Under conditions of impaired T-cell immunity, human cytomegalovirus (HCMV) can reactivate from lifelong latency, resulting in potentially fatal disease. A crucial role for CD8+ T cells has been demonstrated in control of viral replication, and high levels of HCMV-specific cytotoxic T-lymphocytes are seen in immunocompetent HCMV-seropositive individuals despite very low viral loads. Elucidation of the minimum portion of the anti-HCMV T-cell repertoire that is required to suppress viral replication requires further study of clonal composition. The ability of dendritic cells to take up and process exogenous viral antigen by constitutive macropinocytosis was used to study HCMV-specific T-cell memory in the absence of viral replication. The specificity and clonal composition of the CD8+ T-cell responses were evaluated using HLA tetrameric complexes and T-cell receptor β chain (TCRBV) spectratypic analyses. There was a skewed reactivity toward the matrix protein pp65, with up to 40-fold expansion of CD8+ T cells directed toward a single peptide-MHC combination. Individual expansions detected on TCRBV spectratype analysis were HCMV-specific and composed of single or highly restricted numbers of clones. There was preferential TCRBV gene usage (BV6.1/6.2, BV8, and BV13 in HLA-A*0201+ individuals) but lack of conservation of CDR3 length and junctional motifs between donors. While there was a spectrum of TCR repertoire diversity directed toward individual MHC-peptide combinations between donors, a relatively small number of clones appeared to predominate the response in each case. These data provide further insight into the range of anti-HCMV responses and will aid the design and monitoring of adoptive immunotherapy protocols.

Human cytomegalovirus (HCMV) is a ubiquitous β-herpesvirus that infects up to 90% of individuals depending on the population studied. In common with other members of the herpesvirus group, HCMV is able to persist lifelong in a latent or persistent state following primary infection, remaining under the control of the immune system.1 HCMV remains a significant cause of morbidity and mortality following allogeneic bone marrow transplantation, due to reactivation of virus acquired prior to transplantation.2,3 Pressures to expand the potential donor pool to facilitate more widespread application of transplantation have led to the use of conditioning protocols or posttransplantation immunosuppressive regimens that result in more profound immune suppression and enhanced risk of viral infection or reactivation.4 The predisposition to viral reactivation appears to relate to the absence of a sufficient cytotoxic T-lymphocyte (CTL) response,5 because reconstitution of CD8+ HCMV-reactive T cells or supplementation by adoptive transfer of T-cell clones has been shown to correlate with a reduced risk of HCMV disease.5,6 While the development of sensitive surveillance methods to monitor for HCMV reactivation allows available therapies to be targeted to those at greatest risk for the development of HCMV disease,7,8 the limitations of currently available surveillance protocols and antiviral pharmacotherapies has led to the need to develop alternative treatment strategies, particularly because HCMV seropositivity of recipient or donor prior to transplantation remains a significant adverse prognostic factor in some studies.4 

Adoptive transfer of unmanipulated T cells has been demonstrated to mediate both antiviral and antitumor immunity in humans.9 However, the cells' use has been constrained by the low frequency of antigen-specific T cells and by the presence of alloreactive T cells, which have the potential to induce graft versus host disease.9,10 Because of these considerations, the HCMV-specific CD8+ CTLs used for patient infusion in early studies by Riddell and colleagues were cloned from bulk cultures.6,11 These studies demonstrated the ability of infused cells to reconstitute short-term HCMV-specific CTL responses, which appeared sufficient to prevent the development of HCMV disease. In addition, they confirmed the requirement for CD4+ T-cell helper function to restore longer-term immune memory as had been suggested by earlier work in murine models.12 No adverse effects, including graft versus host disease, were documented. However, large numbers (up to 1 × 109/m2) of CD8+ cells were required at regular intervals to maintain in vivo anti-HCMV responses, and this required several weeks of cell culture and large-scale cloning that are both time-consuming and costly. These practical difficulties may partially explain why these studies have not led to the widespread use of adoptive immunotherapy for prevention and treatment of HCMV infection.

The human CTL response to CMV is dominated by structural protein pp65, targeted by 70% to 90% of HCMV-specific CTLs.13,14 Other immunogenic peptides accounting for a smaller part of the overall response include elements of the major immediate-early gene product (IE-1), the matrix protein pp150, and virion envelope glycoprotein B.15,16 Immunodominant nonapeptides from the pp65 matrix glycoprotein that are restricted to specific HLA molecules are being identified. In addition, the development of techniques to detect and quantify antigen-specific T cells now enable more detailed analysis of culture output cells and provide powerful tools for monitoring the fate of infused cells in immunotherapy protocols.17,18 As more interest focuses on the in vitro manipulation of immune responses in order to dissociate potentially beneficial from harmful effects, the elucidation of the “sufficient T-cell repertoire”19required to enable clinical efficacy (ie, the minimum portion of the overall T-cell response necessary to allow, for example, sustained viral clearance or regression of Epstein-Barr viris [EBV]–associated lymphoproliferative disorders) becomes increasingly important. Studies to more precisely document the overall clonal composition of T-cell responses and any differences or similarities in the responses of different individuals are required as a prelude to clinical studies based on selection of more restricted subpopulations of T cells. We have combined HCMV pp65–specific HLA tetrameric complex analysis with TCRBV spectratype analysis to evaluate HCMV-specific CD8+T-cell memory responses in HLA-A*0201+ and/or HLA-B*0702+ individuals following in vitro restimulation by dendritic cells (DCs) pulsed with exogenous viral antigen.

Generation of human monocyte-derived DCs and CTL culture

Generation of human monocyte-derived DCs and CTL culture has been described previously.20 Briefly, fresh peripheral blood mononuclear cells from 8 HLA-A*0201+ and/or HLA-B*0702+ HCMV-seropositive donors were prepared by Ficoll-Paque density centrifugation (Pharmacia, St Albans, United Kingdom), suspended in X Vivo 20 medium (Biowhittaker, Wokingham, United Kingdom), and allowed to adhere to tissue culture flasks for 2 to 3 hours. Nonadherent cells were removed by washing 2 times with phosphate-buffered saline. Adherent cells were differentiated into immature DCs in X Vivo 20 medium supplemented with 10% autologous human serum, 100 ng/mL interleukin-4 (Insight Biotechnology, Wembley, United Kingdom), and 100 ng/mL granulocyte-macrophage colony-stimulating factor (Hoescht, Hounslow, United Kingdom) for 7 days in a 37°C 5% CO2 humidified incubator.

Autologous peripheral blood lymphocytes (PBLs) and monocyte-derived DCs were cocultured for 14 to 21 days in X Vivo 20 medium supplemented with 10% autologous human serum and 1 mg/mL CMV antigen (Dade Behring, Marburg, Germany) in tissue culture flasks. The cocultures were restimulated on day 7 with further autologous DCs and 0.5 mg/mL CMV antigen. From day 10, cocultures were supplemented with 20 U/mL interleukin-2 (Sigma, Poole, United Kingdom) every 2 days. For cocultures maintained longer than 14 days, there was an additional restimulation with autologous CMV antigen–pulsed DCs on day 14.

Characterization of the coculture system

The proliferative and cytolytic characteristics of the cocultures have been detailed elsewhere.20 Briefly, in 15 donor cocultures studied, T-cell proliferation (as detected in [3H]thymidine incorporation assays) in response to the control antigen was significantly less than in response to the HCMV antigen (P = .0007, Wilcoxon signed rank test) but often slightly greater than proliferation without any antigen (P = .01, Wilcoxon signed rank test). There was a median 2.1-fold expansion in total lymphocyte numbers over the 2-week culture period. Unseparated mononuclear cells obtained from the culture were used as effectors in cytotoxicity assays. Cells from 12 of 14 cocultures from HCMV-seropositive donors tested showed HCMV-specific cytotoxicity against HCMV-infected fibroblasts/DCs or antigen-pulsed DCs (median 14.7% specific lysis, range 6.1%-42% at maximal effector:target ratios of 20:1, vs median 0%, range 0%-5.1% for uninfected/unpulsed targets; P = .002, Wilcoxon signed rank test). In general, therefore, the culture conditions promoted the development of HCMV-specific cytotoxicity. In addition, killing was HLA-restricted because only autologous but not allogeneic HLA-mismatched target cells were lysed (median 0.8%, range 0%-5.1% specific lysis, P = .005, Wilcoxon signed rank test), and lysis was prevented by preincubation of targets with HLA class I blocking antibodies.

