Dendritic cells (DCs) are professional antigen-presenting cells that are highly effective adjuvants for immunizing against pathogens and tumor antigens. The potential merit of genetic approaches to loading DCs with antigens is to express high and sustained levels of proteins that can be subsequently processed and presented to T lymphocytes. Replication-defective oncoretroviruses are able to efficiently transduce CD34+ progenitor-derived DCs but not monocyte-derived DCs. Here, it is shown that efficient gene transfer is obtained using a human immunodeficiency virus-1–derived lentiviral vector deleted of all structural and accessory genes. Infection of immature DCs with the lentiviral vector at a multiplicity of infection of 20 resulted in stable gene expression in 30% to 40% of the matured DCs. Proviral DNA was detectable by Alu polymerase chain reaction for the lentiviral but not the oncoretroviral vector. Most importantly, it is demonstrated that lentivirus-transduced DCs were fully functional and effectively activated autologous HLA A2.1+ peripheral blood cytotoxic T lymphocytes (CTLs). DCs expressing lentiviral vector-encoded Flu peptide were at least as efficient as DCs pulsed with the same peptide in stimulating specific CTLs. The efficacy of the lentivirus-transduced DCs was further demonstrated by their ability to directly activate freshly harvested peripheral blood Flu-specific CTLs in the absence of CD4+ T-cell help and exogenous cytokines. The availability of a stable gene delivery system based on a multiply attenuated lentivirus that does not encode any viral protein and that allows sustained antigen presentation by DCs derived from blood monocytes will be very useful for the biologic investigation of DCs and the improvement of immunotherapeutic strategies involving DCs.

Dendritic cells (DCs) are specialized antigen-presenting cells with a remarkable ability to stimulate naive T lymphocytes and generate memory T lymphocytes. Immature DCs effectively take up antigen and upon maturation up-regulate expression of a number of molecules critical for effective antigen presentation.1DCs are thus currently viewed as ideal adjuvants for immunizing against pathogens or tumor antigens.2,3 There are nonetheless a number of unanswered questions about how to best use DCs for these purposes. The source of DCs, their requirements for differentiation and activation, and approaches to load them with antigen are important biologic parameters that will determine the efficacy of DC-based immunization.

Genetic approaches to antigen loading, based on the transduction of DCs with complementary DNA or messenger RNA encoding tumor antigens, offer several advantages over pulsing with peptide or cellular extracts. A set of selected antigens can be expressed at high levels in the DCs; antigens can be processed into HLA-restricted peptides irrespective of the HLA type of each individual and without prior knowledge of the peptide best suited for that particular HLA; and antigen presentation should be more sustained if the antigen is expressed endogenously rather than pulsed. Stable gene transfer can be achieved with viral vectors that integrate in host cell chromosomes. We and others have successfully used oncoretroviral vectors based on murine leukemia viruses (MLVs) to genetically modify CD34+ DC progenitors from various sources (eg, bone marrow, cord blood, and cytokine-elicited peripheral blood).4-9 Because oncoretroviral vectors require cell division for efficient transduction,10-12 their efficacy is restricted to DC progenitor cells that proliferate before terminal differentiation. This approach is thus not applicable to DCs generated from peripheral blood mononuclear cells (PBMCs), a common and practical source of DCs, because they do not show any substantial proliferation.13-15 Lentiviruses, on the other hand, are able to stably integrate their provirus in certain nondividing cells.16,17 Wild-type human immunodeficiency virus (HIV)-1 in particular is capable of infecting blood-derived DCs in the absence of cell proliferation.13 Thus, lentivirus-mediated gene transfer could offer the unique opportunity to investigate the biologic activity of DCs differentiating from blood monocytes if efficient transduction can be accomplished without impairing DC biologic activity.

However, the safe use of recombinant lentivirus requires attenuated vectors containing a minimum of viral sequences. We have therefore investigated whether efficient gene transfer in human DCs derived from PBMCs could be obtained using an HIV-1–derived lentiviral vector deleted of gag, pol, env, tat, rev, nef,vif, vpu, and vpr. We compared the lentiviral vector to an MLV-based oncoretroviral vector, both pseudotyped with the vesicular stomatitis virus G protein (VSV-G) and used under the same conditions. The lentiviral vector mediated stable gene transduction of human monocyte-derived DCs, whereas the oncoretroviral vector did not. Most importantly, we show that transduction with the lentiviral vector did not impair DC function in terms of induction of allogeneic T-cell proliferation and of a peptide-specific cytolytic response. To further demonstrate that lentivirus-transduced DCs could efficiently present a defined antigen to autologous T lymphocytes, we compared DCs pulsed with the HLA A2.1-restricted Flu peptide to DCs transduced with a vector encoding the corresponding sequence. In this manner, the vector-encoded peptide did not require antigen processing to be presented and was not part of a larger protein that could encode additional T-cell epitopes including T-helper epitopes. Here we show that the lentivirus-transduced DCs stimulated CD8+ T cells thoroughly depleted of CD4+ T cells at least as efficiently as the peptide-pulsed DCs. Thus, the Flu-specific T-cell response was not dependent on CD4+ T-helper cells, demonstrating the potential of DCs transduced with a lentiviral vector to directly stimulate cyotoxic T lymphocytes.

Lentiviral vector construction and production

The lentiviral vector pHR'NTP was derived from pHR'CMVlacZ 18 by replacing theXbaI-XhoI fragment encoding the lacZ gene with the 0.8-kilobase (kb) XbaI-SalI fragment encoding the truncated human low-affinity nerve growth factor receptor, termed NTP.19 The vector HIG was constructed by inserting theBamHI-NcoI fragment containing the internal ribosomal entry site (IRES) of the encephalomyocarditis virus20 and the NcoI-XhoI fragment encoding the enhanced green fluorescence protein (eGFP; Clontech, Palo Alto, CA) into the BamHI and XhoI sites of pHR'CMVlacZ. The dicistronic vector HFIG derived from HIG by inserting the NcoI-EcoRI fragment containing the gene encoding the Flu peptide fused to the human CD8α leader sequence21 into the NcoI and EcoRI sites between the cytomegalovirus (CMV) promoter and the IRES element.

The lentiviral vector particles were produced by calcium phosphate transfection into 293T cells (American Type Culture Collection [ATCC], Rockville, MD) as previously described.22,23Briefly, 1 × 107 293T cells were seeded in 15-cm diameter dishes 24 hours prior to transfection in Dulbecco's modified Eagle's medium (DMEM; Mediatech, Herndon, VA) with 10% heat-inactivated fetal calf serum (FCS; Hyclone, Logan, UT), penicillin (100 IU/mL), and streptomycin (100 μg/mL) in a 5% CO2incubator. A total of 58 μg plasmid DNA was used for the transfection of one dish: 12 μg pMD.G,24 24 μg packaging plasmid pCMVΔR8.925 and 22 μg vector plasmid. The precipitate was formed by adding the plasmids to a final volume of 630 μL of 1 mM Tris and 630 μL of 0.5 M CaCl2, mixing well. The DNA mix was then added dropwise to 630 μL of 2 × HEPES-buffered saline (281 mM NaCl, 100 mM HEPES, 1.5 mM Na2HPO4 [pH 7.07-7.12]) while vortexing. The precipitate was immediately added to the medium with gently swirling. The medium was replaced after 8 to 16 hours with fresh DMEM (20 mL) lacking FCS. The viral supernatants were collected 24 hours later, centrifuged for 10 minutes at 1200 rpm, and filtered through 0.45 μm low-protein binding filters (Millipore, Bedford, MA). The viral supernatants were concentrated 100 × by ultracentrifugation (50 000g, 1.5 hours, 4°C) as previously described,19 and the viral pellets were resuspended in DMEM or TNE (50 mM Tris, pH7.8; 130 mM NaCl; 1 mM ethylenediaminetetraacetic acid). The titer of the concentrated viral vectors was determined by infecting 1 × 105 HeLa cells in 6-well plates with serial dilutions of each vector in the presence of polybrene (Sigma, St. Louis, MO) at 8 μg/mL in a volume of 2 mL. Efficiency of transduction was determined by flow cytometry analysis (FACS) as previously described.19 

