You are on page 1of 6

A critical role of T cell antigen receptor-transduced

MHC class I-restricted helper T cells


in tumor protection
Emma C. Morris*†, Aristotle Tsallios*, Gavin M. Bendle*, Shao-an Xue*, and Hans J. Stauss*
Department of Immunology, Imperial College, Du Cane Road, London W12 0NN, United Kingdom

Edited by N. Avrion Mitchison, University College London, London, United Kingdom, and approved April 20, 2005 (received for review January 14, 2005)

Adoptive transfer of antigen-specific CD4ⴙ and CD8ⴙ T cells is one provides help for CTL-mediated rejection of NP-expressing EL4
of the most efficient forms of cancer immunotherapy. However, tumors, whereas adoptive transfer of helper T cells producing high
the isolation of antigen-specific CD4ⴙ T cells is limited because only levels of IFN-␥ was ineffective.
few tumor-associated helper epitopes are identified. Here, we used
T cell antigen receptor gene transfer to target CD4ⴙ T cells against Materials and Methods
an MHC class I-presented epitope of a model tumor antigen. Mice. C57BL兾6 mice were purchased from Harlan (Harlan UK,
IFN-␥-producing CD4ⴙ T cells were unable to expand in vivo and to Loughborough, United Kingdom) and maintained in the animal
provide help for tumor rejection. In contrast, CD4ⴙ T cells produc- facility of Imperial College London. All procedures were carried
ing high levels of IL-2 expanded in vivo, provided help for cytotoxic out according to United Kingdom Home Office Regulations.
T lymphocyte-mediated tumor rejection, and developed T cell
memory. The data demonstrate in vivo synergy between T cell Peptides, Cells, Cell Lines, and Transfectants. The influenza virus A
antigen receptor-transduced CD4ⴙ and CD8ⴙ T cells specific for the NP-derived peptide (ASNENDAM, pNP366), the control peptides
same epitope resulting in long-term tumor protection. pMDM100 (YAMIYRNL) derived from the murine double
minute (MDM) 2 protein, and pSV9 (FAPGNYPAL) derived from
T cell antigen receptor gene transfer 兩 tumor immunotherapy the Sendai virus (12), were synthesized by ProImmune (Oxford).
Murine splenocytes derived from C57BL兾6 mice were cultured in

T he infusion of allogeneic T cells into patients with leukemia can


provide long-term leukemia-free survival. Similarly, the adop-
RPMI medium 1640, 10% FCS兾1% penicillin兾1% streptomycin兾2
mM L-glutamine. Murine bone marrow-derived dendritic cells
Downloaded from https://www.pnas.org by 143.208.36.172 on November 12, 2022 from IP address 143.208.36.172.

tive transfer of Epstein–Barr virus-specific donor-derived T cells (DCs) were isolated from C57BL兾6 mice and cultured for 6–7 days
has been successfully used to treat Epstein–Barr virus-induced in the presence of granulocyte兾macrophage colony-stimulating
malignancies after allogeneic stem cell transplantation (1, 2) and factor. The murine lymphoma EL4 tumor cell line expresses H2-Db
solid organ transplants (3–5). More recently, it was demonstrated MHC class I molecules but not MHC class II molecules (13). The
that adoptive T cell transfer into patients conditioned by lymphoa- stably transfected EL4 NP tumor cells express the influenza virus
blative chemotherapy resulted in strong antitumor effects in mel- A NP and were a kind gift from B. Stockinger (National Institute
anoma patients (6, 7). for Medical Research, London). Phoenix-Eco (PhEco) adherent
Indirect evidence suggests that the success of the adoptive packaging cells (Nolan Laboratory, Stanford University, Stanford,
immunotherapy treatments described above may be related to the CA) were transiently transfected with retroviral vectors for the
use of antigen-specific CD4⫹ and CD8⫹ T lymphocytes. For generation of cell supernatant containing the recombinant retro-
example, in melanoma the clinical response to infusion of CD8⫹ T virus required for infection of target cells.
cell clones was much less impressive than the effect of adoptive
transfer of mixed CD4⫹ and CD8⫹ T cells, although a direct Retroviral Transduction of Murine Splenocytes. The retroviral vector
comparison in patients after lymphoablative conditioning has not pMX-TCR␣-IRES-TCR␤ (pMX-F5) was a kind gift from T.
yet been performed (6, 8). Administration of IL-2 can improve the Schumacher (Nederlands Kanker Instituut, Amsterdam). The ␣-
antimelanoma efficacy of transferred CD8⫹ T cells (9), suggesting and ␤-chain TCR genes were cloned from the NP-specific CD8⫹
an important role of this cytokine that is typically produced by CTL clone F5 (14). The F5 TCR recognizes the influenza virus A
CD4⫹ T helper cells. Superiority of mixed CD4⫹ and CD8⫹ T cells NP (NP366–379) peptide in the context of murine Db MHC class
was also suggested by adoptive cell transfer experiments in immu- I. The PhEco packaging cell line was cotransfected by calcium
nocompromised patients at risk of CMV disease. The cell dose of phosphate precipitation with the pMX-F5 construct and the pCL-
cloned CD8⫹ T cells used to control CMV was ⬇1,000-fold higher Eco construct. Splenocytes were harvested from C57BL兾6 (H2-Db)
than the dose of CMV-specific CD8⫹ T cells given together with female mice and activated with conconavalin A (2 ␮g兾ml final
CMV-specific CD4⫹ T cells (10, 11). concentration) and IL-7 (1 ng兾ml final concentration) for 48–72 h.
The generation of tumor antigen-specific CD4⫹ helper T cells is After 48 h, the PhEco supernatant was harvested and used to
limited by the paucity of known MHC class II-binding tumor transduce activated splenocytes by coculture on fibronectin-treated
epitopes and the lack of expression of MHC class II molecules on tissue culture plates in the presence of IL-2 (100 units兾ml). The
most tumor cells. In contrast, a large number of CD8⫹ T cell- murine CD8␣ gene [a kind gift from R. Zamoyska (National
recognized tumor epitopes presented by MHC class I molecules has
been identified, and in many cases cytotoxic T lymphocyte (CTL)
clones against such epitopes were isolated. In this study, we ex- This paper was submitted directly (Track II) to the PNAS office.
plored in a murine model system whether the T cell antigen Abbreviations: TCR, T cell antigen receptor; TCR-td, TCR-transduced; mock-td, mock-
receptor (TCR) genes of tumor-specific CTL can be transferred to transduced; CTL, cytotoxic T lymphocyte; NP, nucleoprotein; pNP, peptide NP; PE, phyco-
CD4⫹ T cells to generate MHC class I-restricted, tumor-specific T erythrin; DC, dendritic cell; PhEco, Phoenix-Eco; MDM, murine double minute.
helper cells. We show that the TCR specific for an H2-Db-presented *Present address: Department of Immunology and Molecular Pathology, Royal Free Hos-
epitope of influenza nucleoprotein (NP) can be used to produce pital, University College London, London NW3 2QG, United Kingdom.

