Abstract
The avidity of the T-cell receptor (TCR) for antigenic peptides presented by the peptide–MHC (pMHC) on cells is a key parameter for cell-mediated immunity. Yet a fundamental feature of most tumor antigen-specific CD8+ T cells is that this avidity is low. In this study, we addressed the need to identify and select tumor-specific CD8+ T cells of highest avidity, which are of the greatest interest for adoptive cell therapy in patients with cancer. To identify these rare cells, we developed a peptide–MHC multimer technology, which uses reversible Ni2+-nitrilotriacetic acid histidine tags (NTAmers). NTAmers are highly stable but upon imidazole addition, they decay rapidly to pMHC monomers, allowing flow-cytometric–based measurements of monomeric TCR–pMHC dissociation rates of living CD8+ T cells on a wide avidity spectrum. We documented strong correlations between NTAmer kinetic results and those obtained by surface plasmon resonance. Using NTAmers that were deficient for CD8 binding to pMHC, we found that CD8 itself stabilized the TCR–pMHC complex, prolonging the dissociation half-life several fold. Notably, our NTAmer technology accurately predicted the function of large panels of tumor-specific T cells that were isolated prospectively from patients with cancer. Overall, our results demonstrated that NTAmers are effective tools to isolate rare high-avidity cytotoxic T cells from patients for use in adoptive therapies for cancer treatment. Cancer Res; 75(10); 1983–91. ©2015 AACR.
Introduction
Binding of T-cell receptor (TCR) to peptide–MHC (pMHC) complex is the key step for T-cell activation and cellular immune responses (1). Efficient triggering of T-cell responses critically depends on the strength of TCR-mediated antigen recognition, namely it has been shown that strong binding to pMHC confers superior effector function than weak interactions (2–7). This is of particular interest for immunotherapy based on adoptive T-cell transfer, aiming to convey immune reactivity against tumor-associated antigens, for which endogenous T-cell responses are usually weak. Many tumor antigens are in fact self-antigens that are expressed in the thymus, and accordingly, most tumor-reactive T cells of high avidity become negatively selected, in contrast with pathogen-specific T cells (8). Therefore, there is a need for a robust technology that allows rapid identification and isolation of CD8+ T cells expressing TCRs capable to efficiently activate and enhance T-cell function against malignant cells.
TCR–pMHC binding parameters are typically assessed by surface plasmon resonance (SPR), which requires laborious and expensive production of soluble TCRs and ignores the TCR–pMHC avidity effects related to CD8 coreceptor binding. On the other hand, kinetic measurements using pMHC tetramers or multimers have been extensively studied on the surface of antigen-specific CD8+ T cells, but due to the multivalent and heterogeneous composition of these multimers, this approach does not allow to accurately determine TCR–pMHC dissociation rates (9). Binding and dissociation measurements should ideally be performed using monomeric pMHC complexes. Because TCR–pMHC interactions typically exhibit weak affinities and fast dissociation rates, this has long precluded conclusive measurements with monomeric pMHCs (10).
Nauerth and colleagues (11) recently measured monomeric TCR–pMHC dissociation kinetics using reversible Streptamers and reported that virus-specific CD8+ T cells with longer half-lives (low koff) conferred increased functional avidity and better in vivo protection than T cells exhibiting shorter t1/2 (high koff). However, the Streptamer assay (11) needs a significant lag time until monomeric TCR–pMHC dissociation starts to become detectable, limiting thereby off-rate analyses to antigen-specific T cells of relative long half-lives, typically found in immune responses against pathogens. Moreover, accurate measurements of TCR–pMHC binding parameters on living T cells (11–14) require specialized equipment, which is currently not available for the high-throughput screen of antigen-specific T-cell populations.
To overcome these limitations, we here applied pMHC multimers built on reversible chelate complexes of Ni2+-nitriloacetic acid (NTA) with oligohistidines (15) allowing the efficient and direct cytometry-based assessment of monomeric TCR–pMHC dissociation kinetics on the surface of (self) tumor-specific CD8+ T cells. We found that the NTAmer technology accurately predicted T-cell biologic responses within a large panel of tumor-specific T-cell clones, providing novel means for the direct isolation of rare functionally relevant CD8+ T cells for adoptive cell transfer therapy.
Materials and Methods
Ethics statement
The three HLA-A*0201–positive patients had stage III/IV metastatic melanoma and were included in immunotherapy studies (patient LAU 50, NCT00112242; patient LAU 155, NCT00002669; and patient LAU 618, NCT00112229; www.clinicaltrials.gov). The studies were designed and conducted according to the relevant regulatory standards, upon approval by the ethical commissions and regulatory agency of Switzerland. Patient recruitment, study procedures, and blood withdrawal were done upon written informed consent.
Primary bulk CD8+ T cells and generation of tumor-specific CD8+ T-cell clones
Human primary HLA-A*0201pos CD8+ T lymphocytes were obtained following positive enrichment using anti–CD8-coated magnetic microbeads (Miltenyi Biotec), and cultured in RPMI supplemented with 8% human serum (HS) and 150 U/mL recombinant human IL2. HLA-A*0201-NTAmerpos (NY-ESO-1157–165–specific or Melan-AMART-1-26–35–specific) T cells from melanoma patient LAU 50 and LAU 618, respectively, were sorted by flow cytometry, cloned by limiting dilution and expanded in RPMI-1640 medium supplemented with 8% HS, 150 U/mL recombinant human IL2, 1 μg/mL phytohemagglutinin (PHA; Oxoid), and irradiated (30 Gy) allogeneic PBMCs as feeder cells. For patient LAU 155, twenty HLA-A*0201/NY-ESO-1157–165–specific T-cell clones expressing dominant TCR BV1, BV8, and BV13 clonotypes and a nondominant TCR BV2 clonotype were selected from our previously generated database of T-cell clones (16), thawed and in vitro expanded before further use. Primary bulk CD8+ T cells and tumor-specific T-cell clones were expanded by periodic restimulation with 30-Gy irradiated allogeneic PBMCs and 1 μg/mL PHA.
