Effector T cells (TEFF) are a barrier to booster vaccination because they can rapidly kill Ag-bearing APCs before memory T cells are engaged. We report in this study that i.v. delivery of rhabdoviral vectors leads to direct infection of follicular B cells in the spleen, where the earliest evidence of secondary T cell responses was observed. This allows booster immunizations to rapidly expand CD8+ central memory T cells (TCM) during the acute phase of the primary response that is dominated by TEFF. Interestingly, although the ablation of B cells before boosting with rhabdoviral vectors diminishes the expansion of memory T cells, B cells do not present Ags directly. Instead, depletion of CD11c+ dendritic cells abrogates secondary T cell expansion, suggesting that virus-infected follicular B cells may function as an Ag source for local DCs to subsequently capture and present the Ag. Because TCM are located within B cell follicles in the spleen whereas TEFF cannot traffic through follicular regions, Ag production and presentation by follicular APCs represent a unique mechanism to secure engagement of TCM during an ongoing effector response. Our data offer insights into novel strategies for rapid expansion of CD8+ T cells using prime-boost vaccines by targeting privileged sites for Ag presentation.

This article is featured in In This Issue, p.4421

The CD8+ T cell subset plays an essential role in host defense against viruses, intracellular bacteria, and malignancies. Generation of protective CD8+ T cell immunity has been a central focus for the development of Ag-specific vaccines. Although numerous strategies have been designed to elicit high frequencies of circulating Ag-specific CD8+ T cells, sequential immunizations with viral vectors (conventionally known as prime-boost immunization) is one of the most effective (1). The acute phase of the primary response is dominated by highly differentiated CD8+ effector T cells (TEFF) that display robust cytotoxicity and production of inflammatory cytokines, necessary for initial control of fatal infections and other severe diseases. However, TEFF have a limited capacity to proliferate and booster immunizations need to optimally stimulate memory T cells, especially central memory T cells (TCM), which proliferate rapidly and vigorously upon encounter with Ags (2). Although a small number of TCM can be identified during the acute phase of a typical primary CD8+ T cell response, subsequent boosting is limited by pre-existing TEFF that rapidly clear Ag-loaded cells, preventing adequate Ag presentation to TCM (35). Thus, prime-boost immunizations in a prophylactic setting are typically spread across a period of weeks to months to allow time for the TCM to dominate the available T cell pool and enable maximal secondary expansion. In the case of therapeutic vaccination for patients with lethal (e.g., Ebola) or chronic infection (e.g., HIV, hepatitis C virus) or cancer, where immediate and high levels of the TEFF are needed for protection, boosting strategies that can bypass TEFF-mediated negative feedback regulation would be desirable.

It is established that activation of naive T cells requires a coordinated process between migratory dendritic cells (DCs) from peripheral tissues and lymphoid-resident APCs (68), but little is known about the role of different APCs in Ag presentation to memory T cells. It was originally thought that memory T cells had a reduced threshold for activation and, thus, could be triggered to proliferate by both professional and nonprofessional APCs (9, 10). Recent work, however, has changed that perspective, as hematopoietic APCs have been shown to be required for secondary expansion (11); one study has suggested that DCs, in particular, may be required for maximal secondary responses (12). However, Ag-carrying DCs are sensitive to TEFF-mediated killing and they can be rapidly eliminated prior to their engagement with memory cells in the secondary lymphoid organs (1315).

Interestingly, we have recently demonstrated that vaccination with recombinant rhabdoviruses, such as vesicular stomatitis virus (VSV) and Maraba virus, could provoke massive secondary expansion of T cells as early as 4 d after priming or at the height of the primary CD8+ T cell response (7–14 d after priming) (1618), suggesting that rhabdoviral boosting can overcome the TEFF barrier, allowing Ag-loaded APCs to escape TEFF-mediated killing. Understanding the mechanisms of Ag presentation that facilitate rapid boosting by rhabdoviruses will provide important information that can be used to design optimal vaccination strategies for rapid generation of protective responses.

In the present study, we demonstrate that a robust secondary CD8+ T cell expansion following booster immunization with VSV occurs primarily in the spleen. Inducing maximal secondary CD8+ T cell responses at the peak of the primary response requires i.v. delivery of VSV, which results in direct infection of follicular B cells and subsequent Ag presentation by neighboring DCs in the follicular region. Escape of Ag-loaded APCs from TEFF-mediated killing is consistent with evidence that TCM are located within B cell follicles in the spleen whereas TEFF cannot traffic through follicular regions (1921). These data reveal a distinct role of splenic APCs with regard to secondary CD8+ T cell activation, which may not only offer insights into engineering prime-boost vaccines for rapid expansion of CD8+ T cells but may also provide new fundamental mechanistic insight into the immune response to viremia.

C57BL/6 and BALB/c mice were purchased from Charles River Laboratories (Wilmington, MA). B6.PL-Thy1a/CyJ (Thy1.1 congenic), B6.PL-Thy1a/CyJ, B6.129S2-Igh-6tm1Cgn/J (referred to as B−/− mice), and B6/JiKbtm1Dbtm1N12 (referred to as KbDb−/− mice) were obtained from The Jackson Laboratory (Bar Harbor, ME). CD11c–diphtheria toxin receptor (DTR) mice were bred in the Central Animal Facility at McMaster University. All animal experimentation was approved by McMaster University’s Animal Research Ethics Board and complied with the Canadian Council on Animal Care guidelines.

The recombinant human adenovirus (Ad)-BHG was a replication-deficient recombinant human serotype 5 Ad with no transgene inserted and served as a control vector. The Ad–human dopachrome tautomerase (hDCT) expressed the full-length hDCT gene. The Ad-SIINFEKL-luciferase (Luc) (Ad-SIIN) vector expressed the immunodominant epitope of chicken OVA (OVA257–264) coupled to firefly luciferase. The Ad-gp33 expressed the immunodominant eptiope from the gp33 of lymphocytic choriomeningitis virus (LCMV). Recombinant VSV-hDCT and VSV-GFP have been described (22). VSV-SIIN (expressing OVA257–264 and luciferase) and VSV-gp33 (expressing gp33–41 from LCMV) were engineered by an identical construction strategy. The VSV-MT was a control vector lacking a transgene. Recombinant vaccinia virus (VV, Western Reserve strain) expressing SIINFEKL-Luc (VV-SIINFEKL-Luc) has been described (23). Recombinant Maraba virus expressing hDCT (Maraba-hDCT) was constructed based on the attenuated strain MG1 of Maraba virus (24).

Kb-restricted DCT (DCT180–188, SVYDFFVWL) and OVA (OVA257–264, SIINFEKL) peptides were synthesized by Pepscan Systems (Lelystad, the Netherlands). Using a peptide library of 15-mers spanning the full length of the DCT protein (Pepscan Systems) we identified several CD8+ T cell epitope–containing peptides, which were pooled for evaluating immune responses in BALB/c mice.

Abs and kits used for surface or intracellular staining were purchased from BD Biosciences (Mississauga, ON, Canada). The tetramers Kb-DCT180–188-allophycocyanin, Kb-OVA257–264-allophycocyanin, and Db-gp33–41-allophycocyanin were obtained from Baylor College of Medicine.