Tetrameric complex formation

Soluble MHC-peptide tetramers were produced using standard approaches.18 Briefly, recombinant HLA-A*0201 and HLA-B*0702 heavy chain and β2-microglobulin protein were produced in Escherichia coli cells transformed with the relevant expression vectors. Expression of the HLA heavy chain was limited to the extracellular domain, and the C-terminus of this domain was modified by the addition of a substrate sequence for the biotinylating enzyme BirA. The peptide epitopes were the HLA-A*0201–restricted NLVPMVATV epitope (amino acids 495-503 of the lower matrix protein pp65) and the HLA-B*0702 epitope, TPRVTGGGAM (amino acids 417-426 of the lower matrix protein pp65). Monomeric HLA-peptide complexes were folded in vitro by addition of HLA protein to β2-microglobulin in the presence of appropriate peptide. The MHC complexes were biotinylated using purified recombinant BirA enzyme and were then purified by gel filtration and anion exchange chromatography. HLA-peptide tetramers were made by mixing the biotinylated protein complex with streptavidin-phycoerythrin (PE) (Leinco, St Louis, MO) at a molar ratio of 4:1. Tetramers were purified by gel filtration on a Sephadex S-200 column (Amersham Pharmacia, St Albans, United Kingdom).

Flow cytometric analysis

Lymphocytes from day 14 or day 21 cocultures were both triple-stained with fluorescein isothiocyanate–conjugated anti-CD4 mAb (Dako, Ely, United Kingdom), PE-conjugated anti-CD8 mAb (Dako), and PE Cy5–conjugated anti-CD3 mAb (Dako) and dual-stained with the appropriate HLA-restricted HCMV-specific tetrameric complex and PE Cy5–conjugated anti-CD8 mAb (Dako) in 20 to 30 μL phosphate-buffered saline, 2% bovine serum albumin, and azide for 60 minutes on ice. Cells were washed twice in the same buffer and analyzed immediately in a flow cytometer (BeckmanCoulter, High Wycombe, United Kingdom). Between 4 × 106 and 1 × 107 cells were analyzed in the samples stained with tetrameric complex. Negative controls included cells from HCMV-seronegative individuals expressing the appropriate HLA antigen and from HCMV-seropositive individuals not expressing the appropriate HLA antigen. In addition, the HLA-A*0201 tetrameric complex provided a negative control for HLA-B*0702+ individuals and the HLA-B*0702 tetrameric complex for the HLA-A*0201+ individuals. These additional controls were performed for donors 01 to 04 but not thereafter in order to maximize the number of cells available for subsequent analysis. Cells demonstrating dual staining with CD8 mAb and tetrameric complex were electronically sorted on a BeckmanCoulter EPICS Elite flow cytometer.

TCR CDR3 spectratyping

RNA was extracted from preculture and postculture PBLs using Ultraspec RNA (BiotecX Laboratories, Houston, TX) according to the manufacturer's protocol. Complementary DNA (cDNA) was generated from 1 μg RNA in a 30 μL reaction using random hexanucleotide primers for reverse transcription with reverse transcriptase (Superscript, Gibco BRL, Paisley, United Kingdom). In the case of cells sorted according to dual staining with CD8 mAb and tetrameric complex (4000-17 000 cells), RNA was extracted in a similar manner but using glycogen (Boehringer Mannheim, Lewes, United Kingdom) as a carrier. The total RNA extracted was used to generate cDNA in a 15 μL reaction. Each of 22 functionally rearranged T-cell receptor (TCR) β chain variable (BV) gene subfamilies was amplified across the complementarity determining region 3 (CDR3)–encoding regions using the 24 BV subfamily-specific primers described previously by Maslanka et al21 and a fluorescent dye–conjugated (FAM, Perkin Elmer, Cambridge, United Kingdom) β chain constant (BC) region-specific primer (Table1). The polymerase chain reaction (PCR) product lengths using this technique reflect the CDR3 lengths of the input TCR RNA, being dependent upon joining (BJ) and diversity (BD) gene segment usage along with the balance of exonuclease activity and N nucleotide addition by terminal transferase at the junctional regions. Peaks corresponding to in-frame transcripts are detected at 3 nucleotide intervals. The appearance of a dominant peak suggests the presence of an oligoclonal or clonal T-cell population, while the absence of peaks or entire subfamily spectratypes suggests the absence of T cells of the given CDR3 length or BV subfamily, respectively. BV primers were combined in duplex PCR reactions as follows: BV 5.1 plus 1; BV2 plus 12; BV13 plus 3; BV4 plus 5.3; BV8 plus 7; BV9 plus 14; BV11 plus 20; BV17 plus 15; BV16 plus 21; BV18 plus 23; and BV24 plus 22. BV6.1 and BV6.2 were used unpaired.

Table 1.

Primer sequences of TCR BV and BC chains

TCR primerSequenceSize (mer)
BV1 CAGTTCCCTGACTTGCACTC 20 
BV2 GCTTCTACATCTGCAGTGC 19 
BV3 GAGAGAAGAAGGAGCGCTTC 20 
BV4 GCAGCATATATCTCTGCAGC 20 
BV5.1 CTCGGCCCTTTATCTTTGCG 20 
BV5.3 CCCTAACTATAGCTCTGAGC 20 
BV6.1 GATCCAGCGCACACAGC 17 
BV6.2 GATCCAGCGCACAGAGC 17 
BV7 CCTGAATGCCCCAACAGC 18 
BV8 GAACCCAGGGACTCAGCTG 19 
BV9 GGAGCTTGGTGACTCTGCTG 20 
BV11 CAGGCCCTCACATACCTCTCA 21 
BV12 CAAAGACAGAGGATTTCCTCC 21 
BV13 GTCGGCTGCTCCCTCCC 17 
BV14 GTCTCTCGAAAAGAGAAGAGG 21 
BV15 GTCTCTCGACAGGCACAGGC 20 
BV16 GAACTGGAGGATTCTGGAGTT 21 
BV17 CCAAAAGAACCCGACAGCTTTC 22 
BV18 GTGCGAGGAGATTCGGCAGC 20 
BV20 CACACCCCAGGACCGGCAG 19 
BV21 GGCTCAAAGGAGTAGACTCC 20 
BV22 GTTGAAAGGCCTGATGGATC 20 
BV23 CAGTTCAGTGACTATCATTCTG 22 
BV24 GGGGACGCAGCCATGTACC 19 
BC CTGTGTTTGAGCCATCAGAAGC 22 
TCR primerSequenceSize (mer)
BV1 CAGTTCCCTGACTTGCACTC 20 
BV2 GCTTCTACATCTGCAGTGC 19 
BV3 GAGAGAAGAAGGAGCGCTTC 20 
BV4 GCAGCATATATCTCTGCAGC 20 
BV5.1 CTCGGCCCTTTATCTTTGCG 20 
BV5.3 CCCTAACTATAGCTCTGAGC 20 
BV6.1 GATCCAGCGCACACAGC 17 
BV6.2 GATCCAGCGCACAGAGC 17 
BV7 CCTGAATGCCCCAACAGC 18 
BV8 GAACCCAGGGACTCAGCTG 19 
BV9 GGAGCTTGGTGACTCTGCTG 20 
BV11 CAGGCCCTCACATACCTCTCA 21 
BV12 CAAAGACAGAGGATTTCCTCC 21 
BV13 GTCGGCTGCTCCCTCCC 17 
BV14 GTCTCTCGAAAAGAGAAGAGG 21 
BV15 GTCTCTCGACAGGCACAGGC 20 
BV16 GAACTGGAGGATTCTGGAGTT 21 
BV17 CCAAAAGAACCCGACAGCTTTC 22 
BV18 GTGCGAGGAGATTCGGCAGC 20 
BV20 CACACCCCAGGACCGGCAG 19 
BV21 GGCTCAAAGGAGTAGACTCC 20 
BV22 GTTGAAAGGCCTGATGGATC 20 
BV23 CAGTTCAGTGACTATCATTCTG 22 
BV24 GGGGACGCAGCCATGTACC 19 
BC CTGTGTTTGAGCCATCAGAAGC 22 

The primers were as previously published by Maslanka et al.21 

Hot-start PCR amplifications were performed in a total volume of 20 μL containing Genamp PCR buffer (Perkin Elmer), 2 mM MgCl2, 0.2 mM each dNTP, 1 mM of each primer, and 1 μL cDNA (equivalent to approximately 25 000 cells). In the case of the cells sorted according to dual staining with CD8 mAb and tetrameric complex, the total cDNA was divided equally between the PCR reactions. After a 5-minute denaturation step at 95°C, 0.5 U Amplitaq DNA polymerase (Perkin Elmer) was added. Optimal cycling conditions were 95°C for 30 seconds, 58°C for 30 seconds, and 72°C for 45 seconds, for 30 cycles, followed by a final extension at 72°C for 5 minutes. One microliter of PCR product was denatured in 12 μL formamide and electrophoresed through Performance Optimized Polymer 4 (Perkin Elmer) on an ABI 110 automated sequencer (Perkin Elmer) in the presence of Tamra 500 size standard (Perkin Elmer). Genescan software 2.1 (Perkin Elmer) was used to analyze the data.