Lentiviral production and cell transduction
Full-length codon-optimized TCR AV23.1 and TCR BV13.1 chain sequences of a dominant HLA-A*0201/NY-ESO-1157–165 specific T-cell clone of patient LAU 155 (16) were cloned in the pRRL third-generation lentiviral vectors as an hPGK-AV23.1-IRES-BV13.1 construct and structure-based amino acid substitutions were introduced into the WT TCR sequence as described previously (5). Lentiviral production was performed using the calcium-phosphate method and concentrated supernatant of lentiviral-transfected 293T cells was used to infect TCRα knockout CD8+ SUP-T1 cells (ATCC number CRL-1942, mycoplasma free), CD8−null Jurkat T cells (ATCC number TIB-152; mycoplasma free), or primary CD8+ T cells overnight. Levels of transduced TCR expression on SUP-T1, Jurkat cells, and primary bulk CD8+ T cells were monitored with PE-labeled HLA-A*0201/NY-ESO-1157–165 specific multimers (TCMetrix Sàrl) and FITC-conjugated BV13.1 antibody (Beckman Coulter).
NTAmer production and staining
NTAmers were synthetized by TCMetrix Sàrl (www.tcmetrix.ch) as described in ref. 15. NTAmers are composed of streptavidin-phycoerythrin (SA-PE; Invitrogen) complexed with biotinylated peptides carrying four Ni2+-nitrilotriacetic acid (NTA4) moieties and noncovalently bound to His-tagged HLA-A*0201 monomers containing β2m or a Cy5-labeled β2m. Monomers were obtained by refolding of the HLA-A*0201 heavy chain in the presence of β2m or Cy5-labeled β2m containing the S88C mutation with Cy-5-maleimide (GE Healthcare) and the analog NY-ESO-1157–165 [SLLMWITQA] or Melan-AMART-126–35 [ELAGIGILTV] tumor antigenic peptide, optimized for enhanced HLA-A*0201 binding. After purification on a Superdex S75 column, pMHC monomers were mixed at a 10-fold ratio with SA-PE-NTA4 in the presence of Ni2+ as described in ref. 15. The same procedure was used to prepare CD8-binding deficient HLA-A*0201 monomers bearing the D227K/T228A mutations in the HLAα3 domain (17). Dually labeled NTAmers containing SA-PE and Cy5-labeled monomers were used for dissociation kinetic measurements as described below. Single labeled NTAmers containing SA-PE and unlabeled monomers were used for flow-cytometric-based sorting of tumor-specific T cells. Once sorted, tumor-specific T cells were treated with 100 mmol/L imidazole at 4°C, allowing the rapid dissociation of the SA-PE-NTA4 scaffold and pMHC monomers before in vitro T-cell cloning by limiting dilution.
Dissociation kinetic measurements
TCR-transduced CD8+ SUP-T1 or CD8− Jurkat cells (5 × 105 cells) and tumor-specific CD8+ T-cell clones (2 × 105 cells) derived from patients LAU 155, LAU 50, and LAU 618 were incubated for 40 minutes at 4°C with HLA-A*0201/NY-ESO-1157–165 or HLA-A*0201/Melan-AMART-126–35 NTAmers containing streptavidin-phycoerythrin and Cy5-labeled monomers in 50 μL FACS buffer (PBS supplemented with 0.5% BSA, 15 mmol/L HEPES, and 0.02% NaN3). After a washing step, cells were suspended in 500 μL FACS buffer at 15°C (for SUP-T1 and Jurkat cells) or 200 μL FACS buffer at 4°C (for primary T-cell clones from melanoma patients) and cell surface-associated mean fluorescence was measured under constant temperature using a thermostat device (15°C for SUP-T1 and Jurkat cells and 4°C for primary T-cell clones) on a SORP-LSR II flow cytometer (BD Biosciences) following gating on living cells. PE-NTA4 and Cy5-pMHC monomer fluorescence was measured before (between 30 seconds to 1 minute; baseline) and during 5 to 10 minutes after the addition of imidazole (100 mmol/L). High-resolution microscopy flow analysis was performed on an Amnis ImageStreamx Mark II instrument (Merck Millipore) using a ×40 objective. Staining was performed as described above for SUP-T1 cells. PE-NTA4 and Cy5-pMHC monomer fluorescence was measured before (t = 0) and during 5 minutes upon the addition of imidazole (100 mmol/L) at 20°C. A 60 seconds lag time due to the automated handling of cell suspension by the Amnis instrument precluded earlier time-point measurements.
Dissociation kinetic data analysis
Flow-cytometric–based data were processed using the FlowJo software (v.9.6, Tree Star, Inc.). After gating on living cells, PE or Cy5 mean fluorescence intensity was derived using the kinetic module of the FlowJo software. Gates of 6 seconds period were created following addition of imidazole at the following time points: 15 seconds, 30 seconds, 45 seconds, 60 seconds, 90 seconds, 120 seconds, and then every minute for 10 minutes. Geometric MFI was measured at each time point after gating on HLA-A*0201/NY-ESO-1157–165– or HLA-A*0201/Melan-AMART-126–35–specific staining. Irrelevant Flu-specific gMFI values were systematically subtracted at the various time points. Corrected gMFI values were plotted and analyzed using the GraphPad Prism software (v.6, GraphPad).
Calcium mobilization assays
A total of 5 × 104 TCR-transduced SUP-T1 or primary bulk CD8+ T cells were loaded with 2 μmol/L Indo 1-AM (Sigma-Aldrich) for 45 minutes at 37°C. Cells were washed and resuspended in 250 μL prewarmed RPMI containing 2% FCS. Baseline was recorded for 30 seconds before 1 μg/mL of undissociated HLA-A*0201/NY-ESO-1157–165 NTAmers (for SUP-T1) or 1 μg/mL HLA-A*0201/NY-ESO-1157–165–specific multimers (for primary CD8+ T cells) were added to the cells allowing specific stimulation. Intracellular Ca2+ flux was assessed over 5 minutes under UV excitation and constant temperature of 37°C using a thermostat device on a LSR II SORP (BD Biosciences) flow cytometer. Indo-1 (violet)/Indo-1 (blue) 405/525 nm emission ratio was analyzed by FlowJo kinetics module software (TreeStar).