Immunized mice received i.p. injections of 1 mg BrdU and BrdU-containing drinking water (0.8 mg/ml) 24 h prior to harvesting tissues. To block T cell trafficking from lymphoid organs in some experiments, mice also received an i.p. injection of FTY720 (Cayman Chemical, Ann Arbor, Michigan) at 4 mg/kg body weight 24 h before boosting with VSV. After blocking Fc receptors, lymphocytes in single-cell suspensions from various tissues were stained with a tetramer and then Abs against surface CD8 and intranuclear BrdU (BD Biosciences).

Negatively selected splenic CD8+ T cells (Stemcell Technologies, Vancouver, BC, Canada) from Ad-SIIN–immunized congenic (Thy1.1+) mice were sorted into TEFF (CD127CD62L), effector memory T cell (TEM, CD127+CD62L), and TCM (CD127+CD62L+) populations. Sorted subsets (>90% purity) were adoptively transferred into naive wild-type (WT) C57BL/6 recipients (Thy1.2+) 1 d prior to vaccination with VSV-SIIN. Five days after administration of VSV, blood, bone marrow, and spleens were harvested to determine the frequency of congenic tetramer+ cells.

C57BL/6 mice received 1 × 109 PFU VSV-GFP i.v. Spleens were harvested 1.5 h later to minimize the potential for the confounding effect of cell trafficking between tissues, while providing enough time for VSV to infect cells. Splenocytes were cultured for an additional 4.5 h to allow GFP to accumulate within infected cells for detection by flow cytometry.

For depletion of B cells, mice were given 250 μg anti-CD20 mAb (clone 18B12, Biogen Idec, San Diego, CA) by the i.v. route 7 d prior to vaccination and 2 wk later to maintain depletion. By flow cytometry, >98% of B cells were removed from the blood and spleen. Sham-treated mice received an isotype control.

C57BL/6 recipient mice were irradiated (2 × 550 rads; 48-h interval) and then received i.v. injections of T cell–depleted bone marrow–derived cells. Four sets of chimeric mice were made: group 1 received 5 × 106 WT cells; group 2 received 4 × 106 cells from B−/− mice and 2 × 106 cells from WT mice; group 3 received 4 × 106 cells from B−/− mice and 2 × 106 cells from KbDb−/− mice; and group 4 received 5 × 106 cells from B−/− donors. CD11c-DTR chimeric mice were similarly prepared by transferring CD11c-DTR transgenic mouse-derived bone marrow into lethally irradiated C57BL/6 recipients. Mice were given 3 mo to reconstitute their hematopoietic systems, which was confirmed by flow cytometry prior to experimentation.

C57BL/6 or KbDb−/− mice were injected i.v. with 1 × 109 VSV-GFP or VSV-gp33. Seventeen hours later, splenic B cells were purified using negative selection kits according to the manufacturer’s instructions (Miltenyi Biotec, Teterow, Germany). Naive P14 cells were enriched by positive selection from the spleens of P14 mice (EasySep mouse CD8a positive selection kit, Stemcell Technologies) and labeled with 1 mM CFSE (Sigma-Aldrich, Oakville, ON, Canada). Bone marrow–derived DCs were generated and matured as described previously (25).

Purified WT B cells (2 × 107) were cocultured with 2 × 106 bone marrow–derived DCs for 18 h at 37°C. DCs were separated using CD11c microbeads (Miltenyi Biotec) and cultured with CFSE-labeled P14 cells at a 1:1 ratio. Cultures were analyzed for proliferation after 72 h. Additionally, 5 × 106 KbDb−/− B cells were directly mixed with 5 × 105 CFSE-labeled P14 cells in a 24-well plate or separated by Transwell inserts (Corning, Tewksbury, MA) in the presence or absence of 5 × 105 DCs for 72 h prior to flow cytometric analysis.

Mice primed with Ad-hDCT received splenectomies or sham surgeries 12 d later. At 48 h after surgery, mice were boosted with 1 × 109 PFU i.v. VSV-hDCT. Five days after VSV, DCT180–188-specific CD8+ T cell responses were quantified by flow cytometric analysis of intracellular cytokine staining following in vitro peptide restimulation.

GraphPad Prism for Windows 6.0 (GraphPad Software, San Diego, CA) was used for graphing and statistical analyses. When required, data were normalized by log transformation. A Student two-tailed t test, Mann–Whitney U test, or one- or two-way ANOVA was used to query immune response data. Differences between means were considered significant at p ≤ 0.05. Means with SE bars are shown. Survival data were analyzed using the Kaplan–Meier method and the log-rank test.

The choice of i.v. administration of rhabdoviruses (i.e., VSV and Maraba) in our previous studies was driven by our desire to exploit their oncolytic potential to target both primary and metastatic tumors (16, 18). Our observation that i.v. injection of rhabdoviruses also resulted in a rapid and robust secondary expansion of Ag-specific CD8+ T cells prompted us to determine whether i.v. delivery was a particularly efficient route for booster vaccinations. Mice were immunized with an Ad vector expressing an OVA-derived CD8+ T cell epitope (OVA257– 264, SIINFEKL) coupled to luciferase (Ad-SIIN) (26). Fourteen days following immunization, OVA-specific CD8+ T cells had expanded to 12% of total circulating CD8+ T cells (Fig. 1A, lower panel) and primarily displayed a TEFF phenotype (CD62LloCD127loKLRG1higranzyme Bhi; data not shown). Despite the high frequencies of TEFF, i.v. administration of a VSV vector encoding the same Ag (VSV-SIIN) provoked a massive expansion of OVA-specific cells, reaching a mean of 88% of circulating CD8+ T cells (Fig. 1A, lower panel) or a 58-fold increase by absolute numbers (Fig. 1A, upper panel). However, other routes, including i.m., i.p., intranasal, and s.c., were not as efficient as i.v. (Fig. 1A, upper panel).

FIGURE 1.

VSV can rapidly induce robust CD8+ T cells responses in a route- and dose-dependent manner. (A) C57BL/6 mice were primed with Ad-SIIN (1 × 108 PFU, i.m.). Fourteen days later, they were treated with PBS or boosted with VSV-SIIN (1 × 109 PFU) by various routes. Five days after treatment with VSV, OVA-specific CD8+ T cells in blood were quantified by tetramer staining. (B) C57BL/6 mice were primed i.m. with Ad-hDCT and boosted 14 d later with VSV-hDCT via different routes. DCT-specific CD8+ T cells in blood were measured 5 d after treatment with VSV. (C) C57BL/6 mice primed with Ad-hDCT were boosted with VSV-hDCT or Maraba-hDCT after a 14-d interval. Five days after boosting, CD8+ T cell responses to DCT180–188 were quantified by detection of intracellular cytokine staining. All graphs show means with SE bars; n = 5 per group; one-way ANOVA. Each experiment was replicated a minimum of three times. **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 1.

