Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • My alerts
  • Log in
  • Log out
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

MHC Class II-Transfected Tumor Cells Directly Present Antigen to Tumor-Specific CD4+ T Lymphocytes1

Todd D. Armstrong, Virginia K. Clements and Suzanne Ostrand-Rosenberg
J Immunol January 15, 1998, 160 (2) 661-666;
Todd D. Armstrong
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Virginia K. Clements
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Suzanne Ostrand-Rosenberg
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

We have developed and shown to be efficacious an immunotherapeutic strategy to enhance the generation of tumor-specific CD4+ T helper lymphocytes. The approach uses autologous tumor cells genetically modified to express syngeneic MHC class II genes as cell-based immunogens and is based on the hypothesis that tumor cells directly present tumor Ags to CD4+ T cells. Since the conventional pathway for CD4+ T cell activation is indirect via professional APC, induction of immunity following immunization with class II-transfected tumor cells was examined in bone marrow chimeric mice. Both tumor and host-derived cells are APC for tumor Ags, suggesting that the efficacy of tumor cell vaccines can be significantly improved by genetic modifications that enhance tumor cell Ag presentation.

Many of the recently developed strategies for enhancing immunity to autologous tumors involve immunization with genetically modified tumor cells. These approaches are based on the reasoning that tumor cells present potentially immunogenic tumor peptides, and that if modified appropriately, they could directly present tumor Ag and activate T lymphocytes, by-passing the requirement for transfer of tumor Ag (peptide) to host-derived professional APC.

Most of these studies have focussed on the direct activation of tumor-specific CD8+ T lymphocytes, either by providing appropriate costimulatory signals such as B7 (1, 2, 3, 4) or by providing cytokines normally produced by CD4+ T helper lymphocytes, such as IL-2 or IL-4 (5, 6). In contrast, we and others have reasoned that optimal tumor-specific CD8+ T cells and long term immune memory will only be generated if sufficient CD4+ Th cells are also stimulated (7, 8, 9, 10). As a result, we have targeted the activation of tumor-specific CD4+ T cells. We have hypothesized that class II-transfected tumor cells will present endogenously encoded peptides in the context of the transfected class II molecules and, therefore, directly present tumor peptides to CD4+ T helper lymphocytes. Reports from our laboratory as well as studies by others demonstrate that such genetically modified tumor cells are potent inducers of tumor-specific immunity in naive mice (7, 9, 11) as well as in tumor-bearing mice (12), demonstrating their potential as immunotherapeutic agents. Although these therapeutic results are consistent with the hypothesis that the genetically modified tumor cells directly present Ag to CD4+ T cells during the immunization process, there is no direct evidence demonstrating that the modified tumor cells directly present Ag to responding CD4+ T lymphocytes. Such a pathway would be unconventional, since CD4+ T cells are usually activated by the process of indirect Ag presentation or cross-priming, in which Ag is taken up and presented by host-derived professional APC (i.e., dendritic cells, macrophages, or B lymphocytes) rather than by the Ag-expressing cells themselves (13, 14). Since optimal exploitation of this immunotherapeutic approach will depend on a full understanding of the mechanism through which the genetically modified tumor cells activate T lymphocytes, the present studies were undertaken to identify the cell population(s) that serves as APC during the activation of tumor-specific CD4+ T cells following immunization with class II-transfected tumor cells.

Materials and Methods

Mice

C57BL/6, A/J, and (C57BL/6 × A/J) ((B6 × A/J)F1) mice were purchased from The Jackson Laboratory (Bar Harbor, ME) or bred in the University of Maryland Baltimore County animal facility. Bone marrow donors and recipients were female mice, and chimeras were generated as previously described (15). Briefly, recipient (B6 × A/J)F1 mice were maintained on tetracycline water (0.2%) for 1 wk before and 5 wk after reconstitution and were given gentamicin sulfate s.c. (500 μg) for 7 consecutive days beginning 1 day before irradiation/reconstitution. Approximately 24 h before irradiation/reconstitution, recipients were taken off food. Recipients were irradiated with 875 rad total body irradiation using a 137Cs source (Kewaunee Scientific, Statesville, NC) and reconstituted i.v. with one femur-equivalent of donor bone marrow within 2 to 3 h of irradiation. Chimeras were maintained in a pathogen-free environment for 6 wk before use. All chimeras were tested by indirect immunofluorescence to ascertain bone marrow genotype and assure chimeric status as follows. Concurrent with the in vitro APC assays, spleens of chimeras were removed and stained for MHC class I (H-2Kk for A/J (mAb 16-3-1) (16); H-2KbDb for C57BL/6 (mAb 28-13-3) (17)) Ags. Positively staining cells were gated relative to conjugate alone controls, and positive cells were compared with wild-type A/J and C57BL/6 splenocytes stained under identical conditions. The percentage of the donor phenotype was calculated by comparing the percentage of positive chimeric spleen cells vs that of wild-type cells. For example, if 98 and 2% of A/J→F12 chimeric splenocytes were stained with the 16-3.1 and 28-13-3 mAbs, respectively, and 99 and 1% of A/J wild-type splenocytes were stained with the 28-13-3 and 16-3.1 mAbs, respectively, then the A/J→F1 chimeras were considered 99% donor phenotype.