H2-Db-restricted, NP-specific CD4⫹ T cells. Adoptive transfer of †To whom correspondence should be addressed. E-mail: emma.morris@medsch.ucl.ac.uk.
CD4⫹ T cells producing high levels of IL-2 and low levels of IFN-␥ © 2005 by The National Academy of Sciences of the USA

7934 –7939 兩 PNAS 兩 May 31, 2005 兩 vol. 102 兩 no. 22 www.pnas.org兾cgi兾doi兾10.1073兾pnas.0500357102
Institute for Medical Research, London)] was cloned into the were added to 50 ␮l of harvested supernatant and incubated for 24 h
pMP71-pre retroviral vector [received from W. Uckert (Humboldt before [3H]thymidine pulsing and counting as above.
University, Berlin) and originally made by C. Baum (Medical
School Hannover, Hannover, Germany)]. Adoptive Immunotherapy. C57BL兾6 mice (Thy1.2) were condi-
tioned with 600 rad on day ⫺2. Twelve hours later (day ⫺1), the
Purification of TCR-Transduced (TCR-td) CD4ⴙ Cells and TCR-td CD8ⴙ mice were injected s.c. with a tumorigenic dose of EL4 NP cells (3 ⫻
Cells. Transduced splenocytes were stained with antimurine CD4, 106 cells in 200 ␮l of PBS). After a further 24 h, mice were injected
CD8, NP tetramers, and V␤11 antibodies [antimurine CD4-FITC, i.v. with Thy1.1⫹ TCR-td T cells, Thy1.1⫹ Mock-transduced (mock-
antimurine CD8-allophycocyanin, and antimurine V ␤ 11- td) T cells, or PBS alone (day 0). Mice were inspected daily and
phycoerythrin (PE) antibodies were obtained from Pharmingen killed when tumor burden exceeded 1 cm2 or if tumor ulceration
and used as directed]. The PE-labeled NP tetramer (Db兾 occurred regardless of tumor size. Representative surviving tumor-
ASNENMDAM) was synthesized by Proimmune and was used as free mice, were rechallenged with tumor cells on day ⫹90. These
directed. Forty-eight hours after transduction cells were sorted into mice were injected in the right lower leg with 1 ⫻ 106 irradiated EL4
CD4⫹ and CD8⫹ populations using PE-labeled anti-CD4 antibod- NP cells. After 5 days, mice were killed, and right and left inguinal
ies and anti-PE beads (Miltenyi Biotec, Bergisch Gladbach, Ger- and popliteal lymph nodes were stained with anti-Thy1.1 FITC,
many). Further enrichment for V␤11-expressing cells was per- anti-CD4-allophycocyanin, and anti-V␤11 PE antibodies before
formed 72 h after transduction. analysis by flow cytometry.

Functional Assays of TCR-td T Cells. For IFN-␥ release assays, 106 Results
DCs were incubated for 30 min in 200 ␮l of assay medium (RPMI Generation of TCR-td CD4ⴙ, CD4ⴙ8ⴙ, and CD8ⴙ T Cell Populations.
medium 1640 with 5% heat-inactivated FCS) with synthetic pep- Retroviral transduction of C57BL兾6 murine splenocytes with the
tides. TCR-td CD4⫹ or TCR-td CD8⫹ T cells (5 ⫻ 104) were H2-Db-restricted, NP-specific F5 TCR resulted in successful intro-
incubated with 5 ⫻ 104 peptide loaded DCs (1:1 ratio) in duplicate duction and surface expression of the TCR into CD8⫹ T cells and
CD4⫹ T cells. Typically, after transduction, 6–12% of viable CD3⫹

IMMUNOLOGY
or triplicate. After 24 h, the supernatant was harvested and tested
in an IFN-␥ ELISA assay by using anti-IFN-␥ antibodies (Pharm- T cells were V␤11⫹ CD4⫹, and 10–15% were V␤11⫹ CD8⫹, as
ingen). Further IFN-␥ release assays used peptide-loaded EL4 cells determined by flow cytometry (Fig. 1 A and B). In each experiment,
or EL4NP cells as targets. mock-td T cells were used to determine the percentage of CD4⫹
For proliferation assays, 106 stimulator cells were incubated for and CD8⫹ T cells expressing endogenous V␤11 (Fig. 1 D and E).
1 h in 200 ␮l of assay medium (as above) with 100 ␮M synthetic Tetramer analysis revealed that the F5 TCR-td splenocytes bound
peptide at 37°C. 5 ⫻ 104 TCR-td CD4⫹, or TCR-td CD8⫹ T cells the NP (366–374) H2-Db tetramers, whereas the mock-td spleno-
were incubated with 5 ⫻ 104 peptide-loaded DCs (1:1 ratio) or cytes did not (Fig. 1 C and F). Typically, only 40–50% of transduced
Downloaded from https://www.pnas.org by 143.208.36.172 on November 12, 2022 from IP address 143.208.36.172.