VSV can rapidly induce robust CD8+ T cells responses in a route- and dose-dependent manner. (A) C57BL/6 mice were primed with Ad-SIIN (1 × 108 PFU, i.m.). Fourteen days later, they were treated with PBS or boosted with VSV-SIIN (1 × 109 PFU) by various routes. Five days after treatment with VSV, OVA-specific CD8+ T cells in blood were quantified by tetramer staining. (B) C57BL/6 mice were primed i.m. with Ad-hDCT and boosted 14 d later with VSV-hDCT via different routes. DCT-specific CD8+ T cells in blood were measured 5 d after treatment with VSV. (C) C57BL/6 mice primed with Ad-hDCT were boosted with VSV-hDCT or Maraba-hDCT after a 14-d interval. Five days after boosting, CD8+ T cell responses to DCT180–188 were quantified by detection of intracellular cytokine staining. All graphs show means with SE bars; n = 5 per group; one-way ANOVA. Each experiment was replicated a minimum of three times. **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

We next examined the route dependence of the boosting immunization using another Ag. In this case, mice were primed with an Ad expressing the weakly immunogenic melanocyte-differentiation Ag DCT (Ad-hDCT) and boosted with a VSV expressing the same Ag (VSV-hDCT) (22). Interestingly, not only was i.v. injection the best route for the booster vaccination, it was the only route that generated a significant boost against this self-epitope (Fig. 1B). Similar boosting responses were achieved with Maraba virus, suggesting that rhabdoviruses share similar biological properties (Fig. 1C). Intravenous administration of VSV-hDCT also induced a dramatic expansion of CD8+ T cells in BALB/c mice, indicating that VSV-mediated boosting was not strain specific (Fig. 2A). We also determined that the VSV boost was effective in the same time frame (i.e., 7–14 d after primary immunization) in mice primed with DCs or VV expressing the same Ag (Fig. 2B, 2C). Thus, the remarkable potency of the VSV boost was not limited to a single strain, Ag, or priming method but did require i.v. delivery of the boosting virus.

FIGURE 2.

Efficient boosting is not limited to a single mouse strain, Ag, or priming method. (A) BALB/c mice were primed with Ad-hDCT and then treated i.v. with PBS or boosted with VSV-hDCT at a 14-d interval. Five days after boosting, DCT-specific CD8+ T cells were measured in blood by flow cytometry after in vitro restimulation with pooled immunoreactive peptides and intracellular cytokine staining. ****p < 0.0001. (B) C57BL/6 mice that received DCs pulsed with the gp33 peptide were boosted 14 d later with VSV-gp33. (C) C57BL/6 mice primed with VV-SIINFEKL-Luc were boosted with VSV-SIINFEKL-Luc. Five days after treatment with VSV, SIINFEKL-specific CD8+ T cells were quantified by intracellular cytokine staining. (D) C57BL/6 mice carrying 5-d-old B16-gp33 tumors were treated with DC/gp33 or Ad-gp33 vaccine alone or in combination with VSV-gp33 at a 9-d interval. PBS or VSV-gp33 alone was included as controls. Means plus SE bars are shown; n = 5 per group; two-way ANOVA. Most of these groups were repeated more than once, with a similar result.

FIGURE 2.

Efficient boosting is not limited to a single mouse strain, Ag, or priming method. (A) BALB/c mice were primed with Ad-hDCT and then treated i.v. with PBS or boosted with VSV-hDCT at a 14-d interval. Five days after boosting, DCT-specific CD8+ T cells were measured in blood by flow cytometry after in vitro restimulation with pooled immunoreactive peptides and intracellular cytokine staining. ****p < 0.0001. (B) C57BL/6 mice that received DCs pulsed with the gp33 peptide were boosted 14 d later with VSV-gp33. (C) C57BL/6 mice primed with VV-SIINFEKL-Luc were boosted with VSV-SIINFEKL-Luc. Five days after treatment with VSV, SIINFEKL-specific CD8+ T cells were quantified by intracellular cytokine staining. (D) C57BL/6 mice carrying 5-d-old B16-gp33 tumors were treated with DC/gp33 or Ad-gp33 vaccine alone or in combination with VSV-gp33 at a 9-d interval. PBS or VSV-gp33 alone was included as controls. Means plus SE bars are shown; n = 5 per group; two-way ANOVA. Most of these groups were repeated more than once, with a similar result.

Close modal

The therapeutic relevance of this prime-boost approach was demonstrated by an aggressive murine melanoma model (B16) where a single vector alone only slightly slowed down the growth of pre-established B16 tumors (5 d old) and mice reached the endpoint rapidly in 12 d after treatment (Fig. 2D). However, priming animals bearing 5-d-old tumors with either Ad or DCs was sufficient to allow a boost by VSV 9 d after priming, leading to complete tumor regression (Fig. 2D).

To learn more about the mechanism of the VSV-mediated boost, we first determined the locations of secondary expansion of T cells following i.v. inoculation of VSV. Mice were primed with Ad-hDCT and boosted 14 d later by i.v. VSV-hDCT. As a control, a subset of the primed mice received only VSV-MT, which does not express any transgene. The frequency of DCT-specific CD8+ T cells was assessed in various lymphoid tissues on days 1, 2, 3, 4, and 7 following administration of VSV. To monitor local proliferation, BrdU was administered to the mice for 24 h prior to lymphocyte harvest. As shown in Fig. 3A, the proliferative response peaked around days 3–4 following boosting and returned to baseline by day 7. Notably, the earliest evidence of a proliferative response was observed in the spleen at day 2 after boosting, suggesting that the secondary expansion began in the spleen somewhere between 24 and 48 h after boost. Ultimately, proliferation was observed in all tissues, likely as a consequence of migratory activated CD8+ T cells. To further confirm that the spleen was the primary site where the proliferation of secondary T cells started, we carried out two more experiments. First, we treated mice with FTY720 to inhibit lymphocyte egress from lymphoid organs during boosting, and T cell proliferation was monitored by BrdU incorporation. Compared to the VSV-MT control, the earliest proliferation of hDCT-specific CD8+ T cells was evident only in the spleen 48 h after boosting with VSV-hDCT (Fig. 3A, 3B, Supplemental Fig. 1). Second, spleens in Ad-hDCT–primed mice were surgically removed 2 d before VSV-hDCT boosting (i.e., 12 d after priming) and secondary expansion was determined on day 5 after boosting. Data in Fig. 3C show that CD8+ T cell expansion was abrogated in the absence of a spleen, reinforcing a critical role of this organ in initiating the secondary T cell responses.

FIGURE 3.

The spleen is the primary site for secondary expansion of central memory T cells. (A) Ad-hDCT–primed C57BL/6 mice were boosted 14 d later with VSV-hDCT. Each group received BrdU via i.p. injections and drinking water 24 h prior to tissue harvest, at which time BrdU+ DCT-specific CD8+ T cells were enumerated (n = 3 per group per time point; mean with SE bars; two-way ANOVA; *p < 0.05). (B) C57BL/6 mice primed with Ad-hDCT were boosted with VSV-hDCT 14 d later. Twenty-four hours after treatment with VSV, mice received BrdU and FTY720 via i.p. injections. At 24 h after BrdU/FTY720 treatment, harvested tissues (spleen, lymph nodes, and bone marrow) were assessed by tetramer staining to quantify DCT180–188-specific CD8+ T cells. (C) Mice primed with Ad-hDCT received splenectomies (Splx) or sham surgeries 12 d later. At 48 h after surgery, mice were boosted with VSV-hDCT. Five days after treatment with VSV, DCT180–188-specific CD8+ T cell responses were quantified by intracellular cytokine staining. Means plus SE bars are shown; n = 5 per group; unpaired t tests (B) and two-way ANOVA (C). This experiment was replicated three times.

FIGURE 3.