Cells, transfectants, and hybridomas

The SaI sarcoma and its transfectants were cultured as previously described (7). SaI sarcoma cells transfected with syngeneic MHC class II Aαk and Aβk genes (SaI/Ak cells) have been previously described (7). SaI cells expressing I-Ab class II molecules (SaI/Ab cells) were generated by transfecting SaI cells with the Aαb and Aβb cDNAs contained in the pKCR3 plasmid (18) plus the pSV2.neo plasmid using the transfection procedure previously described (7). SaI, SaI/Ak, or SaI/Ab cells expressing an endoplasmic reticulum-retained hen egg lysozyme gene (HEL; SaI/HEL, SaI/Ak/HEL, SaI/Ab/HEL cells) were generated as previously described by transfection with the BCMG-HEL plasmid containing the hygromycinR gene (19). Transfectants expressing MHC class II genes or HEL were maintained in medium supplemented with 400 μg/ml G418 or 400 μg/ml hygromycin, respectively. Double transfectants were maintained on both drugs. 3A9 is an I-Ak-restricted HEL46–61-specific T cell hybridoma (20) and was maintained as previously described (19); JK1290 is an I-Ab-restricted HEL specific hybridoma (21) and was maintained in Iscove’s modified Dulbecco’s medium supplemented with 10% Fetalclone I (Hyclone, Logan, UT), 1% penicillin, 1% streptomycin, and 1% gentamicin.

Tumor challenges

For tumorigenicity studies, mice were inoculated i.p. with live tumor cells, observed daily for survival, and killed when they became moribund. The inoculation dose was chosen based on previous titration studies (7). For immunization studies, chimeric mice were inoculated i.p. with 5 × 105 to 106 live tumor cells and killed 6 or 7 days later. For tumor challenge studies, mice were inoculated i.p. with the indicated number of tumor cells and examined three times per week for tumor growth. Ascites tumors usually became palpable within 10 to 14 days of inoculation and grew progressively. Based on our previous experience with the SaI tumor, if mice do not develop palpable tumor within 2 mo of challenge, they will remain tumor free during their lifetime (7). Tumor incidence is the number of mice with progressively growing tumors divided by the total number of mice challenged. Tumor-bearing mice were killed according to University of Maryland Baltimore County institutional animal care and use committee guidelines when they became moribund. The mean survival time is the time between inoculation and sacrifice.

In vitro Ag presentation assays

Splenocytes from immunized mice were prepared from mechanically dissociated spleens, and B lymphocytes were removed by panning as previously described (22). Resulting T cells (5 × 106 cells/well) were cocultured in flat-bottom 96-well plates with fresh naive A/J or C57BL/6 splenocytes in a final volume of 250 μl/well containing 1 mg/ml lysozyme. Responder to stimulator ratios were 10:1 and/or 50:1. Supernatants were harvested after 24 h and assayed for IL-2 content using ELISA kits as described by the manufacturer (Endogen, Boston, MA). Samples were run in triplicate, and the mean ± SD determined for each sample. In most cases, SDs were ≤5% of test values. Background values (IL-2 release in the absence of HEL) were subtracted from experimental values (IL-2 release in the presence of HEL) to obtain specific IL-2 release. Values were converted to picograms per milliliter using a standard curve incorporated into the IL-2 assay. In some experiments splenocytes were depleted of CD4+ or CD8+ T lymphocytes in vitro (22) or in vivo (12) before use in an APC assay. APC assays using splenocytes from chimeric mice plus T cell hybridomas 3A9 and JK1290 were performed as previously described (19).

Indirect immunofluorescence and flow cytometry

Tumor cells, transfectants, and splenocytes were monitored for cell surface Ag expression by indirect immunofluorescence as previously described (7) and analyzed on an Epics XL flow cytometer (Coulter, Hialeah, FL). The following mAbs were used: I-Ak (10-3-6 or 10-2-16) (23), I-Ab (34-5-3S) (17), Kk (16-3-1) (16), Dd (34-5-8) (24), and lysozyme (HyHEL 7 and 10) (25). Cells monitored for intracellular lysozyme were fixed with paraformaldehyde and stained with a mixture of the HyHEL 7 and 10 mAbs (25) as previously described (19). Isotype controls were performed for surface and cytoplasmically stained cells, and staining was essentially identical with that in fluorescent conjugate alone controls.