Oncoretroviral vector construction and production

The oncoretroviral vector myeloproliferative sarcoma virus (MPSV)-NTP was generated by subcloning the NTP gene into an MFG-derived vector that contains a chimeric MLV/MPSV long terminal repeat (LTR).26,27 A high-titer producer clone was generated by transfection of gpg29 packaging cells24 with the MPSV-NTP vector DNA as described.28 

Generation of dendritic cells

Peripheral blood was obtained from normal HLA A2.1+donors in heparinized tubes. HLA typing was performed by polymerase chain reaction (PCR) in the HLA laboratory at Memorial Sloan-Kettering Cancer Center (New York, NY). PBMCs were isolated by density gradient centrifugation on lymphocyte separation medium (Accurate Chemical & Scientific, Westbury, NY). DCs were generated as described.29,30 Briefly, the T-cell–depleted (ER) population was prepared by rosetting with sheep red blood cells (Colorado Serum Company, Denver, CO) as described.14 A total of 2 × 106ER cells per well were plated in 6-well plates. Granulocyte-macrophage colony-stimulating factor (GM-CSF; Immunex, Seattle, WA) and interleukin (IL)-4 (R & D Systems, Minneapolis, MN) were added at 1000 U/mL every second day for 7 days. Monocyte-conditioned medium was prepared by adding 50 × 106 ER cells on Petri dishes coated with human gamma globulins (Sigma) at 10 mg/mL. Nonadherent cells were removed and the monocyte-conditioned medium, which was collected after 24 hours, was added (one half or one third of the final volume) to the cells for 4 days to get fully mature DCs.

T-cell purification and CD4 depletion of purified T cells

T cells were purified as described.21,31 Briefly, the T-cell–enriched (ER+) population was collected from the same donors. After lysis of the rosetted sheep red blood cells and 3 washes in phosphate-buffered saline with 2% FCS, B cells, natural killer cells, monocytes-macrophages, and activated T cells were depleted. This was accomplished by incubating cells with mouse immunoglobulin G (IgG) monoclonal antibodies directed against CD11b, CD16, and HLA-DP, -DQ, -DR (Pharmingen, San Diego, CA) at 1 μg per 1 × 106 cells for 30 minutes, followed by a panning on Petri dishes coated with goat antimouse IgG (Caltag, Burlingame, CA) as described.32 After 3 washes in phosphate-buffered saline with 2% FCS, the T cells were resuspended at a final concentration of 1 × 107 cells/mL. These resulting T cells were more than 99% pure (data not shown). For the experiments with CD4-depleted T cells, purified T cells were incubated with mouse IgG1 monoclonal antibody directed against CD4 (Leu-3a; Monoclonal Antibody Core Facility at Memorial Sloan-Kettering Cancer Center) at 1 μg per 1 × 106 cells for 30 minutes. Panning on Petri dishes was performed as described.32 CD4 depletion, after 3 or 4 rounds of panning, was more than 99%.

Culture conditions

DCs and T lymphocytes were maintained in RPMI 1640 (Mediatech) with 10% FCS. Penicillin at 100 U/mL and streptomycin at 100 μg/mL were added to all the cultures.

Viral transduction of dendritic cells

DCs were infected at a multiplicity of infection (MOI) of 2 to 20 in 6-well plates with 1 × 106 to 2 × 106 DCs per well in a volume of 2 mL per well. The cells were infected for 8 to 16 hours at 37°C in the presence of 4 μg/mL polybrene. GM-CSF and IL-4 were added at 1000 U/mL. After infection the DCs were washed and resuspended in fresh RPMI with 10% FCS and cytokines. Four days after infection, genomic DNA was extracted for Southern blot analysis.28 

Alu PCR

Genomic DNA was extracted from the HeLa cells or DCs 4 days after exposure to the viral vectors pHR'NTP and MPSV-NTP. Junctions between viral and genomic sequences were amplifed by using oligonucleotides specific for the conserved sequences of the human Alu element (3′Alu: 5′-TGAGCCGAGATCGCGCCACTGCAC-3′) and for the vector-encoded NTP reporter gene (NTP3: 5′-TCCCTGGCCGTTGGATTACACGGTC-3′). The PCRs were carried out with 300 ng DNA in a volume of 50 μL. The cycling conditions were 94°C for 5 minutes, followed by 30 cycles of 94°C for 30 seconds, 66°C for 30 seconds, and 72° C for 5 minutes, with 10 minutes at 72°C for the final extension step. The PCR products were analyzed on 0.8% agarose gels, and Southern blot hybridization was performed using theXhoI-BamHI fragment of the NTP gene (448 bp) as a probe.

Flow cytometry analysis

The cell surface phenotype of DCs was analyzed using phycoerythrin (PE)- or fluorescein isothiocyanate (FITC)–labeled monocloal antibodies against CD14, CD80, CD40, HLA-DR (Becton Dickinson, Mountain View, CA), and a monoclonal antibody against CD83 (Immunex, Marseille, France). To measure gene transfer, cells were stained with the anti-LNGFR antibody 20.4 (ATCC). Anti-CD83 antibody and 20.4 were detected by a phycoerythrin-labeled polyclonal goat antimouse antibody (Caltag, South San Francisco, CA).

Allogeneic and syngeneic mixed leukocyte reactions

Stimulatory cells (untransduced DCs, transduced DCs, or total PBMCs) were irradiated (1500 rad) and plated in triplicates in round-bottomed 96-well tissue culture plates at graded concentrations, each well containing 1 × 105 allogeneic or syngeneic T cells, in a total volume of 200 μL regular medium. PBMCs, T cells, and DCs were obtained as described. T cells were thoroughly depleted of B cells, natural killer cells, monocytes-macrophages, and preactivated T cells as described. Five or six days after the start of the cocultures, 37 × 102 Bq (1 μCi) of3H-thymidine (3H-TdR; New England Nuclear, Boston, MA) was added to each well for 12 hours. Incorporation of3H-TdR is given as the mean cpm ± SD of triplicates. Wells containing only stimulators or T cells always incorporated less than 100 cpm 3H-TdR.

Stimulation of specific cytotoxic T lymphocytes

Untransduced and HIG-transduced DCs were pulsed with the Flu peptide (10 μM) for 2 hours at room temperature in RPMI without serum. In vitro stimulation of the T cells was carried out at the ratio 10 T lymphocytes to 1 DC in 24-well plates, with 1 × 106T cells per well for 8 to 10 days in RPMI with 10% FCS. Ten days later the T cells were restimulated by adding DCs following the same procedure. Then, 20 IU/mL IL-2 (Chiron, St. Louis, MO) was added every third day.