The spleen is the primary site for secondary expansion of central memory T cells. (A) Ad-hDCT–primed C57BL/6 mice were boosted 14 d later with VSV-hDCT. Each group received BrdU via i.p. injections and drinking water 24 h prior to tissue harvest, at which time BrdU+ DCT-specific CD8+ T cells were enumerated (n = 3 per group per time point; mean with SE bars; two-way ANOVA; *p < 0.05). (B) C57BL/6 mice primed with Ad-hDCT were boosted with VSV-hDCT 14 d later. Twenty-four hours after treatment with VSV, mice received BrdU and FTY720 via i.p. injections. At 24 h after BrdU/FTY720 treatment, harvested tissues (spleen, lymph nodes, and bone marrow) were assessed by tetramer staining to quantify DCT180–188-specific CD8+ T cells. (C) Mice primed with Ad-hDCT received splenectomies (Splx) or sham surgeries 12 d later. At 48 h after surgery, mice were boosted with VSV-hDCT. Five days after treatment with VSV, DCT180–188-specific CD8+ T cell responses were quantified by intracellular cytokine staining. Means plus SE bars are shown; n = 5 per group; unpaired t tests (B) and two-way ANOVA (C). This experiment was replicated three times.

Close modal

The population of Ad-induced CD8+ T cells within the spleen on the day of the boost was composed primarily of TEFF, although a small population of TCM was also detectable (Fig. 4A). To determine which CD8+ T cell subset was contributing to the secondary expansion, OVA257–264-specific splenic CD8+ T cells (Thy1.1) were sorted into TEFF, TEM, and TCM subsets based on CD127 and CD62L expression (Fig. 4B) (27). These subsets were adoptively transferred into naive congenic recipients (Thy1.2) that were subsequently immunized with VSV-SIIN. Owing to the low frequencies of Ag-specific CD8+ T cells in the TCM population, we did not attempt to normalize the number of Ag-specific T cells that were transferred. Rather, we normalized the total number of adoptively transferred cells using naive splenocytes to ensure that equal numbers of total T cells were delivered to each recipient (106 cells per mouse). Tetramer staining indicated that each bolus of 106 cells contained 1.9 × 105 OVA257–264-specific TEFF, 6.6 × 104 TEM, or 4 × 103 TCM. Frequencies of adoptively transferred cells (Thy1.1) were determined 5 d after boosting (Fig. 4C). The transgene-specific TCM were the primary responders, as they underwent dramatic expansion 5 d after exposure to VSV-SIIN (i.e., 135-, 425-, and 206-fold increases in frequency relative to day −1 in the spleen, bone marrow, and blood, respectively). In contrast, TEM contributed little to the expansion (∼50% increase over input). As expected, TEFF did not contribute to the secondary expansion and actually contracted during the 5-d period. Our observation is consistent with previous reports that the TCM possess the highest proliferative potential. The results also demonstrated that the TCM subset was preferentially and effectively expanded by the VSV vaccine.

FIGURE 4.

TCM are preferentially and effectively expanded by the VSV vaccine. (A) A typical CD127 versus CD62L staining profile of Kb-OVA257–264 (Tet)–specific CD8+ T cells from the blood of a C57BL/6 mouse 14 d after Ad-SIINFEKL-Luc vaccination. (B) Splenocytes from congenic Ad-SIINFEKL-Luc–immunized mice (Thy1.1) were sorted 14 d later into TEFF (CD127CD62L), TEM (CD127+CD62L), and TCM (CD127+CD62L+) T cell subsets. (C) Pooled splenocytes from Ad-SIIN–immunized congenic mice (Thy1.1) were used to sort OVA-specific T cells into TEFF, TEM, and TCM subsets. Total numbers of cells were normalized using naive splenocytes and then these subsets were injected i.v. into naive C57BL/6 (Thy1.2) recipient mice. Twenty-four hours later, mice received 1 × 109 PFU VSV-SIIN i.v. Five days after treatment with VSV the frequencies of OVA-specific Thy1.1 T cells were determined in various tissues and expressed as a fold change over the frequencies present in the cells that were originally transferred (n = 5 per group; mean with SE bars; one-way ANOVA). These experiments were replicated twice.

FIGURE 4.

TCM are preferentially and effectively expanded by the VSV vaccine. (A) A typical CD127 versus CD62L staining profile of Kb-OVA257–264 (Tet)–specific CD8+ T cells from the blood of a C57BL/6 mouse 14 d after Ad-SIINFEKL-Luc vaccination. (B) Splenocytes from congenic Ad-SIINFEKL-Luc–immunized mice (Thy1.1) were sorted 14 d later into TEFF (CD127CD62L), TEM (CD127+CD62L), and TCM (CD127+CD62L+) T cell subsets. (C) Pooled splenocytes from Ad-SIIN–immunized congenic mice (Thy1.1) were used to sort OVA-specific T cells into TEFF, TEM, and TCM subsets. Total numbers of cells were normalized using naive splenocytes and then these subsets were injected i.v. into naive C57BL/6 (Thy1.2) recipient mice. Twenty-four hours later, mice received 1 × 109 PFU VSV-SIIN i.v. Five days after treatment with VSV the frequencies of OVA-specific Thy1.1 T cells were determined in various tissues and expressed as a fold change over the frequencies present in the cells that were originally transferred (n = 5 per group; mean with SE bars; one-way ANOVA). These experiments were replicated twice.

Close modal

Previous reports have indicated that Ag presentation is rapidly shut off by CD8+ TEFF through a feedback mechanism that involves killing Ag-loaded APCs (14, 28). To determine which cell types were infected by VSV, a vector expressing GFP (VSV-GFP) was injected i.v. into mice and spleens were harvested 90 min later to minimize the migration of infected cells between tissues. Single-cell suspensions were incubated for an additional 4.5 h to provide sufficient time for the VSV to express GFP and then infected cells were analyzed by flow cytometry (Fig. 5A). Strikingly, most infected cells within the spleen were B220+CD19+ B cells (∼82%; Fig. 5B). Further characterization of the VSV-infected B cells revealed most were CD21/35intCD23hiCD27, which is indicative of follicular B cells (Fig. 5B) (29). Ag expression by B cells within the follicular region may represent a unique mechanism to avoid TEFF-mediated clearance because TEFF cannot circulate through this area (30). The GFP expression pattern in the spleen, together with the fact that TCM were primarily reactivated in the spleen, appeared to suggest that follicular B cells might be serving as APCs for the secondary expansion.

FIGURE 5.

Intravenous delivery of VSV results in B cell infection in the spleen. (A) Spleens were harvested from C57BL/6 mice that received i.v. injection of PBS (not shown) or 1 × 109 PFU live or UV-inactivated VSV-GFP for 1.5 h. Splenocytes were cultured for an additional 4.5 h so that enough GFP could be produced by infected cells to allow detection by flow cytometry. (B) Infected (GFP+) cells were stained for B220 and CD19 or CD11b and CD11c to detect B and myeloid cells, respectively. VSV-GFP–infected splenic B cells were further characterized based on their expression of CD21/35 and CD23. This experiment was replicated five times.

FIGURE 5.