TCR CDR3 cloning and sequencing

Fresh spectratype PCR products were cloned using TA Cloning Kit Dual Promoter Version B (pCRII vector) with TOP10F′ One Shot chemically competent E coli (Invitrogen, Groningen, The Netherlands) according to the manufacturer's instructions. Transformed cells were selected by ampicillin resistance and blue/white colony screening and then plucked and cultured in 96-well plates in the presence of 50 μg/mL ampicillin (Gibco BRL). Cells from each well were further screened by PCR amplification of TCR CDR3 sequence as previously described using primers appropriate for the expected BV family. Those expressing a product of the appropriate size as determined by agar gel electrophoresis with ethidium bromide staining were amplified in 50 μL PCR reactions and the amplicons purified using Wizard Plus Minipreps DNA Purification System (Promega, Southhampton, United Kingdom) according to the manufacturer's instructions. The DNA yield and purity were determined by spectrophotometry. A total of 90 ng of the purified PCR product was used as the template for sequencing using the ABI PRISM Dye Terminator Cycle Sequencing Ready Reaction Kit (Perkin Elmer). The primers used were the same as those used for the TCR CDR3 spectratype PCR reactions. Cycle sequencing was performed on the GeneAmp PCR Systems 2400 (Perkin Elmer) (96°C for 10 seconds, 50°C for 5 seconds, 60°C for 4 minutes for 25 cycles with rapid thermal ramping). Following ethanol precipitation of the extension products, the samples were resuspended in 25 μL Template Suppression Reagent (Perkin Elmer), denatured for 95°C for 2 minutes, and electrophoresed through Performance Optimized Polymer 6 (Perkin Elmer) on an ABI 110 automated sequencer (Perkin Elmer). The data were processed using ABI Prism DNA Sequencing Analysis software (Perkin Elmer).

Phenotypic analysis of coculture output cells

The coculture output cells consisted of mixed populations of both CD4+ and CD8+ cells. The percentage of CD8-expressing cells varied between 10% and 47% (Table 1). HCMV-specific tetrameric complex staining of donor peripheral blood mononuclear cells prior to culture ranged from less than 0.1% to 0.6% of the CD8+ population. Following coculture, this increased to 0.3% to 8.0% of the CD8+ population, demonstrating a maximal 40-fold relative expansion. In 6 of 6 evaluable donors, there was at least a 10-fold expansion with 1 of the 2 HCMV-specific tetrameric complexes (Table 2 and Figure1). Donors 06 and 08 did not have preculture tetrameric complex studies performed and, thus, the degree of expansion in these donors was impossible to enumerate. The proliferation assay results for donor 08 were close to the median for the group (15 606 cpm; range, 11 875-56 574 cpm), suggesting that the low level of staining after the culture was not a result of a failure of the culture system to stimulate proliferation. Specificity of the tetrameric complexes was confirmed by the absence of staining of cells from HCMV-seronegative donors of the appropriate HLA type (both before the culture and after the culture) and the absence of staining of cells from HCMV-seropositive donors not expressing the appropriate HLA molecules (data not shown).

Table 2.

Phenotypic analysis of postculture T cells

DonorPostculture CD8 positivity (% CD3*cells)Tetramer binding (% CD8* cells)
HLA-A*0201 tetramerHLA-B*0702 tetramer
PreculturePostculturePreculturePostculture
01 25 <0.1 0.3 0.6 6.5 
02 10 0.2 8.0   
03 12 0.1 1.6   
04 35 0.1 5.4   
05 26 0.1 1.3   
06 47 NA 0.3 NA 7.9 
07 21 0.1 1.1   
08 39 NA 0.4   
DonorPostculture CD8 positivity (% CD3*cells)Tetramer binding (% CD8* cells)
HLA-A*0201 tetramerHLA-B*0702 tetramer
PreculturePostculturePreculturePostculture
01 25 <0.1 0.3 0.6 6.5 
02 10 0.2 8.0   
03 12 0.1 1.6   
04 35 0.1 5.4   
05 26 0.1 1.3   
06 47 NA 0.3 NA 7.9 
07 21 0.1 1.1   
08 39 NA 0.4   

NA indicates not available.

Fig. 1.

Phenotypic analyses of preculture and postculture cells from donor 01.

Cells were dual-stained with PE Cy5–conjugated anti-CD8 mAb and PE-conjugated HCMV pp65–specific HLA-B*0702 tetrameric complexes. The single parameter histograms are gated onto the CD8+ cell population and demonstrate an increase in the population of cells binding the tetrameric complexes over the period of culture.

Fig. 1.

Phenotypic analyses of preculture and postculture cells from donor 01.

Cells were dual-stained with PE Cy5–conjugated anti-CD8 mAb and PE-conjugated HCMV pp65–specific HLA-B*0702 tetrameric complexes. The single parameter histograms are gated onto the CD8+ cell population and demonstrate an increase in the population of cells binding the tetrameric complexes over the period of culture.

Close modal

TCR CDR3 spectratyping

Preculture spectratypic analysis revealed typically complex appearances, with some skewing of a basic background Gaussian distribution of size classes evident in some of the BV subfamilies. Comparison of preculture and postculture unselected TCR CDR3 spectratypes showed that postculture T-cell repertoires were still polyclonal, ie, most BV spectratypes displayed a complete set of size class peaks. While most spectratypes were similar to the preculture spectratypes, some BV families contained one or more predominant size classes after the culture. When analysis was restricted to the CD8+ subset of cells, 7 of the 8 donors showed much more highly restricted postculture T-cell repertoires with fewer peaks and more irregular profiles (Figure 2). This became more pronounced when the culture period was prolonged to 21 days (Figure 3). The spectratypes of the cells sorted on the basis of dual staining with anti-CD8 mAb and HCMV-specific tetrameric complex showed even more highly restricted T-cell repertoires (Figures 3 and 4). In general, either no size classes were represented within a given BV subfamily (Figure 4, BV12, BV3, and BV9), or between 1 and 4 size classes were represented with a predominance of 1 or 2 members (Figure3, BV6.1, and Figure 4, BV13 and BV14). In 5 donors a minority of the BV subfamilies (range, 2-4; median, 3) demonstrated more than 5 peaks following tetrameric complex–guided sorting, but these were generally within a restricted subset of subfamilies (mainly BV6.1/6.2, BV13, and BV2), and again there was a predominance of 1 or 2 members (Figure 4, BV2 and BV6.1). The predominant peaks selected following tetrameric complex–guided sorting often corresponded to the expansions demonstrated in the unselected CD8+ subset, suggesting that the peptide specificity of the anti-HCMV response was skewed toward pp65 (Figure 3, BV6.1). However, examples were seen of both tetrameric complex–sorted cell size class peaks without correspondingly expanded size classes in the unselected CD8+ subset and expanded size classes in the unselected CD8+ subset, which were not present following tetrameric complex–guided sorting (Figure5). The former can be explained by selection of HCMV-specific CTLs that account quantitatively for less of the overall anti-HCMV immune response. The latter may represent expansions to other pp65 epitopes (associated with the same or different HLA molecules), to alternate HCMV proteins, or to antigens unrelated to HCMV.

Fig. 2.

TCRBV spectratypic profiles from donor 04.

