Adoptive TCR transfer against rapidly mutating targets, such as HIV-1 or cancer, must counteract corresponding immune escape. Hence, we generated T cells expressing two additional receptors (TETARs) specific for HIV-1 by TCR mRNA electroporation. An HLA-A2–restricted gag-specific TCR and an HLA-B13–restricted nef-specific TCR were chosen. When both TCRs were transfected simultaneously, strong competitive effects occurred that were overcome by replacing the human constant domains of one TCR with murine counterparts and adapting the amounts of TCR-RNA used for transfection. The resulting TETAR responded to both epitopes with cytokine secretion and cytotoxic function. Cell sorting revealed that one individual cell indeed recognized both epitopes. The T cells diminished their reactivity to each epitope after stimulation but sequentially killed targets that presented the gag epitope and then targets that presented the nef epitope, or vice versa. Taken together, TETARs represent a sophisticated tool to study TCR functionality and might be a useful strategy in immunotherapy.

For an intended cellular therapy approach, T cells can be reprogrammed with a new specificity by T-cell receptor (TCR) transfer. This was performed successfully by retroviral transduction of virus- and tumor-specific TCRs1,2  or alternatively by transient transfection with TCR-encoding mRNA.3-8 

Mutation-prone targets such as HIV-1 or tumors can evade immune surveillance by CD8+ T cells through introduction of escape mutations within CTL epitopes9-13  or complete antigen loss,14  respectively. Therefore, it would be of great advantage to target two epitopes simultaneously. The most direct approach would be to generate T cells expressing two additional receptors (TETARs) specific for different epitopes. Transferred TETARs with two different HIV-1 specificities could still recognize target cells even if one of the CTL epitopes was lost by mutation. To date, dual-specific human T cells have been generated by retroviral transfer of one TCR into T cells with a defined endogenous TCR specificity (eg, CMV, influenza), not to target the pathogen with two TCRs but to provide a constant low-dose triggering to promote T-cell survival in vivo.15,16 

Here, we generate HIV-1–specific human TETAR by simultaneous introduction of two TCRs by TCR mRNA electroporation. We used TCRs against the HLA-A2–restricted gag epitope SLYNTVATL (SL9)17  and the HLA-B13–restricted nef epitope RQDILDLWI (RI9).18  We demonstrate that TETARs are generated by transfer of a human TCR together with a murinized TCR, that both TCRs are functional in the same CD8+ T cell, and that TETARs could sequentially kill targets that presented the gag epitope and then targets that presented the nef epitope, or vice versa. To the best of our knowledge, this is the first time that human T cells have been artificially equipped with two new specificities.

Cells and reagents

CD8+ T cells were isolated and cultured as described previously.6  The EBV-transformed B-cell line e_0361MS (HLA-A2+/HLA-B13+) was cultured in R20 medium.19  The HLA-A2–restricted HIV-1-p17 (gag) SLYNTVATL peptide (SL9) and the HLA-B13–restricted HIV-1-nef RQDILDLWI peptide (RI9) were used.

TCR RNA electroporation

TCR-encoding sequences were cloned, transcribed in vitro, and transfected as described previously.4,5  See supplemental Methods for details (available on the Blood Web site; see the Supplemental Materials link at the top of the online article).

Flow cytometry

The gagTCR was detected with PE-labeled gag/HLA-A2 Dextramer according to the manufacturer's instructions (Immundex). Cell sorting was performed with a FACSAria II.

Functional T-cell assays

EBV-transformed B cells were loaded with 2 μg/mL of peptide for 1 hour at 37°C. Stimulations and cytokine detection were performed as described previously.20  Cytolytic activity was tested in standard 4- to 6-hour 51Cr-release assays as described previously,5,6  with peptide-loaded B cells as targets. Alternatively, TCR-RNA–transfected T cells were prestimulated for 2 hours with peptide-loaded B cells. The mixture of T and B cells was used as effector cells in a cytotoxicity assay.