tumor cells in duplicate or triplicate. After 6, 24, or 48 h, 0.5 ␮Ci T cells expressing the introduced V␤11 chain were able to bind NP
(1 Ci ⫽ 37 GBq) [3H]thymidine was added to each well. The cells tetramers (Fig. 1, compare B, E, and C). TCR-td bulk T cells were
were harvested by using a 96-well plate harvester (Amersham sorted 48 h after transduction into CD4⫹ and CD8⫹ cells by using
Pharmacia), and thymidine incorporation was measured by using a PE-labeled anti-CD4 antibody staining and anti-PE magnetic
gamma counter (Wallace, Milton Keynes, United Kingdom). beads. After 24 h, the sorted populations were further enriched for
IL-2 secretion of TCR-td T cells was measured by using IL-2- V␤11⫹ cells, again by using magnetic bead selection. The TCR-td
dependent CTLL cells. The stimulation phase of the IL-2 assay was CD4⫹ T cells used in all subsequent experiments contained ⬎99%
performed exactly as the IFN-␥ and proliferation assays, and after CD4⫹ T cells, always with ⬍1% contaminating CD8⫹ T cells (Fig.
24, 48, or 72 h, the supernatant was harvested. CTLL cells (5 ⫻ 103) 1G). After enrichment, ⬎60% of the CD4⫹ T cells expressed the

Fig. 1. Retroviral gene transfer of TCR


chains, purification of TCR-expressing CD4⫹
and CD8⫹ T cells, and retroviral gene trans-
fer of CD8␣. (A–F) Flow cytometric analysis
of viable CD3⫹ murine splenocytes 2 days
after retroviral transfer with F5 TCR␣ and
TCR␤ genes (A–C) or mock transduction (D–
F). Cells were stained with PE-anti-V␤11,
PE-labeled NP tetramer, FITC-anti-CD4, or
allophycocyanin-anti-CD8. (G and H) Flow
cytometric analysis of purified CD4⫹ and
CD8⫹ T cells. Forty-eight hours after trans-
duction, TCR-td bulk T cells were sorted into
CD4⫹ and CD8⫹ T cells by using magnetic
beads, followed by enrichment for V␤11⫹
cells 1 day later. Cells were stained with
PE-anti-V␤11, FITC-anti-CD4, and兾or allo-
phycocyanin-anti-CD8, and quadrants
were set based on anti-CD4 and anti-CD8
stained mock-td cells. (I and J) Flow cyto-
metric analysis of purified TCR-td CD4⫹ T
cells (I) and TCR-td CD4⫹8⫹ T cells (J) after
retroviral cotransfer of F5 TCR␣, TCR␤, and
murine CD8␣ genes. After cotransfer of
CD8␣, ⬎95% of CD4⫹ T cells coexpressed
CD8␣ (TCR-td CD4⫹8⫹ T cells).

Morris et al. PNAS 兩 May 31, 2005 兩 vol. 102 兩 no. 22 兩 7935
V␤11. The TCR-td CD8⫹ T cell population was typically ⱖ95%
pure (Fig. 1H). In some experiments, we introduced the CD8␣ gene
together with the F5 TCR genes and purified CD4⫹ T cells
expressing TCR and CD8. Most CD4⫹ T cells were found to
coexpress CD8, as determined by flow cytometry (Fig. 1J). This
expression pattern allowed us to assess the functional activity of the
NP-specific, class I-restricted F5 TCR in CD4⫹ T cells and in CD4⫹
cells that also expressed CD8, referred to as CD4⫹8⫹ T cells. Each
functional experiment was preceded by flow cytometric analysis to
confirm purity of the TCR-td T cell populations.

In Vitro Functional Activity of TCR-td CD4ⴙ and CD8ⴙ T Cells. TCR-td


bulk T cells secrete IFN-␥ in response to syngeneic bone marrow-
derived DCs coated with NP peptide (pNP) but not in response to
an irrelevant peptide (pMDM-100) or no peptide. Similarly, with
peptide-coated EL4 tumor cells as stimulators, the TCR-td bulk T
cells produced IFN-␥ in response to pNP but not pMDM-100 or no
peptide. Mock-td T cells did not secrete IFN-␥ in response to any
of the above targets. (Fig. 2A). Functional assays were repeated with
purified TCR-td CD4⫹ T cells and TCR-td CD8⫹ T cells. Using
peptide-coated syngeneic DCs as stimulator cells, 10 pM of pNP
peptide was required to induce IFN-␥ secretion by CD8⫹ T cells,
whereas a 10-fold higher peptide concentration (100 pM) was
required to trigger IFN-␥ secretion by the CD4⫹ T cells (Fig. 2B).
Once triggered, TCR-td CD4⫹ T cells secreted amounts of IFN-␥
equivalent to the TCR-Td CD8⫹ T cells when stimulated with DC.
However, the TCR-td CD4⫹ T cells produced little IFN-␥ in
response to peptide-coated EL4 tumor cells (Fig. 2C) or to EL4
cells transfected with NP (data not shown), whereas the purified
TCR-td CD8⫹ T cells mounted an efficient IFN-␥ response to
peptide-coated EL4 cells and EL4-NP cells. We explored whether
Downloaded from https://www.pnas.org by 143.208.36.172 on November 12, 2022 from IP address 143.208.36.172.