Results

In our previous studies the A/J-derived (H-2k/d) SaI sarcoma, when transfected with syngeneic MHC class II Aαk and Aβk genes (SaI/Ak tumor cells), induced potent CD4+ T cell-dependent, tumor-specific immunity in syngeneic A/J and semisyngeneic (C57BL/6 × A/J)F1 mice. In vitro experiments using HEL-specific, I-Ak-restricted T cell hybridomas showed that SaI/Ak tumor cells transfected with a gene encoding an endoplasmic reticulum-retained HEL (SaI/Ak/HEL tumor cells) present endogenously synthesized HEL peptide to CD4+ T cells (19). The genetically modified tumor cells, therefore, are capable of presenting tumor-encoded Ag directly to T cells. To determine whether during the immunization process the tumor cells themselves are APC for endogenously encoded tumor Ags (direct Ag presentation) or if host-derived cells are the APC (indirect Ag presentation or cross-priming), we used the following genetic approach. (C57BL/6 × A/J)F1 mice are lethally irradiated and reconstituted with either C57BL/6 (H-2KbAbDb) or A/J (H-2KkAkDd) bone marrow so that the resulting chimeric mice (B6→F1 or A/J→F1, respectively) have host-derived APC of either the C57BL/6 or the A/J genotype, respectively. The chimeras are subsequently challenged with either SaI/Ak/HEL or SaI/Ab/HEL tumor cells, respectively, and the MHC restriction pattern of the response is determined. If the tumor cells are the exclusive APC for the tumor-encoded Ag (HEL), then the response will be restricted to the MHC class II genotype of the tumor cells. However, if host-derived cells are the exclusive APC for tumor-encoded HEL, then the T cell response will be restricted to the genotype of the reconstituting bone marrow in the chimeras. If both tumor cells and host cells are APC for tumor-encoded Ags, then the response will be restricted to both the tumor and bone marrow genotypes.

SaI sarcoma cells transfected with MHC class II and/or HEL genes express these gene products at the cell surface or intracellularly

SaI/Ak and SaI/Ak/HEL cells were available from previous experiments (7, 19). SaI/Ab and SaI/Ab/HEL cells were generated by gene transfection as described in Materials and Methods. The resulting transfectants were stained for cell surface expression of MHC class II molecules (live cells) or for intracellular expression of lysozyme (paraformaldehyde-fixed and saponin-permeabilized cells). As shown in Figure 1⇓, cells transfected with the MHC class II I-Ak and I-Ab genes expressed comparable levels of these molecules, as measured by staining with the 10-2-16 and 34-5-3S mAbs, respectively (Fig. 1⇓, i and j, and e and f for SaI/Ak and SaI/Ab cells, respectively). Similarly, cells transfected with the HEL construct expressed comparable levels of intracellular lysozyme as measured by staining with the mixture of HyHEL 7 and HyHEL 10 mAbs (Fig. 1⇓, n, p, and r for SaI/HEL, SaI/Ak/HEL, and SaI/Ab/HEL cells, respectively.), while untransfected cells were negative for lysozyme (Fig. 1⇓, m, o, and q). HEL transfectants were also stained for cell surface HEL expression and were negative (data not shown). Supernatants of the transfectants were assayed by ELISA for HEL secretion and showed low levels of soluble HEL (1–5 ng/ml/6.7 × 105 cells/24 h). The transfectants were also stained for MHC class I H-2Kk, H-2Dd, and H-2Ld Ag expression, and these levels were approximately equivalent among all transfectants and parental SaI cells (data not shown).

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

Indirect immunofluorescence staining of SaI (a, g, and m), SaI/HEL (b, h, and n), SaI/Ak (c, i, and o), SaI/Ak/HEL (d, j, and p), SaI/Ab (e, k, and q), and SaI/Ab/HEL (f, l, and r) cells. Tumor cells were stained for I-Ab with the 28-13-3 mAb (17) (a–f), for I-Ak with the 10-2-16 mAb (23) (g–l), or for lysozyme with the HyHEL 7 plus 10 mAbs (25) (m–r). MHC class II staining was performed on live tumor cells; lysozyme staining was performed on fixed, permeabilized cells. The second-step, fluorescent conjugate FITC-goat anti-mouse IgG was used with all Abs. The solid line represents staining by fluorescent conjugate alone; the dotted line represents staining by specific Ab plus fluorescent conjugate.

Lysozyme peptides are presented by both I-Ak and I-Ab MHC class II molecules

Numerous in vitro studies have demonstrated that both I-Ak and I-Ab MHC class II molecules present HEL-derived peptides to CD4+ T cells (21, 26, 27, 28). To ascertain that HEL peptides are presented by both class II alleles when the alleles are expressed by SaI sarcoma cells, semisyngeneic (C57BL/6 × A/J)F1 mice were challenged i.p. with parental SaI, SaI/Ak, SaI/Ab, SaI/HEL, SaI/Ak/HEL, and SaI/Ab/HEL tumor cells and followed for tumor incidence. As shown in Table I⇓, wild-type SaI and SaI/HEL tumor cells were lethal in ≥94% of F1 mice, indicating that HEL expression alone does not cause tumor rejection. SaI/Ak and SaI/Ab cells were significantly less lethal than SaI cells; however, they still caused tumors in a significant number of F1 mice (50 and 40% lethal, respectively). In contrast, SaI/Ak/HEL and SaI/Ab/HEL tumor cells were rejected by 100% of F1 mice, suggesting that HEL peptides are presented by the I-Ak and I-Ab MHC class II molecules of the tumor cells and function as nominal Ag for T cell recognition.

View this table:
  • View inline
  • View popup
Table I.

