  Published November 2003
  • Article
  • Info & Metrics
  • PDF
Loading

Abstract

Allogeneic lymphocytes are potent mediators of leukemia and lymphoma remission. The goal of this study was to determine whether single MHC class I locus-mismatched lymphocytes could generate an antilymphoma activity in the absence of graft-versus-host-disease (GVHD) and to understand the underlying mechanisms. Immunoincompetent Scid or lethally irradiated mice were challenged i.v. with a lethal dose of A20 lymphoma cells together with an infusion of single MHC class I locus mismatched splenocytes. Mice that were challenged with A20 cells alone succumbed to lymphoma between 34 and 50 days after infusion. In contrast, >75% of mice that were coinfused with single class I MHC locus mismatched splenocytes survived indefinitely (n = 20) in the absence of GVHD. Interestingly, the number of CD3+CD4−CD8− double-negative (DN) T cells increased 15-fold in mice that did not develop lymphoma. Both DN T cells isolated from the spleens of lymphoma-free mice and DN T cells cloned from naïve mice were cytotoxic to A20 lymphoma cells in vitro. When DN T cell clones were infused into naïve mice i.v. together with A20 lymphoma cells, 86% of recipient mice were protected from lymphoma onset and did not develop GVHD (n = 22). To assess whether the systemic injection of DN T cells can also suppress local tumor development, A20 cells were infused i.m., and at the same time DN T cell clones were infused either i.v. or i.m. Results indicated that DN T cells infused systemically (i.v.) could not prevent local tumor outgrowth, but DN T cells coinfused locally (i.m.) prevented local tumor development in 91% of animals (n = 11). Furthermore, we demonstrate that primary DN T cells were also able to prevent tumor growth in 75% of mice when infused together with A20 cells i.m. (n = 12). Together, these results demonstrate that an antilymphoma activity can be generated in mice without causing GVHD. Furthermore, DN T cells can suppress lymphoma cells in vivo and in vitro, suggesting that DN T cells could be used as a novel strategy for the treatment of lymphoma.

INTRODUCTION

The transplantation of stem cells after high-dose chemotherapy and irradiation is an effective treatment for many hematological malignancies (1) . The success of this therapy was historically attributed to the conditioning regimen that was able to eliminate tumor cells. However, several studies recognized that antihost reactivity by transplanted grafts was associated with a decreased relapse of leukemia and increased survival (2, 3, 4) . Moreover, this effect was not apparent in patients given T cell-depleted allografts (4, 5, 6, 7) or patients who were given syngeneic bone marrow grafts (4) . These early findings lead to the now widely accepted view that donor T cells can be induced to kill malignant cells, which is known as the GVL 3 effect.

Although donor lymphocytes are beneficial in preventing lymphoma relapse, their usefulness is hampered by their ability to respond to MHC and minor histocompatibility antigens on host cells and destroy host tissue. This effect, which is known as GVHD, represents a major factor responsible for the morbidity and mortality of allogeneic stem cell transplant recipients (8) . To reduce the severity of GVHD, donor lymphocytes are often matched for MHC antigens with recipients, and recipients are given nonspecific immunosuppression. Although these approaches decrease the incidence and severity of GVHD, they are also associated with an increased leukemia/lymphoma relapse (9, 10, 11) .

Several strategies have been developed to induce GVL in the absence of GVHD. Less toxic nonmyeloablative stem cell transplants (12 , 13) and donor lymphocyte infusions (14) have been successfully used in patients with malignancies, and the adoptive transfer of lymphocytes mismatched with the host for minor histocompatibility antigens has been successful in eradicating tumors in mice (15) . Additionally, accumulating evidence indicates that Treg lymphocytes play an important role in the down-regulation of immune responses to self or allogeneic antigens (16, 17, 18, 19, 20, 21, 22, 23, 24, 25) . Several groups have shown that Treg cells may be involved in suppressing GVHD (26, 27, 28, 29, 30, 31, 32) , opening the door to a new strategy for treating or preventing GVHD. However, Treg cells have also been shown to inhibit antitumor cytotoxic T-lymphocyte responses (33, 34, 35) .

We have identified recently a novel CD3+CD4−CD8− DN T-cell population from recipients who have permanently accepted donor skin allografts. This population of DN T cells differs from other T cells in their surface marker expression, cytokine profile, and mechanism of suppression (16) , and can specifically suppress CD8+ and CD4+ T cells that are primed against the same alloantigen (16 , 36 , 37) . Moreover, we have shown that DN T cells can attenuate GVHD caused by an infusion of single class I mismatched lymphocytes (32) . The goal of this study was to determine whether GVL activity could also be established in mice that were resistant to GVHD. We show that the infusion of single class I locus mismatched lymphocytes into immunodeficient mice leads to an antilymphoma activity in the absence of GVHD. DN T cells increase in the periphery of recipient mice and are able to directly kill lymphoma cells in vitro. Furthermore, the infusion of DN T cells is able to prevent both systemic and local lymphoma outgrowth. Together, these data indicate the possibility of using DN T cells as a novel cellular therapy for tumors.

MATERIALS AND METHODS

Mice.

C57BL/6 (B6), (B6xBALB/c)F1, BALB/c H-2-dm2 (dm2), and B6.MRL-Faslpr lymphoproliferative (lpr) mice were purchased from The Jackson Laboratory (Bar Harbor, ME). Breeding stocks of 2C and H-Y transgenic mice were kindly provided by Drs. Dennis Y. Loh (Howard Hughes Medical Institute, St. Louis, MO; 38 ) and Hung-Sia Teh (University of British Columbia, Vancouver, British Columbia, Canada), respectively. 2C transgenic mice (on B6 background) carry functionally rearranged TCR α- and β-chain transgenes, which are specific for class I MHC Ld (38) and are recognized by the mAb 1B2 (39) . 2C transgenic mice were bred with dm2 mice (a BALB/c Ld loss mutant). The offspring, either (2Cxdm2)F1 mice (H-2b/d, Ld-, 1B2+) or (B6xdm2)F1 mice (H-2b/d, Ld−, or 1B2−), were used as lymphocyte donors. C.B-17 Scid mice (effectively BALB/c congenic to B6 at the IgH locus; H-2d/d) were bred with B6 Scid mice. The resulting ScidF1 mice (Db/d, Kb/d, and Ld+) were sublethally irradiated (2.5Gy) and used as recipients. Lethally irradiated (8.5 Gy; B6xBALB/c)F1 mice (H-2b/d, Ld+) were also used as recipients.

Generation and Maintenance of 1B2+DN T-Cell Clones.

Spleen cells were collected from naïve (2Cxdm2)F1 mice and used to generate T-cell clones using standard cloning and subcloning procedures as described elsewhere (16) . We have demonstrated previously that IL-2 and IL-4 are required for DN T-cell survival and function (16 , 40) . Therefore, to maintain the DN T-cell clones, 5 × 104 cells were cultured in a 24-well plate containing 5 × 105 irradiated Ld+ cells in an α-MEM supplemented with 10% FCS and 30 IU/ml recombinant IL-2 and 30 IU/ml recombinant IL-4. The cells were incubated at 37°C with 5% CO2. The T-cell clones were stimulated in the above manner every 3–4 days.