transcostimulation and兾or the CD8 molecule could rescue the


antitumor IFN-␥ response of CD4⫹ T cells. The addition of Fig. 2. In vitro functional analysis of TCR-td bulk T cells, CD8⫹ T cells, CD4⫹
T cells, and CD4⫹8⫹ T cells. (A) Peptide-specific IFN-␥ secretion of TCR-td bulk
syngeneic DCs to the stimulation cultures restored the peptide-
T cells (black bars) in response to peptide-loaded DCs and MHC class-II negative
specific IFN-␥ secretion of CD4⫹ T cells to EL4-NP tumor cells peptide-loaded EL4 tumor cells. Mock-td bulk T cells (gray bars) did not secrete
(Fig. 2D). The same effect was observed with allogeneic DCs, IFN-␥ in response to any of the targets. (B) Purified populations of TCR-td CD8⫹
indicating that cross-presentation was not involved, and as few as T cells (black bars) and TCR-td CD4⫹ T cells (gray bars) secreted IFN-␥ in
one DC per 1,000 tumor cells was sufficient (data not shown). The response to peptide-loaded syngeneic DCs. TCR-td CD8⫹ T cells recognized 10
introduction of CD8 into TCR-td CD4⫹ T cells abolished the DC pM peptide, whereas a 10-fold higher concentration of peptide (100 pM) was
dependence of the IFN-␥ response. Purified TCR-td CD4 ⫹8⫹ T required to trigger IFN-␥ secretion by the TCR-td CD4⫹ T cells. (C) Using
cells produced IFN-␥ when stimulated with EL4-NP tumor cells in peptide-loaded EL4 tumor cells as targets, TCR-td CD8⫹ T cells (black bars)
the absence of DC. (Fig. 2E). mounted an efficient IFN-␥ response, whereas the TCR-td CD4⫹ T cells (gray
bars) did not. (D) Transcostimulation by means of the addition of syngeneic
Surprisingly, we found that the ability of T cells to produce IFN-␥
DCs rescued the peptide-specific IFN-␥ response of TCR-td CD4⫹ T cells (gray
did not correlate with their ability to proliferate after stimulation bars) to MHC class II negative NP-expressing EL4 tumor cells (EL4 NP) compared
with tumor cells. Despite the antitumor IFN-␥ response of TCR-td with that observed with TCR-td CD8⫹ T cells (black bars). (E) Coexpression
CD4 ⫹ 8⫹ T cells, these cells proliferated poorly when stimulated of murine CD8␣ abolished the DC dependence of the IFN-␥ response. TCR-td
with peptide presenting tumor cells (Fig. 3A). The opposite pattern CD4⫹8⫹ T cells (gray bars) and TCR-td CD8⫹ T cells (black bars) respond to
was observed for TCR-td CD4⫹ T cells, which proliferated effi- antigen presented by DCs and EL4 NP.
ciently after stimulation with tumor cells, although they produced
little IFN-␥ (Figs. 2C and 3A). The TCR-td CD8⫹ T cells efficiently
proliferated and produced IFN-␥ after stimulation with tumor cells. also received a small dose of 104 TCR-td CD8⫹ T cells, which was
There was a correlation between the proliferative potential of insufficient to cause tumor rejection in the absence of help. Tumor
helper T cells and their IL-2 production. Stimulation with tumor protection by large numbers of CTL (i.e., 106 TCR-td CD8⫹ T cells)
cells triggered high levels of IL-2 production in CD4⫹ T cells and was T helper cell-independent (data not shown).
reduced levels in CD4⫹8⫹ T cells (Fig. 3B). However, the CD4⫹8⫹ Fig. 4A shows poor survival of EL4-NP tumor-challenged mice
T cells were not defective in IL-2 production, suggesting that the that were treated with PBS (n ⫽ 5) or mock-td CD4⫹ T cells (n ⫽
poor proliferation may be related to an antiproliferative effect of 5). A small improvement was seen in the group of mice (n ⫽ 5) that
IFN-␥ produced at high levels by these cells (15, 16). was treated with 106 TCR-td CD4⫹8⫹ helper T cells together with
104 TCR-td CD8⫹ T cell. In contrast, substantial protection was
TCR-Td CD4ⴙ T Cells Provide in Vivo Help for CD8-Mediated Tumor seen in the five mice treated with 106 TCR-td CD4⫹ helper T cells
Rejection. We performed adoptive immunotherapy experiments in in combination with 104 TCR-td CD8⫹ T cells. To explore whether
a syngeneic setting to determine whether CD4⫹ T cells with an the NP specificity of the CD4⫹ T cells was required, tumor-
IFN-␥low兾proliferationhigh phenotype or CD4⫹8⫹ T cells with an challenged mice (n ⫽ 5) were treated in a separate experiment with
IFN-␥high兾proliferationlow phenotype were more efficient in pro- 106 mock-td CD4⫹ T cells combined with 104 TCR-td CD8⫹ T cells.
viding help for tumor rejection. Two groups of 5 C57BL兾6 mice This resulted in progressive tumor growth in all mice (Fig. 4B).
received a tumorigenic dose of EL4-NP cells followed by adoptive Similarly, treatment of five more mice with TCR-td CD4⫹ T cells
transfer of 106 purified TCR-td CD4⫹ or CD4⫹8⫹ helper T cells. in the absence of CD8⫹ T cells was ineffective, indicating that
To assess the helper function in vivo, a further group of five mice protection required the combination of antigen-specific CD4⫹ T

7936 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.0500357102 Morris et al.