MHC class II/HEL-transfected tumor cells are rejected by semisyngeneic (C57BL/6 × A/J)F1 micea

Immunization of chimeric mice with tumor cell transfectants generates T cells restricted to both the tumor genotype and the host genotype

If the genetically modified tumor cells are the exclusive APC for tumor Ags, then A→F1 bone marrow chimeras immunized with B tumor cells will have HEL-specific T cells restricted to the tumor (B) MHC genotype. In contrast, if host-derived cells are the APC, then the chimeras will have HEL-specific T cells restricted to the genotype of the host (A) regardless of the genotype of the tumor. To test these alternatives, A/J→ F1 and B6→F1 chimeras were generated and challenged i.p. with SaI/Ab/HEL or SaI/Ak/HEL tumor cells, respectively. Seven days after tumor cell immunization, spleens were removed and depleted of B cells and adherent cells by panning, and the remaining T cells were incubated in vitro with lysozyme plus fresh A/J or C57BL/6 splenocytes as APC. Supernatants were harvested 24 h later and tested for IL-2 content. Since the goal of these experiments is to characterize the earliest Ag-specific response, T cells were tested 1 wk postimmunization. Likewise, T cells were assayed for IL-2 secretion without an in vitro or in vivo boost to the Ag (HEL), so that the primary or initial response would be measured.

A/J→F1 and B6→F1 bone marrow chimeras were generated as described in Materials and Methods. The efficiency of chimera formation was determined using indirect immunofluorescence to measure the percentage of donor genotype cells in the recipients’ spleens. Table II⇓ lists the chimeras used in subsequent experiments and shows the MHC class I genotype (H-2Kk for A/J vs H-2Db for C57BL/6) of splenocytes from the chimeras as measured by immunofluorescence. As shown, all the chimeras used in the following experiments were ≥97.9%, and most chimeras were ≥99.5%, of the donor genotype. Chimeras were also tested functionally for hemopoietic reconstitution. Splenocytes of representative chimeras were used as APC for intact HEL to I-Ak- and I-Ab-restricted HEL-specific hybridomas, 3A9 and JK1290, respectively. A/J→F1 and A/J splenocytes presented HEL to 3A9 hybridoma cells, but not to JK1290 hybridoma cells. In contrast, B6→F1 and C57BL/6 splenocytes presented Ag to JK1290 cells, but not to 3A9 hybridoma cells (data not shown). APC of the bone marrow chimeras, therefore, are phenotypically and functionally the donor genotype.

View this table:
  • View inline
  • View popup
Table II.

Efficiency of chimera formation and T cell depletion in A/J→F1 and B6→F1 bone marrow chimeric micea

Table III⇓ shows the results of three representative, independent experiments assessing IL-2 production by total T cells from tumor-immunized, chimeric mice. In two experiments (Expt. 1 and 2) A/J→F1 chimeras were immunized with SaI/Ab/HEL tumor cells, while in one experiment (Expt. 4), B6→F1 chimeras were immunized with SaI/Ak/HEL tumor cells. In all three experiments HEL-specific T cells restricted to both the tumor and the host genotype were present. IL-2 release by immune T cells cocultured in vitro with lysozyme plus irrelevant genotype APC (SWR, H-2q) was at background levels (data not shown). Therefore, at 1 wk postimmunization with genetically modified tumor cells, both tumor cells and host bone marrow-derived cells were APC for tumor- encoded Ags.

View this table:
  • View inline
  • View popup
Table III.

Both tumor cells and host-derived APC present tumor-encoded Ag to CD4+ T cells in A/J→F1 and B6→F1 bone marrow chimeric micea

The genetically modified tumor cells constitutively express MHC class I (H-2Kk, H-2Dd, and H-2Ld) as well as the transfected MHC class II (I-Ak or I-Ab) molecules. Since CD4+ and CD8+ T cells are activated by Ag in the context of MHC class II and class I molecules, respectively, and since both T cell types can synthesize IL-2, it is possible that the read-out of the splenic T cell experiments reflects HEL-specific, class I-restricted CD8+ T cells. To identify the responding splenic T cells, chimeric mice were depleted of CD4+ or CD8+ T cells before and during immunization, and the remaining T cells were tested for IL-2 secretion following HEL presentation in vitro. Two representative experiments are shown in Table III⇑, one experiment using A/J→F1 chimeric mice immunized with SaI/Ab/HEL tumor cells (Expt. 3) and a second experiment using B6→F1 chimeric mice immunized with SaI/Ak/HEL tumor cells (Expt. 5). As shown in Table II⇑, mice depleted for CD4+ or CD8+ T cells had ≤1.2 or 0.6% of these cells, respectively, demonstrating functional depletion of these populations. As shown in Table III⇑, in both the depletion experiments most of the IL-2 activity was produced by CD4+ T cells, since depletion of CD4+ T cells reduced IL-2 activity to negligible levels. Consistent with the results of Expt. 1, 2, and 4, in Expt. 3, both tumor genotype and host genotype APC stimulated HEL-specific IL-2 release, indicating that both tumor cells and host-derived cells are APC for tumor-encoded Ag (HEL). In contrast, in Expt. 5, only host genotype (H-2b) cells stimulated IL-2 secretion, suggesting that in these mice only host-derived cells are the APC for tumor-encoded Ags.