TETARs can be generated with a murinized nefTCR in combination with the human gagTCR

To generate TETARs, we electroporated RNAs that encoded a gag/HLA-A2–specific TCR (gagTCR) and an nef/HLA-B13–specific TCR (nefTCR) into CD8+ T cells either alone or together at different ratios (Figure 1A). These cells were stimulated with peptide-loaded EBV-immortalized B cells, and cytokine secretion was determined (Figure 1A). T cells transfected with each individual TCR recognized their cognate peptide specifically (Figure 1). In contrast, transfection of both TCRs simultaneously resulted in considerably lower specific cytokine production (Figure 1A). The use of a 5-fold excess of 1 TCR restored recognition of the cognate peptide but further reduced or abolished recognition via the other TCR (Figure 1A). The same was the case for the reverse experiment (Figure 1A). Hence, the mere coexpression of two additional TCRs in one cell does not result in reactivity to both epitopes, which was not expected given the assumption that either TCR can signal independently, and rather indicates that some mechanism of competition or mutual interference exists.

Figure 1

Simultaneous transfection of two human TCRs, but not of one human TCR combined with one murinized TCR, leads to a competitive effect that impairs their function. (A) CD8+ T cells were transfected with the α and β chains of the human gagTCR, the human nefTCR, or a combination of the human gagTCR and the human nefTCR by RNA electroporation (RNA quantities as indicated), or (B) T cells were electroporated with 10 μg of TCR RNA encoding the gagTCR, the human (hum) nefTCR, or the murinized (mur) nefTCR, or 2 μg of the human or the murinized nefTCR combined with 12 μg of the gagTCR. These T cells were used as effector cells in a cytokine-production assay 2-4 hours after electroporation. The transfection volume was equalized in all transfections with water. EBV-transformed B cells, either loaded with the gag peptide (solid bars) or with the nef peptide (open bars), were used as stimulator cells at a ratio of 1:1 to the effector cells. TNF secretion into the supernatant was measured with a cytometric bead array after overnight incubation. Cytokine production by CD8+ T cells transfected with 10 μg of the human gagTCR and stimulated with the gag peptide (mean value TNF secretion 15.6 ng/mL) was set to 100%, and other values were normalized to that. Average values of 3 independent standardized experiments ± SEM are shown.

Figure 1

Simultaneous transfection of two human TCRs, but not of one human TCR combined with one murinized TCR, leads to a competitive effect that impairs their function. (A) CD8+ T cells were transfected with the α and β chains of the human gagTCR, the human nefTCR, or a combination of the human gagTCR and the human nefTCR by RNA electroporation (RNA quantities as indicated), or (B) T cells were electroporated with 10 μg of TCR RNA encoding the gagTCR, the human (hum) nefTCR, or the murinized (mur) nefTCR, or 2 μg of the human or the murinized nefTCR combined with 12 μg of the gagTCR. These T cells were used as effector cells in a cytokine-production assay 2-4 hours after electroporation. The transfection volume was equalized in all transfections with water. EBV-transformed B cells, either loaded with the gag peptide (solid bars) or with the nef peptide (open bars), were used as stimulator cells at a ratio of 1:1 to the effector cells. TNF secretion into the supernatant was measured with a cytometric bead array after overnight incubation. Cytokine production by CD8+ T cells transfected with 10 μg of the human gagTCR and stimulated with the gag peptide (mean value TNF secretion 15.6 ng/mL) was set to 100%, and other values were normalized to that. Average values of 3 independent standardized experiments ± SEM are shown.

Close modal

Competitive effects strongly depend on the chosen TCRs (unpublished observation), and here, the nefTCR appeared dominant over the gagTCR. To overcome this problem, we used an excess of the gagTCR but improved the proper pairing and the CD3 binding of the nefTCR by replacing the human constant domains with murine ones,21  which resulted in a murinized nefTCR. Consequently, T cells transfected with adapted amounts of mRNA of the human gagTCR and the murinized nefTCR were able to recognize both epitopes with similar efficiency as T cells transfected with only one receptor, as indicated by TNF production (Figure 1B). In contrast, the completely human nefTCR lost its function when combined with an excess of the gagTCR (Figure 1B). These results indicate that it was possible to generate TETARs by cotransfection of a human TCR and a murinized TCR.