Infusion of Allogeneic Lymphocytes and Tumor Cells.

Single viable cell suspensions were prepared from the spleen and lymph nodes of donor mice, and injected into sex-matched recipient mice via the tail vein (4 × 107 cells/mouse). DN T-cell clones were injected either i.v. (5 × 106 cells/mouse) or i.m. (1 × 106 cells/mouse). The A20 B cell lymphoma line (BALB/c origin; Ref. 41 ) was obtained from American Type Culture Collectino (Manassas, VA), and 105 A20 cells were injected either i.v. or i.m. per mouse.

Evaluation of GVHD and Tumor Burden.

Clinical signs of GVHD (42) such as weight loss, diarrhea, ruffled fur, hunched posture, and scaled ears were monitored several times weekly. In addition, tissue samples from major GVHD lesion sites (liver, skin, and intestine) were harvested, stained with H&E, and evaluated by light microscopy for GVHD according to standard methods (42) . For evaluation of i.v.-injected tumor burden, major organs such as liver, spleen, lung, heart, kidney, intestine, and lymph nodes were examined for the presence of tumor cell infiltration. Morbidity was also monitored daily. For evaluation of i.m.-injected tumor, mice were monitored daily and graded from levels 1–4 based on tumor size, level 4 being the largest and causing impaired movement at which point the animal was sacrificed.

Cell Surface Marker Staining.

Splenocytes were collected at various time points after reconstitution and triple stained with fluorescence-conjugated mAbs specifically recognizing the αβ-TCR (1B2), CD4, and CD8. In some mice staining was also performed using mAbs that recognized CD3, NK1.1, and Fas. All of the mAbs except 1B2 were purchased from BD PharMingen. Data were acquired and analyzed on an EPICS XL-MCL flow cytometer.

Cytotoxicity Assays.

Purified DN T cells from lpr mice were stimulated with irradiated (B6xBALB/c)F1 splenocytes for 2–3 days in the presence of 30 units/ml IL-2 and 50 units/ml IL-4 before their use. Murine DN T cells were purified from the spleens of reconstituted mice by fluorescence activated cell sorting as described previously (16) . Viable cells were harvested and seeded at various dilutions into 96-well plates and used as effector cells. Tumor target cells were labeled with 10 μCi/ml of [3H]thymidine at 37°C for 18 h and cultured for another 18 h in the presence of IL-2 and IL-4 either together or in the absence of DN T cells. Cytotoxicity was measured using a liquid scintillation counter as described previously (16) . Specific cell killing was calculated using the equation: % specific killing = (S − E)/S ×100, where E (experimental) is cpm of retained DNA in the presence of effector cells and S (spontaneous) is cpm of retained DNA in the absence of effector cells.

Statistical Analyses.

All of the statistical analyses were performed using Student’s t test and were calculated using Microsoft Excel XP. Error bars represent SD.

RESULTS

Infusion of Transgenic Anti-Ld Splenocytes Protects Immunodeficient Mice from Developing Lymphoma Without Causing GVHD.

We have demonstrated previously that infusion of MHC class I Ld mismatched (2Cxdm2)F1 splenocytes into ScidF1 mice leads to an unresponsiveness of anti-Ld CD8+ T cells in the host (43 , 44) and GVHD-free survival of recipient animals (32) . To investigate whether the infused allogeneic lymphocytes could kill tumor cells, ScidF1 mice were sublethally irradiated and infused i.v. with 4 × 106 viable spleen cells from Ld-mismatched (2Cxdm2)F1 mice, together with 105 A20 lymphoma cells, which were syngeneic to the host and mismatched at the Ld locus with donor lymphocytes. As controls, ScidF1 mice were infused with A20 lymphoma cells alone. All of the ScidF1 mice that were infused with tumor cells alone developed lymphoma and were sacrificed between 28 and 42 days (median survival time 32 days; n = 11; Fig. 1A ⇓ ). Autopsies and histopathologic studies confirmed that lymphoma was the cause of death (Fig. 1B ⇓ , middle). In striking contrast, 11 of 12 mice that were infused with (2Cxdm2)F1 splenocytes and a lethal dose of A20 lymphoma cells remained healthy during the course of the study (>150 days; Fig. 1A ⇓ ). Neither tumor infiltration nor GVHD was found in any organ of these animals (Fig. 1B ⇓ , bottom). These data demonstrate that infusion of splenocytes mismatched for a single class I locus antigen does not result in GVHD but can protect immunodeficient recipients from dying of lymphoma.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Infusion of Ld-mismatched splenocytes prevents death caused by lymphoma in the absence of GVHD. A, ScidF1 mice were infused with a lethal dose of A20 lymphoma cells alone (♦, n = 11) or together with viable spleen cells from (2Cxdm2)F1 mice (▾, n = 42). Survival of recipient mice were monitored. B, livers harvested at 4 weeks after infusion of A20 alone were greatly enlarged (top, left), and H&E staining (×300) shows diffuse infiltration of the liver by tumor cells, which completely replace the normal liver architecture (middle). The liver from recipients of A20 and (2Cxdm2)F1 splenocytes is normal in size, shape, and color (top, right), and no histopathological lesions were observed in hepatocytes, liver cell cords, or portal and venous structures (bottom).

An Antilymphoma Activity Is Established in the Absence of GVHD After the Infusion of Nontransgenic Ld Mismatched Splenocytes into Naïve Mice.

The above observations are based on a transgenic-Scid model and, thus, may not be representative of true pathophysiological responses. To validate the findings in a nontransgenic model, (B6xBALB/c)F1 mice were lethally irradiated, and infused with a lethal dose of A20 lymphoma cells and 4 × 107 viable spleen cells from (B6xdm2)F1 mice. As a control, a group of mice were infused with A20 lymphoma alone. All of the control mice succumbed to lymphoma within 50 days of injection of A20 cells. In contrast, 6 of the 8 (B6xBALB/c)F1 mice that were infused with both (B6xdm2)F1 spleen cells and A20 tumor cells remained free from both GVHD and tumor invasion for >150 days (Fig. 2) ⇓ . The remaining 2 mice were sacrificed at the indicated periods of time, and histopathological analyses revealed that the mice died of lymphoma and had no signs of GVHD (data not shown). These results confirm the findings in the transgenic-ScidF1 model, and demonstrate that infusion of MHC class I Ld-mismatched lymphocytes allow immune-incompetent animals to reject tumors without causing GVHD.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Infusion of nontransgenic Ld-mismatched splenocytes prevents lymphoma onset in the absence of GVHD. (B6xBALB/c)F1 mice were given a lethal dose of A20 lymphoma alone (♦, n = 8), or lethally irradiated and infused with (B6xdm2)F1 lymphocytes (▾, n = 8). Survival and signs of GVHD and tumor were monitored.