Chromium release and tumor recognition assays
The functional avidity of antigen recognition was analyzed in a 4-hour 51Cr-release assay using TAP−/−-deficient T2 (HLA-A*0201pos) target cells pulsed with serial dilutions of the natural NY–ESO-1157–165 peptide [SLLMWITQC] for tumor-specific T-cell clones derived from patient LAU 155 and LAU 50 or the natural Melan-AMART-126–35 peptide [EAAGIGILTV] for tumor-specific T-cell clones derived from patient LAU 618. The NY-ESO-1 peptide was preincubated for 1 hour at room temperature with the disulfide-reducing agent Tris [2-carboxyethyl] phosphine (TCEP; 2 mmol/L, Pierce Biotechnology) before functional assays. The percentage of specific lysis was calculated as follows: 100 × (experimental − spontaneous release)/(total − spontaneous release). For T-cell clones of defined TCRα,β clonotypes previously derived from patient LAU 155 (16), antigen-specific recognition and lytic activity were further assessed against the melanoma cell lines Me 275 (HLA-A2pos/NY-ESO-1pos) and NA8 (HLA-A2pos/NY-ESO-1neg).
Statistical analysis
Statistical analyses were done with the GraphPad Prism software. A minimum of three independent experiments were performed to ensure a statistical power of 100% at α = 0.05 (Figs. 2 and 3), and a sample size ≥ 23 to ensure a statistical power of 80% at α = 0.05 (Fig. 5). Correlation analyses were performed using Pearson (Figs. 2 to 4) or Spearman (Fig. 5) coefficient r. The associated P value (two-tailed, α = 0.05) quantifies the likelihood that the correlation is due to random sampling. Two different investigators performed each TCR–pMHC dissociation kinetic experiment independently, and experiments described in Fig. 5 were performed blinded for both sample allocation and outcome assessment.
Results
Direct measurements of monomeric TCR–pMHC dissociation kinetics by NTAmers
We previously developed a method for the isolation and analysis of antigen-specific cytotoxic T cells by reversible NTAmers (10, 15). Upon addition of imidazole at low, nontoxic concentration, the SA-PE-NTA4 moieties rapidly decay (average dissociation half-life of 2.5 seconds), thereby releasing the pMHC monomers bound at the cell surface. Consequently, NTAmers allow FACS-sorting of CD8+ T cells without inducing adverse effects on the cell integrity (e.g., activation-induced cell death; refs. 10, 15). Taking advantage of the fact that NTAmers can be switched from stable binding to rapid dissociation, a two-color NTAmer was engineered to assess monomeric TCR–pMHC dissociation kinetics directly on living CD8+ T cells (Fig. 1A). We first evaluated this novel approach by direct visualization of the dissociation process on individual human CD8+ SUP-T1 cells expressing TCRs of increasing affinities for the HLA-A*0201–restricted tumor epitope NY-ESO-1157–165 (18), using a flow cytometer generating simultaneous high-resolution microscopy imaging (ImageStreamX MarkII; Fig. 1B). As predicted, we observed strongest and most sustained fluorescence levels for T cells of very high TCR affinities (e.g., QMα and wtc51m). Because of a 60-second lag time due to the automated handling of cells following the addition of imidazole, the imaging approach precluded the visualization of labeled monomeric pMHC dissociation for T cells expressing TCRs ranging within the physiologic range (e.g., V49I, wild-type and DMβ). To solve this issue, we used a conventional flow cytometer (LSRII-SORP) equipped with a thermostat, which drastically reduced the lagging time to less than 5 seconds (Fig. 1C), thereby allowing the accurate assessment of monomeric pMHC dissociation kinetics on the surface of CD8+ T cells across a wide TCR affinity spectrum (Fig. 1D and Supplementary Fig. S1A). To validate the NTAmer technology, we next compared these dissociation values with SPR kinetics data obtained with the corresponding soluble TCRs (Table 1). Robust correlations were found between NTAmer-based cell surface monomeric pMHC dissociation kinetics (half-live t1/2 and koff), with both koff rates and affinities measured by SPR, contrasting with the weaker correlations obtained with pMHC tetramers (Fig. 2A and Supplementary Fig. S2A).
Assessment of monomeric dissociation kinetics by reversible NTAmers. A, basic illustration of the off-rate (koff) dissociation assay. Human CD8+ antigen-specific T cells were first stained with HLA-A*0201/tumor antigen-specific NTAmers containing PE-labeled backbone (green) and Cy5-labeled monomers (red) carrying imidazole-sensitive Ni2+-NTA4 moieties. Upon addition of imidazole, the NTAmer multimeric complex rapidly dissociated into Cy5-labeled pMHC monomers releasing the SA-PE NTA4 molecules. Decay of Cy5 fluorescence was visualized by high-resolution microscopy flow cytometry (B) or conventional flow cytometry over time (C) using SUP-T1 cells engineered with TCRs of incremental affinity for the tumor antigen A2/NY-ESO-1157–165 (Table 1; ref. 20). B, differential interference contrast (DIC), PE, Cy5, and PE/Cy5 composite images are shown at the indicated time points. C, representative dot plots from FACS-based dissociation curves (Cy5-labeled pMHC monomers) from TCR-transduced SUP-T1 cells. Decay of PE-labeled NTA4 scaffold moieties is depicted alongside. D, temperature-controlled (15°C) TCR–pMHC monomeric dissociation off-rates (Cy5-monomers, blue circles) were assessed upon addition of imidazole (t = 0) within the entire panel of TCR-transduced SUP-T1 cells. Data best fitted a one-phase exponential decay equation after subtraction of nonspecific background and are expressed as percentage of maximal binding, normalized to 100%, and plotted over time. Decay of SA-PE-NTA4 fluorescence (white circles) from NTAmers occurred within the first 2 to 3 seconds upon imidazole addition and was independent TCR–pMHC affinity. Time for half maximal binding (t1/2) was determined and average half-life value (t1/2) of >3 independent experiments is depicted in second (s) for each TCR engineered SUP-T1 cell variant. Untransduced SUP-T1 cells; no TCR. N/A, not applicable.