The five experiments shown in Table III⇑ are representative of 25 similar experiments performed using chimeric or (C57BL/6 × A/J)F1 mice. Results similar to those of Expt. 5, in which only host genotype APC stimulated IL-2 secretion, were noted in only two of these experiments, while the remaining experiments all showed either host and tumor genotype presentation, or tumor genotype presentation alone. It was occasionally observed that tumor-encoded Ags were exclusively presented by host-derived cells; however, such presentation was a relatively rare event. Similar experiments using class II− SaI/HEL tumor cells showed no direct Ag presentation (data not shown), indicating that direct Ag presentation by tumor cells to CD4+ T cells requires MHC class II expression by the tumor. Therefore, as measured at 1 wk postimmunization, CD4+ T cells specific for tumor-encoded Ags were activated via Ag presentation by both tumor and host-derived cells, indicating that both direct and indirect (cross-priming) pathways are used.

Discussion

During an in vivo immune response, CD4+ T lymphocytes are activated by an Ag-specific signal plus a second or costimulatory signal (29). The Ag-specific signal consists of antigenic peptide bound to a MHC class II molecule, which interacts with the corresponding TCR and CD4 complex on the responding T cell. The second signal consists of a costimulatory molecule, such as B7, which binds to its cognate receptor, CD28, on the responding T cell. Since only certain cells, such as dendritic cells, macrophages, activated B lymphocytes, or Langerhans cells, express MHC class II and either constitutively express or are inducible for costimulatory molecules (29), only these so-called professional APC are thought to activate CD4+ T lymphocytes. In our recently developed immunotherapeutic strategy, tumor cells are transfected with genes encoding syngeneic MHC class II molecules or MHC class II plus B7 molecules (4, 7). The class II transfectants are excellent immunogens for vaccinating tumor-free mice against subsequent challenge with wild-type tumor, and during the immunization process the transfectants are induced to express B7-1 and B7-2 molecules (30). Although the single transfectants (MHC class II) are not effective immunogens in tumor-bearing mice, double transfectants (MHC class II plus B7) are potent immunotherapeutic agents for the treatment of established solid tumors (12) and metastatic disease3 (31). We have hypothesized that such genetically modified tumor cells are APC for tumor-encoded Ags because they deliver both the Ag-specific signal and the costimulatory signal to CD4+ T cells. The studies reported here confirm this hypothesis by demonstrating that during the immunization process, class II-transfected tumor cells are APC for endogenously encoded tumor molecules. Our immunotherapeutic approach, therefore, provides an alternative pathway for activation of CD4+ T lymphocytes. Presumably, the ability of the class II transfected tumor cells to function as APC and directly activate CD4+ T cells is responsible for their potent immunotherapeutic effect.

Our data indicate that both tumor cells and host-derived cells are APC for tumor-encoded HEL. These experiments were designed to test Ag presentation of an intracellular, surrogate tumor Ag; however, ELISA assays of SaI/Ak/HEL supernatants show low levels of secreted HEL. Since the SaI/Ak/HEL transfectants are “leaky” for HEL, one cannot determine whether intracellular tumor Ag presentation is normally indirect via host-derived APC, or if cross-priming occurs because soluble HEL is available. If leakiness is the reason for the observed cross-priming, then the relative roles of direct vs indirect presentation for bonafide tumor Ags will be dependent on the cellular locale and behavior of tumor Ag.

Cross-priming or indirect Ag presentation is the usual route for activation of CD4+ T cells. In this process, tumor Ag (in this case, lysozyme) is released from tumor cells, trafficks via the lymphatic and/or circulatory system to the closest regional lymph node(s), and is internalized by professional APC within the lymph node. Alternatively, Ag may be internalized in the periphery by professional APC and brought to the regional lymph node by the APC. In either case, internalized Ag is processed by the APC and presented as peptide in the context of MHC class II molecules on the surface of the APC (32). Since only a small percentage of CD4+ T cells has the appropriate receptor for a given Ag, localization of Ag to the regional lymph node allows exposure to the maximal number of CD4+ T cells. Trafficking of Ag to the regional lymph node during a conventional immune response, therefore, optimizes contact of Ag with the appropriate T cells.

If tumor cells are the APC, Ag presentation could occur either at the tumor site or in the regional lymph node. Presentation at the tumor site is unlikely, since unless a very large number of CD4+ T cells traffick to the site, it is improbable that T cells with an appropriate TCR will be exposed to Ag. Trafficking of the transfectants to the regional nodes is more likely. Tumors are known to metastasize via the lymphatic circulation, and a recent study using genetically modified, nonmetastatic tumor cells demonstrated tumor cells in draining nodes (33). The class II-transfected tumor cells, therefore, may migrate to the draining lymph nodes; however, the precise logistics of the Ag presentation process remain unclear.

Although the activation of CD4+ T cells by genetically modified tumor cells has not been previously studied, other investigators have examined the activation of CD8+ T cells during immunization with transfected tumor cells. Three studies have examined Ag presentation by cytokine modified tumor cells (granulocyte-macrophage CSF, IL-4, IL-7, or IL-3) and have found that cytokine expression facilitates cross-priming (34, 35) or that tumor-specific CD8+ T cells are exclusively activated by host-derived APC (36). Two of these studies also investigated whether B7-transfected tumor cells directly or indirectly presented Ag to tumor-specific CD8+ T cells. In one study, Ag presentation was exclusively via cross-priming (37), while in the other report, Ag presentation was directly via the B7-transfected tumor cells (34). Most of these results, therefore, were unexpected, since cytokine and B7-transfected tumor cells were presumed to induce tumor-specific immunity by direct presentation of tumor Ags to T cells (1, 3, 5).