Profiles are for both (A) the unselected preculture PBLs and (B) the postculture CD8+ population of cells. The postculture profile showed a more restricted repertoire, with prominent skewing evident in a number of families.

Fig. 2.

TCRBV spectratypic profiles from donor 04.

Profiles are for both (A) the unselected preculture PBLs and (B) the postculture CD8+ population of cells. The postculture profile showed a more restricted repertoire, with prominent skewing evident in a number of families.

Close modal
Fig. 3.

TCRBV spectratypic profiles from donor 02 at 2 and 3 weeks of culture.

BV6.1 profiles are shown for (A) preculture, (B) postculture unselected, (C) postculture CD8+ selected, and (D) postculture HCMV pp65–specific HLA-A*0201 tetrameric complex–selected cells. The tetramer-selected population showed a highly restricted repertoire. The major size class remained the same and became more apparent in the unselected population after 3 weeks of culture.

Fig. 3.

TCRBV spectratypic profiles from donor 02 at 2 and 3 weeks of culture.

BV6.1 profiles are shown for (A) preculture, (B) postculture unselected, (C) postculture CD8+ selected, and (D) postculture HCMV pp65–specific HLA-A*0201 tetrameric complex–selected cells. The tetramer-selected population showed a highly restricted repertoire. The major size class remained the same and became more apparent in the unselected population after 3 weeks of culture.

Close modal
Fig. 4.

TCRBV spectratypic profiles from donor 05.

Profiles are for (A) the unselected preculture PBLs and (B) the postculture HCMV pp65–specific HLA-A*0201 tetrameric complex–selected cells. Many BV families had no representative members following tetramer-guided sorting. There was a predominance of 1 or 2 size classes in those families in which representatives were present.

Fig. 4.

TCRBV spectratypic profiles from donor 05.

Profiles are for (A) the unselected preculture PBLs and (B) the postculture HCMV pp65–specific HLA-A*0201 tetrameric complex–selected cells. Many BV families had no representative members following tetramer-guided sorting. There was a predominance of 1 or 2 size classes in those families in which representatives were present.

Close modal
Fig. 5.

TCRBV spectratypic profiles from donor 04 for BV1 and BV13.

Profiles are for (A) preculture, (B) postculture CD8+selected, and (C) postculture HCMV pp65–specific HLA-A*0201 tetrameric complex–selected cells. Examples are shown of populations that were expanded after the culture that were selected based on tetramer-guided sorting (BV13—the larger of the 2 major size classes in panel B) along with those that were not selected (BV1 and BV13). In addition, the size class selected in panel C for BV1 showed no correspondingly expanded size class peak in the CD8+ selected population.

Fig. 5.

TCRBV spectratypic profiles from donor 04 for BV1 and BV13.

Profiles are for (A) preculture, (B) postculture CD8+selected, and (C) postculture HCMV pp65–specific HLA-A*0201 tetrameric complex–selected cells. Examples are shown of populations that were expanded after the culture that were selected based on tetramer-guided sorting (BV13—the larger of the 2 major size classes in panel B) along with those that were not selected (BV1 and BV13). In addition, the size class selected in panel C for BV1 showed no correspondingly expanded size class peak in the CD8+ selected population.

Close modal

The number of BV subfamilies in which at least one size class was represented following HLA-A*0201 tetrameric complex–guided sorting was variable between donors (range, 5-20; median, 10). Due to variations in amplification efficiency between individual PCR reactions, strict quantitative comparison of the PCR products between subfamilies for any given individual is not valid. Allowing for this caveat, a normal spectratypic profile is composed of 180 to 190 size class peaks with no individual peak accounting for more than 5% of the total peak heights and/or area under the peaks. The spectratypes of all 8 donors whose postculture cells were sorted on the basis of dual staining with anti-CD8 mAb and HCMV-specific HLA-A*0201 tetrameric complex showed at least 3 (range, 3-5; median, 4) “major” peaks accounting for more than 5% of the total area under the peaks. In 5 of these donors (all with 4 major peaks), the combined area of these major peaks accounted for more than 50% of the total area under the peaks (51%-71%, median 61%), demonstrating a highly focused response to the single HCMV pp65 epitope. In these donors the combined area of the 10 largest peaks accounted for 63% to 91% of the total. In the other 3 donors, the combined area of the major peaks accounted for 31% to 42% and of the 10 largest peaks, 50% to 62% of the total area under the peaks. These donors therefore appeared to show a slightly less focused immune response, although overall a continuous spectrum of response was evident in terms of diversity of size class peaks represented.

There was clear evidence of preferential BV gene usage among donors. Major peaks from HLA-A*0201 tetrameric complex–sorted cells were present in BV6.1/6.2 and BV8 in 6 of 8 donors and in BV13 in 4 of 8 donors (Figure 6). At least one size class peak was present in BV6.1/6.2 and BV8 in all 8 donors and in BV13 in 7 of 8 (Figure 7).

Fig. 6.

BV gene usage of the major size class peaks selected on the basis of HCMV pp65–specific HLA-A*0201 tetrameric complex staining.

A major peak was defined as accounting for more than 5% of the total area under the peaks in the TCRBV spectratypic profile of an individual donor.

Fig. 6.

BV gene usage of the major size class peaks selected on the basis of HCMV pp65–specific HLA-A*0201 tetrameric complex staining.

A major peak was defined as accounting for more than 5% of the total area under the peaks in the TCRBV spectratypic profile of an individual donor.

Close modal
Fig. 7.

TCRBV spectratypic profiles for BV8 and BV13 of all 8 donors following HCMV pp65–specific HLA-A*0201 tetrameric complex–guided sorting.

The profiles are aligned according to PCR product size with a control profile for comparison and show a lack of conservation of a single TCR CDR3 size among the donors.

Fig. 7.

TCRBV spectratypic profiles for BV8 and BV13 of all 8 donors following HCMV pp65–specific HLA-A*0201 tetrameric complex–guided sorting.

The profiles are aligned according to PCR product size with a control profile for comparison and show a lack of conservation of a single TCR CDR3 size among the donors.

Close modal

The results following sorting on the basis of dual staining with anti-CD8 mAb and HLA-B*0702 tetrameric complex in donors 01 and 06 were similar to those obtained with the HLA-A*0201 tetrameric complex. In both donors there were 20 to 30 times more CD8+ cells that bound the HLA-B*0702 tetrameric complex as compared with the HLA-A*0201 tetrameric complex following culture (Table2). Postculture spectratypes following tetrameric complex–guided sorting demonstrated 4 and 6 major size class peaks, respectively, which accounted for 78% and 46% of the total area under the peaks. Representatives were present in 8 BV subfamilies in donor 01 and 23 in donor 06, showing the same extremes of the spectrum of immune response focusing seen with the HLA-A*0201 tetrameric complexes. Major peaks were present in BV6.2 and BV14 in donor 01 (2 major peaks in each) and in BV1, BV4, BV9, BV14, BV6.1, and BV6.2 in donor 06.

Overall there appeared to be a lack of TCR CDR3 length conservation of the tetrameric complex–sorted T cells for a given BV family when compared between different donors (Figure 7).

Sorting based on HLA-A*0201 tetrameric complex binding was performed on 3 separate occasions on cells cultured from donor 02. Comparison of the spectratypic analysis from cells selected after 2 and 3 weeks of culture showed that the most abundant size classes remained identical (Figure 3). Analysis of cells recultured for 2 weeks from fresh peripheral blood mononuclear cells also demonstrated that the same size classes were selected. These data suggest that HCMV pp65 epitope-specific CTLs with the same CDR3 size are reproducibly expanded in the culture system and that the effects of stochastic events on the postsorting spectratype appearances remain minimal despite the relatively low numbers of cells used for RNA preparation following sorting.