Cytotoxicity assays

Standard chromium release assays were performed. As target cells, we used TAP-deficient HLA A2.1+ T2 cells (kind gift of Dr J. W. Young) loaded with the Flu or the MART-1 peptide (10 μM, 1 hour at room temperature, in RPMI without serum) before pulsing with 51Cr for 1 hour at 37°C. A total of 5000 T2 cells per well were used in 96 V-bottom plates at different effector-to-target cell (E:T) ratios for 4 hours. Specific51Cr release was calculated using the formula [(51Cr release − spontaneous release)/(maximum release − spontaneous release)] × 100.

Peptide synthesis

The peptides were synthesized in the Peptide Synthesis Facility at Memorial Sloan-Kettering Cancer Center, resuspended in 50% (vol:vol) RPMI/dimethylsulfoxide (Sigma), and stored at −20°C. The following peptides were used in this study: the influenza matrix protein-derived peptide58-66 GILGFVFTL (Flu peptide); and the MART-1 protein-derived peptide27-35 AAGIGILTV (MART-1 peptide).33 

Recombinant lentiviruses efficiently transduce blood monocyte-derived DCs

To compare lentivirus- and MLV-mediated gene transfer in human blood-derived DCs, cultured DCs were transduced under identical conditions using 2 vectors encoding an inactive cell surface marker termed NTP.20 NTP expression is easily and accurately monitored by FACS analysis, allowing enumeration of DC transduction on a single-cell basis. The HIV- and MLV-based vectors, respectively termed MPSV-NTP and pHR'NTP (Figure 1A), were pseudotyped with the VSV-G envelope providing both vectors with the same mode of cell entry via receptor-mediated endocytosis. Viral titers of both vectors were measured on HeLa cells, and viral stocks were set at a concentration of 3 × 107 IU/mL. DCs were generated by differentiation from blood monocytes with GM-CSF and IL-4. On day 8, monocyte-conditioned medium was added to the cultures to induce terminal maturation (Figure 2A). The DCs were infected on either day 3, 7 (immature DCs), or 12 (mature DCs). The MPSV-NTP or the pHR'NTP particles were admixed at an MOI ranging from 2 to 20. Infection with the lentiviral vector at MOI 20 on day 3 led to NTP expression in 35% of the DCs as shown in one representative experiment in Figure 2B. The highest levels of NTP expression were achieved at an MOI of 20 on day 3 or day 7 of the culture, when the DCs are immature (18%-35% NTP+, data not shown). At lower MOIs (2-4), 15% to 17% of the HLA-DR++ population was NTP+ (data not shown). In contrast, susceptibility of the DCs to lentiviral vector transduction was reduced considerably, although not abolished, when infection was carried out after the DCs have matured (5%-10% NTP+ at MOI 20 on day 12, data not shown), corroborating observations by others using a less attenuated lentiviral vector.34 The MLV vector, on the other hand, did not lead to significant transduction efficiencies (0%-3% NTP+) at any time during DC differentiation (Figure 2B and data not shown). Infection of DCs at MOI of 10 to 20 was toxic, reducing cell yields by 50% or more after several days (data not shown). In the functional studies, DCs were therefore infected at MOI 2 to 4 on day 7 to obtain undiminished yields of mature cells.

Fig. 1.

Schematic representation of the viral vectors.

(A) The lentiviral vector pHR'NTP expresses NTP under the transcriptional control of the CMV promoter and/or the HIV LTR. The MLV-vector, MPSV-NTP, contains a chimeric MPSV/MLV-LTR in which the U3 region of MLV was replaced by the U3 region of the myeloproliferative sarcoma.27 (B) In the bicistronic vector HFIG, the Flu peptide and the eGFP reporter gene are linked by the IRES of the encephalomyocarditis virus. In the vector HIG, the Flu peptide has been deleted. Ψ indicates packaging signal of HIV-1; Ψ+, extended packaging signal of MLV; Δgag, truncated gag gene containing an out-of-frame mutation; RRE, Rev-responsive element.

Fig. 1.

Schematic representation of the viral vectors.

(A) The lentiviral vector pHR'NTP expresses NTP under the transcriptional control of the CMV promoter and/or the HIV LTR. The MLV-vector, MPSV-NTP, contains a chimeric MPSV/MLV-LTR in which the U3 region of MLV was replaced by the U3 region of the myeloproliferative sarcoma.27 (B) In the bicistronic vector HFIG, the Flu peptide and the eGFP reporter gene are linked by the IRES of the encephalomyocarditis virus. In the vector HIG, the Flu peptide has been deleted. Ψ indicates packaging signal of HIV-1; Ψ+, extended packaging signal of MLV; Δgag, truncated gag gene containing an out-of-frame mutation; RRE, Rev-responsive element.

Close modal
Fig. 2.

Maturation and transduction of monocyte-derived DCs.

(A) On day 18 the cells had the typical phenotype of mature DCs with high expression levels of HLA-DR, expression of CD80 and CD83, but loss of expression of the monocyte-macrophage marker CD14. (B) Transduction efficiency of DCs infected on day 3 with the oncoretroviral vector MPSV-NTP or the lentiviral vector pHR'NTP at an MOI of 20. On day 18 (15 days after infection) the cells were double stained with anti–HLA-DR and anti-LNGFR monoclonal (20.4) antibodies. NTP expression: y-axis; HLA-DR: x-axis. Data from 1 representative experiment of 3.

Fig. 2.

Maturation and transduction of monocyte-derived DCs.

(A) On day 18 the cells had the typical phenotype of mature DCs with high expression levels of HLA-DR, expression of CD80 and CD83, but loss of expression of the monocyte-macrophage marker CD14. (B) Transduction efficiency of DCs infected on day 3 with the oncoretroviral vector MPSV-NTP or the lentiviral vector pHR'NTP at an MOI of 20. On day 18 (15 days after infection) the cells were double stained with anti–HLA-DR and anti-LNGFR monoclonal (20.4) antibodies. NTP expression: y-axis; HLA-DR: x-axis. Data from 1 representative experiment of 3.

Close modal

Stability of transgene expression in DCs

Expression of lentiviral vector-encoded NTP was maintained for at least 2 weeks, suggesting that intact reverse transcription of the viral RNA into DNA, transport of the preintegration complex into the nucleus, and proviral integration had occurred. To address whether viral DNA was present after viral transduction, Southern blot analysis was performed on genomic DNA from DCs infected with either pHR'NTP or MPSV-NTP. Three days after transduction, infection with the pHR'NTP vector system resulted in a detectable band of the predicted size (3.6 kb) representing integrated or episomal vector sequence (Figure3A). The predicted 1.8-kb band corresponding to the MPSV-NTP vector DNA was not detectable even after prolonged exposures, suggesting that the low level of NTP+cells measured by FACS analysis (0%-3%) corresponds to either very low transduction efficiency or pseudotransduction.19 We used Alu PCR to resolve whether transduction of DCs with the lentiviral vector leads to integration of the provirus.35,36Alu-repeats appear on average every 4 kb in the human genome and therefore allow PCR amplification of viral sequences integrated at random sites within the genome. Two primers were designed—one specific for the 3′ end of the consensus Alu sequence, the other specific for the NTP reporter gene of the viral vectors. Alu PCR carried out on genomic DNA from untranduced (U) and transduced (H, M) HeLa cells showed that both viral vectors gave the characteristic smear of NTP+ bands representing multiple integration sites (Figure3B,C). Alu PCR performed on genomic DNA from DCs resulted in an NTP+ smear only in case of transduction with the vector pHR'NTP (H), demonstrating that the lentiviral vector was capable of proviral integration in DCs in absence of proliferation whereas the MLV-based vector (M) was not (Figure 3B,C). The intensity of the signal detected by Southern blot hybridization with the NTP probe was proportional to the fraction of NTP+ cells detected by flow cytometry (HeLa, pHR'NTP: 50%; HeLa, MPSV-NTP: 5%; DC, pHR'NTP: 10%; data not shown).