TETARs functionally express both TCRs on the same cell and can successively kill target cells via each TCR

To prove that one cell expresses both TCRs, we stained gag- and nef-specific TETARs with a gag/HLA-A2 Dextramer and sorted them by FACS. Those T cells that bound the gag/HLA-A2 Dextramer were subsequently able to recognize target cells loaded with gag peptide, a combination of gag and nef peptide, but also target cells loaded only with the nef peptide, as indicated by IL-2 secretion (Figure 2A). T cells transfected with only the gagTCR recognized only target cells that were loaded with gag peptide or the combination of gag and nef peptide (Figure 2A). These results clearly show that both TCRs were functional in the same cell. This could counteract the immune escape by antigen mutation, because both antigens would have to mutate simultaneously. HIV-1 reverse transcriptase mutates 1 per 1700 nucleotides.22  In theory, the chance that a 9-mer CTL epitope is altered is roughly 1/100, disregarding amino acids conserved for functional properties; however, the chance that a viral variant arises that bears mutations in 2 recognized epitopes is much lower (ie, 1/100 × 1/100 = 1/10 000). If, in contrast, two single-specific T-cell populations were present, they would not necessarily encounter the target simultaneously, so the mutations could occur slightly after each other, making escape mutant selection presumably more effective. The same would apply to tumor-specific TETARs.

Figure 2

TETARs functionally expresses both introduced TCRs simultaneously and retains the ability to kill target cells with the second TCR after antigen-specific down-modulation of the first TCR. (A) CD8+ T cells were transfected with the gagTCR or with a combination of the gagTCR and the murinized (mur) nefTCR (quantities as indicated). RNA-transfected CD8+ T cells were stained with the gag/HLA-A2 dextramer and sorted for dextramer-positive cells 2-3 hours after electroporation. These positive cells were used as effector cells in a cytokine production assay 6 hours after electroporation. Irradiated EBV-transformed B cells loaded with gag peptide, nef peptide, a combination of gag and nef peptide, or, as a negative control, an influenza nucleoprotein (INP) peptide (CTELKLSDY) were used as stimulator cells at a ratio of 1:1 to the effector cells. IL-2 release was measured by a cytometric bead array after overnight stimulation. Cytokine production by CD8+ T cells transfected with gagTCR and stimulated with gag peptide (mean value IL-2 secretion 3.5 ng/mL) was set to 100%, and other values were normalized to that. Average values of 4 standardized independent experiments ± SEM are shown. (B-C) CD8+ T cells were transfected with the human gagTCR, the murinized nefTCR, or a combination of the gagTCR and the murinized nefTCR (as indicated). Twelve to 16 hours after electroporation, these reprogrammed T cells were cocultured at a ratio of 1:1 with irradiated EBV-transformed B cells loaded with gag (open bars) or nef (solid bars) peptide. Two hours after stimulation, these cells were used in a standard 4-6–hour cytotoxicity assay in which additionally Cr51-labeled EBV-transformed B cells were added. These labeled B cells were loaded with the gag peptide (B) or the nef peptide (C) and served as target cells at a ratio of 1:60 (target-to-effector ratio). Data of 1 representative experiment of 3 are shown. Error bars indicate the SD of triplicate values. Negative values were set to zero.