Cloning and sequencing of CDR3 spectratype PCR products

Individual size class peaks of the CDR3 spectratype analysis may represent the CDR3-encoding RNA of a single T-cell clone or may represent the CDR3-encoding RNA of multiple clones with identical CDR3 length. To test whether the immune responses to HCMV resulted in preferential expansions of multiple clones with a particular CDR3 length or of a single clone within a given BV family, we cloned the spectratype PCR products and sequenced the corresponding DNA regions. The postsorting spectratypes of donor 01 showed an identical size peak selected in the BV6.1 subfamily with both the HLA-A*0201 and HLA-B*0702 tetrameric complexes (Figure 8). The sequence of 7 clones from the HLA-B*0702–sorted population was identical. That of 10 clones from the HLA-A*0201–sorted population was also identical but differed from that of the HLA-B*0702–sorted cells in both J region usage and VDJ region sequence (Figure 8). These results confirm the specificity of the 2 distinct HLA tetrameric complexes and suggest that each size peak is composed of either a single clone or a very limited number of clones. Sequence comparison with the HLA-A*0201 tetrameric complex–sorted major size class peak from BV6.1 of donor 02 (5 of 5 clones with identical sequence) showed different J region usage and different CDR3 length (Figure9). Thus, T-cell clones sharing the same BV region and selected by the same HLA tetrameric complex in different individuals display markedly different CDR3 nucleotide and predicted amino acid sequences. Similarly, comparison of the sequences derived from size class peaks in the same or different BV families from a single donor selected with the same HLA-A*0201 tetrameric complex (BV6.1 and BV15 [10 of 10 clones with identical sequence] in Figure9) demonstrates that a single HLA molecule–nonapeptide combination can be recognized by a number of TCRs with differing TCR CDR3 lengths and differing predicted amino amino acid sequences within the same individual.

Fig. 8.

Sequence analysis of tetramer-selected populations.

The panel on the left shows the postculture TCRBV spectratypic profiles for BV6.1 of donor 01, including the unselected, the HLA-B*0702 tetramer–selected, and HLA-A*0201 tetramer–selected populations. Seven and 10 clones representing the major peaks (shaded) of the HLA-B*0702 tetramer–selected and HLA-A*0201 tetramer–selected populations, respectively, showed identical sequence, indicating that each size class peak is composed of a single or very limited number of clones. The sequence data confirm a lack of conservation of CDR3 motifs between the clones selected by the different tetrameric complexes.

Fig. 8.

Sequence analysis of tetramer-selected populations.

The panel on the left shows the postculture TCRBV spectratypic profiles for BV6.1 of donor 01, including the unselected, the HLA-B*0702 tetramer–selected, and HLA-A*0201 tetramer–selected populations. Seven and 10 clones representing the major peaks (shaded) of the HLA-B*0702 tetramer–selected and HLA-A*0201 tetramer–selected populations, respectively, showed identical sequence, indicating that each size class peak is composed of a single or very limited number of clones. The sequence data confirm a lack of conservation of CDR3 motifs between the clones selected by the different tetrameric complexes.

Close modal
Fig. 9.

TCR CDR3 sequence data for clones corresponding to the size class peaks indicated for donor 01 and donor 02 (selected with the HLA-A*0201 tetrameric complex).

There was a lack of conservation of CDR3 length and no evidence of frequent usage of public clonotypes both between the same BV family in different donors (BV6.1 donor 01 and donor 02), different size class peaks from the same BV family in a single donor (BV6.1 donor 02), and different BV families in a single donor (BV6.1 and BV15 donor 02).

Fig. 9.

TCR CDR3 sequence data for clones corresponding to the size class peaks indicated for donor 01 and donor 02 (selected with the HLA-A*0201 tetrameric complex).

There was a lack of conservation of CDR3 length and no evidence of frequent usage of public clonotypes both between the same BV family in different donors (BV6.1 donor 01 and donor 02), different size class peaks from the same BV family in a single donor (BV6.1 donor 02), and different BV families in a single donor (BV6.1 and BV15 donor 02).

Close modal

The sequences of the TCR CDR3 regions corresponding to the major peaks in BV15 and BV6.1 of donor 02 following culture and tetrameric complex–guided sorting on 2 separate occasions were also identical (Figure 10), confirming that the expanded T-cells with conserved TCR CDR3 lengths were indeed due to reproducible expansions of the same clones in these cases.

Fig. 10.

Reproducibility of clonal expansion and tetrameric complex–guided selection.

The spectratype appearances of BV15 and BV6.1 are shown following coculture and tetrameric complex–guided sorting on 2 separate occasions for donor 02. The major peaks selected were identical in size and sequence.

Fig. 10.

Reproducibility of clonal expansion and tetrameric complex–guided selection.

The spectratype appearances of BV15 and BV6.1 are shown following coculture and tetrameric complex–guided sorting on 2 separate occasions for donor 02. The major peaks selected were identical in size and sequence.

Close modal

We have studied immune responses to HCMV antigen in a culture system that is free of live HCMV virions and that uses monocyte-derived DCs as antigen-presenting cells (APCs). DCs are potent APCs that are able to process exogenously supplied antigen and present peptide fragments on both class I and class II HLA molecules, along with an array of costimulatory molecules.22,23 This in turn allows stimulation of lymphocytes expressing either CD8 or CD4 in combination with an appropriate TCR. We have previously demonstrated that cells derived from such a culture system that has been used to stimulate a recall response to HCMV antigen can be shown to possess HCMV-specific HLA-restricted cytotoxic activity, although the system has not allowed the expansion of HCMV-specific T cells from HCMV-naive donors.20 The development of techniques to allow identification of antigen-specific populations of T cells such as the enzyme-linked immunospot assay and HLA tetrameric complexes now allows more detailed analyses of immune responses.17,18 These developments are in part based on an increasing knowledge of the immunodominant epitopes involved in specific responses. Tetrameric complex analysis is particularly suited to cases in which restricted numbers of defined epitopes dominate the response, allowing direct quantification of the antigen-specific cells. Studies in healthy virus carriers of the immune response to HCMV pp65 have shown that in some individuals the peptide specificity of the pp65-specific CTLs is highly focused to a single peptide while in others multiple pp65 peptides are recognized.14 These studies have helped to define the minimal CD8+ T-cell epitopes of HCMV and their HLA restrictions,14,24 which in turn have allowed determination of which of these combinations can be formed into stable HLA tetrameric-peptide complexes. This study has therefore used 2 peptide-HLA combinations that have been shown to allow formation of such stable tetrameric complexes,25 which appear to be immunodominant in at least some individuals in a number of previous studies,14,24,26 and which allow examination of most individuals in view of the prevalence of these HLA alleles in our study population. However, evidence is also emerging that in some individuals the major immediate-early protein (IE-1) may be the major target for CD8+ T cells with only a minor or undetectable response to pp65.27 

In the present study we show an increase in the number of HCMV pp65–specific T cells in donors expressing HLA-A*0201 and/or HLA-B*0702 over the time period of the culture. This confirms that the culture conditions result in expansion of a population of cells expressing CD8 that are able to recognize a single HLA molecule–peptide combination in agreement with earlier functional studies20 and adds to the previous literature suggesting that DCs derived from a variety of sources are able to present exogenous antigen constitutively on their MHC class I molecules following antigen capture by constitutive macropinocytosis.28 The relative importance of this mode of antigen presentation in vivo compared with classical MHC class I pathway presentation of pathogen-encoded proteins synthesized de novo within DCs remains unclear. Similarly, any differences in the antigenic profiles that are presented by these alternate pathways, as has been suggested for the EBV-encoded nuclear antigen EBNA1,29 are unknown. In the context of presentation of exogenous antigen, the results of this study suggest that none of the donors demonstrated a recall response completely polarized toward IE-1 to the exclusion of pp65, although a possible explanation for the relatively low level of tetrameric complex binding after the culture in donor 08 could relate to either a broader antigen or pp65 peptide–directed immune response in this donor. These findings may have implications for the mechanism of host control over viral replication in vivo. The persistently high levels of HCMV-specific CTLs in immunocompetent individuals suggest ongoing stimulation of the immune response.30 HCMV possesses numerous strategies to evade the immune response. These include resistance to natural killer cell–mediated lysis through surface expression of a MHC class I–like molecule31 and disruption of inducible MHC class I and class II expression resulting in reduced presentation of HCMV-associated antigens.32,33 However, nested PCR for HCMV DNA suggests very low viral loads in immunocompetent individuals (at least for the analytes studied, which include peripheral blood leucocytes, plasma, and serum).34,35 One possible explanation for the paradoxical existence of high circulating levels of HCMV-specific CTLs but low viral load could be that viral proteins expressed in mononuclear cells, the proposed “latent” reservoir of HCMV, continually stimulate CTLs. Our data show that oligoclonal pp65-specific CD8+ T-cell responses are restimulated following indirect presentation of CMV antigen and suggest that active viral replication may not be required to maintain the high level of CMV-specific CTLs seen in vivo.