Assessment of monomeric dissociation kinetics by reversible NTAmers. A, basic illustration of the off-rate (koff) dissociation assay. Human CD8+ antigen-specific T cells were first stained with HLA-A*0201/tumor antigen-specific NTAmers containing PE-labeled backbone (green) and Cy5-labeled monomers (red) carrying imidazole-sensitive Ni2+-NTA4 moieties. Upon addition of imidazole, the NTAmer multimeric complex rapidly dissociated into Cy5-labeled pMHC monomers releasing the SA-PE NTA4 molecules. Decay of Cy5 fluorescence was visualized by high-resolution microscopy flow cytometry (B) or conventional flow cytometry over time (C) using SUP-T1 cells engineered with TCRs of incremental affinity for the tumor antigen A2/NY-ESO-1157–165 (Table 1; ref. 20). B, differential interference contrast (DIC), PE, Cy5, and PE/Cy5 composite images are shown at the indicated time points. C, representative dot plots from FACS-based dissociation curves (Cy5-labeled pMHC monomers) from TCR-transduced SUP-T1 cells. Decay of PE-labeled NTA4 scaffold moieties is depicted alongside. D, temperature-controlled (15°C) TCR–pMHC monomeric dissociation off-rates (Cy5-monomers, blue circles) were assessed upon addition of imidazole (t = 0) within the entire panel of TCR-transduced SUP-T1 cells. Data best fitted a one-phase exponential decay equation after subtraction of nonspecific background and are expressed as percentage of maximal binding, normalized to 100%, and plotted over time. Decay of SA-PE-NTA4 fluorescence (white circles) from NTAmers occurred within the first 2 to 3 seconds upon imidazole addition and was independent TCR–pMHC affinity. Time for half maximal binding (t1/2) was determined and average half-life value (t1/2) of >3 independent experiments is depicted in second (s) for each TCR engineered SUP-T1 cell variant. Untransduced SUP-T1 cells; no TCR. N/A, not applicable.
Contribution of CD8 binding to TCR–pMHC dissociation kinetics. A, positive correlations (Pearson coefficient r and P value) obtained between surface-based monomeric half-lives (t1/2) with wild-type NTAmers (monomer, blue line) or CD8-binding deficient NTAmers227–228 (monomer227–228, red line) on CD8+ TCR-transduced SUP-T1 cells, and monomeric half-lives (t1/2) and affinities (equilibrium dissociation constant KD) as measured by SPR on soluble TCRs. Weaker correlations were found when assessing half-lives using a pure grade tetramer assay (opened circles). B, direct comparison of wild-type (blue circles) and CD8-binding deficient (red diamonds) monomeric dissociation half-lives by NTAmers (>3 independent experiments). The impact of CD8 binding is shown as fold change (gray histograms) for each TCR-transduced SUP-T1 variant. C, representative first-order monomeric TCR–pMHC dissociation curves detected upon addition of imidazole at 15°C (t = 0) for CD8-null Jurkat T cells engineered with the indicated TCR variants (expressed as % of maximal binding over time) and labeled with wild-type NTAmers (monomer, blue circles) or CD8-binding deficient NTAmers227–228 (monomer227–228, red circles). Decay of PE-NTA4 fluorescence (white circles) is also represented. Average dissociation half-life value (of three independent experiments) with wild-type NTAmers or NTAmers227–228 is shown for each TCR-transduced Jurkat T-cell variant. D, positive correlations (Pearson coefficient r and P values) between surface-based monomeric half-lives (t1/2) with wild-type NTAmer (monomer, blue line) or CD8-binding deficient NTAmer227–228 (monomer227–228, red line) on TCR-transduced CD8-null Jurkat T cells, and monomeric half-lives (t1/2) by SPR on soluble TCR variants.
Contribution of CD8 binding to TCR–pMHC dissociation kinetics. A, positive correlations (Pearson coefficient r and P value) obtained between surface-based monomeric half-lives (t1/2) with wild-type NTAmers (monomer, blue line) or CD8-binding deficient NTAmers227–228 (monomer227–228, red line) on CD8+ TCR-transduced SUP-T1 cells, and monomeric half-lives (t1/2) and affinities (equilibrium dissociation constant KD) as measured by SPR on soluble TCRs. Weaker correlations were found when assessing half-lives using a pure grade tetramer assay (opened circles). B, direct comparison of wild-type (blue circles) and CD8-binding deficient (red diamonds) monomeric dissociation half-lives by NTAmers (>3 independent experiments). The impact of CD8 binding is shown as fold change (gray histograms) for each TCR-transduced SUP-T1 variant. C, representative first-order monomeric TCR–pMHC dissociation curves detected upon addition of imidazole at 15°C (t = 0) for CD8-null Jurkat T cells engineered with the indicated TCR variants (expressed as % of maximal binding over time) and labeled with wild-type NTAmers (monomer, blue circles) or CD8-binding deficient NTAmers227–228 (monomer227–228, red circles). Decay of PE-NTA4 fluorescence (white circles) is also represented. Average dissociation half-life value (of three independent experiments) with wild-type NTAmers or NTAmers227–228 is shown for each TCR-transduced Jurkat T-cell variant. D, positive correlations (Pearson coefficient r and P values) between surface-based monomeric half-lives (t1/2) with wild-type NTAmer (monomer, blue line) or CD8-binding deficient NTAmer227–228 (monomer227–228, red line) on TCR-transduced CD8-null Jurkat T cells, and monomeric half-lives (t1/2) by SPR on soluble TCR variants.