DN T Cells Increase in Immunodeficient Mice After Reconstitution.

Both T cells and NK cells have been shown to have an antilymphoma activity. To identify the type of cells that might be involved in the observed antilymphoma activity, we first analyzed the proportions of T cells and NK cells in recipient mice. Splenocytes were collected from recipient mice 60 days after reconstitution and stained for either NK1.1 or CD3. As shown in Fig. 3, A and B ⇓ , only 1.9% of the total splenocytes were NK1.1+, whereas 22.7% were CD3+. Although not completely eliminating the possibility that NK cells are involved in preventing tumor onset, these results led us to additionally study the potential role of T cells in the prevention of lymphoma. The fate of anti-Ld 1B2+ T lymphocytes was monitored after the infusion of sublethally irradiated ScidF1 mice with (2Cxdm2)F1 lymphocytes and A20 lymphoma cells (Fig. 3C) ⇓ . Consistent with previous reports (43 , 45) , a vigorous expansion followed by a massive depletion of antihost 1B2+CD8+ T cells was observed in the spleen of recipients. 1B2+CD4+ T cells also increased in the first 35 days after infusion of donor lymphocytes but remained relatively unchanged thereafter. Interestingly, we observed a steady increase in the number of donor-derived 1B2+DN T cells in these recipient mice, which became the dominant population of T cells over time (Fig. 3C) ⇓ . A similar change was also observed in sublethally irradiated ScidF1 mice that had been given an infusion of Ld mismatched lymphocytes alone (32) , although the increase observed here was greater. These results support the possibility that DN T cells might be involved in the prevention of lymphoma onset.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

The number of DN T cells increased after infusion of Ld-mismatched splenocytes. ScidF1 mice were infused with (2Cxdm2)F1 spleen cells in combination with A20 lymphoma cells. A and B, 60 days later, splenocytes were collected from recipient mice, and stained with anti-NK1.1 (A) and anti-CD3 (B) mAbs. C, the total number of donor-derived splenic 1B2+CD8+ (▪) and 1B2+CD4+ (♦), and 1B2+CD8− T cells was determined on the days indicated in the figure. The number of 1B2+DN T cells (▴) was calculated by subtracting the number of 1B2+CD4+ T cells from 1B2+CD8− T cells. Each time point contains the data from 3–5 mice, and shown as the mean; bars, ±SD.

DN Treg Cells Possess an Antilymphoma Activity.

To investigate whether DN T cells have an antitumor activity, we first studied whether mice that did not develop lymphoma after an initial coinjection of Ld-mismatched donor lymphocytes and A20 lymphoma cells could mount a secondary antitumor response. At 8 weeks after the initial infusion of allogeneic lymphocytes and A20 tumor cells, at which time donor-derived DN T cells have formed the dominant lymphocyte population in the recipients (Fig. 3C) ⇓ , ScidF1 mice were given a second inoculation of A20 tumor cells. As shown in Fig. 4A ⇓ , these mice survived indefinitely and showed no histopathological signs of tumor or GVHD when sacrificed at 125 days.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

DN T cells are cytotoxic to A20 lymphoma cells in vitro. A, ScidF1 mice were infused with (2Cxdm2)F1 splenocytes and a lethal dose of A20 lymphoma (▪, n = 5) or left untreated(▴, n = 4). Both groups of mice were inoculated with lymphoma cells 8 weeks later, and percentage of survival of recipients is shown. B and C, lethally (8.5Gy) irradiated (B6xBALB/c)F1 mice were coinfused with (2Cxdm2)F1 (B) or (B6xdm2)F1 (C) lymphocytes and A20 lymphoma cells. Eight weeks later, 1B2+DN (B) or CD3+DN (C) T cells were purified from the spleens and used as effector cells in a cytotoxicity assay at the indicated ratios. A20 (▪) and EL4 (▴) tumor cells were used as targets. Results shown are the percentage of specific killing and are representative of two independent experiments each with five replicates; bars, ±SD.

To evaluate directly the antitumor activity of donor-derived DN T cells, DN T cells were purified from the spleens of tumor-free mice 8 weeks after coinjection with (2Cxdm2)F1 and A20 cells, and assessed for their ability to kill A20 lymphoma cells in vitro. As shown in Fig. 4B ⇓ , purified 1B2+DN T cells were able to lyse allogeneic A20 tumor cells in a dose-dependent manner but could not kill syngeneic EL4 tumor targets. To assess whether nontransgenic DN T cells were also able to kill A20 cells, DN T cells were purified from (B6xBALB/c)F1 mice 8 weeks after infusion of (B6xdm2)F1 splenocytes and A20 lymphoma cells. As shown in Fig. 4C ⇓ , purified nontransgenic DN T cells were also able to kill A20 lymphoma cells in a dose-dependent manner. Together, these data indicate that in addition to suppressing syngeneic antihost T cells (32) , both transgenic and nontransgenic DN T cells from reconstituted mice are capable of killing allogeneic A20 tumor cells in vitro.

DN T Cell-Mediated Cytotoxicity to Lymphoma Cells Requires Fas/FasL Interactions.

We have demonstrated previously that DN T-cell clones are able to kill activated syngeneic CD8+ T cells through a Fas/FasL-mediated pathway (16) . To gain an insight into the mechanism involved in DN T cell-mediated killing of allogeneic lymphoma cells, we evaluated the role of Fas/FasL in the cytotoxicity to tumor cells. DN T-cell clones generated from (2Cxdm2)F1 mice (16) were used as effector cells. Tumor cell lines that are Ld+ (Fig. 5A ⇓ , left) and either Fashigh (A20; Fig. 5A ⇓ , right) or Faslow (P815; Fig. 5A ⇓ , right) were used as targets in a killing assay. Fashigh tumor cell lines were killed by DN T cell clones in a dose-dependent manner, whereas killing of Faslow tumor targets was much less efficient (Fig. 5B) ⇓ , suggesting that Fas/FasL interactions are involved for DN T cell-mediated cytotoxicity of tumor cells. To additionally test the role of Fas/FasL interactions in DN T cell-mediated killing of tumor cells, FasL was blocked on DN T cells using Fas-Fc Fusion protein. As shown in Fig. 5C ⇓ , Fas-Fc partially inhibited the killing of A20 lymphoma cells mediated by DN Treg clones. These findings indicate that Fas/FasL interactions are important for DN T cell-mediated cytotoxicity to A20 lymphoma cells.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Fas/FasL interactions are required for DN T cell mediated cytotoxicity. A, A20 (gray) and EL4 (white) tumor cell lines were stained with biotinylated anti-Ld mAb followed by streptavidin-FITC (left), or anti-Fas phycoerythrin-conjugated mAb (right). Negative controls are shown in ----. B, varying numbers of 1B2+DN T cell clones were used as effector cells in cytotoxicity assays as described previously (68) . A20 (▪, Ld+Fashigh) and P815 (♦, Ld+Faslow) tumor cells were used as targets. The data show the mean percentage of specific killing of 3 replicates and are representative of three independent experiments; bars, ±SD. C, 1B2+DN T-cell clones were preincubated with various concentrations of Fas-Fc fusion protein before coincubation with A20 tumor cells or left untreated as controls. The percentage of inhibition of killing of tumor cells by Fas-Fc-treated DN T cells compared with controls is shown. The experiment was repeated two times each with three replicates, and similar results were obtained. Bars, ±SD.