Fig. 3.

Gene transfer in human monocyte-derived DCs with MPSV-NTP or pHR'NTP.

(A) Detection of vector sequence in DCs by Southern blot analysis. DCs were infected on day 4 at an MOI of 4 with the oncoretroviral vector MPSV-NTP (MPSV) or the lentiviral vector pHR'NTP (pHR'). Four days after infection genomic DNA was extracted and digested with eitherNheI (MPSV-NTP) or ScaI (pHR'NTP), which cut in the LTR. Southern blot analysis was performed using a radiolabeled NTP complementary DNA probe. The left 3 lanes represent copy controls obtained from MEL clones bearing 0, 1, or 2 copies of the MLV vector SFG-NTP digested with NheI. Position of the signals expected for the vector DNAs are indicated (copy control = 2.8 kb; MPSV = 1.8 kb; pHR' = 3.6 kb). EB indicates endogenous bands; −, untransduced; +, transduced. No band was observed in the MPSV+ lane, even after extended exposure (24 hours in Figure 3; 10 days for extended exposure). (B,C) Detection of integrated lentiviral vector DNA by Alu PCR. HeLa cells and DCs were either untreated (U) or infected with the HIV-based vector pHR'NTP (H) or the MLV-based vector MPSV-NTP (M). Proviral junctions were amplified using the primers 3′Alu and NTP3. PCR products were analyzed by gel electrophoresis (B) and Southern blot hybridization with an NTP-specific probe (C).

Fig. 3.

Gene transfer in human monocyte-derived DCs with MPSV-NTP or pHR'NTP.

(A) Detection of vector sequence in DCs by Southern blot analysis. DCs were infected on day 4 at an MOI of 4 with the oncoretroviral vector MPSV-NTP (MPSV) or the lentiviral vector pHR'NTP (pHR'). Four days after infection genomic DNA was extracted and digested with eitherNheI (MPSV-NTP) or ScaI (pHR'NTP), which cut in the LTR. Southern blot analysis was performed using a radiolabeled NTP complementary DNA probe. The left 3 lanes represent copy controls obtained from MEL clones bearing 0, 1, or 2 copies of the MLV vector SFG-NTP digested with NheI. Position of the signals expected for the vector DNAs are indicated (copy control = 2.8 kb; MPSV = 1.8 kb; pHR' = 3.6 kb). EB indicates endogenous bands; −, untransduced; +, transduced. No band was observed in the MPSV+ lane, even after extended exposure (24 hours in Figure 3; 10 days for extended exposure). (B,C) Detection of integrated lentiviral vector DNA by Alu PCR. HeLa cells and DCs were either untreated (U) or infected with the HIV-based vector pHR'NTP (H) or the MLV-based vector MPSV-NTP (M). Proviral junctions were amplified using the primers 3′Alu and NTP3. PCR products were analyzed by gel electrophoresis (B) and Southern blot hybridization with an NTP-specific probe (C).

Close modal

Dendritic cells transduced with a multiply attenuated lentiviral vector retain normal allostimulatory functions and efficiently stimulate Flu-specific primary cytotoxic T cells

The function of lentivirus-transduced DCs was assessed in 2 ways: induction of an allogeneic proliferative T-cell response and stimulation of an antigen-specific autologous cytotoxic T-cell response. In the mixed leukocyte reactions, DCs were 100- to 300-fold more potent than DCs in inducing proliferation of allogeneic T cells (Figure 4), demonstrating the high allostimulatory potential expected from fully mature DCs.9Transduction of DCs neither reduced nor augmented the proliferative response of the same T cells when compared with untransduced DCs (Figure 4).

Fig. 4.

Transduction of DCs with multiply attenuated lentiviral vector does not alter the allostimulatory capacity of these DCs.

Untransduced DCs (DC.utr) or transduced DCs (in this case with the HIG vector, DC.HIG) were cultured in graded doses with allogeneic (allo) or syngeneic (syn) T cells. Incorporation of 3H-TdR was measured after 6 days of coculture. PBMCs were used as a control to assess DC potency. X-axis: number of stimulating cells per 105 T cells (logarithmic scale); y-axis: 3H-TdR incorporation by T cells (logarithmic scale).

Fig. 4.

Transduction of DCs with multiply attenuated lentiviral vector does not alter the allostimulatory capacity of these DCs.

Untransduced DCs (DC.utr) or transduced DCs (in this case with the HIG vector, DC.HIG) were cultured in graded doses with allogeneic (allo) or syngeneic (syn) T cells. Incorporation of 3H-TdR was measured after 6 days of coculture. PBMCs were used as a control to assess DC potency. X-axis: number of stimulating cells per 105 T cells (logarithmic scale); y-axis: 3H-TdR incorporation by T cells (logarithmic scale).

Close modal

Transduced DCs and peptide-pulsed DCs were then compared in their ability to recall a specific CTL response using autologous peripheral blood T cells as the responder population. To avoid the use of a precursor protein that would require processing and eventually introduce additional epitopes including T-helper epitopes into the DC, we turned to an established model system based on the peptide58-66 GILGFVFTL derived from the influenza matrix protein, termed Flu peptide. The peptide is presented in the context of the A2.1 major histocompatibility complex (MHC) class I molecule and has previously been shown to stimulate T cells efficiently.37,38 It is well established that HLA A2.1-restricted Flu peptide–specific CTLs are capable to lyse with the same efficiency both Flu peptide–pulsed and influenza virus–infected lymphoid or monocytic target cells.39-44 Therefore, the use of HLA-A2.1+ T2 cells pulsed with the Flu peptide as target cells provides a convenient and reliable system to study a biologically relevant HLA-A2.1+-restricted CD8+response.

To generate the lentiviral vector HFIG, the Flu peptide coding sequence was fused to that of the CD8α leader to target the endoplasmic reticulum21,45 and cloned in place of the NTP complementary DNA in pHR'NTP (Figure 1B). The HIG vector lacks the Flu peptide and served as a negative control. Immature DCs were infected on day 7 at an MOI of 2 to 4 with the lentiviral vectors HFIG or HIG. Expression of the reporter gene eGFP was evaluated on day 11 and showed that the DCs were transduced with an efficiency of 15% to 18%, consistent with the efficiency of the NTP gene transfer achieved with pHR'NTP (data not shown). DCs transduced with the HFIG vector were tested for their ablility to present the Flu peptide in the context of HLA A2.1 and induce a specific cytotoxic T-cell response. T-cell stimulation was achieved by incubating mature transduced or peptide-pulsed DCs with autologous T cells at a 1:10 stimulator-to-responder ratio. Untransduced DCs, either pulsed (DC+) or nonpulsed (DC) with the Flu peptide, served as controls. The HFIG-transduced DCs (DC.HFIG) were able to activate a strong cytotoxic T-cell response against the Flu peptide, albeit with slightly lower efficiency than the peptide-pulsed, untransduced DCs (DC+) (Figure5). The high potential of the genetically modified DCs is underlined by the fact that only a fraction of the DCs was transduced by the HFIG vector (about 15%-17% of the DCs in these experiments). DCs transduced with the control vector HIG pulsed with the Flu peptide (DC.HIG+) were comparable to untransduced DCs in their ability to stimulate Flu-specific T cells, indicating that lentivirus-mediated transduction did not perturb DC function.