Kinetic characteristics of HLA-A*0201/NY-ESO-1157–165–specific TCR variants
. | SPR TCR–pMHC kineticsb . | NTAmer dissociation kinetics . | NTAmer227–228 dissociation kinetics . | ||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
. | Soluble TCRs . | CD8+c . | CD8−c . | CD8+c . | CD8−c . | ||||||||
. | . | kon . | koff . | . | koff . | . | koff . | . | koff . | . | koff . | . | |
TCR variantsa . | KD μmol/L . | 1/[(mol/L)× s] (×104) . | 1/s (×10−2) . | t1/2 s . | 1/s (×10−2) . | t1/2 s . | 1/s (×10−2) . | t1/2 s . | 1/s (×10−2) . | t1/2 s . | 1/s (×10−2) . | t1/2 s . | |
V49I | n.a. | n.a. | n.a. | n.a. | 21.21 | 3 | n.a. | n.a. | n.a. | n.a. | n.a. | n.a. | |
Wild-type | 21.4 | 1.1 | 23.0 | 3 | 4.08 | 17 | 10.2 | 7 | 11.2 | 6 | 15.4 | 5 | |
G50A | 4.62 | 1.5 | 6.9 | 10 | 1.47 | 47 | n.d. | n.d. | 5.28 | 13 | n.d. | n.d. | |
A97L | 2.69 | 2.3 | 6.1 | 11 | 1.60 | 44 | n.d. | n.d. | 4.70 | 15 | n.d. | n.d. | |
DMβ | 1.91 | 2.4 | 4.5 | 15 | 0.78 | 90 | 2.76 | 25 | 3.02 | 23 | 3.15 | 22 | |
TMβ | 0.91 | 1.4 | 1.3 | 53 | 0.28 | 247 | 1.03 | 67 | 0.87 | 79 | 0.97 | 72 | |
TMα | 0.40 | 12.1 | 4.8 | 14 | 0.44 | 158 | n.d. | n.d. | 1.76 | 40 | n.d. | n.d. | |
QMα | 0.14 | 10.9 | 1.5 | 46 | 0.21 | 341 | 0.80 | 87 | 0.74 | 94 | 0.75 | 92 | |
Wtc51m | 0.015 | 8.5 | 0.13 | 533 | 0.05 | 1505 | 0.14 | 497 | 0.15 | 475 | 0.14 | 496 |
. | SPR TCR–pMHC kineticsb . | NTAmer dissociation kinetics . | NTAmer227–228 dissociation kinetics . | ||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
. | Soluble TCRs . | CD8+c . | CD8−c . | CD8+c . | CD8−c . | ||||||||
. | . | kon . | koff . | . | koff . | . | koff . | . | koff . | . | koff . | . | |
TCR variantsa . | KD μmol/L . | 1/[(mol/L)× s] (×104) . | 1/s (×10−2) . | t1/2 s . | 1/s (×10−2) . | t1/2 s . | 1/s (×10−2) . | t1/2 s . | 1/s (×10−2) . | t1/2 s . | 1/s (×10−2) . | t1/2 s . | |
V49I | n.a. | n.a. | n.a. | n.a. | 21.21 | 3 | n.a. | n.a. | n.a. | n.a. | n.a. | n.a. | |
Wild-type | 21.4 | 1.1 | 23.0 | 3 | 4.08 | 17 | 10.2 | 7 | 11.2 | 6 | 15.4 | 5 | |
G50A | 4.62 | 1.5 | 6.9 | 10 | 1.47 | 47 | n.d. | n.d. | 5.28 | 13 | n.d. | n.d. | |
A97L | 2.69 | 2.3 | 6.1 | 11 | 1.60 | 44 | n.d. | n.d. | 4.70 | 15 | n.d. | n.d. | |
DMβ | 1.91 | 2.4 | 4.5 | 15 | 0.78 | 90 | 2.76 | 25 | 3.02 | 23 | 3.15 | 22 | |
TMβ | 0.91 | 1.4 | 1.3 | 53 | 0.28 | 247 | 1.03 | 67 | 0.87 | 79 | 0.97 | 72 | |
TMα | 0.40 | 12.1 | 4.8 | 14 | 0.44 | 158 | n.d. | n.d. | 1.76 | 40 | n.d. | n.d. | |
QMα | 0.14 | 10.9 | 1.5 | 46 | 0.21 | 341 | 0.80 | 87 | 0.74 | 94 | 0.75 | 92 | |
Wtc51m | 0.015 | 8.5 | 0.13 | 533 | 0.05 | 1505 | 0.14 | 497 | 0.15 | 475 | 0.14 | 496 |
Abbreviations: n.a., not applicable; n.d., not done.
aWild-type TCR (BC1; AV23.1/BV13.1) was isolated from melanoma patient LAU 155 (16) and a panel of TCR variants of progressive increasing affinities against HLA-A*0201/NY-ESO-1157–165 was established as described in ref. 20.
bTCR–pMHC affinity, koff and kon values were previously measured by SPR as reported in ref. 20.
cCD8+ T cells, TCR-transduced SUP-T1 cells; CD8− T cells, TCR-transduced Jurkat cells.
CD8 coreceptor stabilized the TCR–pMHC complex by prolonging the dissociation half-life by a factor of 3- to 4-fold
To precisely quantify the contribution of CD8 coreceptor to monomeric TCR–pMHC dissociation, we generated NTAmer227–228 variants containing the HLA-A*0201 D227K/T228A mutations in the MHC α3 domain that abolish CD8 binding to pMHC (17). Using the same panel of TCR-transduced CD8+ SUP-T1 cells, we show that abrogating CD8–MHC binding drastically diminished dissociation half-lives (t1/2) over the whole spectrum of TCR affinities (Fig. 2B and Supplementary Fig. S1B and S1C). Monomeric off-rate (koff and t1/2) correlations between NTAmers227–228 and SPR were highly significant (Fig. 2A and Supplementary Fig. S2A). Comparison between wild-type and CD8-binding deficient NTAmer-based half-lives revealed that CD8 stabilized by a factor of 3- to 4-fold the TCR-pMHC complex, and this was irrespective of the TCR affinity (Fig. 2B). A similar effect of CD8 binding to pMHC was observed when using TCR transduced Jurkat T cells that lack CD8 expression (Fig. 2C and D and Supplementary Fig. S2B) with koff and t1/2 values that were highly comparable with those found by NTAmers227–228 or by SPR on CD8+ T cells (Table 1). Altogether, the NTAmer technology permits (i) the precise measurements of monomeric TCR–pMHC dissociation kinetics directly on living T cells expressing a wide TCR affinity range, including those typically found within the self/tumor-specific repertoires, and (ii) the quantification of the contribution of CD8 binding on the TCR–pMHC complex stability, which is not possible in SPR measurements.
Monomeric TCR–pMHC dissociation kinetics correlate with enhanced T-cell responsiveness
Next we compared koff rates obtained using NTAmers and the intracellular calcium mobilization on CD8+ SUP-T1 cells (Fig. 3A and B) and primary CD8+ T cells (Fig. 3D and E) expressing the panel of affinity-optimized TCRs against NY-ESO-1 tumor antigen. In line with our previous reports (19, 20), the Ca2+ flux was enhanced with increasing TCR affinity, up to reaching a plateau, while further affinity increase (KD < 1 μmol/L) resulted in reduced calcium signaling. Within the physiologic TCR–pMHC affinity range, however, both SUP-T1 cells and primary T cells exhibited strong correlations between dissociation rates and Ca2+ mobilization (Fig. 3C and F); there was an enhanced Ca2+ flux in T cells of longer half-lives (low koff), contrasting with the reduced signaling observed in T cells of shorter t1/2 (high koff). An overall reduction in Ca2+ flux was observed when using mutant multimer/HLA-A*0201227–228 for specific stimulation, but this effect was less stringent for T cells expressing very high TCR affinities (e.g., TMα, QMα, and wtc51m; Fig. 3A and B). As previously reported (20–22), these data indicate that the degree of CD8 dependence for T-cell activation inversely depends on the affinity and dissociation half-life of the TCR–pMHC interaction.