DN T-Cell Clones Can Prevent Systemic Lymphoma Outgrowth.

To determine the antilymphoma activity of DN T cells in vivo and to study the therapeutic potential of using in vitro generated DN T cells for the treatment of lymphoma, nonirradiated (B6xBALB/c)F1 mice were coinjected with various doses of 1B2+DN T cell clones and a lethal dose of A20 tumor cells. All of the mice that received A20 cells alone died of lymphoma between 30 and 40 days after injection (Fig. 6A ⇓ ; Fig. 6B ⇓ , top). Interestingly, infusion of 5 × 106 DN T-cell clones with A20 lymphoma cells prevented >80% of the mice from developing lymphoma. These mice lived for >150 days (Fig. 6A) ⇓ with no detectable clinical or histopathological signs of lymphoma or GVHD (Fig. 6B ⇓ , bottom). The infusion of 10 × 106 DN T cells similarly protected mice from lymphoma and did not cause GVHD (data not shown). These data demonstrate that in vitro generated DN T cells can prevent the onset of lymphoma and suggest that DN Treg cells may be used as a novel cellular therapy for lymphoma.

Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

DN T-cell clones prevent systemic lymphoma outgrowth. (A; B6xBALB/c) mice were infused with either a lethal dose of A20 cells alone (▾, n = 10), or together with either L12.2 (▪, n = 8) or CN04 (▴, n = 14) DN T-cell clones. Survival and clinical signs of GVHD were monitored for >150 days. B, mice injected with A20 alone showed dense tumor infiltrates in the liver at 30 days (B, top). Mice that were coinjected with DN T-cell clones had normal liver structure with no infiltrating tumor cells or signs of GVHD at 100 days (B, bottom).

DN T Cell Clones Can Prevent the Outgrowth of Solid Tumor.

To determine whether systemically injected DN T cells are also able to prevent solid tumor outgrowth, (B6xBALB/c)F1 mice were injected i.v. with DN T-cell clones and at the same time given an i.m. infusion of A20 lymphoma cells into their right hind leg. As controls, mice were given an i.m. infusion of A20 lymphoma cells with no other treatment. Eight of 9 mice that received A20 lymphoma alone developed large tumors in their hind legs within 30 days and were sacrificed when their tumors reached a level 4 (Fig. 7A) ⇓ . Similarly, 75% of the animals that were infused with A20 i.m. together with DN T-cell clones injected i.v. developed lesions within 30 days (Fig. 7A) ⇓ . Together, these results indicate that systemically infused DN T cells are unable to prevent local solid tumor outgrowth.

Fig. 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 7.

Local tumor outgrowth can be prevented by locally injected DN T cells. A, (B6xBALB/c)F1 mice were i.m. injected with 105 A20 tumor cells alone (▪, n = 9) or along with i.v. injection of 5 × 106 CN04 DN T-cell clone (n = 4). B, (B6xBALB/c)F1 mice were injected i.m. with 105 A20 tumor cells either alone (▪, n = 10) or together with 1 × 106 CN04 DN T-cell clone (▴, n = 10). Mice were monitored and sacrificed when the tumor appearance was a level 4, as described in “Materials and Methods.”

The inability of DN T cells to kill local tumor cells could be explained by DN T cells being unable to migrate to this tissue or exert their effect in the muscle. To test these possibilities, DN T cells were coinjected with lymphoma cells i.m., and recipient mice were monitored for tumor growth. As seen previously, 89% of control mice developed tumor lesions in their hind legs within 30 days of tumor infusion. Interestingly, when DN T-cell clones were coinfused with A20 tumor cells, tumor outgrowth was prevented in 91% of recipient animals (Fig. 7B) ⇓ . These results indicate that DN T-cell clones are able to prevent the outgrowth of tumor locally when infused directly into the muscle and demonstrate that DN T cells are able to exert their effect locally.

Primary DN T Cells Have an Antilymphoma Activity.

DN T cells are difficult to isolate from naïve nontransgenic mice because of their low numbers and the lack of specific markers on DN T cells. We have demonstrated previously that DN T cells from lpr mice are able to suppress syngeneic CD8+ T cells as well as enhance skin graft survival (37) . Therefore, these mice provide a source of primary DN T cells, which can be used to verify our findings using DN T clones. To ensure that DN T cells from lpr mice are not NK T cells, splenocytes from lpr mice were stained using CD3, CD4, CD8, and NK1.1. Fig. 8A ⇓ shows that the vast majority of DN T cells are NK1.1− (66.2% versus 0.49%). DN T cells were subsequently isolated as described in “Materials and Methods,” and were tested for their ability to kill A20 lymphoma cells. Fig. 8B ⇓ shows that lpr DN T cells were able to dose-dependently kill A20 tumor cells in vitro. Next, (B6xBALB/c)F1 mice were infused i.m. with A20 tumor cells together or in the absence of 1 × 106 lpr DN T cells. All of the mice that received A20 lymphoma cells alone developed lesions within 30 days and were sacrificed. In contrast, 75% of mice that were coinfused with lpr DN T cells showed no signs of tumor growth and survived tumor free indefinitely (Fig. 8C) ⇓ . Together, these results verify our finding with transgenic DN T cell clones and additionally demonstrate that DN T cells have an antilymphoma activity in vivo.

Fig. 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 8.

Primary DN T cells are NK1.1− and can prevent tumor outgrowth. A, LN and spleen cells were isolated from lpr mice and stained with anti-CD4/8 FITC, anti-NK1.1 PhycoErythrin, and anti-CD3 CyChrome. The histogram is gated on the CD3+ T-cell population, and the percentages of the CD4+/CD8+ and DN T cells are explicitly indicated. B, DN T cells were purified from the lymph nodes and spleen of lpr mice as described in “Materials and Methods” and stimulated with irradiated (B6xBALB/c)F1 splenocytes for 4 days. The proportion of DN T cells was then determined by flow cytometry and used as effector cells in various dilutions as indicated. A20 tumor cells were labeled overnight with 10 μCi of [3H]thymidine, and used as target cells. The data shown is the percentage of specific killing for three replicates, and the experiment is representative of two independent experiments. C, (B6xBALB/c)F1 mice were injected i.m. with A20 tumor cell either alone (▪, n = 10) or together with 1 × 106 purified lpr DN T-cell clones (▾, n = 12). Tumor growth and recipient survival were monitored.