There are several potential explanations for the observed differences between previously reported studies and the results of the present report. 1) The present report addresses Ag presentation to CD4+ T cells, while earlier studies examined presentation to CD8+ T cells. The varying results could be due to differences in Ag presentation to CD4+ vs CD8+ T cells. 2) In this report, Ag presentation during the primary response (7 days postpriming) was studied, while earlier reports examined later time points (35) and/or secondary responses (34, 36, 37). Since tumor cells remain intact in vivo for only a limited time, direct Ag presentation may occur during the early stage of the immunization process while tumor cells remain intact, and indirect Ag presentation may dominate during later stages when tumor debris is available. 3) In two of these studies (36, 37), the transfected tumor cells were irradiated, and secretion of the surrogate tumor Ag was not assessed. If soluble Ag was available, cross-priming would be the expected result. 4) One of these studies (35) was not designed to measure direct Ag presentation, so even if direct presentation by genetically modified tumor cells occurred, it would not have been detected.

In contrast, studies by Kündig and colleagues (38) demonstrated that fibroblasts transfected with a viral Ag, if injected into a lymph node, directly present Ag to CD8+ T cells, thereby stimulating potent viral-specific CD8+ CTL. The logistics of CD8+ T cell activation by genetically modified therapeutic cells are, therefore, unclear, with data supporting both direct and indirect pathways.

The finding that genetically modified tumor cells directly activate CD4+ T cells has clear implications for the design of immunizing and immunotherapeutic agents. If cell-based immunogens are to be considered as vaccines and/or immunotherapeutic agents, then the cells should be engineered to optimally present Ag to CD4+ T cells. If Ag presentation activity correlates with immunization potential, measurements of in vitro Ag presentation activity may be prognostic of therapeutic efficacy. Depending on the desired type of Th cell (Th1 vs Th2), immunizing cells could be tailored to facilitate activation of a particular subpopulation, perhaps via coexpression or simultaneous bolus administration of cytokines favoring differentiation of one or the other helper population (i.e., IL-12, IL-10, etc.).

Although concern has been voiced about using tumor cell-based immunogens in a clinical setting, a significant number of phase I or II clinical trials using tumor cell material have been completed or are in progress (http://cancernet.nci.nih.gov). Since these trials have not identified any significant safety issues and in some cases have shown modest therapeutic responses (despite overwhelming tumor load), the clinical use of tumor cell-based immunogens is feasible. All the trials using cell-based strategies have been exclusively aimed at enhancing CD8+ T cell responses. The data presented in this report combined with published therapeutic studies (12) strongly argue that an optimal cell-based vaccine should also target the activation of CD4+ T lymphocytes.

Acknowledgments

We are very appreciative of the excellent care given to our mice by Ms. Sandy Mason. We also thank Drs. D. Mathis, H. Pelham, D. Pardoll, S. Adams, and N. Shastri for the I-Ab plasmids, erHEL construct, BCMG vector, and 3A9 and JK1290 hybridomas, respectively.

Footnotes

  • ↵1 Address correspondence and reprint requests to Dr. S. Ostrand-Rosenberg, Department of Biological Sciences, University of Maryland, 1000 Hilltop Circle, Baltimore, MD 21250.

  • ↵2 Abbreviations used in this paper: A/J→F1, lethally irradiated (C57BL/6 × A/J)F1 mouse reconstituted with A/J bone marrow; SaI/Ak, SaI tumor cells transfected with Aαk and Aβk genes; SaI/Ab, SaI tumor cells transfected with Aαb and Aβb genes; HEL, hen egg lysozyme; SaI/HEL, SaI tumor cells transfected with the hen egg lysozyme gene; B6→F1, lethally irradiated (C57BL/6 × A/J)F1 mouse reconstituted with C57BL/6 bone marrow; F1, (C57BL/6 × A/J)F1 mouse.

  • ↵3 B. Pulaski and S. Ostrand-Rosenberg. Reduction of established spontaneous mammary carcinoma metastases following immunotherapy with MHC class II and B7.1 cell-based tumor vaccines. Submitted for publication.

  • Received May 20, 1997.
  • Accepted October 1, 1997.
  • Copyright © 1998 by The American Association of Immunologists