Relationship between cell-surface monomeric dissociation kinetics and calcium flux. A–C, intracellular calcium mobilization of CD8+ SUP-T1 cells expressing TCRs of incremental affinities for pMHC before (baseline) and over time after stimulation with 1 μg/mL A2/NY-ESO-1157–165–specific multimers (blue circles) or 1 μg/mL CD8-binding deficient A2/NY-ESO-1157–165–specific multimers227–228 (red diamonds). A representative kinetic analysis of calcium mobilization is depicted in A and the mean calcium flux values of four independent experiments are plotted as Ca2+ peak MFI (×104) in B together with maximal calcium flux induced after ionomycin stimulation. C, correlation (Pearson coefficient r and P value) between calcium flux peak MFI and dissociation koff rates for SUP-T1 cells expressing TCRs within the physiological affinity range (blue circles). Calcium flux values for SUP-T1 cells expressing supraphysiological affinity TCRs (TMα, QMα, and wtc51m; blue stars), and those obtained after stimulation with CD8-binding deficient multimers227–228 (red diamonds) are shown, but were not included in the correlative analysis. D, a representative kinetic analysis of calcium mobilization in primary TCR-transduced CD8+ T cells before (no antigen-presenting cells; APC) and after stimulation with T2 cells pulsed with graded concentration of the analog NY-ESO-1157–165 peptide (SLLMWITQA) or ionomycin as positive control. E, the mean calcium flux values were plotted as Ca2+ peak MFI, with varying peptide concentration for each primary CD8+ TCR-transduced T-cell variant. No calcium flux was detected upon stimulation of wild-type (WT) NY-ESO-1–transduced T cells with Flu-specific peptide (Flu) or no APCs. F, correlation (Pearson coefficient r and P value) between half-maximal calcium mobilization capacity (Ca2+ EC50) and NTAmer-based dissociation koff values for T cells expressing TCRs within the physiologic affinity range (blue circles). Calcium flux values obtained for CD8+ T cells expressing TCRs of supraphysiological affinities (TMα, QMα, and wtc51m) are shown (blue stars), but were not included in the correlative analysis.
Relationship between cell-surface monomeric dissociation kinetics and calcium flux. A–C, intracellular calcium mobilization of CD8+ SUP-T1 cells expressing TCRs of incremental affinities for pMHC before (baseline) and over time after stimulation with 1 μg/mL A2/NY-ESO-1157–165–specific multimers (blue circles) or 1 μg/mL CD8-binding deficient A2/NY-ESO-1157–165–specific multimers227–228 (red diamonds). A representative kinetic analysis of calcium mobilization is depicted in A and the mean calcium flux values of four independent experiments are plotted as Ca2+ peak MFI (×104) in B together with maximal calcium flux induced after ionomycin stimulation. C, correlation (Pearson coefficient r and P value) between calcium flux peak MFI and dissociation koff rates for SUP-T1 cells expressing TCRs within the physiological affinity range (blue circles). Calcium flux values for SUP-T1 cells expressing supraphysiological affinity TCRs (TMα, QMα, and wtc51m; blue stars), and those obtained after stimulation with CD8-binding deficient multimers227–228 (red diamonds) are shown, but were not included in the correlative analysis. D, a representative kinetic analysis of calcium mobilization in primary TCR-transduced CD8+ T cells before (no antigen-presenting cells; APC) and after stimulation with T2 cells pulsed with graded concentration of the analog NY-ESO-1157–165 peptide (SLLMWITQA) or ionomycin as positive control. E, the mean calcium flux values were plotted as Ca2+ peak MFI, with varying peptide concentration for each primary CD8+ TCR-transduced T-cell variant. No calcium flux was detected upon stimulation of wild-type (WT) NY-ESO-1–transduced T cells with Flu-specific peptide (Flu) or no APCs. F, correlation (Pearson coefficient r and P value) between half-maximal calcium mobilization capacity (Ca2+ EC50) and NTAmer-based dissociation koff values for T cells expressing TCRs within the physiologic affinity range (blue circles). Calcium flux values obtained for CD8+ T cells expressing TCRs of supraphysiological affinities (TMα, QMα, and wtc51m) are shown (blue stars), but were not included in the correlative analysis.
We extended these observations to a clinically relevant setting, and characterized koff rates and functional avidity of T cells obtained from a large set of HLA-A*0201/NY-ESO-1157–165–specific CD8+ T-cell clones expressing well-defined TCRαβ clonotypes derived from LAU 155, a melanoma patient with long lasting antitumor T-cell responses (16). The dissociation rates of those cytotoxic T cell clones clustered within a narrow range (Fig. 4A and Supplementary Fig. S3A), and strongly correlated with EC50 target cell killing (50% maximal lysis; Fig. 4C) and tumor cell recognition (Fig. 4E), with TCR clonotypes having long half-lives (low koff) exhibiting increased T-cell cytotoxicity (Fig. 4B and D). Conversely, NY-ESO-1–specific TCR clonotypes with short half-lives (high koff) showed reduced T-cell killing capacities.
Relationship between cell-surface monomeric dissociation kinetics and target cell killing. Experiments were performed on A2/NY-ESO-1157–165–specific CD8+ T-cell clones (n = 20) expressing eight well-defined TCRαβ clonotypes, derived from melanoma patient LAU 155 as previously described (16). A, representative first-order monomeric dissociation curves obtained upon addition of imidazole at 4°C (t = 0) for tumor-specific T-cell clones stained with A2/NY-ESO-1157–165–specific-NTAmers. Average half-lives (t1/2) determined from each distinct TCRαβ clonotypes (n = 2 to 3 independent sister T-cell clones) are indicated and were highly reproducible in two independent experiments (data not shown). B, the relative functional avidity of tumor-specific T-cell clones expressing distinct TCRαβ clonotypes was assessed by measuring their lytic capacity for T2 target cells (A2pos; TAPneg/neg) pulsed with graded concentration of the natural NY-ESO-1157–165 peptide (SLLMWITQC). C, correlations (Pearson coefficient r and P value) between relative functional avidity (EC50, peptide concentration used to achieve 50% of maximal lysis) and monomeric TCR–pMHC dissociation koff values. Mean EC50 values of three independent experiments. D, tumor reactivity by tumor-specific T-cell clonotypes for the melanoma cell line Me275 (A2pos/NY-ESO-1157–165pos) at the indicated effector:target (E:T) ratio. E, correlations (Pearson coefficient r and P value) between melanoma cell killing (mean % of specific lysis at the E:T ratio of 10:1 from two independent experiments) and monomeric TCR–pMHC dissociation koff values.