Figure 2

TETARs functionally expresses both introduced TCRs simultaneously and retains the ability to kill target cells with the second TCR after antigen-specific down-modulation of the first TCR. (A) CD8+ T cells were transfected with the gagTCR or with a combination of the gagTCR and the murinized (mur) nefTCR (quantities as indicated). RNA-transfected CD8+ T cells were stained with the gag/HLA-A2 dextramer and sorted for dextramer-positive cells 2-3 hours after electroporation. These positive cells were used as effector cells in a cytokine production assay 6 hours after electroporation. Irradiated EBV-transformed B cells loaded with gag peptide, nef peptide, a combination of gag and nef peptide, or, as a negative control, an influenza nucleoprotein (INP) peptide (CTELKLSDY) were used as stimulator cells at a ratio of 1:1 to the effector cells. IL-2 release was measured by a cytometric bead array after overnight stimulation. Cytokine production by CD8+ T cells transfected with gagTCR and stimulated with gag peptide (mean value IL-2 secretion 3.5 ng/mL) was set to 100%, and other values were normalized to that. Average values of 4 standardized independent experiments ± SEM are shown. (B-C) CD8+ T cells were transfected with the human gagTCR, the murinized nefTCR, or a combination of the gagTCR and the murinized nefTCR (as indicated). Twelve to 16 hours after electroporation, these reprogrammed T cells were cocultured at a ratio of 1:1 with irradiated EBV-transformed B cells loaded with gag (open bars) or nef (solid bars) peptide. Two hours after stimulation, these cells were used in a standard 4-6–hour cytotoxicity assay in which additionally Cr51-labeled EBV-transformed B cells were added. These labeled B cells were loaded with the gag peptide (B) or the nef peptide (C) and served as target cells at a ratio of 1:60 (target-to-effector ratio). Data of 1 representative experiment of 3 are shown. Error bars indicate the SD of triplicate values. Negative values were set to zero.

Close modal

Because T cells transiently lose their ability to bind to their specific epitope after stimulation,23  we examined whether TETARs could successively kill targets via their two specificities. We stimulated T cells that expressed the human gagTCR, the murinized nefTCR, or both for 2 hours with peptide-loaded B cells. Then, Cr51-labeled B cells, loaded with the same epitope or the other epitope, were added. T cells transfected with only one TCR displayed an abrogated or reduced lytic activity against their epitope when preincubated with cold targets that presented the same epitope. The same effect was observed with TETARs. In contrast, when TETARs were prestimulated with nef-peptide–loaded targets, they still lysed gag-peptide–loaded B cells. In turn, when TETARs were prestimulated with gag-peptide–loadedtargets, their ability to lyse a second wave of nef-loaded cells was better than that of TETARs prestimulated with nef peptide (Figure 2B-C). These data provide evidence that TETARs obtained the ability to kill target cells via both of the newly introduced receptors and that the down-modulation of a TCR after antigen encounter is a process that acts in cis on a TCR complex but not in trans from one TCR complex to another on the same cell. The fact that TETARs can kill via the second TCR after down-modulation of the first one should also clearly improve their ability to obliterate the intended targets. These findings confirm in the human system the results of Gladow and coworkers,24  who showed that TCR down-modulation on murine transgenic dual-specific T cells depends primarily on binding of the specific ligand, and the results of Hah and coworkers,25  who showed indirectly that tolerization is receptor specific in a related murine model.

To the best of our knowledge, this is the first work describing the reprogramming of human T cells with two new specificities, but whether this approach will result in better in vivo functionality needs to be addressed in further experiments. If not efficient against HIV-1, the treatment of cancer with TETARs appears promising, because tumors can also escape immune surveillance by generation of mutation variants.14 

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank Wolfgang Uckert for the murine TCR constant domains; Kris Thielemans for the pGEM4Z-5′UTR-sig-huSurvivin-DC.LAMP-3′UTR vector; Stefanie Baumann, Ina Müller, Tanja Moritz, Verena Wellner, and Kathrin Zitzelsberger for technical assistance; Barbara Schmidt for performing accompanying experiments not included in the manuscript; Kathrin Pritschet and Philipp Schuster for providing reagents; Stefan Schliep, Michael Erdmann, Stina Rosenheinrich, Sandra Schiemann, Margit Lamm, and Doris Schuster for collection of blood; and Katrin Birkholz, Jennifer Etschel, Stefanie Böhm, Christian Krug, Sandra Müller-Schmucker, Isabell Pfeiffer, and Sabrina Prommersberger for fruitful discussions.

Financial support was provided by the ELAN (Erlanger Leistungsbezogene Anschubfinanzierung und Nachwuchsförderung) fund of the Friedrich-Alexander-University of Erlangen-Nuremberg (DE-06.03.29.1), the DFG (German Research Foundation) Graduiertenkolleg 1071 (Viruses of the Immune System, project B1), DFG grant HA 2331/2-1, DFG grant SCHA1247/1-1, the German Competence Network for HIV/AIDS (HIVNET), the Interdisciplinary Center for Clinical Research (IZKF) Erlangen (T.H., project A27), and the Hector Foundation (T.H.).