References

  1. ↵
    Chen, L., S. Ashe, W. A. Brady, I. Hellstrom, K. E. Hellstrom, J. A. Ledbetter, P. McGowan, P. S. Linsley. 1992. Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell 71: 1093
    OpenUrlCrossRefPubMed
  2. ↵
    Chen, L., P. S. Linsley, K. E. Hellstrom. 1993. Co-stimulation of T cells for tumor immunity. Immunol. Today 14: 483
    OpenUrlCrossRefPubMed
  3. ↵
    Townsend, S. E., J. P. Allison. 1993. Tumor rejection after direct costimulation of CD8+ T cells by B7-transfected melanoma cells. Science 259: 368
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Baskar, S., S. Ostrand-Rosenberg, N. Nabavi, L. M. Nadler, G. J. Freeman, L. H. Glimcher. 1993. Constitutive expression of B7 restores immunogenicity of tumor cells expressing truncated major histocompatibility complex class II molecules. Proc. Natl. Acad. Sci. USA 90: 5687
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Fearon, E., D. Pardoll, T. Itaya, P. Golumbek, H. Levitsky, J. Simons, H. Karasuyama, B. Vogelstein, P. Frost. 1990. IL-2 production by tumor cells bypasses T helper function in the generation of an antitumor response. Cell 60: 397
    OpenUrlCrossRefPubMed
  6. ↵
    Golumbek, P., A. Lazenby, H. Levitsky, L. Jaffee, H. Karasuyama, M. Baker, D. Pardoll. 1991. Treatment of established renal cancer by tumor cells engineered to secrete interleukin-4. Science 254: 713
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Ostrand-Rosenberg, S., A. Thakur, V. Clements. 1990. Rejection of mouse sarcoma cells after transfection of MHC class II genes. J. Immunol. 144: 4068
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Ostrand-Rosenberg, S.. 1994. Tumor immunotherapy: the tumor cell as an antigen-presenting cell. Curr. Opin. Immunol. 6: 722
    OpenUrlCrossRefPubMed
  9. ↵
    James, R., S. Edwards, K. Hui, P. Bassett, F. Grosveld. 1991. The effect of class II gene transfection on the tumorigenicity of the H-2K negative mouse leukemia cell line K36.16. Immunology 72: 213
    OpenUrlPubMed
  10. ↵
    Chen, P., S. Ullrich, H. Ananthaswamy. 1994. Presentation of endogenous tumor antigens to CD4+ T lymphocytes by murine melanoma cells transfected with major histocompatibility complex class II genes. J. Leukocyte Biol. 56: 469
    OpenUrlAbstract
  11. ↵
    Chen, P., H. Ananthaswamy. 1993. Rejection of K1735 murine melanoma in syngeneic hosts requires expression of MHC class I antigens and either class II antigens or IL-2. J. Immunol. 151: 244
    OpenUrlAbstract
  12. ↵
    Baskar, S., L. Glimcher, N. Nabavi, R. T. Jones, S. Ostrand-Rosenberg. 1995. Major histocompatibility complex class II+B7-1+ tumor cells are potent vaccines for stimulating tumor rejection in tumor-bearing mice. J. Exp. Med. 181: 619
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Bevan, M.. 1976. Cross-priming for a secondary cytotoxic response to minor H antigens with H-2 congenic cells which do not cross-react in the cytotoxic assay. J. Exp. Med. 143: 1283
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Bevan, M.. 1976. Minor H antigens introduced on H-2 different stimulating cells cross-react at the cytotoxic T cell level during in vivo priming. J. Immunol. 117: 2233
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Ostrand-Rosenberg, S., A. Cohn. 1981. H-2 antigen expression on teratocarcinoma cells passaged in genetically resistant mice is regulated by lymphoid cells. Proc. Natl. Acad. Sci. USA 78: 7106
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Ozato, K., N. Mayer, D. Sachs. 1980. Hybridoma cell lines secreting monoclonal antibodies to mouse H-2 and Ia antigens. J. Immunol. 124: 533
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Ozato, K., D. Sachs. 1981. Monoclonal antibodies to mouse MHC antigens. J. Immunol. 126: 317
    OpenUrlAbstract
  18. ↵
    Landais, D., B. Beck, J. Buerstedde, S. Degraw, D. Klein, N. Koch, D. Murphy, M. Pierres, T. Tada, K. Yamamoto, C. Benoist, D. Mathis. 1986. The assignment of chain specificities for anti-Ia monoclonal antibodies using L cell transfectants. J. Immunol. 137: 3002
    OpenUrlAbstract
  19. ↵
    Armstrong, T., V. Clements, B. Martin, J. P.-Y. Ting, S. Ostrand-Rosenberg. 1997. Major histocompatibility complex class II-transfected tumor cells present endogenous antigen and are potent inducers of tumor-specific immunity. Proc. Natl. Acad. Sci. USA 120: 123
    OpenUrl
  20. ↵
    Johnson, N., A. Cavland, P. Allen, L. Glimcher. 1989. T cell receptor gene segment usage in a panel of hen-egg white lyosozyme specific I-Ak-restricted T helper hybridomas. J. Immunol. 142: 3298
    OpenUrlAbstract
  21. ↵
    Shastri, N., A. Oki, A. Miller, E. Sercarz. 1985. Distinct recognition phenotypes exist for T cell clones specific for small peptide regions of proteins. J. Exp. Med. 162: 332
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Ostrand-Rosenberg, S., V. Clements, L. Marr. 1986. 402AX teratocarcinoma MHC class I antigen expression is regulated in vivo by Lyt1, Lyt2, and L3T4 expressing splenic T cells. Cell. Immunol. 98: 257
    OpenUrlCrossRefPubMed
  23. ↵
    Oi, V., P. Jones, J. Goding, L. Herzenberg, L. Herzenberg. 1978. Properties of monoclonal antibodies to mouse Ig allotypes, H-2, and Ia antigens. Curr. Top. Microbiol. Immunol. 81: 115
    OpenUrlPubMed
  24. ↵
    Ozato, K., N. Mayer, D. Sachs. 1982. Monoclonal antibodies to mouse major histocompatibility complex antigens. Transplantation 34: 113
    OpenUrlCrossRefPubMed
  25. ↵
    Smith-Gill, S., T. Lavoie, C. Mainhart. 1984. Antigenic regions defined by monoclonal antibodies correspond to structural domains of avian lysozyme. J. Immunol. 133: 384
    OpenUrlAbstract
  26. ↵
    Allen, P., D. Strydom, E. Unanue. 1984. Processing of lysozyme by macrophages: identification of determinant recognized by two T-cell hybridomas. Proc. Natl. Acad. Sci. USA 81: 2489
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Gammon, G., H. Geysen, R. Apple, E. Pickett, M. Palmer, A. Ametani, E. Sercarz. 1991. T cell determinant structure: cores and determinant envelopes in three mouse MHC haplotypes. J. Exp. Med. 173: 609
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Moudgil, K., E. Sercarz. 1993. Dominant determinants in hen eggwhite lysozyme correspond to the cryptic determinants within its self-homologue, mouse lysozyme. J. Exp. Med. 178: 2131
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Lenschow, D., T. Walunas, J. Bluestone. 1996. CD28/B7 system of T cell costimulation. Annu. Rev. Immunol. 14: 233
    OpenUrlCrossRefPubMed
  30. ↵
    Baskar, S., V. Clements, L. Glimcher, N. Nabavi, S. Ostrand-Rosenberg. 1996. Rejection of MHC class II-transfected tumor cells requires induction of tumor-encoded B7-1 and/or B7-2 costimulatory molecules. J. Immunol. 156: 3821
    OpenUrlAbstract
  31. ↵
    Ostrand-Rosenberg, S., S. Baskar, N. Patterson, V. Clements. 1996. Expression of MHC class II and B7-1 and B7-2 costimulatory molecules accompanies tumor rejection and reduces the metastatic potential of tumor cells. Tissue Antigens 47: 414
    OpenUrlCrossRefPubMed
  32. ↵
    Lanzavecchia, A.. 1996. Mechanisms of antigen uptake for presentation. Curr. Opin. Immunol. 8: 348
    OpenUrlCrossRefPubMed
  33. ↵
    Yang, G., M. Mizuno, K. Hellstrom, L. Chen. 1997. B7-negative versus B7-positive P815 tumor: differential requirements for priming of an antitumor immune response in lymph nodes. J. Immunol. 158: 851
    OpenUrlAbstract
  34. ↵
    Cayeux, S., G. Richter, G. Noffz, B. Dorken, T. Blankenstein. 1997. Influence of gene-modified (IL-7, IL-4, and B7) tumor cell vaccines on tumor antigen presentation. J. Immunol. 158: 2834
    OpenUrlAbstract
  35. ↵
    Pulaski, B., K. Yeh, N. Shastri, K. Maltby, D. Penney, E. Lord, J. Frelinger. 1996. IL-3 enhances CTL development and class I MHC presentation of exogenous antigen by tumor-infiltrating macrophages. Proc. Natl. Acad. Sci. USA 93: 3669
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Huang, A., P. Golumbek, M. Ahmadzadeh, E. Jaffee, D. Pardoll, H. Levitsky. 1994. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science 264: 961
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Huang, A., A. Bruce, D. Pardoll, H. Levitsky. 1996. Does B7-1 expression confer antigen-presenting cell capacity to tumors in vivo?. J. Exp. Med. 183: 769
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Kundig, T., M. Bachmann, C. DiPaolo, J. Simard, M. Battegay, H. Lother, A. Gessner, K. Kuhlcke, P. Ohashi, H. Hengartner, R. Zinkernagel. 1995. Fibroblasts as efficient antigen-presenting cells in lymphoid organs. Science 268: 1343
    OpenUrlAbstract/FREE Full Text
View Abstract
PreviousNext
Back to top