An alternative explanation for those cases with low postculture tetrameric complex binding would be to invoke the possibility of greater in vitro deletion (ie, activation-induced cell death [AICD]) of lymphocytes in these cases. It has been demonstrated in animal models that peripheral T cells may be induced to die following high-intensity TCR signaling36 and that the result of viral infection in terms of viral clearance and induction of AICD may be determined by viral load alone.37 However, various costimulatory signals have been described that appear to reduce the potential for AICD. Probably the best described of these is CD28, which is activated by CD80 and CD86 (both expressed on APCs),38resulting in up-regulation of Bcl-xL39 and FLIP40 (both antiapoptotic) and down-regulation of FasL.41 Thus, DCs may be capable of inducing strong T-cell activation while protecting the cells from AICD. Subsequent antigen encounter (ie, in pathologically infected cells) and induction of apoptosis following appropriate effector function could provide one form of homeostatic control. We have previously published data showing that increasing antigen concentration in the DC/T-cell coculture system results in reducing levels of T-cell proliferation measured on day 6 of coculture and data on the variability in expansion of T cells between individuals under the same conditions.20 However, there was no evidence that those cases with the lowest postculture tetrameric complex binding exhibited either greater or lesser proliferation than those with higher levels of binding, nor that there was a greater degree of cell death as assessed by flow cytometry (data not shown) in these donors. Further studies into the degree of protection from AICD that is afforded by coculture and repeated restimulation with DCs and, in particular, comparison between differing approaches using alternate stimulants (eg, HCMV peptides versus proteins, mature versus immature DCs, and DCs versus infected fibroblasts) are warranted.

Analysis of TCRBV spectratypes gives a more detailed picture of total T-cell repertoire diversity than is available from phenotypic studies of either CD antigen or antigen-specific TCR (as assessed by tetramer complex binding) expression alone. While not strictly quantitative when compared across different primer pairs due to variations in amplification efficiency, the relative amounts of each size class PCR product within a given BV family do give some idea of the relative contributions of clones with differing CDR3 lengths to the diversity of the individual family. Attempts to ascribe clinical correlates to temporal alterations in the spectratypic profiles of PBLs have been strengthened by the demonstration of expansions of lymphocytes using the same BV genes and with identical CDR3 length within disease lesions (eg, tumor-infiltrating lymphocytes21 or those within the skin lesions of graft versus host disease42,43 or infiltrating the target organs in autoimmune disorders44). However, the lack of evidence that cells of a given CDR3 length have a defined antigen specificity has meant that the biological significance of alterations in such profiles detected in PBLs has remained speculative. In the environment of evolving immune reconstitution that prevails following allogeneic transplantation, changes in spectratypic patterns are to be expected. By combining a technique that allows isolation of antigen-specific T cells with TCRBV spectratypic analysis, we have been able to demonstrate the BV gene and CDR3 length usage of HCMV pp65 epitope-specific clones. This provides the strongest direct evidence to date that temporal alterations in spectratypic profiles relate to expansions of T cells of known antigen specificity, providing a tool for monitoring adoptive immunotherapy protocols.

Advances allowing more detailed analyses of the clonal composition of immune responses to defined antigens are giving increasing insight into the degree of diversity of T-cell responses, which in part reflect the degeneracy of the TCR–MHC-peptide interaction. It is well established that the interaction between TCR and MHC-peptide shows some degree of degeneracy in both directions, both for MHC class I–CD8 and MHC class II–CD4 interactions.45,46 A single TCR can recognize structurally distinct MHC-peptide complexes,47 and ontogenetically thymocytes bearing a specific TCR can be positively selected by a single MHC molecule in combination with a variety of peptides. Conversely, a single MHC-peptide combination can positively select thymocytes bearing many distinct TCRs,48-50 albeit based on interactions with relatively low avidity (because high-avidity interactions result in negative selection during this stage of development). It has been estimated in a murine model that these low-avidity interactions may result in a minimum of 105different BV rearrangements being selected by a single peptide-MHC class II complex.51 The diversity of the T-cell repertoire selected for the higher-avidity reactions required for antigen recognition and subsequent triggering of cellular response and proliferation as part of an immune response to exogenous antigen has been studied for a number of viral antigens and/or epitopes, including those from influenza A, HTLV-1, and EBV.52-56 Preferential usage of certain BV families by the TCR of T cells responding to specified peptides has been reported in these studies. Our study suggests that this is also true for responses to HCMV in HLA-A*0201+ donors, confirming the BV gene usage (BV8, BV6.1/6.2, and BV13) previously demonstrated by multiple independently derived epitope-specific CTL clones generated by formal single-cell cloning or from clonal CTL microculture.24 Although we have fewer results for HLA-B*0702+ donors, the possible preferential usage of BV14 and BV6.1/6.2 by CTL clones defined by binding of tetrameric complexes generated with the HLA-B*0702–restricted peptide similarly correlates with those documented in the previous study.24 Sequencing of clones selected according to their ability to lyse pp65-pulsed target cells following limiting dilution analysis has suggested that the memory CTL response to individual HCMV pp65 epitopes is highly focused.24 Tetrameric complex selection would not be predicted to select specifically for those clones with the greatest lytic capacity but rather to select all cells bearing TCRs that bind the complexes with sufficient avidity. Thus, while tetrameric complexes have been found to select for the highest-avidity T cells and would indeed be expected to select the clones with greatest lytic capacity, they would not do so selectively.57 The prediction that tetrameric complex–guided sorting would identify a larger number of T-cell clones than techniques based on assessment of lytic activity may in part explain the higher frequencies of antigen-specific T cells reported by this technique compared with previous studies using limiting dilution analysis.58,59 Indeed, T cells selected by single-specificity soluble HLA tetrameric-peptide complexes have been shown to be heterogeneous in terms of cell surface immunophenotype (CD11a, CD45RO/RA, and CD28 expression) and intracellular cytokine production,18,30,60-62suggesting some possible heterogeneity of function. In addition, the ability of tetramers to select a population of cells that are functionally unresponsive despite having many of the hallmarks of effector T cells (ie, cells that appear to have been selectively rendered anergic in vivo) has been demonstrated in the setting of a tumor-specific target.60 While our data do demonstrate a larger number of clonal T-cell expansions to a single pp65 epitope-MHC combination than the study by Weekes et al,24 they confirm that the response to specific HCMV pp65 epitopes is highly focused, apparently to a greater degree in some donors compared with others.

The general lack of conservation of other TCR gene segments and CDR3 length in HCMV pp65–specific T cells, both when comparing between different donors and between different CTL clones derived from the same donor, is also in agreement with the results of Weekes et al.24 These findings differ from those published for a dominant HLA-A2–restricted EBV epitope reported recently by Lim et al,56 where there was a frequent contribution of T-cell clonotypes with public TCR features (conserved CDR3 length and predicted TCR motifs) detected following selection with peptide-specific HLA tetrameric complexes. The suggestion that this feature may permit direct molecular follow-up of T-cell responses by, for example, use of primers specific for public clonotype junctions in BV spectratypic analysis (as elegantly demonstrated for EBV) may therefore not hold true for other antigenic responses such as that to HCMV. Analysis of HTLV-1 Tax-specific CTLs from different donors has also demonstrated largely unrelated TCR usage,55 and class II–restricted tetanus toxin peptide-specific T cells similarly have been shown to display highly heterogeneous CDR3 regions.63While analysis of larger numbers of donors would be required to fully exclude any common structural motifs within the CDR3 encoding region, our data do add to an the increasing literature suggesting that distinct TCRs may recognize a peptide-MHC complex by binding to it with differing primary TCR contact residues.14,24,55,61,63 

In summary, we have combined the use of 2 techniques that are able to demonstrate differing facets of the immune response to gain further insight into the human immune response to HCMV in an in vitro culture system that has potential for application in immunotherapeutic protocols. We have shown that there is diversity between individuals in terms of the number of TCR clones that recognize single HLA-restricted viral epitopes, but that a small number of clones appear to predominate, and that there is preferential usage of certain BV gene segments within this restricted part of the overall immune response. Unlike the case for EBV, there does not appear to be conservation of CDR3 length or sequence (including J region usage) between donors for the HLA-peptide combinations we have studied. The ability to determine the BV gene usage and CDR3 size of the pp65 epitope-specific CTLs will allow comparison of in vitro culture systems with responses detected in vivo during periods of HCMV reactivation following allogeneic bone marrow transplantation and allow monitoring of the fate of infused cells during adoptive immunotherapy protocols. Correlation with quantitative techniques for evaluation of HCMV load will allow investigation of the relationships between immune response and viral dynamics in early infection. Attempts to further enrich infused cells for HCMV specificity (for example, by selection of cells expressing TCRs with a restricted range of BV genes or those binding specific HLA-tetramer-peptide combinations) may help to determine the “sufficient T-cell repertoire” for reestablishment of host control over viral replication, because the heterogeneity of responses (both in terms of the degree of focusing toward specific pp65 peptides and in terms of the clonal diversity directed toward individual HLA-tetramer-peptide combinations) suggests that any such enrichment based on positive selection is unlikely to encompass the entirety of the human anti-HCMV immune response.