Fig. 5.

Flu-specific CTL activation by DCs transduced with the lentiviral vector HFIG.

DCs were infected with HIG or HFIG or left uninfected. The ability of HFIG-transduced DCs to activate Flu-specific CTLs (DC.HFIG) was compared with untransduced or HIG-transduced DCs either nonpulsed (DC, DC.HIG) or pulsed (DC+, DC.HIG+) with the Flu peptide. CTL assays were performed after incubation of the DCs with autologous T cells for 8 to 10 days. T2 cells pulsed with the Flu peptide (T2+) or with the irrelevant MART-1 peptide (T2) were used as targets. Data are shown for 1 of 3 independent experiments.

Fig. 5.

Flu-specific CTL activation by DCs transduced with the lentiviral vector HFIG.

DCs were infected with HIG or HFIG or left uninfected. The ability of HFIG-transduced DCs to activate Flu-specific CTLs (DC.HFIG) was compared with untransduced or HIG-transduced DCs either nonpulsed (DC, DC.HIG) or pulsed (DC+, DC.HIG+) with the Flu peptide. CTL assays were performed after incubation of the DCs with autologous T cells for 8 to 10 days. T2 cells pulsed with the Flu peptide (T2+) or with the irrelevant MART-1 peptide (T2) were used as targets. Data are shown for 1 of 3 independent experiments.

Close modal

Lentivirus-transduced DCs effectively restimulate primed CTLs

To increase the specific response against the Flu peptide, the T cells were restimulated for another 8 days with newly generated DCs from the same donor. As expected, the restimulation with either transduced or peptide-pulsed DCs led to a strong increase of the Flu-specific response (Figure 6). The primary induction resulted in 75% to 80% Flu-specific cytotoxicity at E:T ratios of 20:1, whereas the restimulation of the T cells led to generation of CTLs with at least 4-fold higher activity (75%-100% cytotoxicity at the 5:1 E:T ratio). Thus, lentivirus-transduced DCs were able to efficiently restimulate the cytotoxic lymphocytes without loss of antigen specificity.

Fig. 6.

Restimulation of Flu-specific CTLs with monocyte-derived DCs.

Ten days after primary induction the T cells in each group (as for Figure 5) were restimulated with newly generated untransduced DCs (DC, DC+), HIG (DC.HIG, DC.HIG+), or HFIG (DC.HFIG)-transduced DCs.

Fig. 6.

Restimulation of Flu-specific CTLs with monocyte-derived DCs.

Ten days after primary induction the T cells in each group (as for Figure 5) were restimulated with newly generated untransduced DCs (DC, DC+), HIG (DC.HIG, DC.HIG+), or HFIG (DC.HFIG)-transduced DCs.

Close modal

Lentivirus-transduced DCs do not require CD4+ T cell help to stimulate Flu-specific CTLs

To further assess the potential of DCs transduced with the lentiviral vector, we investigated whether the stimulation of Flu-specific CTLs in vitro required CD4+ T-cell help. T cells were therefore depleted of CD4+ T cells to high purity (< 0.2% CD4+, Figure7A,B) and stimulated with DCs as described above, in the absence of any exogenous cytokine. DCs transduced with the HFIG vector (DC.HFIG) were compared with Flu peptide–pulsed untransduced (DC+) and HIG-transduced DCs (DC.HIG+). As shown in Figure 7C, DCs+ were able to stimulate Flu-specific T cells in the absence of CD4+ T-cell help and without addition of any exogenous cytokine. Interestingly, DC.HIG+ stimulated specific CTLs the same way as untransduced DCs. With DC.HFIG, the specific response was comparable or even stronger (3- to 5-fold higher in Figure 7C, right panel). These results obtained with CD4+-depleted peripheral blood T cells further demonstrate that lentiviral transduction does not alter DC function and enables DCs to efficiently present a specific MHC class I peptide to cytotoxic T cells.

Fig. 7.

Transduced DCs induce a specific cytotoxic response against the Flu peptide in the absence of CD4+ T cells and in the absence of exogenous cytokines.

(A,B) Purified HLA A2.1+ T cells before and after depletion of CD4+ T cells. (A) T cells (gate R, left panel) were purified as described in “Materials and methods” and stained with anti-CD3 and anti-CD19 antibodies (middle panel) or anti-CD8 and anti-CD4 antibodies (right panel). (B) Depletion of CD4+ T cells was accomplished as described in “Materials and methods.” After depletion, T cells (gate R, left panel) were stained with the same antibodies (middle and right panels). Background staining given by mouse isotype IgG controls was always lower than 0.25% and subtracted. (C) HLA A2.1+ DCs were cultured with autologous CD4-depleted T cells for 8 to 10 days in the absence of exogenous cytokines. CTL assays were performed on T2 cells as described in Figures 5 and 6. The following populations of DCs were compared: DCs pulsed with the Flu peptide (DC+), DCs transduced with the HIG vector and pulsed with the Flu peptide (DC.HIG+), and DCs transduced with the HFIG vector (DC.HFIG). Without CD4 depletion and using the same absolute number of T cells at the start of the cocultures, cytotoxic activity was comparable to that shown in Figure 5(data not shown).

Fig. 7.

Transduced DCs induce a specific cytotoxic response against the Flu peptide in the absence of CD4+ T cells and in the absence of exogenous cytokines.

(A,B) Purified HLA A2.1+ T cells before and after depletion of CD4+ T cells. (A) T cells (gate R, left panel) were purified as described in “Materials and methods” and stained with anti-CD3 and anti-CD19 antibodies (middle panel) or anti-CD8 and anti-CD4 antibodies (right panel). (B) Depletion of CD4+ T cells was accomplished as described in “Materials and methods.” After depletion, T cells (gate R, left panel) were stained with the same antibodies (middle and right panels). Background staining given by mouse isotype IgG controls was always lower than 0.25% and subtracted. (C) HLA A2.1+ DCs were cultured with autologous CD4-depleted T cells for 8 to 10 days in the absence of exogenous cytokines. CTL assays were performed on T2 cells as described in Figures 5 and 6. The following populations of DCs were compared: DCs pulsed with the Flu peptide (DC+), DCs transduced with the HIG vector and pulsed with the Flu peptide (DC.HIG+), and DCs transduced with the HFIG vector (DC.HFIG). Without CD4 depletion and using the same absolute number of T cells at the start of the cocultures, cytotoxic activity was comparable to that shown in Figure 5(data not shown).