IMMUNOLOGY
Fig. 3. In vitro proliferative potential and IL-2 production of TCR-td CD4⫹ T
cells and TCR-td CD4⫹8⫹ T cells. (A) TCR-td CD4⫹8⫹ T cells proliferated poorly Fig. 4. TCR-td CD4⫹ T cells provide in vivo help for CD8 T cell-mediated tumor
in response to peptide-presenting tumor cells (white bars) compared with rejection. C57BL兾6 mice received nonmyeloablative radiation (600 rad) and
TCR-td CD4⫹ T cells (gray bars) and TCR-td CD8⫹ T cells (black bars). The then 12 h later were injected s.c. with 3 ⫻ 106 EL4 NP tumor cells. (A) A further
proliferative potential of the TCR-td T cell populations correlated well with 24 h later, mice received 106 TCR-td CD4⫹ T cells with 104 TCR-td CD8⫹ T cells
IL-2 production. (B) TCR-td CD4⫹8⫹ T cells (white bars) secreted reduced levels (open triangles), 106 TCR-td CD4⫹8⫹ T cells with 104 TCR-td CD8⫹ T cells
of IL-2 in response to EL4 NP tumor cells as compared with TCR-td CD4⫹ T cells (crosses), 106 mock-td bulk T cells (open squares), or PBS (diamonds). (B) In a
(gray bars). TCR-td CD8⫹ T cells (black bars) did not secrete IL-2 in response to separate tumor protection experiment, mice were conditioned and tumor-
challenged as above, followed by transfer of 106 mock-td CD4⫹ T cells with 104
Downloaded from https://www.pnas.org by 143.208.36.172 on November 12, 2022 from IP address 143.208.36.172.

any of the targets. Proliferation of the IL-2-dependent cell line CTLL was used
to measure the IL-2 content in the supernatant taken from the stimulated T TCR-td CD8⫹ T cells (open squares), 106 TCR-td CD4⫹ T cells with anti-CD8-
cells. blocking antibody (filled diamonds), or 106 TCR-td CD4⫹ T cells with 2 ⫻ 104
TCR-td CD8⫹ T cells (open triangles).

cells together with small numbers of TCR-td CD8⫹ T cells. In-


creasing the dose of TCR-td CD8⫹ T cells from 104 to 2 ⫻ 104 provides for the production of tumor-specific T cell populations for
improved the level of tumor protection (Fig. 4, compare A with B). adoptive immunotherapy without the need for in vivo immunization
and in vitro T cell expansion. In a previous study (20) and in this
TCR-td CD4ⴙ T Cells Develop Long-Term T Cell Memory. Spleens of all report T cells were transduced once on day 2–3 after polyclonal
mice used in the tumor challenge experiments in Fig. 4A were activation and subsequently used on days 5–7 for adoptive transfer
analyzed by FACS to determine the number of adoptively trans- without any further in vitro expansion. TCR gene transfer also
ferred Thy1.1⫹兾V␤11⫹ TCR-td T cells at the time of antigen provides a strategy to generate antigen-specific helper T cells for
exposure in tumor-bearing mice and at later time points to analyze adoptive cancer immunotherapy. This strategy can be achieved by
T cell persistence and recall memory responses in tumor-free mice. transfer of MHC class II-restricted TCRs into CD4⫹ cells (19, 27)
No Thy1.1⫹兾V␤11⫹ T cells were identified in the control PBS- or, as shown recently, by transfer of MHC class I-restricted TCRs
treated mice (Fig. 5A). In tumor-bearing mice treated with mock-td
CD4⫹ T cells, between 0.5% and 1.7% of the gated V␤11⫹ cells into CD4⫹ T cells (28). One recent publication showed that TCRs
were the transferred T cells (Fig. 5B). A similar proportion of isolated from CD8-independent CTL can be functionally active in
transferred T cells, 0.5–3.3% of the gated V␤11⫹ cells, was seen in CD4⫹ T cells, and another study showed that the cotransfer of
three tumor-bearing mice treated with TCR-td CD4 ⫹ 8⫹ T cells, CD8␣ and a TCR兾CD3␨ hybrid construct can be used to produce
suggesting that there was no antigen-driven expansion of these cells HLA class I-restricted CD4⫹ T cells (29, 30). To date, a possible in
compared with mock-td CD4⫹ T cells (Fig. 5C). Analysis of a vivo role of CD4⫹ T cells transduced with a class I-restricted TCR
tumor-bearing mouse treated with TCR-td CD4⫹ T cells showed a in tumor protection has not yet been explored.
large expansion of transferred CD4⫹ cells (34% of gated V␤11⫹ T Here, we have generated MHC class I-restricted helper T cells
cells) and an expansion of the cotransferred TCR-td CD8⫹ T cells that respond to antigen-presentation by MHC class II-negative
(13.6% of V␤11⫹ cells) (Fig. 5D). EL4-NP tumor cells. TCR-td CD4⫹ T cells proliferated and pro-
Analysis of tumor-free mice 35 and 90 days after adoptive T cell duced high levels of IL-2 but little IFN-␥, whereas CD4⫹ T cells
transfer showed persistence and memory development of TCR-td transduced with the same TCR and CD8␣ produced high levels of
CD4⫹ T cells. On day 35, the transferred CD4⫹ T cells were present IFN-␥ and reduced levels of IL-2 and proliferated poorly when
at a higher frequency than CD4⫹8⫹ cells (Fig. 6 A and B), and
stimulated with tumor cells. The mechanism by which CD8␣
CD4⫹ but not CD4⫹8⫹ T cells responded to tumor rechallenge at
day 90 (Fig. 6 C–H). The memory response of the TCR-td CD4⫹ expression modifies the T cell response is currently unknown.
T cells at day 90 was associated with a recall response of the TCR-td Altered cytokine production might be due to the ability of CD8 to
CD8⫹ T cells (Fig. 6 F and H). enhance the avidity of TCR兾MHC peptide interaction. For exam-
ple, a recent study demonstrated that high avidity TCR兾MHC
Discussion peptide interactions selectively impaired IL-2 production, which is
Several groups have used retroviral TCR gene transfer to produce similar to our observation with CD8-expressing helper T cells (31).
antigen-specific human and murine T cells (17–27). This strategy Alternatively, the CD8 molecule may alter signal transduction in