Intravenous delivery of VSV results in B cell infection in the spleen. (A) Spleens were harvested from C57BL/6 mice that received i.v. injection of PBS (not shown) or 1 × 109 PFU live or UV-inactivated VSV-GFP for 1.5 h. Splenocytes were cultured for an additional 4.5 h so that enough GFP could be produced by infected cells to allow detection by flow cytometry. (B) Infected (GFP+) cells were stained for B220 and CD19 or CD11b and CD11c to detect B and myeloid cells, respectively. VSV-GFP–infected splenic B cells were further characterized based on their expression of CD21/35 and CD23. This experiment was replicated five times.

Close modal

To characterize the requirement of B cells in vivo for the secondary expansion of TCM, we tested our prime-boost regimen in B cell–deficient (B−/−) mice (31). WT and B−/− mice were immunized with Ad-hDCT and boosted 14 d later with VSV-hDCT. The absence of B cells had no effect on the primary response to the DCT transgene carried by the Ad vector (Fig. 6A, 1°). In contrast, the secondary response to hDCT was severely attenuated in the absence of B cells (Fig. 6A, 2°), suggesting that B cells were required for secondary CD8+ T cell expansion. Because B cells are necessary for the organogenesis of lymphoid tissues (32), we repeated the same experiment in WT mice where B cells were depleted with an anti-CD20 mAb. Similar to the results in B−/− mice, the ablation of B cells before boosting with VSV had a dramatic negative effect on the expansion of the memory DCT-specific T cells (Fig. 6B).

FIGURE 6.

B cells are required for maximal CD8+ T cell expansion but DCs mediate direct Ag presentation. (A) C57BL/6 or B−/− mice were immunized i.m. with 1 × 108 PFU Ad-hDCT and then boosted i.v. with 1 × 109 PFU VSV-hDCT 14 d later. DCT-specific CD8+ T cells were quantified in blood at the peak of the primary (1o) and secondary (2o) immune responses (12 and 5 d after treatment with Ad and VSV, respectively). (B) C57BL/6 mice with or without B cell depletion received Ad-hDCT followed by VSV-hDCT 14 d later. DCT-specific T cells were quantified in blood 5 d after treatment with VSV. (C) Lethally irradiated C57BL/6 mice received bone marrow from WT and/or B−/− and/or KbDb−/− donors. Reconstituted mice were subsequently vaccinated i.m. with 1 × 108 PFU Ad-hDCT and boosted i.v. 14 d later with 1 × 109 PFU VSV-hDCT. DCT-specific CD8+ T cells were quantified in blood at the peak of the primary (1o; 12 d after Ad) and secondary (2o; 5 d after VSV) immune responses. (D and E) Chimeras that had received bone marrow grafts from WT (controls) or CD11c-DTR–transgenic mice were primed with 1 × 108 PFU Ad-hDCT i.m. and boosted 14 d later with 1 × 109 PFU i.v. VSV-hDCT. Two days prior to injection of VSV, DCT-specific T cells isolated from spleens of congenic (i.e., Thy1.1+) Ad-hDCT–immunized mice were adoptively transferred into the chimeras that had received CD11c-DTR bone marrow. One day prior to treatment with VSV and every other day thereafter, half of the chimeric mice received 100 ng i.p. DT to deplete CD11c+ cells. Endogenous [Thy1.1 (D)] and adoptively transferred [Thy1.1+ (E)] DCT-specific T cell responses in blood were quantified 6 d after VSV. In all cases, T cell responses were measured by detection of intracellular cytokines following in vitro stimulation with peptides; n = 5 per group; means with SE bars; one-way ANOVA (B), two-way ANOVA (A and C), or unpaired t tests (D and E). These experiments were replicated three times.

FIGURE 6.

B cells are required for maximal CD8+ T cell expansion but DCs mediate direct Ag presentation. (A) C57BL/6 or B−/− mice were immunized i.m. with 1 × 108 PFU Ad-hDCT and then boosted i.v. with 1 × 109 PFU VSV-hDCT 14 d later. DCT-specific CD8+ T cells were quantified in blood at the peak of the primary (1o) and secondary (2o) immune responses (12 and 5 d after treatment with Ad and VSV, respectively). (B) C57BL/6 mice with or without B cell depletion received Ad-hDCT followed by VSV-hDCT 14 d later. DCT-specific T cells were quantified in blood 5 d after treatment with VSV. (C) Lethally irradiated C57BL/6 mice received bone marrow from WT and/or B−/− and/or KbDb−/− donors. Reconstituted mice were subsequently vaccinated i.m. with 1 × 108 PFU Ad-hDCT and boosted i.v. 14 d later with 1 × 109 PFU VSV-hDCT. DCT-specific CD8+ T cells were quantified in blood at the peak of the primary (1o; 12 d after Ad) and secondary (2o; 5 d after VSV) immune responses. (D and E) Chimeras that had received bone marrow grafts from WT (controls) or CD11c-DTR–transgenic mice were primed with 1 × 108 PFU Ad-hDCT i.m. and boosted 14 d later with 1 × 109 PFU i.v. VSV-hDCT. Two days prior to injection of VSV, DCT-specific T cells isolated from spleens of congenic (i.e., Thy1.1+) Ad-hDCT–immunized mice were adoptively transferred into the chimeras that had received CD11c-DTR bone marrow. One day prior to treatment with VSV and every other day thereafter, half of the chimeric mice received 100 ng i.p. DT to deplete CD11c+ cells. Endogenous [Thy1.1 (D)] and adoptively transferred [Thy1.1+ (E)] DCT-specific T cell responses in blood were quantified 6 d after VSV. In all cases, T cell responses were measured by detection of intracellular cytokines following in vitro stimulation with peptides; n = 5 per group; means with SE bars; one-way ANOVA (B), two-way ANOVA (A and C), or unpaired t tests (D and E). These experiments were replicated three times.

Close modal

Having established a critical role for B cells in secondary expansion of TCM, we next investigated whether B cells were required for Ag presentation. We generated chimeric mice using bone marrow cells from B−/− and KbDb−/− mice to reconstitute lethally irradiated WT mice. In these chimeras, all B cells could only be derived from the KbDb−/− bone marrow whereas a large proportion of other APCs (from the B−/− bone marrow) had intact MHC class I molecules (Supplemental Fig. 2). Similarly prepared recipient mice reconstituted with WT and/or B−/− bone marrow were included as controls (Fig. 6C). Surprisingly, in contrast to the results in B−/− and B cell–depleted mice, the secondary expansion was not attenuated when the B cells were unable to present Ag on MHC class I (Fig. 6C), suggesting that B cells do not have a direct role as an APC.

The above observations prompted us to determine whether splenic DCs were responsible for Ag presentation. To address this possibility, we generated chimeric mice with bone marrow from CD11c-DTR transgenic mice as described by Zammit et al. (12). These mice tolerated multiple injections of DT to remove CD11c+ DCs (Supplemental Fig. 3). To ensure that CD8+ T cells were not affected by DT treatment, we transferred Ag-primed CD8+ T cells from WT congenic mice to CD11c-DTR chimeric mice prior to boosting with VSV. Fig. 6D shows that injections of DT diminished expansion of endogenous T cells (Thy1.1) in CD11c-DTR chimeric mice but not WT controls, suggesting that host DCs may mediate Ag presentation to trigger memory T cell proliferation. DT injections also reduced the secondary response of transferred WT CD8+ T cells (Thy1.1+), confirming the requirement of CD11c+ DCs instead of removal of certain CD8+ T cells that may also express CD11c (Fig. 6E) (33, 34).