Relationship between cell-surface monomeric dissociation kinetics and target cell killing. Experiments were performed on A2/NY-ESO-1157–165–specific CD8+ T-cell clones (n = 20) expressing eight well-defined TCRαβ clonotypes, derived from melanoma patient LAU 155 as previously described (16). A, representative first-order monomeric dissociation curves obtained upon addition of imidazole at 4°C (t = 0) for tumor-specific T-cell clones stained with A2/NY-ESO-1157–165–specific-NTAmers. Average half-lives (t1/2) determined from each distinct TCRαβ clonotypes (n = 2 to 3 independent sister T-cell clones) are indicated and were highly reproducible in two independent experiments (data not shown). B, the relative functional avidity of tumor-specific T-cell clones expressing distinct TCRαβ clonotypes was assessed by measuring their lytic capacity for T2 target cells (A2pos; TAPneg/neg) pulsed with graded concentration of the natural NY-ESO-1157–165 peptide (SLLMWITQC). C, correlations (Pearson coefficient r and P value) between relative functional avidity (EC50, peptide concentration used to achieve 50% of maximal lysis) and monomeric TCR–pMHC dissociation koff values. Mean EC50 values of three independent experiments. D, tumor reactivity by tumor-specific T-cell clonotypes for the melanoma cell line Me275 (A2pos/NY-ESO-1157–165pos) at the indicated effector:target (E:T) ratio. E, correlations (Pearson coefficient r and P value) between melanoma cell killing (mean % of specific lysis at the E:T ratio of 10:1 from two independent experiments) and monomeric TCR–pMHC dissociation koff values.
Identification of rare high-avidity, tumor-specific CD8+ T cells by monomeric TCR–pMHC dissociation kinetic measurements
To find out whether NTAmers allow direct identification of tumor-specific CD8+ T cells with high tumor killing capacity, we derived 147 tumor-specific T-cell clones from two other melanoma patients with detectable ex vivo T-cell responses against the HLA-A*0201–restricted tumor antigens NY-ESO-1157–165 and Melan-AMART-126–35, respectively, and screened them for monomeric dissociation rates (Fig. 5A and D and Supplementary Fig. S3B and S3C). Tumor-specific T-cell clones were distributed according to their TCR Vβ family expression using monoclonal antibodies and koff rate analyses revealed large differences in half-lives between specific TCRs (Supplementary Fig. S4). Next, we performed killing assays with 57 clones selected for relatively low or high koff values (Fig. 5B and E). We observed, for both antigenic specificities, a robust correlation between off-rates and the functional avidity (EC50) determined with the cytotoxicity assay (Fig. 5C and F), demonstrating that NTAmers allow reliable assessment of surface-based TCR–pMHC dissociation kinetics and rapid selection of highly potent tumor-specific T-cell clones derived from different patients with cancer.
High-throughput screen of functionally relevant tumor-specific CD8+ T cells by NTAmers. Relationship between functional avidity and monomeric TCR–pMHC koff rates of a large panel of tumor-specific CD8+ T-cell clones specific for A2/NY-ESO-1157–165 (A–C) or A2/Melan-AMART-126–35 (D–F) tumor antigen and derived from melanoma patient LAU 50 and LAU 618, respectively. A and D, representative first-order dissociation curves obtained after addition of imidazole at 4°C for CD8+ A2/NY-ESO-1157–165–specific T-cell clones (LAU 50; n = 67) and A2/Melan-AMART-126–35–specific T-cell clones (LAU 618, n = 80), respectively, stained with specific NTAmers and arbitrarily separated into short (white circles) or long (black squares) half-lives according to their t1/2 values. B and E, relative functional avidity on a selection of A2/NY-ESO-1157–165–specific T-cell clones (n = 23) or A2/Melan-AMART-126–35–specific T-cell clones (n = 34) of short or long half-lives using T2 target cells pulsed with graded concentration of natural NY-ESO-1157–165– or Melan-AMART-126–35–specific peptide. C and F, positive correlations (Spearman coefficient r and P value) obtained between relative functional avidity by EC50 (50% of maximal target cell killing) and monomeric TCR–pMHC dissociation koff values. Each data point represents the result of an individual tumor-specific T-cell clone, averaged from two independent experiments.
High-throughput screen of functionally relevant tumor-specific CD8+ T cells by NTAmers. Relationship between functional avidity and monomeric TCR–pMHC koff rates of a large panel of tumor-specific CD8+ T-cell clones specific for A2/NY-ESO-1157–165 (A–C) or A2/Melan-AMART-126–35 (D–F) tumor antigen and derived from melanoma patient LAU 50 and LAU 618, respectively. A and D, representative first-order dissociation curves obtained after addition of imidazole at 4°C for CD8+ A2/NY-ESO-1157–165–specific T-cell clones (LAU 50; n = 67) and A2/Melan-AMART-126–35–specific T-cell clones (LAU 618, n = 80), respectively, stained with specific NTAmers and arbitrarily separated into short (white circles) or long (black squares) half-lives according to their t1/2 values. B and E, relative functional avidity on a selection of A2/NY-ESO-1157–165–specific T-cell clones (n = 23) or A2/Melan-AMART-126–35–specific T-cell clones (n = 34) of short or long half-lives using T2 target cells pulsed with graded concentration of natural NY-ESO-1157–165– or Melan-AMART-126–35–specific peptide. C and F, positive correlations (Spearman coefficient r and P value) obtained between relative functional avidity by EC50 (50% of maximal target cell killing) and monomeric TCR–pMHC dissociation koff values. Each data point represents the result of an individual tumor-specific T-cell clone, averaged from two independent experiments.