DISCUSSION

It has been well established that allogeneic lymphocytes play a role in the eradication of leukemia and lymphoma. Here we demonstrate that the reconstitution of immunoincompetent mice with single class I mismatched lymphocytes leads to tumor- and GVHD-free survival of recipients. We have shown that DN T cells expand in the periphery of reconstituted mice and form the majority of anti-Ld lymphocytes. These cells are able to eliminate allogeneic tumor cells in vitro through a mechanism that involves Fas/FasL. In addition, DN T cells that were cloned from (2Cxdm2)F1 mice or purified from lpr mice have a similar ability to eliminate lymphoma cells in vitro and are also able to prevent the outgrowth of both systemic and solid tumor in vivo. These data indicate that the infusion of DN T cells are sufficient to prevent lymphoma onset. Whereas these data do not preclude a role for other lymphocytes, such as NK or CD4+ T cells, in mediating GVL in reconstituted animals, they indicate the importance of DN T cells in mediating GVL and their potential use as a novel cellular antitumor therapy.

Although allogeneic lymphocytes are an effective treatment for eliminating cancer cells, this therapy is limited by the risk of developing GVHD (46) . For example, a recent study demonstrated that more than half of patients with chronic myeloid leukemia, who were given a nonmyeloablative stem cell transplant, developed acute GVHD despite being given immunosuppressive drugs (13) . Also, in a study conducted by Childs et al. (12) , nonmyeloablative stem cell transplants successfully induced tumor regression only in those patients that developed GVHD. Furthermore, patients who did not develop GVHD also did not develop an antitumor effect (12) . These data demonstrate that despite modern immunotherapeutic regimens, GVHD remains one of the largest barriers to successful treatment of cancer using allogeneic lymphocytes.

Several recent studies have been designed to assess the possibility of infusing lymphocytes with an antitumor activity into patients without causing GVHD, especially as part of a nonmyeloablative regimen (12 , 25 , 47) . One such approach involved the insertion of a thymidine kinase suicide gene into allogeneic lymphocytes so that infused lymphocytes could be eliminated at the onset of GVHD (48) . Whereas thymidine kinase was successful in controlling GVHD, less than half of the patients had a complete remission. Other groups have attempted to induce T-cell responses against tumor-associated antigens (49) such as MART-1, gp100, and tyrosinase on melanoma cells (50) . However, many of these tumor-associated antigens are also expressed on normal melanocytes, resulting in autoimmunity and depigmentation (51) . Autoimmunity can be avoided by using low avidity T cells (52 , 53) ; however, successful tumor rejection will only be achieved if the level of self-antigen expression on the tumor is higher than that expressed on the healthy tissues. Hence a dichotomy exists between successfully eradicating tumor cells, and avoiding GVHD and autoimmune diseases. We demonstrate here that although DN T cells are allogeneic to the host, they do not cause GVHD or autoimmune disease in recipient mice. Most of the recipient mice that have been injected with either DN T cells or clones survived indefinitely, and none showed signs of GVHD. Moreover, unlike most CD4+ and CD8+ T cells, which are susceptible to activation-induced cell death (54) , DN Treg cells are resistant to TCR cross-linking-induced apoptosis in vitro (40) and activation-induced cell death in vivo (Fig. 3C) ⇓ . Whether DN T cells can also mediate a GVL activity in humans without causing GVHD is currently under study, and the results derived from these studies will determine the efficacy of using DN T cells as an antitumor therapy.

Both DN T cells (32) and CD4+CD25+ Treg cells (27 , 28) have been shown to suppress GVHD. For example, DN T cells have been shown to attenuate GVHD caused by the infusion of allogeneic CD8+ T cells (32) , and CD4+CD25+ Treg cells suppress acute GVHD after the infusion of allogeneic CD4+CD25− T cells (27 , 28) . However, there is evidence that CD4+CD25+ Treg cells additionally inhibit antitumor T-cell responses and could ultimately contribute to the progression of cancer (33, 34, 35 , 55, 56, 57, 58) . For example, it has been shown recently that autologous antitumor T cells are inhibited by CD4+CD25+ Treg cells in both mouse and human models (33, 34, 35 , 55) . Large numbers of CD4+CD25+ Treg cells have also been shown to infiltrate the tumor microenvironment (34 , 55 , 59) and have been found in patients immunized with known melanoma antigens (56) . Furthermore, tumors were efficiently rejected by the host immune system in mice that were depleted of CD4+CD25+ Treg cells using antiCD25 mAb (35 , 57 , 58) . In contrast to CD4+CD25+ Treg cells, which inhibit antitumor immune responses, we show here that DN T cells augment the antitumor immune responses.

The dual role of DN T cells to inhibit GVHD and promote GVL can be explained as follows. DN T cells are activated after their infusion into Ld+ recipient mice (60) . After activation, DN T cells expand and become the major subset of T cells (Fig. 3C) ⇓ . These cells are then able to acquire allo-MHC molecules from antigen-presenting cells, and present these molecules on their surface. Antihost T cells with the same TCR specificity as DN T cells can recognize the acquired MHC molecules on the DN T-cell surface and are subsequently killed by the DN T cells through Fas/FasL interaction (32) . Hence, GVHD is prevented. On the other hand, DN T cells can also kill Ld+ A20 tumor cells through the Fas/FasL pathway (Fig. 4, B and C ⇓ ; Fig. 5C ⇓ ), therefore mediating an antilymphoma response. The finding that Fas-Fc fusion protein does not completely block DN T cell-mediated killing indicates that there may be other pathways involved in the elimination of tumors by DN T cells. However, it is not clear why host tissues, which express both Ld and Fas, are not damaged by DN T cells. Several studies have shown that although endothelial cells express Fas they are normally resistant to Fas-induced apoptosis (61 , 62) . Additionally, evidence suggests that IFN-γ can cause T-cell immunodeficiency (63) and protect mice from GVHD (64, 65, 66, 67) by indirectly inhibiting donor T-cell activation and expansion (64) . We found that both in vitro and in vivo activated DN Treg cells produce large amounts of IFN-γ ( 16 ; data not shown), and may therefore protect hosts from tissue damage. In conclusion, we demonstrate here that DN T cells have a novel antitumor function both in vitro and in vivo. This potent antitumor activity makes DN T cells a prospective candidate for cellular immune therapy.

Footnotes

  • Grant support: National Cancer Institutes of Canada 12160 (L. Z.) and Canadian Institutes of Health Research MT14431 (L. Z.).

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • K. J. Y. and L. S. K. contributed equally to this work.

  • Requests for reprints: Li Zhang, Department of Laboratory Medicine and Pathobiology, Multi Organ Transplantation Program, Toronto General Research Institute, University Health Network, 621 University Avenue, Toronto, Ontario M5G 2C4, Canada. Phone: (416) 340-4915; Fax: (416) 597-9749; E-mail lzhang@transplantunit.org.