Supported by the Leukaemia Research Fund, London, United Kingdom, and by a grant from the Golden Charitable Trust, London.

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 U.S.C. section 1734.

1
Borysiewicz
LK
Graham
S
Hickling
JK
Mason
PD
Sissons
JG
Human cytomegalovirus-specific cytotoxic T cells: their precursor frequency and stage specificity.
Eur J Immunol.
18
1988
269
275
2
Meyers
JD
Flournoy
N
Thomas
ED
Nonbacterial pneumonia after allogeneic marrow transplantation: a review of ten years' experience.
Rev Infect Dis.
4
1982
1119
1132
3
Meyers
JD
Flournoy
N
Thomas
ED
Risk factors for cytomegalovirus infection after human marrow transplantation.
J Infect Dis.
153
1986
478
488
4
Broers
AE
van Der
HR
van Esser
JW
et al
Increased transplant-related morbidity and mortality in CMV-seropositive patients despite highly effective prevention of CMV disease after allogeneic T-cell–depleted stem cell transplantation.
Blood.
95
2000
2240
2245
5
Reusser
P
Riddell
SR
Meyers
JD
Greenberg
PD
Cytotoxic T-lymphocyte response to cytomegalovirus after human allogeneic bone marrow transplantation: pattern of recovery and correlation with cytomegalovirus infection and disease.
Blood.
78
1991
1373
1380
6
Riddell
SR
Watanabe
KS
Goodrich
JM
Li
CR
Agha
ME
Greenberg
PD
Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones.
Science.
257
1992
238
241
7
Einsele
H
Ehninger
G
Hebart
H
et al
Polymerase chain reaction monitoring reduces the incidence of cytomegalovirus disease and the duration and side effects of antiviral therapy after bone marrow transplantation.
Blood.
86
1995
2815
2820
8
Boeckh
M
Gooley
TA
Myerson
D
Cunningham
T
Schoch
G
Bowden
RA
Cytomegalovirus pp65 antigenemia-guided early treatment with ganciclovir versus ganciclovir at engraftment after allogeneic marrow transplantation: a randomized double-blind study.
Blood.
88
1996
4063
4071
9
Papadopoulos
EB
Ladanyi
M
Emanuel
D
et al
Infusions of donor leukocytes to treat Epstein-Barr virus-associated lymphoproliferative disorders after allogeneic bone marrow transplantation.
N Engl J Med.
330
1994
1185
1191
10
Riddell
SR
Greenberg
PD
Principles for adoptive T cell therapy of human viral diseases.
Annu Rev Immunol.
13
1995
545
586
11
Greenberg
PD
Reusser
P
Goodrich
JM
Riddell
SR
Development of a treatment regimen for human cytomegalovirus (CMV) infection in bone marrow transplantation recipients by adoptive transfer of donor-derived CMV-specific T cell clones expanded in vitro.
Ann N Y Acad Sci.
636
1991
184
195
12
Matloubian
M
Concepcion
RJ
Ahmed
R
CD4+ T cells are required to sustain CD8+ cytotoxic T-cell responses during chronic viral infection.
J Virol.
68
1994
8056
8063
13
McLaughlin-Taylor
E
Pande
H
Forman
SJ
et al
Identification of the major late human cytomegalovirus matrix protein pp65 as a target antigen for CD8+ virus-specific cytotoxic T lymphocytes.
J Med Virol.
43
1994
103
110
14
Wills
MR
Carmichael
AJ
Mynard
K
et al
The human cytotoxic T-lymphocyte (CTL) response to cytomegalovirus is dominated by structural protein pp65: frequency, specificity, and T- cell receptor usage of pp65- specific CTL.
J Virol.
70
1996
7569
7579
15
Riddell
SR
Rabin
M
Geballe
AP
Britt
WJ
Greenberg
PD
Class I MHC-restricted cytotoxic T lymphocyte recognition of cells infected with human cytomegalovirus does not require endogenous viral gene expression.
J Immunol.
146
1991
2795
2804
16
Borysiewicz
LK
Hickling
JK
Graham
S
et al
Human cytomegalovirus-specific cytotoxic T cells. Relative frequency of stage-specific CTL recognizing the 72-kD immediate early protein and glycoprotein B expressed by recombinant vaccinia viruses.
J Exp Med.
168
1988
919
931
17
Miyahira
Y
Murata
K
Rodriguez
D
et al
Quantification of antigen specific CD8+ T cells using an ELISPOT assay.
J Immunol Methods.
181
1995
45
54
18
Altman
JD
Moss
PAH
Goulder
PJR
et al
Phenotypic analysis of antigen-specific T lymphocytes.
Science.
274
1996
94
96
19
Ibisch
C
Saulquin
X
Gallot
G
et al
The T cell repertoire selected in vitro against EBV: diversity, specificity, and improved purification through early IL-2 receptor alpha-chain (CD25)-positive selection.
J Immunol.
164
2000
4924
4932
20
Peggs
KS
Verfuerth
S
Mackinnon
S
Induction of CMV specific T cell responses using dendritic cells pulsed with CMV antigen: a novel culture system that is free of live CMV virions.
Blood.
97
2001
994
1000
21
Maslanka
K
Piatek
T
Gorski
J
Yassai
M
Gorski
J
Molecular analysis of T cell repertoires. Spectratypes generated by multiplex polymerase chain reaction and evaluated by radioactivity or fluorescence.
Hum Immunol.
44
1995
28
34
22
Banchereau
J
Steinman
RM
Dendritic cells and the control of immunity.
Nature.
392
1998
245
252
23
Sigal
LJ
Crotty
S
Andino
R
Rock
KL
Cytotoxic T-cell immunity to virus-infected non-haematopoietic cells requires presentation of exogenous antigen.
Nature.
398
1999
77
80
24
Weekes
MP
Wills
MR
Mynard
K
Carmichael
AJ
Sissons
JG
The memory cytotoxic T-lymphocyte (CTL) response to human cytomegalovirus infection contains individual peptide-specific CTL clones that have undergone extensive expansion in vivo.
J Virol.
73
1999
2099
2108
25
Cwynarski
K
Ainsworth
J
Cobbold
M
et al
Direct visualization of cytomegalovirus-specific T-cell reconstitution after allogeneic stem cell transplantation.
Blood.
97
2001
1232
1240
26
Diamond
DJ
York
J
Sun
JY
Wright
CL
Forman
SJ
Development of a candidate HLA A*0201 restricted peptide-based vaccine against human cytomegalovirus infection.
Blood.
90
1997
1751
1767
27
Kern
F
Surel
IP
Faulhaber
N
et al
Target structures of the CD8(+)-T-cell response to human cytomegalovirus: the 72-kilodalton major immediate-early protein revisited.
J Virol.
73
1999
8179
8184
28
Norbury
CC
Chambers
BJ
Prescott
AR
Ljunggren
HG
Watts
C
Constitutive macropinocytosis allows TAP-dependent major histocompatibility complex class I presentation of exogenous soluble antigen by bone marrow-derived dendritic cells.
Eur J Immunol.
27
1997
280
288
29
Blake
N
Lee
S
Redchenko
I
Thomas
W
et al
Human CD8+ T cell responses to EBV EBNA1: HLA class I presentation of the (Gly-Ala)-containing protein requires exogenous processing.
Immunity.
7
1997
791
802
30
Gillespie
GM
Wills
MR
Appay
V
et al
Functional heterogeneity and high frequencies of cytomegalovirus-specific CD8(+) T lymphocytes in healthy seropositive donors.
J Virol.
74
2000
8140
8150
31
Farrell
HE
Vally
H
Lynch
DM
et al
Inhibition of natural killer cells by a cytomegalovirus MHC class I homologue in vivo [see comments].
Nature.
386
1997
510
514
32
Warren
AP
Ducroq
DH
Lehner
PJ