Discussion
Identifying antigen-specific T cells that confer efficient effector function is critical for successful adoptive cell therapy. The stability of TCR binding for the peptide–MHC complex is a key determinant for T-cell activation. Several lines of evidence argue for a close relationship between cell surface dissociation kinetics (koff) and functional T-cell responses (5, 23–26). Yet, rapid and accurate screening methods to measure TCR–pMHC binding kinetics are still needed to isolate antigen-specific T cells expressing TCRs of high binding avidity. Recently, we developed a novel approach combining reversible peptide–MHC multimers (NTAmers; ref. 15) and real-time flow cytometry (Fig. 1). Using SPR, we had previously determined the TCR–pMHC binding strength of sequence-optimized HLA-A*0201/NY-ESO-1157–165–specific TCRs with increasing affinity of up to 150-fold from the wild-type receptor, including two outliers, a very low- and a very high-affinity TCR (5, 20). This TCR panel provides a unique model for validating monomeric TCR–pMHC dissociation kinetics at the surface of T cells by NTAmers. Here, we demonstrate that NTAmer-based off-rates (koff, t1/2) followed the same TCR–pMHC binding hierarchy than previously established (20), in excellent agreement to both binding parameters, koff and the dissociation constant KD, obtained by SPR (Fig. 2; Table 1).
Because of their switch ability, that is, high stability and rapid reversibility (<5 sec), NTAmers allowed accurate determination of dissociation rates, even for weak TCR–pMHC interactions, that is, fast off-rates, such as those found for (self) tumor-specific CD8+ T-cell repertoires (Fig. 4). Notably, the NTAmer approach differs from the Streptamer one, which is mostly limited to the detection of CD8+ T cells of high avidity such as virus-specific cells, as it requires a significant lag time (60 seconds) before monomeric TCR–pMHC dissociation becomes detectable (11). Moreover, the NTAmer-based assay represents a rapid and straightforward approach for the quantitative assessment of monomeric koff rates on a large set of cloned antigen-specific CD8+ T cells derived from different patients and tumor epitopes (HLA-A*0201/NY-ESO-1157–165 and HLA-A*0201/Melan-AMART-126–35; Fig. 5). Importantly, we demonstrate robust correlations between dissociation off-rates and the biologic responses (e.g., calcium flux and target cell killing) on a large panel of CD8+ T-cell clones specific for two distinct tumor antigens, indicating that TCR–ligand koff rate is a reliable predictor of T-cell function (Figs. 3–5).
Interactions between TCRs and pMHC are usually measured by SPR or pMHC tetramer or multimer staining, which requires one binding partner in soluble form. Both approaches have caveats. Because of their incomplete dissociation and multivalent nature, accurate off-rates data from pMHC tetramer/multimer staining measurements are imprecise. Conversely, monomeric dissociation koff rates measured by the NTAmer technology were within the range of seconds to minutes, spanning a broad range (2-logs), as compared with a narrow range of minutes observed when using pMHC tetramers. Moreover, SPR fails to take into account rapid rebinding of the TCR to the same pMHC, because one of the two binding partners is constantly moving in the fluid phase, which impacts on the binding kinetics. Increased kon rates have been shown to allow rapid rebinding after TCR–ligand dissociation, resulting in enhanced effective dissociation half-life of the TCR–pMHC interaction (25, 27). This may explain our observation that TCR variants with faster kon (e.g., TMα and QMα) showed prolonged NTAmer-based dissociation half-lives compared with soluble monomeric off-rates measured by SPR (Table 1 and Supplementary Fig. S5).
The NTAmer-based approach further deviates from SPR measurements, as it provides data on living cells and includes contributions of CD8 to TCR–pMHC interactions. The CD8 coreceptor enhances antigen recognition and T-cell activation by stabilizing TCR–pMHC interaction at the cell surface (28–31) and recruiting p56lck to TCR/CD3 complex promoting cell signaling (32, 33). Here, we demonstrate that CD8 strengthened the TCR–pMHC binding mainly by decreasing the TCR–pMHC dissociation by a factor of 3- to 4-fold (Fig. 2), as anticipated by previous tetramer dissociation assays (22, 30). Interestingly, the CD8 stabilization factor was independent of the TCR–pMHC affinity, in contrast with the CD8 dependence for T-cell activation, which can be directly linked to the affinity (3, 20, 22), and allows tuning the sensitivity and specificity of T-cell responses (34).
We recently provided new evidence that T-cell signaling and activation are optimal within a given TCR–pMHC affinity window (20), controlled through TCR affinity-mediated regulatory molecules, involving the inhibitory receptor PD-1 and SHP-1 phosphatase (19). Furthermore, while high-avidity T cells have been shown to control tumor growth, they become preferentially tolerized in the tumor microenvironment (35) or can target normal tissues expressing the cognate antigen (36, 37). Therefore, tumor-specific T cells of high avidity may not always be functionally better, and it remains to be fully determined to which degree intermediate or high-avidity T cells contribute to protective immunity. In this regard, NTAmers constitute a highly valuable tool for assessing the TCR–pMHC avidity and its relation to cell activation, signaling, and function in naturally or therapeutically induced tumor-specific T-cell responses, with TCR–pMHC affinities spanning within the physiologic range.
In summary, NTAmer technology enables efficient and direct interrogation of monomeric TCR–pMHC dissociation kinetics on a large set of living antigen-specific T cells by flow cytometry, and provides novel perspectives for rapid identification of rare functionally relevant tumor-reactive CD8+ T cells. Our approach may also be applicable to the analysis of other weak protein–protein interactions. Precise and widespread characterization of TCR–pMHC avidities will likely improve the development of T-cell–based immunotherapies in patients with cancer.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Authors' Contributions
Conception and design: M. Hebeisen, J. Schmidt, D.E. Speiser, N. Rufer
Development of methodology: M. Hebeisen, J. Schmidt, P. Guillaume, D.E. Speiser
Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M. Hebeisen, J. Schmidt, P. Baumgaertner, N. Rufer
Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M. Hebeisen, N. Rufer
Writing, review, and/or revision of the manuscript: M. Hebeisen, J. Schmidt, D.E. Speiser, N. Rufer
Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): D.E. Speiser, I. Luescher
Study supervision: N. Rufer
Acknowledgments
The authors thank P. Gannon, M. Irving, O. Michielin, P. Romero, S. Siegert, and L. Zhang for essential collaboration and advice, and N. Montandon and M. van Overloop for outstanding technical assistance.
Grant Support
This work was supported by the Department of Oncology of the University of Lausanne and the Ludwig Center for Cancer Research of the University of Lausanne.
The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.