Morris et al. PNAS 兩 May 31, 2005 兩 vol. 102 兩 no. 22 兩 7937
transduced CD4⫹ T cells reflect a feature of CD8␣兾␣ homodimers.
For example, the expression of CD8␣兾␣ homodimers in TCR-td
CD4⫹ cells may result in redirected homing to the gut, as is seen
with CD8␣兾␣ positive intraepithelial lyphocytes, which might result
in low numbers of T cells in lymphoid tissues. In fact, gene transfer
into CD4⫹ T cells will provide an interesting model to dissect
functional activities of CD8␣兾␣ homodimers and CD8␣兾␤ het-
erodimers in mature T cells.
We show that IL-2high兾proliferationhigh CD4⫹ T cells effectively
provide in vivo help for tumor rejection, whereas IFN-␥high兾
proliferationlow CD4⫹ T cells are unable to do so despite having the
same specificity. A similar in vivo protective effect has been
described for IL-2high兾proliferationhigh CD4⫹ T cells in HIV-
infected patients. Large numbers of these CD4⫹ T cells were
observed in patients controlling virus load, whereas the presence of
IFN-␥high兾proliferationlow CD4⫹ T cells was associated with un-
controlled virus replication (34–37).
In addition to the efficient help for tumor rejection, we also
showed that IL-2high兾proliferationhigh CD4⫹ T cells persist and
readily respond to tumor cell rechallenge 90 days after adoptive
Fig. 5. Detection and in vivo persistence of TCR-td T cells in tumor-bearing
transfer. This finding is in line with a recent report showing that
mice. Shown is the analysis of splenocytes prepared from mice in Fig. 4A at the
time they were killed because of tumor burden. Adoptively transferred T cells
murine CD4⫹ T cells producing little IFN-␥ were able to persist in
were identified by triple staining with antibodies specific for Thy1.1, V␤11, vivo and develop into long-term memory cells, whereas CD4⫹ T
and CD4. Representative examples are shown for each group of mice, and all cells producing high levels of IFN-␥ survived poorly in vivo (38).
FACS plots display viable lymphocytes gated on V␤11⫹ T cells. (A) No Thy1.1⫹ Our experiments suggest that an important mechanism by which
V␤11⫹ T cells were identified in four tumor-bearing PBS-treated control mice. proliferation competent helper T cells contribute to tumor immu-
(B) In four mice treated with mock-td CD4⫹ T cells, between 0.5% and 1.7% of nity is by triggering the in vivo expansion of antigen-specific CD8⫹
the gated V␤11⫹ cells were the CD4⫹ Thy1.1⫹ transferred T cells. (C) Similar T cells. Given separately, neither antigen-specific helper T cells nor
numbers of TCR-td CD4⫹8⫹ cells were detected in three mice (0.5–3.3%)
CD8⫹ T cells were able to mediate protection in tumor-challenged
suggesting a lack of antigen-driven expansion of the TCR-td CD4⫹8⫹ as
compared with the Mock-td CD4⫹ T cells. (D) Analysis of the spleen of a
mice. It is likely that antigen-specific IL-2 production by helper T
tumor-bearing animal that received TCR-td CD4⫹ T cells showed a large cells at the site of tumor growth may enhance local proliferation and
Downloaded from https://www.pnas.org by 143.208.36.172 on November 12, 2022 from IP address 143.208.36.172.

expansion of transferred Thy1.1⫹ CD4⫹ T cells (34.0% of gated V␤11⫹), to- retention of CD8⫹ T cells, as described previously in a murine
gether with an expansion of the cotransferred Thy1.1⫹ CD8⫹ T cells (13.6% of tumor model (39).
gated V␤11⫹). Staining of lymph node cells of mice in the four treatment In our experiments, TCR-td CD4⫹ and TCR-td CD8⫹ T cells
groups showed similar levels of transferred Thy1.1⫹ V␤11⫹ CD4⫹ T cells (data expanded and persisted at similar frequencies, even when 100-fold
not shown). more CD4⫹ T cells were adoptively transferred into the mice.
Although this result suggested a competitive advantage for CD8⫹
T cells, it is possible that some cross-regulation may determine the
helper T cells because of its ability to recruit LAT, an adaptor relative expansion rate of CD4⫹ and CD8⫹ cells. For example, the
molecule that preferentially associates with CD8 compared with adoptive transfer of 106 TCR-td CD4⫹ cells along with 104 or 106
CD4 (32, 33). It is possible that the observed functional changes of TCR-td CD8⫹ cells (i.e., 100:1 and 1:1 ratios) resulted in similar size

Fig. 6. TCR-td CD4⫹ T cells persist and


develop long-term memory in vivo.
Shown is the analysis of splenocytes and
lymph node cells prepared from tumor-
free mice in Fig. 4 A on day 35 or day 95
after T cell transfer. Adoptively trans-
ferred T cells were identified by triple
staining with antibodies specific for
Thy1.1, V␤11, and CD4, and all FACS
plots display viable lymphocytes gated
on V␤11⫹ T cells. (A and B) A day-35
comparison of splenocytes of mice
treated with TCR-td CD4 ⫹ (A) and
CD4⫹8⫹ (B) cells. Similar frequencies of
transferred Thy1.1⫹ CD4⫹ T cells were
detected in lymph node samples (data
not shown). (C–H) At day 90, three tu-
mor-free mice were challenged s.c. with
1 ⫻ 106 irradiated EL4 NP tumor cells in
the right lower leg. After 5 days, ex vivo
FACS analysis was used to analyze T
cell responses in the tumor-antigen-
exposed right inguinal and popliteal
lymph nodes, by using the nonexposed
left inguinal and popliteal lymph nodes as a baseline control. The TCR-td CD4⫹8⫹ T cells did not respond to tumor rechallenge at day 90 (C and D), whereas
both mice that received TCR-td CD4⫹ cells demonstrated an expansion of TCR-td CD4⫹ and CD8⫹ T cells in the lymph nodes of the tumor antigen exposed
right side (E–H).