Taken together, these results pointed to a possibility that infected B cells produced and transferred Ags to neighboring DCs, leading to Ag presentation to TCM that were also localized in the follicular region (20, 35). To confirm this possibility, we isolated B cells from KbDb−/− mice 17 h after i.v. inoculation with VSV-gp33 and coincubated them with CFSE-labeled P14 T cells in the presence or absence of bone marrow–derived DCs. Three days after coculture, proliferation of P14 T cells was detected only in the presence of DCs, suggesting that B cell–produced Ags were taken by DCs and presented to T cells (Fig. 7A, upper panel). To confirm this, we used Transwell inserts to separate infected KbDb−/− B cells (top of well) from P14 T cells and DCs (bottom of well). P14 cell proliferation was observed only when DCs were present in the lower compartment, confirming that Ag was transferred from B cells to DCs in a soluble form (Fig. 7A, lower panel). To determine whether infected B cells produce viral progeny that reinfect DCs, we coincubated B cells from VSV-gp33–exposed KbDb−/− mice with Vero cells that are highly sensitive to VSV replication. After 48 h, Vero cells coincubated with B cells (Fig. 7B, middle panel) remained intact (compared with Vero cells alone; Fig. 7B, left panel), whereas cytopathic effect was visibly evident by direct infection with VSV (Fig. 7B, right panel), suggesting that B cells neither permit productive viral replication nor carry viruses on their surface.

FIGURE 7.

Ags, in soluble form, can be transferred from infected splenic B cells to DCs to induce T cell proliferation. (A) KbDb−/− mice were inoculated i.v. with VSV-gp33 for 17 h and splenic B cells were purified. B cells were cultured with CFSE-labeled P14 cells in the presence or absence of bone marrow–derived DCs, either directly (upper panels, Transwell −) or separated by a Transwell (lower panels, Transwell +). Representative flow cytometry profiles of P14 cell proliferation at 72 h after incubation are shown. (B) Five million purified B cells, prepared as described above, were added to Vero cell monolayers and incubated for 48 h at 37°C (middle). Vero cells alone (left) or infected with VSV (right) were included as controls. These experiments were replicated twice. Original magnification ×50.

FIGURE 7.

Ags, in soluble form, can be transferred from infected splenic B cells to DCs to induce T cell proliferation. (A) KbDb−/− mice were inoculated i.v. with VSV-gp33 for 17 h and splenic B cells were purified. B cells were cultured with CFSE-labeled P14 cells in the presence or absence of bone marrow–derived DCs, either directly (upper panels, Transwell −) or separated by a Transwell (lower panels, Transwell +). Representative flow cytometry profiles of P14 cell proliferation at 72 h after incubation are shown. (B) Five million purified B cells, prepared as described above, were added to Vero cell monolayers and incubated for 48 h at 37°C (middle). Vero cells alone (left) or infected with VSV (right) were included as controls. These experiments were replicated twice. Original magnification ×50.

Close modal

TEFF-mediated elimination of Ag-laden DCs has been widely accepted as a homeostatic mechanism to prevent overexuberant activation of CD8+ T cells (5, 14, 28). We report in the present study, however, that i.v. delivery of rhabdoviral vectors could overcome this negative regulation and maximize expansion of CD8+ T cells, even at the peak of the primary response. A unique cooperation between splenic B cells and DCs for Ag presentation and their anatomic location (within the follicle), which is separated from TEFF but proximate to TCM, appears to be a mechanism that enables engagement of TCM during an ongoing effector phase in the presence of circulating viruses (i.e., viremia). Our data revealed a distinct role of splenic APCs during secondary T cell responses, which should be considered for the rational design of booster vaccines to rapidly and effectively expand Ag-specific CD8+ T cells. Our finding may also provide new fundamental mechanistic insight into the immune response to viremia where the primary CD8+ T cell response can be amplified to contend with an uncontrolled virus infection without compromising the regulatory feedback to DCs that limits further naive T cell recruitment and, presumably, minimizes inadvertent presentation of tissue Ags by migrating DCs.

Previous research has demonstrated that the secondary lymphoid organs are the primary sites of naive T cell priming following immunization or pathogen infection. Upon activation, TEFF and TEM enter the circulation and peripheral tissues, whereas TCM home to the secondary lymphoid organs (36). This differential distribution poses a potential barrier to interaction between Ag-laden DCs and TCM because cytolytic TEFF kill migratory DCs in peripheral tissues (13, 15). Moreover, a recent study has shown that TEFF can re-enter reactive lymph nodes and attenuate Ag presentation by killing newly arrived DCs and Ag-loaded residential DCs (37). These data clearly demonstrate that the efficacy of booster immunizations that rely on migratory DCs for Ag presentation will be limited by this TEFF-mediated negative feedback mechanism, especially during primary and chronic immune responses.

In the spleen, CD8+ TCM are located in the T cell zones of the white pulp surrounded by follicular B cells. Using TCR-transgenic CD8+ T cells, several groups have shown that after adoptive transfer to naive recipients, effector CD8+ T cells were only found in the red pulp, whereas memory CD8+ T cells homed to the B cell follicles (19, 30, 38). Subsequent studies confirmed that this localization pattern held true for endogenously activated CD8+ T cells. Lefrançois and colleagues (20) and Jacob and colleagues (21) showed that upon secondary exposure to LCMV or Listeria monocytogenes, memory CD8+ T cells rapidly expanded and mobilized from the B cell follicles to the red pulp via bridging channels. After the immune response subsided, memory CD8+ T cells homed back to the B cell follicles whereas effector cells remained in the splenic red pulp (20, 21). These results point to a possibility that follicular APCs were responsible for memory CD8+ T cell activation. Colocalization with TCM in the follicles confers upon follicular APCs anatomical advantages to interact with TCM while avoiding TEFF-mediated killing. In contrast, migratory DCs, including the CD8α+ subset, primarily reside in the red pulp, making them highly susceptible to killing by TEFF during secondary responses (39). Our results provide unequivocal evidence that this negative regulation can be bypassed when the Ag can be delivered into the follicular region. This discovery has important implications for optimizing therapeutic vaccination strategies where high numbers of TEFF are needed for protection prior to boosting as well as in circumstances where TEFF will inevitably be present for prolonged periods such as during chronic infections.

It was previously demonstrated that delivery of VSV into the flank leads to infection of the lymph nodes where most of the Ag is taken up by macrophages and DCs within the subcapsular sinus, so little of the virus gets into the B cell follicles (40). This may explain why immunization through discrete routes (e.g., s.c., i.m.), which result in Ag uptake through the lymphatics, are not effective routes for boosting. Additionally, the ability of VSV and Maraba virus to infect splenic B cells, especially follicular B cells, not only bypasses circulating TEFF but also increases Ag production due to their enormous number and subsequent Ag presentation by neighboring DCs to TCM, which are also located in the follicular region. Our in vitro analysis confirmed that infected B cells could transfer Ag to DCs without involving direct contact between the two cells although the exact form of the Ag remains to be determined. It was previously reported that B cells could carry viruses including VSV on their surface and transfer them to other cells (41, 42), but our results argue that the Ag is likely in a different soluble form.

Our data suggest a novel strategy for boosting immune responses in the presence of TEFF by delivering Ags to a location that is anatomically separated from the TEFF and in close proximity to TCM. These findings are supported by recent publications revealing differential distributions of TCM and TEFF and offer important insights into how to rapidly boost immunity without the need to compromise the TEFF population induced by primary immunization.