  • ↵3 The abbreviations used are: GVL, graft-versus-leukemia/lymphoma; GVHD, graft-versus-host disease; Treg, regulatory T; DN, double-negative; TCR, T-cell receptor; mAb, monoclonal antibody; Scid, severe combined immunodeficient; IL, interleukin; NK, natural killer; FasL, Fas ligand.

  • Received May 27, 2003.
  • Revision received July 30, 2003.
  • Accepted July 31, 2003.
  • ©2003 American Association for Cancer Research.

References

  1. ↵
    Thomas E. D., Blume K. G. Historical markers in the development of allogeneic hematopoietic cell transplantation. Biol. Blood Marrow Transplant., 5: 341-346, 1999.
    OpenUrlCrossRefPubMed
  2. ↵
    Weiden P. L., Sullivan K. M., Flournoy N., Storb R., Thomas E. D. Antileukemic effect of chronic graft-versus-host disease: contribution to improved survival after allogeneic marrow transplantation. N. Engl. J. Med., 304: 1529-1533, 1981.
    OpenUrlCrossRefPubMed
  3. ↵
    Sullivan K. M., Weiden P. L., Storb R., Witherspoon R. P., Fefer A., Fisher L., Buckner C. D., Anasetti C., Appelbaum F. R., Badger C. Influence of acute and chronic graft-versus-host disease on relapse and survival after bone marrow transplantation from HLA-identical siblings as treatment of acute and chronic leukemia. Blood, 73: 1720-1728, 1989.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Horowitz M. M., Gale R. P., Sondel P. M., Goldman J. M., Kersey J., Kolb H. J., Rimm A. A., Ringden O., Rozman C., Speck B. Graft-versus-leukemia reactions after bone marrow transplantation. Blood, 75: 555-562, 1990.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Apperley J. F., Jones L., Hale G., Waldmann H., Hows J., Rombos Y., Tsatalas C., Marcus R. E., Goolden A. W., Gordon-Smith E. C. Bone marrow transplantation for patients with chronic myeloid leukaemia: T-cell depletion with Campath-1 reduces the incidence of graft-versus-host disease but may increase the risk of leukaemic relapse. Bone Marrow Transplant., 1: 53-66, 1986.
    OpenUrlPubMed
  6. ↵
    Goldman J. M., Gale R. P., Horowitz M. M., Biggs J. C., Champlin R. E., Gluckman E., Hoffmann R. G., Jacobsen S. J., Marmont A. M., McGlave P. B. Bone marrow transplantation for chronic myelogenous leukemia in chronic phase. Increased risk for relapse associated with T-cell depletion. Ann. Intern. Med., 108: 806-814, 1988.
  7. ↵
    Marmont A. M., Horowitz M. M., Gale R. P., Sobocinski K., Ash R. C., van Bekkum D. W., Champlin R. E., Dicke K. A., Goldman J. M., Good R. A. T-cell depletion of HLA-identical transplants in leukemia. Blood, 78: 2120-2130, 1991.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Ferrara J. L., Deeg H. J. Graft-versus-host disease. N. Engl. J. Med., 324: 667-674, 1991.
    OpenUrlCrossRefPubMed
  9. ↵
    Korngold R., Leighton C., Manser T. Graft-versus-myeloid leukemia responses following syngeneic and allogeneic bone marrow transplantation. Transplantation, 58: 278-287, 1994.
    OpenUrlCrossRefPubMed
  10. ↵
    Kolb H. J., Schattenberg A., Goldman J. M., Hertenstein B., Jacobsen N., Arcese W., Ljungman P., Ferrant A., Verdonk L., Niederwieser D., van Rhee F., Mittermuller J., de Witte T., Holler E., Ansari H. Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood, 86: 2041-2050, 1995.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Barrett A. J., Malkovska V. Graft-versus-leukemia: Understanding and using the alloimmune response to treat haematological malignancies. Br. J. Haematol., 93: 754-761, 1996.
    OpenUrlCrossRefPubMed
  12. ↵
    Childs R., Chernoff A., Contentin N., Bahceci E., Schrump D., Leitman S., Read E. J., Tisdale J., Dunbar C., Linehan W. M., Young N. S., Barrett A. J. Regression of metastatic renal-cell carcinoma after nonmyeloablative allogeneic peripheral-blood stem-cell transplantation. N. Engl. J. Med., 343: 750-758, 2000.
    OpenUrlCrossRefPubMed
  13. ↵
    Or R., Shapira M. Y., Resnick I., Amar A., Ackerstein A., Samuel S., Aker M., Naparstek E., Nagler A., Slavin S. Nonmyeloablative allogeneic stem cell transplantation for the treatment of chronic myeloid leukemia in first chronic phase. Blood, 101: 441-445, 2003.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Walter E. A., Greenberg P. D., Gilbert M. J., Finch R. J., Watanabe K. S., Thomas E. D., Riddell S. R. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N. Engl. J. Med., 333: 1038-1044, 1995.
    OpenUrlCrossRefPubMed
  15. ↵
    Fontaine P., Roy-Proulx G., Knafo L., Baron C., Roy D. C., Perreault C. Adoptive transfer of minor histocompatibility antigen-specific T lymphocytes eradicates leukemia cells without causing graft-versus-host disease. Nat. Med., 7: 789-794, 2001.
    OpenUrlCrossRefPubMed
  16. ↵
    Zhang Z. X., Yang L., Young K. J., DuTemple B., Zhang L. Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression. Nat. Med., 6: 782-789, 2000.
    OpenUrlCrossRefPubMed
  17. ↵
    Graca L., Thompson S., Lin C. Y., Adams E., Cobbold S. P., Waldmann H. Both CD4+CD25+ and CD4+CD25− regulatory cells mediate dominant transplantation tolerance. J. Immunol., 168: 5558-5565, 2002.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Shevach E. M. CD4+ CD25+ suppressor T cells: more questions than answers. Nat. Rev. Immunol., 2: 389-400, 2002.
    OpenUrlPubMed
  19. ↵
    Zelenika D., Adams E., Humm S., Lin C. Y., Waldmann H., Cobbold S. P. The role of CD4+ T-cell subsets in determining transplantation rejection or tolerance. Immunol. Rev., 182: 164-179, 2001.
    OpenUrlCrossRefPubMed
  20. ↵
    Sakaguchi S., Sakaguchi N., Shimizu J., Yamazaki S., Sakihama T., Itoh M., Kuniyasu Y., Nomura T., Toda M., Takahashi T. Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol. Rev., 182: 18-32, 2001.
    OpenUrlCrossRefPubMed
  21. ↵
    Roncarolo M. G., Bacchetta R., Bordignon C., Narula S., Levings M. K. Type 1 T regulatory cells. Immunol. Rev., 182: 68-79, 2001.
    OpenUrlCrossRefPubMed
  22. ↵
    Jiang H., Chess L. The specific regulation of immune responses by CD8+ T cells restricted by the MHC class Ib molecule, Qa-1. Annu. Rev. Immunol., 18: 185-216, 2000.
    OpenUrlCrossRefPubMed
  23. ↵
    Hayday A. C. γδ cells: a right time and a right place for a conserved third way of protection. Annu. Rev. Immunol., 18: 975-1026, 2000.
    OpenUrlCrossRefPubMed
  24. ↵
    Palathumpat V., Dejbakhsh-Jones S., Holm B., Strober S. Different subsets of T cells in the adult mouse bone marrow and spleen induce or suppress acute graft-versus-host disease. J. Immunol., 149: 808-817, 1992.