Contribution: C.H., S.H., A.H., S.B., and E.H. performed research; C.H., G.S., J.D, N.S., and T.H. designed research; and C.H., J.D., and N.S. wrote the manuscript.

Conflict-of-interest statement: The authors declare no competing financial interests.

Correspondence: Niels Schaft, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, 91052 Erlangen, Germany; e-mail: niels.schaft@uk-erlangen.de.

1
Hawkins
 
RE
Gilham
 
DE
Debets
 
R
, et al. 
Development of adoptive cell therapy for cancer: a clinical perspective.
Hum Gene Ther
2010
, vol. 
21
 
6
(pg. 
665
-
672
)
2
Schumacher
 
TN
T-cell-receptor gene therapy.
Nat Rev Immunol
2002
, vol. 
2
 
7
(pg. 
512
-
519
)
3
Birkholz
 
K
Hombach
 
A
Krug
 
C
, et al. 
Transfer of mRNA encoding recombinant immunoreceptors reprograms CD4+ and CD8+ T cells for use in the adoptive immunotherapy of cancer.
Gene Ther
2009
, vol. 
16
 
5
(pg. 
596
-
604
)
4
Birkholz
 
K
Hofmann
 
C
Hoyer
 
S
, et al. 
A fast and robust method to clone and functionally validate T-cell receptors.
J Immunol Methods
2009
, vol. 
346
 
1-2
(pg. 
45
-
54
)
5
Hofmann
 
C
Harrer
 
T
Kubesch
 
V
, et al. 
Generation of HIV-1-specific T cells by electroporation of T-cell receptor RNA.
AIDS
2008
, vol. 
22
 
13
(pg. 
1577
-
1582
)
6
Schaft
 
N
Dorrie
 
J
Muller
 
I
, et al. 
A new way to generate cytolytic tumor-specific T cells: electroporation of RNA coding for a T cell receptor into T lymphocytes.
Cancer Immunol Immunother
2006
, vol. 
55
 
9
(pg. 
1132
-
1141
)
7
Zhao
 
Y
Zheng
 
Z
Khong
 
HT
Rosenberg
 
SA
Morgan
 
RA
Transduction of an HLA-DP4-restricted NY-ESO-1-specific TCR into primary human CD4+ lymphocytes.
J Immunother
2006
, vol. 
29
 
4
(pg. 
398
-
406
)
8
Morgan
 
RA
Dudley
 
ME
Wunderlich
 
JR
, et al. 
Cancer regression in patients after transfer of genetically engineered lymphocytes.
Science
2006
, vol. 
314
 
5796
(pg. 
126
-
129
)
9
Moore
 
CB
John
 
M
James
 
IR
, et al. 
Evidence of HIV-1 adaptation to HLA-restricted immune responses at a population level.
Science
2002
, vol. 
296
 
5572
(pg. 
1439
-
1443
)
10
Allen
 
TM
Altfeld
 
M
Geer
 
SC
, et al. 
Selective escape from CD8+ T-cell responses represents a major driving force of human immunodeficiency virus type 1 (HIV-1) sequence diversity and reveals constraints on HIV-1 evolution.
J Virol
2005
, vol. 
79
 
21
(pg. 
13239
-
13249
)
11
Feeney
 
ME
Tang
 
Y
Roosevelt
 
KA
, et al. 
Immune escape precedes breakthrough human immunodeficiency virus type 1 viremia and broadening of the cytotoxic T-lymphocyte response in an HLA-B27-positive long-term-nonprogressing child.
J Virol
2004
, vol. 
78
 
16
(pg. 
8927
-
8930
)
12
Maurer
 
K
Harrer
 
EG
Goldwich
 
A
, et al. 
Role of cytotoxic T-lymphocyte-mediated immune selection in a dominant human leukocyte antigen-B8-restricted cytotoxic T-lymphocyte epitope in Nef.
J Acquir Immune Defic Syndr
2008
, vol. 
48
 