Close modal

A variety of strategies to generate and transduce DCs are currently under investigation. Oncoretroviral vectors derived from MLV have been previously used for stable gene delivery into CD34+ DC progenitors purified from bone marrow, cord blood, or cytokine-elicited peripheral blood stem cells.4-7,9This approach is not applicable to DCs generated from blood monocytes because these cells show little or no cell proliferation, thus precluding oncoretoviral integration as shown here. Lentiviruses, on the other hand, have proven to be extremely efficient at infecting various nondividing cell types.16,47 Recombinant vectors derived from HIV-1 have successfully been used for in vivo or in vitro transduction of terminally differentiated cells such as neurons, hepatocytes, retinocytes, and macrophages.18,25,47-51 

To improve the safety of lentiviral-mediated gene transfer, 1 or more of the 4 accessory genes, vpr, vpu,vif, and nef, which contribute to viral pathogenicity, have been removed in the second generation of lentiviral vectors.25,52 Here, we show that a multiply attenuated vector system based on HIV-1 is efficient in transducing DCs differentiating from blood monocyte precursors. The presence of integrated proviral DNA and detectable expression of the reporter gene up to 2 weeks postinfection demonstrate that gene transfer into DCs is stable. The 4 accessory genes, vpr, vpu,vif and nef, have been removed and are therefore not necessary for stable gene transfer into human DCs. In certain cell types, however, absence of accessory functions can diminish infectivity of wild-type HIV-1 and replication-defective recombinants. Studies on HIV-1 infection of macrophages show that the Vpr protein plays a crucial role in the nuclear import of the preintegration complex.25,53,54 For lentiviral vector-mediated gene delivery into neurons and growth arrested cell lines, Vpr turns out to be completely dispensable, whereas gene delivery into macrophages and hepatocytes is considerably impaired when vpr is deleted.25,49 Interestingly, transduction efficiency with the lentiviral vector was higher in immature DCs than later in the differentiation process. Down-regulation of a viral receptor as a cause for reduced cellular entry in more mature cells is unlikely because infection was mediated by the VSV-G glycoprotein substituted for the HIV-1 envelope. This finding corroborates observations on the infection by wild-type HIV-1 and is consistent with a block in reverse transcription in mature DCs.13 On the other hand, the failure of the MLV-derived vector to efficiently transduce DCs at any time during the culture likely reflects another limitation. Indeed, the oncoretrovirus, which was VSV-G–pseudotyped and admixed at the same MOI as the lentivirus, likely underwent reverse transcription, as did the lentivirus, but then failed to integrate, perhaps because of its inability to translocate to the nucleus.10 

Most importantly, we found that lentivirus-transduced DCs were fully functional and could be used to stimulate specific cytotoxic T cells. First, genetic modification of DCs with a lentiviral vector did not alter the function of mature DCs. Indeed, untransduced and transduced DCs showed comparable stimulatory capacities in the allogeneic mixed leukocyte reaction. Furthermore, DCs transduced with a control vector (HIG) stimulated Flu-specific CTLs with an equal efficiency as untransduced DCs when pulsed with the relevant peptide under the same conditions. Stimulation of Flu-specific CTLs achieved in the absence of CD4+ T-cell help was comparable with either untransduced or control vector-transduced peptide-pulsed DCs. Second, DCs modified with the vector encoding the Flu peptide induced a strong specific T-cell response. Importantly, in vitro stimulation with CD8+ T cells thoroughly depleted of CD4+ T cells showed that Flu-specific CD8+ T cells were also activated in the absence of CD4+ helper T cells and without exogenous cytokine. This observation is consistent with direct activation of CTLs by fully mature DCs of human31,32,55 and mouse56-59 origin. Taken together, our data demonstrate that the efficiency of lentivirus-transduced DCs in stimulating specific CTLs in vitro does not depend on CD4+ T-cell help and that CD4-independent CTL activation did not result from lentivirus activation of DCs.60,61 In the absence of CD4+cells, however, the cytolytic activity was consistently reduced (2- to 4-fold), underscoring the role of CD4+ T cells in sustaining CTL activation under such experimental conditions. Importantly, although CD4+ T cells are dispensable for the activation of CTLs in vitro, T-cell help may be essential to sustain CTL activity and induce immunologic memory against tumor antigens in vivo.62 

The level of Flu-specific cytolytic activity induced by transduced DCs was at least as good as that obtained with pulsed DCs. This demonstrates the highly efficient presentation of the lentivirus-encoded MHC class I–restricted peptide by the transduced DCs. Importantly, a second round of stimulation with transduced DCs further increased Flu-specific activity. Interestingly, background cytotoxicity was consistently higher in the cultures containing CD4+ T cells. Thus, induction of this background activity appears to depend on the recruitment of CD4+ T cells, implying presentation of MHC class II peptides possibly provided by media62,63 or virion components.

A number of clinical trials currently use monocyte-derived DCs as a convenient way to generate DC-based “vaccines” for cancer immunotherapy.3 Antigen presentation is usually achieved by pulsing the DCs with peptide, whole protein, or apoptopic cells. Oncoretroviral vectors can be used to transduce proliferating CD34+ progenitor-derived DCs9 but not monocyte-derived DCs as shown here. Other vector systems have been used to transduce blood-derived DCs, including those based on adenovirus,64 herpesvirus,65 or poxvirus.66-68 One distinguishing feature of recombinant lentiviruses is that efficient and stable gene delivery is achieved with a vector that does not itself encode any immunogenic proteins of viral origin. The efficient generation of immunocompetent lentivirus-transduced DCs derived from blood cells provides a valuable alternative to approaches based on oncoretroviral-mediated gene transfer in CD34+ progenitors. This will allow us to compare genetically modified monocyte- and CD34+-derived DCs and to investigate functionally distinct subsets of DCs.2,69-71 The availability of a stable gene delivery system based on a multiply attenuated lentivirus that does not encode any viral protein and allows efficient and sustained antigen presentation by DCs derived from blood monocytes may ultimately be useful for the improvement of immunotherapies.

We thank Dr D. Trono for providing us with the plasmids PHR'CMVLacZ and pCMVΔR8.9 and C. May for providing us with the pHR'NTP plasmid. We thank Drs A. Krause, M. C. Gong, and I. Rivière for reviewing the manuscript.

Supported by National Institutes of Health grants PO1 CA-59350, RO1 HL-56712, PO1 CA-65930, and the Memorial Sloan-Kettering Cancer Center DeWitt-Wallace Fund.

J.D. and J.-B.L. contributed equally to this work.