7938 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.0500357102 Morris et al.


CD8⫹ T cell populations 5 weeks after T cell transfer. A simple both populations for tumor immunotherapy. In the current exper-
feedback loop may involve IL-2 that is provided by TCR-td CD4⫹ iments, T cells were directed against a model tumor antigen that is
T cells and that controls the extent of expansion of TCR-td CD8⫹ not expressed in normal tissues. In contrast, most human T cell-
T cells with the same specificity. recognized tumor antigens are also expressed in normal cells.
The observation that IFN-␥high helper T cells were unable to High-avidity CTL against such human tumor antigens have been
contribute to tumor protection was surprising, considering the isolated from melanoma patients (6, 43), or they can be isolated
important role of this cytokine in preventing tumor growth by from the allogeneic T cell repertoire in cases in which autologous
inhibition of tumor stroma (40). In addition, IFN-␥ can lead to FAS T cells are tolerant (44, 45). In principle, it is now possible to use
ligand (CD178) up-regulation in T cells and FAS (CD95) up- TCRs isolated from high-avidity CTL to produce antigen-specific
regulation in target cells, thus enhancing CTL killing of target cells CD4⫹ and CD8⫹ T cells for adoptive therapy. It will be interesting
by means of the FAS pathway (41, 42). However, our data do not to assess the antitumor effect of this combination therapy and to
rule out a role of IFN-␥ in tumor immunity. Rather, the data explore whether antigen-presentation by quiescent normal tissues
demonstrate that CD4⫹ T cells, which are unable to expand and poses an autoimmune risk, is ignored, or renders the transferred T
persist in vivo, cannot exert antitumor effects even when they cells unresponsive (46).
produce high levels of IFN-␥. It is possible that the tumor control, This study demonstrates that adoptive tumor immunotherapy
which is mediated by CD8⫹ T cells in our experiments, involves with CD4⫹ and CD8⫹ T cells expressing the same TCR is an
CD8⫹ T cell-derived IFN-␥. attractive concept to harness helper T cell functions and CTL
The ability to produce CD4⫹ and CD8⫹ T cells specific for the effector functions for tumor eradication and long-term immunity.
same tumor antigen provides a powerful model to dissect the
mechanisms of in vivo interaction between these T cell populations This work was supported by the Leukaemia Research Fund, Medical
and provides an opportunity to design strategies to optimally exploit Research Council, and Cancer Research UK.

1. Rooney, C. M., Smith, C. A., Ng, C. Y., Loftin, S., Li, C., Krance, R. A., 23. Schaft, N., Willemsen, R. A., de Vries, J., Lankiewicz, B., Essers, B. W.,

IMMUNOLOGY
Brenner, M. K. & Heslop, H. E. (1995) Lancet 345, 9–13. Gratama, J. W., Figdor, C. G., Bolhuis, R. L., Debets, R. & Adema, G. J. (2003)
2. Rooney, C. M., Smith, C. A., Ng, C. Y., Loftin, S. K., Sixbey, J. W., Gan, Y., J. Immunol. 170, 2186–2194.
Srivastava, D. K., Bowman, L. C., Krance, R. A., Brenner, M. K. & Heslop, 24. Orentas, R. J., Bircher, L. A. & Roskopf, S. (2003) Scand. J. Immunol. 58, 33–42.
H. E. (1998) Blood 92, 1549–1555. 25. Tahara, H., Fujio, K., Araki, Y., Setoguchi, K., Misaki, Y., Kitamura, T. &
3. Khanna, R., Bell, S., Sherritt, M., Galbraith, A., Burrows, S. R., Rafter, L., Yamamoto, K. (2003) J. Immunol. 171, 2154–2160.
Clarke, B., Slaughter, R., Falk, M. C., Douglass, J., et al. (1999) Proc. Natl. 26. Heemskerk, M. H., Hoogeboom, M., Hagedoorn, R., Kester, M. G., Willemze,
Acad. Sci. USA 96, 10391–10396. R. & Falkenburg, J. H. (2004) J. Exp. Med. 199, 885–894.
4. Haque, T., Taylor, C., Wilkie, G. M., Murad, P., Amlot, P. L., Beath, S., 27. Chamoto, K., Tsuji, T., Funamoto, H., Kosaka, A., Matsuzaki, J., Sato, T., Abe, H.,
McKiernan, P. J. & Crawford, D. H. (2001) Transplantation 72, 1399–1402. Fujio, K., Yamamoto, K., Kitamura, T., et al. (2004) Cancer Res. 64, 386–390.
Downloaded from https://www.pnas.org by 143.208.36.172 on November 12, 2022 from IP address 143.208.36.172.