Borysiewicz
LK
Human cytomegalovirus-infected cells have unstable assembly of major histocompatibility complex class I complexes and are resistant to lysis by cytotoxic T lymphocytes.
J Virol.
68
1994
2822
2829
33
Miller
DM
Rahill
BM
Boss
JM
et al
Human cytomegalovirus inhibits major histocompatibility complex class II expression by disruption of the Jak/Stat pathway.
J Exp Med.
187
1998
675
683
34
Hebart
H
Muller
C
Loffler
J
Jahn
G
Einsele
H
Monitoring of CMV infection: a comparison of PCR from whole blood, plasma-PCR, pp65-antigenemia and virus culture in patients after bone marrow transplantation.
Bone Marrow Transplant.
17
1996
861
868
35
Boeckh
M
Gallez-Hawkins
GM
Myerson
D
Zaia
JA
Bowden
RA
Plasma polymerase chain reaction for cytomegalovirus DNA after allogeneic marrow transplantation: comparison with polymerase chain reaction using peripheral blood leukocytes, pp65 antigenemia, and viral culture.
Transplantation.
64
1997
108
113
36
Mixter
PF
Russell
JQ
Morrissette
GJ
Charland
C
Aleman-Hoey
D
Budd
RC
A model for the origin of TCR-αβ+ CD4−CD8− B220+ cells based on high affinity TCR signals.
J Immunol.
162
1999
5747
5756
37
Gallimore
A
Glithero
A
Godkin
A
et al
Induction and exhaustion of lymphocytic choriomeningitis virus-specific cytotoxic T lymphocytes visualized using soluble tetrameric major histocompatibility complex class I-peptide complexes.
J Exp Med.
187
1998
1383
1393
38
Chambers
CA
Sullivan
TJ
Allison
JP
Lymphoproliferation in CTLA-4-deficient mice is mediated by costimulation-dependent activation of CD4+ T cells.
Immunity.
7
1997
885
895
39
Boise
LH
Minn
AJ
Noel
PJ
et al
CD28 costimulation can promote T cell survival by enhancing the expression of Bcl-XL.
Immunity.
3
1995
87
98
40
Kirchhoff
S
Muller
WW
Li-Weber
M
Krammer
PH
Up-regulation of c-FLIPshort and reduction of activation-induced cell death in CD28-costimulated human T cells.
Eur J Immunol.
30
2000
2765
2774
41
Kirchhoff
S
Muller
WW
Krueger
A
Schmitz
I
Krammer
PH
TCR-mediated up-regulation of c-FLIPshort correlates with resistance toward CD95-mediated apoptosis by blocking death-inducing signaling complex activity.
J Immunol.
165
2000
6293
6300
42
Dietrich
PY
Caignard
A
Diu
A
et al
Analysis of T-cell receptor variability in transplanted patients with acute graft-versus-host disease.
Blood.
80
1992
2419
2424
43
Liu
X
Chesnokova
V
Forman
SJ
Diamond
DJ
Molecular analysis of T-cell receptor repertoire in bone marrow transplant recipients: evidence for oligoclonal T-cell expansion in graft-versus-host disease lesions.
Blood.
87
1996
3032
3044
44
Even
J
Lim
A
Puisieux
I
et al
T-cell repertoires in healthy and diseased human tissues analysed by T-cell receptor beta-chain CDR3 size determination: evidence for oligoclonal expansions in tumours and inflammatory diseases.
Res Immunol.
146
1995
65
80
45
Mason
D
A very high level of crossreactivity is an essential feature of the T-cell receptor [see comments].
Immunol Today.
19
1998
395
404
46
Goldrath
AW
Bevan
MJ
Selecting and maintaining a diverse T-cell repertoire.
Nature.
402
1999
255
262
47
Tallquist
MD
Yun
TJ
Pease
LR
A single T cell receptor recognizes structurally distinct MHC/peptide complexes with high specificity.
J Exp Med.
184
1996
1017
1026
48
Ignatowicz
L
Kappler
J
Marrack
P
The repertoire of T cells shaped by a single MHC/peptide ligand.
Cell.
84
1996
521
529
49
Chmielowski
B
Muranski
P
Ignatowicz
L
In the normal repertoire of CD4+ T cells, a single class II MHC/peptide complex positively selects TCRs with various antigen specificities.
J Immunol.
162
1999
95
105
50
Liu
CP
Parker
D
Kappler
J
Marrack
P
Selection of antigen-specific T cells by a single IEk peptide combination.
J Exp Med.
186
1997
1441
1450
51
Gapin
L
Fukui
Y
Kanellopoulos
J
et al
Quantitative analysis of the T cell repertoire selected by a single peptide-major histocompatibility complex.
J Exp Med.
187
1998
1871
1883
52
Moss
PA
Moots
RJ
Rosenberg
WM
et al
Extensive conservation of alpha and beta chains of the human T-cell antigen receptor recognizing HLA-A2 and influenza A matrix peptide.
Proc Natl Acad Sci U S A.
88
1991
8987
8990
53
Lehner
PJ
Wang
EC
Moss
PA
et al
Human HLA-A0201-restricted cytotoxic T lymphocyte recognition of influenza A is dominated by T cells bearing the V beta 17 gene segment.
J Exp Med.
181
1995
79
91
54
Argaet
VP
Schmidt
CW
Burrows
SR
et al
Dominant selection of an invariant T cell antigen receptor in response to persistent infection by Epstein-Barr virus.
J Exp Med.
180
1994
2335
2340
55
Utz
U
Banks
D
Jacobson
S
Biddison
WE
Analysis of the T-cell receptor repertoire of human T-cell leukemia virus type 1 (HTLV-1) Tax-specific CD8+ cytotoxic T lymphocytes from patients with HTLV-1-associated disease: evidence for oligoclonal expansion.
J Virol.
70
1996
843
851
56
Lim
A
Trautmann
L
Peyrat
MA
et al
Frequent contribution of T cell clonotypes with public TCR features to the chronic response against a dominant EBV-derived epitope: application to direct detection of their molecular imprint on the human peripheral T cell repertoire.
J Immunol.
165
2000
2001
2011
57
Yee
C
Savage
PA
Lee
PP
Davis
MM
Greenberg
PD
Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide-MHC tetramers.
J Immunol.
162
1999
2227
2234
58
Murali-Krishna
K
Altman
JD
Suresh
M
et al
Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection.
Immunity.
8
1998
177
187
59
Tan
LC
Gudgeon
N
Annels
NE
et al
A re-evaluation of the frequency of CD8+ T cells specific for EBV in healthy virus carriers.
J Immunol.
162
1999
1827
1835
60
Lee
PP
Yee
C
Savage
PA
et al
Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients.
Nat Med.
5
1999
677
685
61
Bieganowska
K
Hollsberg
P
Buckle
GJ
et al
Direct analysis of viral-specific CD8+ T cells with soluble HLA-A2/Tax11–19 tetramer complexes in patients with human T cell lymphotropic virus-associated myelopathy.
J Immunol.
162
1999
1765
1771
62
Kuroda
MJ
Schmitz
JE
Barouch
DH
et al
Analysis of Gag-specific cytotoxic T lymphocytes in simian immunodeficiency virus-infected rhesus monkeys by cell staining with a tetrameric major histocompatibility complex class I-peptide complex.
J Exp Med.
187
1998
1373
1381
63
Boitel
B
Blank
U
Mege
D
et al
Strong similarities in antigen fine specificity among DRB1* 1302-restricted tetanus toxin tt830–843-specific TCRs in spite of highly heterogeneous CDR3.
J Immunol.
154
1995
3245
3255

Author notes

Stephen Mackinnon, Dept of Haematology, University College Hospital, 98 Chenies Mews, London WC1E 6HX, United Kingdom; e-mail: s.mackinnon@ucl.ac.uk.

Sign in via your Institution