2
(pg. 
133
-
141
)
13
Koenig
 
S
Conley
 
AJ
Brewah
 
YA
, et al. 
Transfer of HIV-1-specific cytotoxic T lymphocytes to an AIDS patient leads to selection for mutant HIV variants and subsequent disease progression.
Nat Med
1995
, vol. 
1
 
4
(pg. 
330
-
336
)
14
Marchand
 
M
Brichard
 
V
van Baren
 
N
Coulie
 
PG
Biological and clinical developments in melanoma vaccines.
Expert Opin Biol Ther
2001
, vol. 
1
 
3
(pg. 
497
-
510
)
15
van Loenen
 
MM
Hagedoorn
 
RS
Kester
 
MG
, et al. 
Kinetic preservation of dual specificity of coprogrammed minor histocompatibility antigen-reactive virus-specific T cells.
Cancer Res
2009
, vol. 
69
 
5
(pg. 
2034
-
2041
)
16
Langerman
 
A
Callender
 
GG
Nishimura
 
MI
Retroviral transduction of peptide stimulated T cells can generate dual T cell receptor-expressing (bifunctional) T cells reactive with two defined antigens.
J Transl Med
2004
, vol. 
2
 
1
pg. 
42
 
17
Brander
 
C
Yang
 
OO
Jones
 
NG
, et al. 
Efficient processing of the immunodominant, HLA-A*0201-restricted human immunodeficiency virus type 1 cytotoxic T-lymphocyte epitope despite multiple variations in the epitope flanking sequences.
J Virol
1999
, vol. 
73
 
12
(pg. 
10191
-
10198
)
18
Harrer
 
EG
Bergmann
 
S
Eismann
 
K
, et al. 
A conserved HLA B13-restricted cytotoxic T lymphocyte epitope in Nef is a dominant epitope in HLA B13-positive HIV-1-infected patients.
AIDS
2005
, vol. 
19
 
7
(pg. 
734
-
735
)
19
Kolowos
 
W
Schmitt
 
M
Herrman
 
M
, et al. 
Biased TCR repertoire in HIV-1-infected patients due to clonal expansion of HIV-1-reverse transcriptase-specific CTL clones.
J Immunol
1999
, vol. 
162
 
12
(pg. 
7525
-
7533
)
20
Birkholz
 
K
Schwenkert
 
M
Kellner
 
C
, et al. 
Targeting of DEC-205 on human dendritic cells results in efficient MHC class II-restricted antigen presentation.
Blood
2010
, vol. 
116
 
13
(pg. 
2277
-
2285
)
21
Cohen
 
CJ
Zhao
 
Y
Zheng
 
Z
Rosenberg
 
SA
Morgan
 
RA
Enhanced antitumor activity of murine-human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability.
Cancer Res
2006
, vol. 
66
 
17
(pg. 
8878
-
8886
)
22
Roberts
 
JD
Bebenek
 
K
Kunkel
 
TA
The accuracy of reverse transcriptase from HIV-1.
Science
1988
, vol. 
242
 
4882
(pg. 
1171
-
1173
)
23
Drake
 
DR
Ream
 
RM
Lawrence
 
CW
Braciale
 
TJ
Transient loss of MHC class I tetramer binding after CD8+ T cell activation reflects altered T cell effector function.
J Immunol
2005
, vol. 
175
 
3
(pg. 
1507
-
1515
)
24
Gladow
 
M
Uckert
 
W
Blankenstein
 
T
Dual T cell receptor T cells with two defined specificities mediate tumor suppression via both receptors.
Eur J Immunol
2004
, vol. 
34
 
7
(pg. 
1882
-
1891
)
25
Hah
 
C
Kim
 
M
Kim
 
K
Induction of peripheral tolerance in dual TCR T cells: an evidence for non-dominant signaling by one TCR.
J Biochem Mol Biol
2005
, vol. 
38
 
3
(pg. 
334
-
342
)

Author notes

*

J.D., N.S., and T.H. share senior authorship.

Sign in via your Institution