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
Banchereau
J
Steinman
RM
Dendritic cells and the control of immunity.
Nature.
392
1998
245
252
2
Bell
D
Young
JW
Banchereau
J
Dendritic cells.
Adv Immunol.
72
1999
255
324
3
Timmerman
JM
Levy
R
Dendritic cell vaccines for cancer immunotherapy.
Annu Rev Med.
50
1999
507
529
4
Bello-Fernandez
C
Matyash
M
Strobl
H
et al
Efficient retrovirus-mediated gene transfer of dendritic cells generated from CD34+ cord blood cells under serum-free conditions.
Hum Gene Ther.
8
1997
1651
1658
5
Chischportich
C
Bagnis
C
Galindo
R
Mannoni
P
Expression of the nlsLacz gene in dendritic cells derived from retrovirally transduced peripheral blood CD34+ cells.
Haematologica.
84
1999
195
203
6
Henderson
RA
Konitsky
WM
Barratt-Boyes
SM
Soares
M
Robbins
PD
Finn
OJ
Retroviral expression of MUC-1 human tumor antigen with intact repeat structure and capacity to elicit immunity in vivo.
J Immunother.
21
1998
247
256
7
Movassagh
M
Baillou
C
Cosset
FL
Klatzmann
D
Guigon
M
Lemoine
FM
High level of retrovirus-mediated gene transfer into dendritic cells derived from cord blood and mobilized peripheral blood CD34+ cells.
Hum Gene Ther.
10
1999
175
187
8
Reeves
ME
Royal
RE
Lam
JS
Rosenberg
SA
Hwu
P
Retroviral transduction of human dendritic cells with a tumor-associated antigen gene.
Cancer Res.
56
1996
5672
5677
9
Szabolcs
P
Gallardo
HF
Ciocon
DH
Sadelain
M
Young
JW
Retrovirally transduced human dendritic cells express a normal phenotype and potent T-cell stimulatory capacity.
Blood.
90
1997
2160
2167
10
Roe
T
Reynolds
TC
Yu
G
Brown
PO
Integration of murine leukemia virus DNA depends on mitosis.
EMBO J.
12
1993
2099
2108
11
Hajihosseini
M
Iavachev
L
Price
J
Evidence that retroviruses integrate into post-replication host DNA.
EMBO J.
12
1993
4969
4974
12
Miller
DG
Adam
MA
Miller
AD
Gene transfer by retrovirus vectors occurs only in cells that are actively replicating at the time of infection.
Mol Cell Biol.
10
1990
4239
4242
13
Granelli-Piperno
A
Delgado
E
Finkel
V
Paxton
W
Steinman
RM
Immature dendritic cells selectively replicate macrophagetropic (M-tropic) human immunodeficiency virus type 1, while mature cells efficiently transmit both M- and T-tropic virus to T cells.
J Virol.
72
1998
2733
2737
14
O'Doherty
U
Steinman
RM
Peng
M
et al
Dendritic cells freshly isolated from human blood express CD4 and mature into typical immunostimulatory dendritic cells after culture in monocyte-conditioned medium.
J Exp Med.
178
1993
1067
1076
15
Schreurs
MW
Eggert
AA
de Boer
AJ
Figdor
CG
Adema
GJ
Generation and functional characterization of mouse monocyte-derived dendritic cells.
Eur J Immunol.
29
1999
2835
2841
16
Lewis
PF
Emerman
M
Passage through mitosis is required for oncoretroviruses but not for the human immunodeficiency virus.
J Virol.
68
1994
510
516
17
Emerman
M
HIV-1, Vpr and the cell cycle.
Curr Biol.
6
1996
1096
1103
18
Naldini
L
Blomer
U
Gallay
P
et al
In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector.
Science.
272
1996
263
267
19
Gallardo
HF
Tan
C
Ory
D
Sadelain
M
Recombinant retroviruses pseudotyped with the vesicular stomatitis virus G glycoprotein mediate both stable gene transfer and pseudotransduction in human peripheral blood lymphocytes.
Blood.
90
1997
952
957
20
Gallardo
HF
Tan
C
Sadelain
M
The internal ribosomal entry site of the encephalomyocarditis virus enables reliable coexpression of two transgenes in human primary T lymphocytes.
Gene Ther.
4
1997
1115
1119
21
Latouche
JB
Sadelain
M
Potent induction of human cytotoxic T lymphocytes against viral and tumor antigens by artificial antigen-presenting cells.
Nat Biotechnol.
18
2000
405
409
22
Chen
C
Okayama
H
High-efficiency transformation of mammalian cells by plasmid DNA.
Mol Cell Biol.
7
1987
2745
2752
23
Dull
T
Zufferey
R
Kelly
M
et al
A third-generation lentivirus vector with a conditional packaging system.
J Virol.
72
1998
8463
8471
24
Ory
DS
Neugeboren
BA
Mulligan
RC
A stable human-derived packaging cell line for production of high titer retrovirus/vesicular stomatitis virus G pseudotypes.
Proc Natl Acad Sci U S A.
93
1996
11400
11406
25
Zufferey
R
Nagy
D
Mandel
RJ
Naldini
L
Trono
D
Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo.
Nat Biotechnol.
15
1997
871
875
26
Ercikan-Abali
EA
Mineishi
S
Tong
Y
et al
Active site-directed double mutants of dihydrofolate reductase.
Cancer Res.
56
1996
4142
4145
27
Riviere
I
Brose
K
Mulligan
RC
Effects of retroviral vector design on expression of human adenosine deaminase in murine bone marrow transplant recipients engrafted with genetically modified cells.
Proc Natl Acad Sci U S A.
92
1995
6733
6737
28
Riviere
I
Sadelain
M
Methods for the Construction of Retroviral Vectors and the Generation of High Titer Producers.
1997
Humana Press
Totowa, NJ
29
Bender
A
Sapp
M
Schuler
G
Steinman
RM
Bhardwaj
N
Improved methods for the generation of dendritic cells from nonproliferating progenitors in human blood.
J Immunol Methods.
196
1996
121
135
30
Romani
N
Reider
D
Heuer
M
et al
Generation of mature dendritic cells from human blood: an improved method with special regard to clinical applicability.
J Immunol Methods.
196
1996
137
151
31
Bhardwaj
N
Bender
A
Gonzalez
N
Bui
LK
Garrett
MC
Steinman
RM
Influenza virus-infected dendritic cells stimulate strong proliferative and cytolytic responses from human CD8+ T cells.
J Clin Invest.
94
1994
797
807
32
Young
JW
Steinman
RM
Dendritic cells stimulate primary human cytolytic lymphocyte responses in the absence of CD4+ helper T cells.
J Exp Med.
171
1990
1315
1332
33
Kawakami
Y
Eliyahu
S
Sakaguchi
K
et al
Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2–restricted tumor infiltrating lymphocytes.
J Exp Med.
180
1994
347
352
34
Schroers
R
Sinha
I
Segall
H
et al
Transduction of human PBMC-derived dendritic cells and macrophages by an HIV-1–based lentiviral vector system.
Mol Ther.
1
2000
171
179
35
Chun
TW
Stuyver
L
Mizell
SB
et al
Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy.
Proc Natl Acad Sci U S A.
94
1997
13193
13197
36
Nelson
DL
Ledbetter
SA
Corbo
L
et al
Alu polymerase chain reaction: a method for rapid isolation of human-specific sequences from complex DNA sources.
Proc Natl Acad Sci U S A.
86
1989
6686
6690
37
Bednarek
MA
Sauma
SY
Gammon
MC
et al
The minimum peptide epitope from the influenza virus matrix protein: extra and intracellular loading of HLA-A2.