In this issue

The Journal of Immunology
Vol. 160, Issue 2
15 Jan 1998
  • Table of Contents
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
MHC Class II-Transfected Tumor Cells Directly Present Antigen to Tumor-Specific CD4+ T Lymphocytes1
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
MHC Class II-Transfected Tumor Cells Directly Present Antigen to Tumor-Specific CD4+ T Lymphocytes1
Todd D. Armstrong, Virginia K. Clements, Suzanne Ostrand-Rosenberg
The Journal of Immunology January 15, 1998, 160 (2) 661-666;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
MHC Class II-Transfected Tumor Cells Directly Present Antigen to Tumor-Specific CD4+ T Lymphocytes1
Todd D. Armstrong, Virginia K. Clements, Suzanne Ostrand-Rosenberg
The Journal of Immunology January 15, 1998, 160 (2) 661-666;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Innate Immunity Together with Duration of Antigen Persistence Regulate Effector T Cell Induction
  • Regulatory Roles of IL-2 and IL-4 in H4/Inducible Costimulator Expression on Activated CD4+ T Cells During Th Cell Development
  • Induction of CD4+ T Cell Apoptosis as a Consequence of Impaired Cytoskeletal Rearrangement in UVB-Irradiated Dendritic Cells
Show more CELLULAR IMMUNOLOGY AND IMMUNE REGULATION

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • Public Access
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2021 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606