We thank Drs. R. Dunn and M. Kehry at Biogen Idec (San Diego, CA) for the gift of mAb against mouse CD20 (clone 18B12).

This work was supported by grants from the Canadian Institutes of Health Research and the Ontario Cancer Research Network (to Y.W.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

Ad

recombinant human adenovirus

DC

dendritic cell

DCT

dopachrome tautomerase

DT

diphtheria toxin

DTR

diphtheria toxin receptor

hDCT

human dopachrome tautomerase

LCMV

lymphocytic choriomeningitis virus

Luc

luciferase

SIIN

SIINFEKL

TCM

central memory T cell

TEFF

effector T cell

TEM

effector memory T cell

VSV

vesicular stomatitis virus

VV

vaccinia virus

WT

wild-type.

1
Woodland
D. L.
2004
.
Jump-starting the immune system: prime-boosting comes of age.
Trends Immunol.
25
:
98
104
.
2
Lanzavecchia
A.
,
Sallusto
F.
.
2005
.
Understanding the generation and function of memory T cell subsets.
Curr. Opin. Immunol.
17
:
326
332
.
3
Wong
P.
,
Lara-Tejero
M.
,
Ploss
A.
,
Leiner
I.
,
Pamer
E. G.
.
2004
.
Rapid development of T cell memory.
J. Immunol.
172
:
7239
7245
.
4
Mueller
S. N.
,
Jones
C. M.
,
Stock
A. T.
,
Suter
M.
,
Heath
W. R.
,
Carbone
F. R.
.
2006
.
CD4+ T cells can protect APC from CTL-mediated elimination.
J. Immunol.
176
:
7379
7384
.
5
Ludewig
B.
,
Bonilla
W. V.
,
Dumrese
T.
,
Odermatt
B.
,
Zinkernagel
R. M.
,
Hengartner
H.
.
2001
.
Perforin-independent regulation of dendritic cell homeostasis by CD8+ T cells in vivo: implications for adaptive immunotherapy.
Eur. J. Immunol.
31
:
1772
1779
.
6
Allan
R. S.
,
Waithman
J.
,
Bedoui
S.
,
Jones
C. M.
,
Villadangos
J. A.
,
Zhan
Y.
,
Lew
A. M.
,
Shortman
K.
,
Heath
W. R.
,
Carbone
F. R.
.
2006
.
Migratory dendritic cells transfer antigen to a lymph node-resident dendritic cell population for efficient CTL priming.
Immunity
25
:
153
162
.
7
Allenspach
E. J.
,
Lemos
M. P.
,
Porrett
P. M.
,
Turka
L. A.
,
Laufer
T. M.
.
2008
.
Migratory and lymphoid-resident dendritic cells cooperate to efficiently prime naive CD4 T cells.
Immunity
29
:
795
806
.
8
Lee
H. K.
,
Zamora
M.
,
Linehan
M. M.
,
Iijima
N.
,
Gonzalez
D.
,
Haberman
A.
,
Iwasaki
A.
.
2009
.
Differential roles of migratory and resident DCs in T cell priming after mucosal or skin HSV-1 infection.
J. Exp. Med.
206
:
359
370
.
9
Crowe
S. R.
,
Turner
S. J.
,
Miller
S. C.
,
Roberts
A. D.
,
Rappolo
R. A.
,
Doherty
P. C.
,
Ely
K. H.
,
Woodland
D. L.
.
2003
.
Differential antigen presentation regulates the changing patterns of CD8+ T cell immunodominance in primary and secondary influenza virus infections.
J. Exp. Med.
198
:
399
410
.
10
Byrne
J. A.
,
Butler
J. L.
,
Cooper
M. D.
.
1988
.
Differential activation requirements for virgin and memory T cells.
J. Immunol.
141
:
3249
3257
.
11
Belz
G. T.
,
Wilson
N. S.
,
Smith
C. M.
,
Mount
A. M.
,
Carbone
F. R.
,
Heath
W. R.
.
2006
.
Bone marrow-derived cells expand memory CD8+ T cells in response to viral infections of the lung and skin.
Eur. J. Immunol.
36
:
327
335
.
12
Zammit
D. J.
,
Cauley
L. S.
,
Pham
Q. M.
,
Lefrançois
L.
.
2005
.
Dendritic cells maximize the memory CD8 T cell response to infection.
Immunity
22
:
561
570
.
13
Luketic
L.
,
Delanghe
J.
,
Sobol
P. T.
,
Yang
P.
,
Frotten
E.
,
Mossman
K. L.
,
Gauldie
J.
,
Bramson
J.
,
Wan
Y.
.
2007
.
Antigen presentation by exosomes released from peptide-pulsed dendritic cells is not suppressed by the presence of active CTL.
J. Immunol.
179
:
5024
5032
.
14
Wong
P.
,
Pamer
E. G.
.
2003
.
Feedback regulation of pathogen-specific T cell priming.
Immunity
18
:
499
511
.
15
Yang
J.
,
Huck
S. P.
,
McHugh
R. S.
,
Hermans
I. F.
,
Ronchese
F.
.
2006
.
Perforin-dependent elimination of dendritic cells regulates the expansion of antigen-specific CD8+ T cells in vivo.
Proc. Natl. Acad. Sci. USA
103
:
147
152
.
16
Pol
J. G.
,
Zhang
L.
,
Bridle
B. W.
,
Stephenson
K. B.
,
Rességuier
J.
,
Hanson
S.
,
Chen
L.
,
Kazdhan
N.
,
Bramson
J. L.
,
Stojdl
D. F.
, et al
.
2014
.
Maraba virus as a potent oncolytic vaccine vector.
Mol. Ther.
22
:
420
429
.
17
Bridle
B. W.
,
Clouthier
D.
,
Zhang
L.
,
Pol
J.
,
Chen
L.
,
Lichty
B. D.
,
Bramson
J. L.
,
Wan
Y.
.
2013
.
Oncolytic vesicular stomatitis virus quantitatively and qualitatively improves primary CD8+ T-cell responses to anticancer vaccines.
OncoImmunology
2
:
e26013
.
18
Bridle
B. W.
,
Stephenson
K. B.
,
Boudreau
J. E.
,
Koshy
S.
,
Kazdhan
N.
,
Pullenayegum
E.
,
Brunellière
J.
,
Bramson
J. L.
,
Lichty
B. D.
,
Wan
Y.
.
2010
.
Potentiating cancer immunotherapy using an oncolytic virus.
Mol. Ther.
18
:
1430
1439
.
19
Weninger
W.
,
Crowley
M. A.
,
Manjunath
N.
,
von Andrian
U. H.
.
2001
.
Migratory properties of naive, effector, and memory CD8+ T cells.
J. Exp. Med.
194
:
953
966
.
20
Khanna
K. M.
,
McNamara
J. T.
,
Lefrançois
L.
.
2007
.
In situ imaging of the endogenous CD8 T cell response to infection.
Science
318
:
116
120
.
21
Dauner
J. G.
,
Williams
I. R.
,
Jacob
J.
.
2008
.
Differential microenvironment localization of effector and memory CD8 T cells.
J. Immunol.
180
:
291
299
.
22
Bridle
B. W.
,
Boudreau