5. Haque, T., Wilkie, G. M., Taylor, C., Amlot, P. L., Murad, P., Iley, A., Dombagoda, 28. Morgan, R. A., Dudley, M. E., Yu, Y. Y., Zheng, Z., Robbins, P. F., Theoret,
D., Britton, K. M., Swerdlow, A. J. & Crawford, D. H. (2002) Lancet 360, 436–442. M. R., Wunderlich, J. R., Hughes, M. S., Restifo, N. P. & Rosenberg, S. A.
6. Rosenberg, S. A. & Dudley, M. E. (2004) Proc. Natl. Acad. Sci. USA 101, (2003) J. Immunol. 171, 3287–3295.
14639–14645. 29. Kuball, J., Schmitz, F. W., Voss, R. H., Ferreira, E. A., Engel, R., Guillaume,
7. Dudley, M. E., Wunderlich, J. R., Robbins, P. F., Yang, J. C., Hwu, P., P., Strand, S., Romero, P., Huber, C., Sherman, L. A. & Theobald, M. (2005)
Schwartzentruber, D. J., Topalian, S. L., Sherry, R., Restifo, N. P., Hubicki, Immunity 22, 117–129.
A. M., et al. (2002) Science 298, 850–854. 30. Willemsen, R., Ronteltap, C., Heuveling, M., Debets, R. & Bolhuis, R. (2005)
8. Dudley, M. E., Wunderlich, J., Nishimura, M. I., Yu, D., Yang, J. C., Topalian, Gene. Ther. 12, 140–146.
S. L., Schwartzentruber, D. J., Hwu, P., Marincola, F. M., Sherry, R., et al. 31. La Gruta, N. L., Turner, S. J. & Doherty, P. C. (2004) J. Immunol. 172, 5553–5560.
(2001) J. Immunother. 24, 363–373. 32. Bosselut, R., Zhang, W., Ashe, J. M., Kopacz, J. L., Samelson, L. E. & Singer,
9. Yee, C., Thompson, J. A., Roche, P., Byrd, D. R., Lee, P. P., Piepkorn, M., Kenyon, A. (1999) J. Exp. Med. 190, 1517–1526.
K., Davis, M. M., Riddell, S. R. & Greenberg, P. D. (2000) J. Exp. Med. 192, 33. Bosselut, R., Kubo, S., Guinter, T., Kopacz, J. L., Altman, J. D., Feigenbaum,
1637–1644. L. & Singer, A. (2000) Immunity 12, 409–418.
10. Walter, E. A., Greenberg, P. D., Gilbert, M. J., Finch, R. J., Watanabe, K. S., 34. Rosenberg, E. S., Billingsley, J. M., Caliendo, A. M., Boswell, S. L., Sax, P. E.,
Thomas, E. D. & Riddell, S. R. (1995) N. Engl. J. Med. 333, 1038–1044. Kalams, S. A. & Walker, B. D. (1997) Science 278, 1447–1450.
11. Peggs, K. S., Verfuerth, S., Pizzey, A., Khan, N., Guiver, M., Moss, P. A. & 35. Palmer, B. E., Boritz, E., Blyveis, N. & Wilson, C. C. (2002) J. Virol. 76,
Mackinnon, S. (2003) Lancet 362, 1375–1377. 5925–5936.
12. Dahl, A. M., Beverley, P. C. & Stauss, H. J. (1996) J. Immunol. 157, 239–246. 36. Younes, S. A., Yassine-Diab, B., Dumont, A. R., Boulassel, M. R., Grossman,
13. Gorer, P. A. (1950) Br. J. Cancer 4, 372–379. Z., Routy, J. P. & Sekaly, R. P. (2003) J. Exp. Med. 198, 1909–1922.
14. Townsend, A. R., Gotch, F. M. & Davey, J. (1985) Cell 42, 457–467. 37. Day, C. L. & Walker, B. D. (2003) J. Exp. Med. 198, 1773–1777.
15. Liu, Y. & Janeway, C. A., Jr. (1990) J. Exp. Med. 172, 1735–1739. 38. Wu, C. Y., Kirman, J. R., Rotte, M. J., Davey, D. F., Perfetto, S. P., Rhee, E. G.,
16. Chu, C. Q., Wittmer, S. & Dalton, D. K. (2000) J. Exp. Med. 192, 123–128. Freidag, B. L., Hill, B. J., Douek, D. C. & Seder, R. A. (2002) Nat. Immunol. 3,
17. Clay, T. M., Custer, M. C., Sachs, J., Hwu, P., Rosenberg, S. A. & Nishimura, 852–858.
M. I. (1999) J. Immunol. 163, 507–513. 39. Shrikant, P. & Mescher, M. F. (1999) J. Immunol. 162, 2858–2866.
18. Cooper, L. J., Kalos, M., Lewinsohn, D. A., Riddell, S. R. & Greenberg, P. D. 40. Qin, Z. & Blankenstein, T. (2000) Immunity 12, 677–686.
(2000) J. Virol. 74, 8207–8212. 41. Mullbacher, A., Lobigs, M., Hla, R. T., Tran, T., Stehle, T. & Simon, M. M.
19. Fujio, K., Misaki, Y., Setoguchi, K., Morita, S., Kawahata, K., Kato, I., Nosaka, (2002) J. Immunol. 169, 145–150.
T., Yamamoto, K. & Kitamura, T. (2000) J. Immunol. 165, 528–532. 42. Roth, E. & Pircher, H. (2004) J. Immunol. 172, 1588–1594.
20. Kessels, H. W., Wolkers, M. C., van den Boom, M. D., van der Valk, M. A. & 43. Yee, C., Thompson, J. A., Byrd, D., Riddell, S. R., Roche, P., Celis, E. &
Schumacher, T. N. (2001) Nat. Immunol. 2, 957–961. Greenberg, P. D. (2002) Proc. Natl. Acad. Sci. USA 99, 16168–16173.
21. Stanislawski, T., Voss, R. H., Lotz, C., Sadovnikova, E., Willemsen, R. A., 44. Stauss, H. J. (1999) Immunol. Today 20, 180–183.
Kuball, J., Ruppert, T., Bolhuis, R. L., Melief, C. J., Huber, C., et al. (2001) Nat. 45. Gao, L., Bellantuono, I., Elsasser, A., Marley, S. B., Gordon, M. Y., Goldman,
Immunol. 2, 962–970. J. M. & Stauss, H. J. (2000) Blood 95, 2198–2203.
22. Heemskerk, M. H., Hoogeboom, M., de Paus, R. A., Kester, M. G., van der Hoorn, 46. Bendle, G. M., Holler, A., Pang, L. K., Hsu, S., Krampera, M., Simpson, E. &
M. A., Goulmy, E., Willemze, R. & Falkenburg, J. H. (2003) Blood 102, 3530–3540. Stauss, H. J. (2004) Cancer Res. 64, 8052–8056.

Morris et al. PNAS 兩 May 31, 2005 兩 vol. 102 兩 no. 22 兩 7939

You might also like