J Immunol.
147
1991
4047
4053
38
Morrison
J
Elvin
J
Latron
F
et al
Identification of the nonamer peptide from influenza A matrix protein and the role of pockets of HLA-A2 in its recognition by cytotoxic T lymphocytes.
Eur J Immunol.
22
1992
903
907
39
Martinon
F
Gomard
E
Hannoun
C
Lévy
J-P
In vitro human cytotoxic T cell responses against influenza A virus can be induced and selected by synthetic peptides.
Eur J Immunol.
20
1990
2171
2176
40
Bender
A
Sapp
M
Schuler
G
Steinman
RM
Bhardwaj
N
Improved methods for the generation of dendritic cells from nonproliferating progenitors in human blood.
J Immunol Methods.
196
1996
121
135
41
Gotch
F
Rothbard
J
Howland
K
Townsend
A
McMichael
A
Cytotoxic T lymphocytes recognize a fragment of influenza virus matrix protein in association with HLA-A2.
Nature.
326
1987
881
882
42
Tussey
LG
Matsui
M
Rowland-Jones
S
Warburton
R
Frelinger
JA
McMichael
A
Analysis of mutant HLA-A2 molecules.
J Immunol.
152
1994
1213
1221
43
McMichael
AJ
Gotch
FM
Santos-Aguado
J
Strominger
JL
Effect of mutations and variations of HLA-A2 on recognition of a virus peptide epitope by cytotoxic T lymphocytes.
Proc Natl Acad Sci U S A.
85
1988
9194
9198
44
Latron
F
Pazmany
L
Morrison
J
et al
A critical role for conserved residues in the cleft of HLA-A2 in presentation of a nonapeptide to T cells.
Science.
257
1992
964
967
45
Anderson
K
Cresswell
P
Gammon
M
Hermes
J
Williamson
A
Zweerink
H
Endogenously synthesized peptide with an endoplasmic reticulum signal sequence sensitizes antigen processing mutant cells to class I-restricted cell-mediated lysis.
J Exp Med.
174
1991
489
492
46
Heemskerk
MH
Hooijberg
E
Ruizendaal
JJ
van der Weide
MM
Kueter
E
Bakker
AQ
Schumacher
TN
Spits
H
Enrichment of an antigen-specific T cell response by retrovirally transduced human dendritic cells.
Cell Immunol.
195
1999
10
17
47
Amado
RG
Chen
IS
Lentiviral vectors—the promise of gene therapy within reach?
Science.
285
1999
674
676
48
Blomer
U
Naldini
L
Kafri
T
Trono
D
Verma
IM
Gage
FH
Highly efficient and sustained gene transfer in adult neurons with a lentivirus vector.
J Virol.
71
1997
6641
6649
49
Kafri
T
Blomer
U
Peterson
DA
Gage
FH
Verma
IM
Sustained expression of genes delivered directly into liver and muscle by lentiviral vectors.
Nat Genet.
17
1997
314
317
50
Miyoshi
H
Takahashi
M
Gage
FH
Verma
IM
Stable and efficient gene transfer into the retina using an HIV-based lentiviral vector.
Proc Natl Acad Sci U S A.
94
1997
10319
10323
51
Naldini
L
Blomer
U
Gage
FH
Trono
D
Verma
IM
Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector.
Proc Natl Acad Sci U S A.
93
1996
11382
11388
52
Kim
VN
Mitrophanous
K
Kingsman
SM
Kingsman
AJ
Minimal requirement for a lentivirus vector based on human immunodeficiency virus type 1.
J Virol.
72
1998
811
816
53
Heinzinger
NK
Bukinsky
MI
Haggerty
SA
et al
The Vpr protein of human immunodeficiency virus type 1 influences nuclear localization of viral nucleic acids in nondividing host cells.
Proc Natl Acad Sci U S A.
91
1994
7311
7315
54
Popov
S
Rexach
M
Ratner
L
Blobel
G
Bukrinsky
M
Viral protein R regulates docking of the HIV-1 preintegration complex to the nuclear pore complex.
J Biol Chem.
273
1998
13347
13352
55
Inaba
K
Young
JW
Steinman
RM
Direct activation of CD8+ cytotoxic T lymphocytes by dendritic cells.
J Exp Med.
166
1987
182
194
56
Lu
Z
Yuan
L
Zhou
X
et al
CD40-independent pathways of T cell help for priming of CD8+ cytotoxic T lymphocytes.
J Exp Med.
191
2000
541
550
57
Buller
RM
Holmes
KL
Hugin
A
Frederickson
TN
Morse
HC
III
Induction of cytotoxic T-cell responses in vivo in the absence of CD4 helper cells.
Nature.
328
1987
77
79
58
Rahemtulla
A
Fung-Leung
WP
Schilham
MW
et al
Normal development and function of CD8+ cells but markedly decreased helper cell activity in mice lacking CD4.
Nature.
53
1991
180
184
59
Hou
S
Mo
XY
Hyland
L
Doherty
PC
Host response to Sendai virus in mice lacking class II major histocompatibility complex glycoproteins.
J Virol.
69
1995
1429
1434
60
Cella
M
Salio
M
Sakakibara
Y
Langen
H
Julkunen
I
Lanzavecchia
A
Maturation, activation, and protection of dendritic cells induced by double-stranded RNA.
J Exp Med.
189
1999
821
829
61
Ridge
JP
Di Rosa
F
Matzinger
P
A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell.
Nature.
393
1998
474
478
62
Schnell
S
Young
JW
Houghton
AN
Sadelain
M
Retrovirally transduced mouse dendritic cells require CD4+ T cell help to elicit antitumor immunity: implications for the clinical use of dendritic cells.
J Immunol.
164
2000
1243
1250
63
Inaba
K
Inaba
M
Romani
N
et al
Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor.
J Exp Med.
176
1992
1693
1702
64
Dietz
AB
Vuk-Pavlovic
S
High efficiency adenovirus-mediated gene transfer to human dendritic cells.
Blood.
91
1998
392
398
65
Coffin
RS
Thomas
SK
Thomas
NS
et al
Pure populations of transduced primary human cells can be produced using GFP expressing herpes virus vectors and flow cytometry.
Gene Ther.
5
1998
718
722
66
Subklewe
M
Chahroudi
A
Schmaljohn
A
Kurilla
MG
Bhardwaj
N
Steinman
RM
Induction of Epstein-Barr virus-specific cytotoxic T-lymphocyte responses using dendritic cells pulsed with EBNA-3A peptides or UV-inactivated, recombinant EBNA-3A vaccinia virus.
Blood.
94
1999
1372
1381
67
Chaux
P
Luiten
R
Demotte
N
et al
Identification of five MAGE-A1 epitopes recognized by cytolytic T lymphocytes obtained by in vitro stimulation with dendritic cells transduced with MAGE-A1.
J Immunol.
163
1999
2928
2936
68
Brown
M
Davies
DH
Skinner
MA
et al
Antigen gene transfer to cultured human dendritic cells using recombinant avipoxvirus vectors.
Cancer Gene Ther.
6
1999
238
245
69
Pulendran
B
Smith
JL
Caspary
G
et al
Distinct dendritic cell subsets differentially regulate the class of immune response in vivo.
Proc Natl Acad Sci U S A.
96
1999
1036
1041
70
Rissoan
MC
Soumelis
V
Kadowaki
N
et al
Reciprocal control of T helper cell and dendritic cell differentiation.
Science.
283
1999
1183
1186
71
Vremec
D
Shortman
K
Dendritic cell subtypes in mouse lymphoid organs: cross-correlation of surface markers, changes with incubation, and differences among thymus, spleen, and lymph nodes.
J Immunol.
159
1997
565
573

Author notes

Michel Sadelain, Box 182, Department of Human Genetics, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10021; e-mail: m-sadelain@ski.mskcc.org.

Sign in via your Institution