    OpenUrlAbstract
  25. ↵
    Lan F., Zeng D., Higuchi M., Huie P., Higgins J. P., Strober S. Predominance of NK1.1+TCRαβ+ or DX5+TCRαβ+ T cells in mice conditioned with fractionated lymphoid irradiation protects against graft-versus-host disease: “natural suppressor” cells. J. Immunol., 167: 2087-2096, 2001.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Johnson B. D., Becker E. E., LaBelle J. L., Truitt R. L. Role of immunoregulatory donor T cells in suppression of graft-versus- host disease following donor leukocyte infusion therapy. J. Immunol., 163: 6479-6487, 1999.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Taylor P. A., Lees C. J., Blazar B. R. The infusion of ex vivo activated and expanded CD4+CD25+ immune regulatory cells inhibits graft-versus-host disease lethality. Blood, 99: 3493-3499, 2002.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Hoffmann P., Ermann J., Edinger M., Fathman C. G., Strober S. Donor-type CD4+CD25+ regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J. Exp. Med., 196: 389-399, 2002.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Tutschka P. J., Hess A. D., Beschorner W. E., Santos G. W. Suppressor cells in transplantation tolerance. III. The role of antigen in the maintenance of transplantation tolerance. Transplantation, 33: 510-514, 1982.
    OpenUrlPubMed
  30. ↵
    Abraham V. S., Sachs D. H., Sykes M. Mechanism of protection from graft-versus-host disease mortality by IL- 2. III. Early reductions in donor T cell subsets and expansion of a CD3+CD4−CD8− cell population. J. Immunol., 148: 3746-3752, 1992.
    OpenUrlAbstract
  31. ↵
    Palathumpat V., Dejbakhsh-Jones S., Holm B., Wang H., Liang O., Strober S. Studies of CD4−CD8− αβ bone marrow T cells with suppressor activity. J. Immunol., 148: 373-380, 1992.
    OpenUrlAbstract
  32. ↵
    Young K. J., Du Temple B., Phillips M. J., Zhang L. Inhibition of graft versus host disease by double negative regulatory T cells. J. Immunol, 171: 134-141, 2003.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Somasundaram R., Jacob L., Swoboda R., Caputo L., Song H., Basak S., Monos D., Peritt D., Marincola F., Cai D., Birebent B., Bloome E., Kim J., Berencsi K., Mastrangelo M., Herlyn D. Inhibition of cytolytic T lymphocyte proliferation by autologous CD4+/CD25+ regulatory T cells in a colorectal carcinoma patient is mediated by transforming growth factor-β. Cancer Res., 62: 5267-5272, 2002.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Liyanage U. K., Moore T. T., Joo H. G., Tanaka Y., Herrmann V., Doherty G., Drebin J. A., Strasberg S. M., Eberlein T. J., Goedegebuure P. S., Linehan D. C. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J. Immunol., 169: 2756-2761, 2002.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Shimizu J., Yamazaki S., Sakaguchi S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J. Immunol, 163: 5211-5218, 1999.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Young K., Zhang L. The nature and mechanisms of DN regulatory T-Cell mediated suppression. Hum. Immunol., 63: 926 2002.
    OpenUrlCrossRefPubMed
  37. ↵
    Ford M. S., Young K. J., Zhang Z. X., Ohashi P. S., Zhang L. The immune regulatory function of lymphoproliferative double negative T cells in vitro and in vivo. J. Exp. Med., 196: 261-267, 2002.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Sha W. C., Nelson C. A., Newberry R. D., Kranz D. M., Russell J. H., Loh D. Y. Selective expression of an antigen receptor on CD8-bearing T lymphocytes in transgenic mice. Nature (Lond.), 335: 271-274, 1988.
    OpenUrlCrossRefPubMed
  39. ↵
    Kranz D. M., Tonegawa S., Eisen H. N. Attachment of an anti-receptor antibody to non-target cells renders them susceptible to lysis by a clone of cytotoxic T lymphocytes. Proc. Natl. Acad. Sci. USA, 81: 7922-7926, 1984.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Khan Q., Penninger J. M., Yang L. M., Marra L. E. K. I., Zhang L. Regulation of apoptosis in mature αβ+ CD4−CD8− antigen-specific suppressor T-cell clones. J. Immunol., 162: 5860-5867, 1999.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Kim K. J., Kannelopoulus-langevin C., Mervin R. M., Sachs D. H., Asofsky R. Establishment and characterization of BALB/c lymphoma lines with B cell properties. J. Immunol., 122: 549-554, 1979.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Fowler D. H., Breglio J., Nagel G., Eckhaus M. A., Gress R. E. Allospecific CD8+ Tc1 and Tc2 populations in graft-versus-leukemia effect and graft-versus-host disease. J. Immunol., 157: 4811-4821, 1996.
    OpenUrlAbstract
  43. ↵
    Zhang L. The fate of adoptively transferred antigen-specific T cells in vivo. Eur. J. Immunol., 26: 2208-2214, 1996.
    OpenUrlCrossRefPubMed
  44. ↵
    Zhang L., Miller R. G., Zhang J. Characterization of apoptosis-resistant antigen-specific T cells in vivo. J. Exp. Med., 183: 2065-2073, 1996.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Dey B., Yang Y. G., Preffer F., Shimizu A., Swenson K., Dombkowski D., Sykes M. The fate of donor T-cell receptor transgenic T cells with known host antigen specificity in a graft-versus-host disease model. Transplantation, 68: 141-149, 1999.
    OpenUrlCrossRefPubMed
  46. ↵
    Raiola A. M., Van Lint M. T., Valbonesi M., Lamparelli T., Gualandi F., Occhini D., Bregante S., di Grazia C., Dominietto A., Soracco M., Romagnani C., Vassallo F., Casini M., Bruno B., Frassoni F., Bacigalupo A. Factors predicting response and graft-versus-host disease after donor lymphocyte infusions: a study on 593 infusions. Bone Marrow Transplant., 31: 687-693, 2003.
    OpenUrlCrossRefPubMed
  47. ↵
    Blazar B. R., Lees C. J., Martin P. J., Noelle R. J., Kwon B., Murphy W., Taylor P. A. Host T cells resist graft-versus-host disease mediated by donor leukocyte infusions. J. Immunol., 165: 4901-4909, 2000.
    OpenUrlAbstract/FREE Full Text
  48. ↵
    Bonini C., Ferrari G., Verzeletti S., Servida P., Zappone E., Ruggieri L., Ponzoni M., Rossini S., Mavilio F., Traversari C., Bordignon C. HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia. Science (Wash. DC), 276: 1719-1724, 1997.
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Rosenberg S. A. Progress in human tumour immunology and immunotherapy. Nature (Lond.), 411: 380-384, 2001.