J. E.
,
Lichty
B. D.
,
Brunellière
J.
,
Stephenson
K.
,
Koshy
S.
,
Bramson
J. L.
,
Wan
Y.
.
2009
.
Vesicular stomatitis virus as a novel cancer vaccine vector to prime antitumor immunity amenable to rapid boosting with adenovirus.
Mol. Ther.
17
:
1814
1821
.
23
Bassett
J. D.
,
Yang
T. C.
,
Bernard
D.
,
Millar
J. B.
,
Swift
S. L.
,
McGray
A. J.
,
VanSeggelen
H.
,
Boudreau
J. E.
,
Finn
J. D.
,
Parsons
R.
, et al
.
2011
.
CD8+ T-cell expansion and maintenance after recombinant adenovirus immunization rely upon cooperation between hematopoietic and nonhematopoietic antigen-presenting cells.
Blood
117
:
1146
1155
.
24
Brun
J.
,
McManus
D.
,
Lefebvre
C.
,
Hu
K.
,
Falls
T.
,
Atkins
H.
,
Bell
J. C.
,
McCart
J. A.
,
Mahoney
D.
,
Stojdl
D. F.
.
2010
.
Identification of genetically modified Maraba virus as an oncolytic rhabdovirus.
Mol. Ther.
18
:
1440
1449
.
25
Boudreau
J. E.
,
Stephenson
K. B.
,
Wang
F.
,
Ashkar
A. A.
,
Mossman
K. L.
,
Lenz
L. L.
,
Rosenthal
K. L.
,
Bramson
J. L.
,
Lichty
B. D.
,
Wan
Y.
.
2011
.
IL-15 and type I interferon are required for activation of tumoricidal NK cells by virus-infected dendritic cells.
Cancer Res.
71
:
2497
2506
.
26
Yang
T. C.
,
Dayball
K.
,
Wan
Y. H.
,
Bramson
J.
.
2003
.
Detailed analysis of the CD8+ T-cell response following adenovirus vaccination.
J. Virol.
77
:
13407
13411
.
27
Huster
K. M.
,
Busch
V.
,
Schiemann
M.
,
Linkemann
K.
,
Kerksiek
K. M.
,
Wagner
H.
,
Busch
D. H.
.
2004
.
Selective expression of IL-7 receptor on memory T cells identifies early CD40L-dependent generation of distinct CD8+ memory T cell subsets.
Proc. Natl. Acad. Sci. USA
101
:
5610
5615
.
28
Ronchese
F.
,
Hermans
I. F.
.
2001
.
Killing of dendritic cells: a life cut short or a purposeful death?
J. Exp. Med.
194
:
F23
F26
.
29
Oliver
A. M.
,
Martin
F.
,
Gartland
G. L.
,
Carter
R. H.
,
Kearney
J. F.
.
1997
.
Marginal zone B cells exhibit unique activation, proliferative and immunoglobulin secretory responses.
Eur. J. Immunol.
27
:
2366
2374
.
30
Potsch
C.
,
Vöhringer
D.
,
Pircher
H.
.
1999
.
Distinct migration patterns of naive and effector CD8 T cells in the spleen: correlation with CCR7 receptor expression and chemokine reactivity.
Eur. J. Immunol.
29
:
3562
3570
.
31
Kitamura
D.
,
Roes
J.
,
Kühn
R.
,
Rajewsky
K.
.
1991
.
A B cell-deficient mouse by targeted disruption of the membrane exon of the immunoglobulin μ chain gene.
Nature
350
:
423
426
.
32
Ngo
V. N.
,
Cornall
R. J.
,
Cyster
J. G.
.
2001
.
Splenic T zone development is B cell dependent.
J. Exp. Med.
194
:
1649
1660
.
33
Lin
Y.
,
Roberts
T. J.
,
Sriram
V.
,
Cho
S.
,
Brutkiewicz
R. R.
.
2003
.
Myeloid marker expression on antiviral CD8+ T cells following an acute virus infection.
Eur. J. Immunol.
33
:
2736
2743
.
34
Huleatt
J. W.
,
Lefrançois
L.
.
1995
.
Antigen-driven induction of CD11c on intestinal intraepithelial lymphocytes and CD8+ T cells in vivo.
J. Immunol.
154
:
5684
5693
.
35
Wakim
L. M.
,
Bevan
M. J.
.
2011
.
Cross-dressed dendritic cells drive memory CD8+ T-cell activation after viral infection.
Nature
471
:
629
632
.
36
Klonowski
K. D.
,
Marzo
A. L.
,
Williams
K. J.
,
Lee
S. J.
,
Pham
Q. M.
,
Lefrançois
L.
.
2006
.
CD8 T cell recall responses are regulated by the tissue tropism of the memory cell and pathogen.
J. Immunol.
177
:
6738
6746
.
37
Guarda
G.
,
Hons
M.
,
Soriano
S. F.
,
Huang
A. Y.
,
Polley
R.
,
Martín-Fontecha
A.
,
Stein
J. V.
,
Germain
R. N.
,
Lanzavecchia
A.
,
Sallusto
F.
.
2007
.
L-selectin-negative CCR7 effector and memory CD8+ T cells enter reactive lymph nodes and kill dendritic cells.
Nat. Immunol.
8
:
743
752
.
38
Jung
Y. W.
,
Rutishauser
R. L.
,
Joshi
N. S.
,
Haberman
A. M.
,
Kaech
S. M.
.
2010
.
Differential localization of effector and memory CD8 T cell subsets in lymphoid organs during acute viral infection.
J. Immunol.
185
:
5315
5325
.
39
Idoyaga
J.
,
Suda
N.
,
Suda
K.
,
Park
C. G.
,
Steinman
R. M.
.
2009
.
Antibody to Langerin/CD207 localizes large numbers of CD8α+ dendritic cells to the marginal zone of mouse spleen.
Proc. Natl. Acad. Sci. USA
106
:
1524
1529
.
40
Hickman
H. D.
,
Takeda
K.
,
Skon
C. N.
,
Murray
F. R.
,
Hensley
S. E.
,
Loomis
J.
,
Barber
G. N.
,
Bennink
J. R.
,
Yewdell
J. W.
.
2008
.
Direct priming of antiviral CD8+ T cells in the peripheral interfollicular region of lymph nodes.
Nat. Immunol.
9
:
155
165
.
41
Junt
T.
,
Moseman
E. A.
,
Iannacone
M.
,
Massberg
S.
,
Lang
P. A.
,
Boes
M.
,
Fink
K.
,
Henrickson
S. E.
,
Shayakhmetov
D. M.
,
Di Paolo
N. C.
, et al
.
2007
.
Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells.
Nature
450
:
110
114
.
42
Zhang
L.
,
Bridle
B. W.
,
Chen
L.
,
Pol
J.
,
Spaner
D.
,
Boudreau
J. E.
,
Rosen
A.
,
Bassett
J. D.
,
Lichty
B. D.
,
Bramson
J. L.
,
Wan
Y.
.
2013
.
Delivery of viral-vectored vaccines by B cells represents a novel strategy to accelerate CD8+ T-cell recall responses.
Blood
121
:
2432
2439
.

B.D.L. is a board member of Turnstone Biologics, which is developing Maraba virus as an oncolytic vaccine. The other authors have no financial conflicts of interest.

Supplementary data