    OpenUrlCrossRefPubMed
  50. ↵
    De Visser K. E., Schumacher T. N., Kruisbeek A. M. CD8+ T Cell Tolerance and Cancer Immunotherapy. J. Immunother., 26: 1-11, 2003.
  51. ↵
    Dudley M. E., Wunderlich J. R., Robbins P. F., Yang J. C., Hwu P., Schwartzentruber D. J., Topalian S. L., Sherry R., Restifo N. P., Hubicki A. M., Robinson M. R., Raffeld M., Duray P., Seipp C. A., Rogers-Freezer L., Morton K. E., Mavroukakis S. A., White D. E., Rosenberg S. A. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science (Wash. DC), 298: 850-854, 2002.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Morgan D. J., Kreuwel H. T. C., Fleck S., Levitsky H. I., Pardoll D. M., Sherman L. A. Activation of low avidity CTL specific for a self epitope results in tumor rejection but not autoimmunity. J. Immunol., 160: 643-651, 1998.
    OpenUrlAbstract/FREE Full Text
  53. ↵
    Melero I., Singhal M. C., McGowan P., Haugen H. S., Blake J., Hellstrom K. E., Yang G., Clegg C. H., Chen L. Immunological ignorance of an E7-encoded cytolytic T-lymphocyte epitope in transgenic mice expressing the E7 and E6 oncogenes of human papillomavirus type 16. J. Virol., 71: 3998-4004, 1997.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    Tan R., Xu X., Ogg G. S., Hansasuta P., Dong T., Rostron T., Luzzi G., Conlon C. P., Screaton G. R., McMichael A. J., Rowland-Jones S. Rapid death of adoptively transferred T cells in acquired immunodeficiency syndrome. Blood, 93: 1506-1510, 1999.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    Woo E. Y., Yeh H., Chu C. S., Schlienger K., Carroll R. G., Riley J. L., Kaiser L. R., June C. H. Cutting edge: Regulatory T cells from lung cancer patients directly inhibit autologous T cell proliferation. J. Immunol., 168: 4272-4276, 2002.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    Javia L. R., Rosenberg S. A. CD4+CD25+ Suppressor lymphocytes in the circulation of patients immunized against melanoma antigens. J. Immunother., 26: 85-93, 2003.
  57. ↵
    Sutmuller R. P., van Duivenvoorde L. M., van Elsas A., Schumacher T. N., Wildenberg M. E., Allison J. P., Toes R. E., Offringa R., Melief C. J. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25+ regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J. Exp. Med., 194: 823-832, 2001.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Onizuka S., Tawara I., Shimizu J., Sakaguchi S., Fujita T., Nakayama E. Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor α) monoclonal antibody. Cancer Res., 59: 3128-3133, 1999.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    Woo E. Y., Chu C. S., Goletz T. J., Schlienger K., Yeh H., Coukos G., Rubin S. C., Kaiser L. R., June C. H. Regulatory CD4+CD25+ T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Res., 61: 4766-4772, 2001.
    OpenUrlAbstract/FREE Full Text
  60. ↵
    Young K. J., Yang L. M., Phillips M. J., Zhang L. Donor-lymphocyte infusion induces tolerance by activating systemic and graft-infiltrating double negative T regulatory cells. Blood, 100: 3408-3414, 2002.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    Richardson B. C., Lalwani N. D., Johnson K. J., Marks R. M. Fas ligation triggers apoptosis in macrophages but not endothelial cells. Eur. J. Immunol., 24: 2640-2645, 1994.
    OpenUrlCrossRefPubMed
  62. ↵
    Sata M., Luo Z., Walsh K. Fas ligand overexpression on allograft endothelium inhibits inflammatory cell infiltration and transplant-associated intimal hyperplasia. J. Immunol., 166: 6964-6971, 2001.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    Krenger W., Falzarano G., Delmonte J., Jr., Snyder K. M., Byon J. C., Ferrara J. L. Interferon-γ suppresses T-cell proliferation to mitogen via the nitric oxide pathway during experimental acute graft-versus-host disease. Blood, 88: 1113-1121, 1996.
    OpenUrlAbstract/FREE Full Text
  64. ↵
    Yang Y. G., Dey B. R., Sergio J. J., Pearson D. A., Sykes M. Donor-derived interferon γ is required for inhibition of acute graft-versus-host disease by interleukin 12. J. Clin. Investig., 102: 2126-2135, 1998.
    OpenUrlCrossRefPubMed
  65. ↵
    Brok H. P., Vossen J. M., Heidt P. J. Interferon-γ-mediated prevention of graft-versus-host disease: development of immune competent and allo-tolerant T cells in chimeric mice. Bone Marrow Transplant., 19: 601-606, 1997.
    OpenUrlCrossRefPubMed
  66. ↵
    Brok H. P., Heidt P. J., van der Meide P. H., Zurcher C., Vossen J. M. Interferon-γ prevents graft-versus-host disease after allogeneic bone marrow transplantation in mice. J. Immunol., 151: 6451-6459, 1993.
    OpenUrlAbstract
  67. ↵
    Baker J., Verneris M. R., Ito M., Shizuru J. A., Negrin R. S. Expansion of cytolytic CD8+ natural killer T cells with limited capacity for graft-versus-host disease induction due to interferon-γ production. Blood, 97: 2923-2931, 2001.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    Matzinger P. The JAM test, A simple assay for DNA fragmentation and cell death. J. Immunol. Methods, 145: 185-192, 1991.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top
Cancer Research: 63 (22)
November 2003
Volume 63, Issue 22
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Antitumor Activity Mediated by Double-Negative T Cells
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Antitumor Activity Mediated by Double-Negative T Cells
Kevin J. Young, Lyndsey S. Kay, M. James Phillips and Li Zhang
Cancer Res November 15 2003 (63) (22) 8014-8021;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Antitumor Activity Mediated by Double-Negative T Cells
Kevin J. Young, Lyndsey S. Kay, M. James Phillips and Li Zhang
Cancer Res November 15 2003 (63) (22) 8014-8021;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • INTRODUCTION
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • Footnotes
    • References
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Abstract 6659: Response to nivolumab and ipilimumab in microsatellite instability-high (MSI-H) cervical carcinoma with acquired resistance to pembrolizumab
  • Abstract 6606: Develop dual-targeted CAR-T cells to achieve RCC cures
  • Abstract 6646: Comprehensive cyclic immunofluorescent analysis reveals new target candidates for CAR T cell based immunotherapy of glioblastoma multiforme
Show more Immunology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement