Download Responses in Peripheral Lymph Nodes Cells and Naive T Cells

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Mitosis wikipedia , lookup

List of types of proteins wikipedia , lookup

Cellular differentiation wikipedia , lookup

Cell culture wikipedia , lookup

Tissue engineering wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

Cell encapsulation wikipedia , lookup

Amitosis wikipedia , lookup

Transcript
Limited Infiltration of Exogenous Dendritic
Cells and Naive T Cells Restricts Immune
Responses in Peripheral Lymph Nodes
This information is current as
of June 17, 2017.
Subscription
Permissions
Email Alerts
J Immunol 2006; 176:4535-4542; ;
doi: 10.4049/jimmunol.176.8.4535
http://www.jimmunol.org/content/176/8/4535
This article cites 39 articles, 20 of which you can access for free at:
http://www.jimmunol.org/content/176/8/4535.full#ref-list-1
Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscription
Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts
The Journal of Immunology is published twice each month by
The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2006 by The American Association of
Immunologists All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Downloaded from http://www.jimmunol.org/ by guest on June 17, 2017
References
David W. Mullins and Victor H. Engelhard
The Journal of Immunology
Limited Infiltration of Exogenous Dendritic Cells and Naive
T Cells Restricts Immune Responses in Peripheral Lymph
Nodes1
David W. Mullins and Victor H. Engelhard2
E
xogenous dendritic cells (DC)3 bearing specific Ag have
been evaluated as adjuvants for the induction of immune
responses. Exogenously administered DC can prime T
cells capable of recognizing and killing tumors in an Ag-specific
manner in animal models (1– 4), and have been subsequently incorporated into clinical trials for immunotherapy of cancers (5–
11). Unfortunately, clinical efficacy has been variable. It is clear
that DC are complex reagents, and that additional insight into the
factors that control their ability to induce effective immunity is
needed.
An important aspect of immunization using exogenous DC is
that their cellular nature and migratory phenotype restrict lymphoid distribution as compared with other types of soluble immunogens (12, 13). i.v.-injected DC efficiently enter spleen (13, 14),
but fail to cross high endothelial venules, and are excluded from
peripheral lymph nodes (LN) (13, 15). s.c. injected exogenous DC
efficiently access peripheral LN via afferent lymphatics in a CCR7dependent manner (16 –18). However, infiltration is restricted to
only those nodes in the injection site drainage (13, 15, 19 –21).
We have previously shown that the constrained distribution of
exogenous DC significantly impacts immunologic control of tumor
outgrowth (13). Control of s.c. growing tumor requires that DC
prime Ag-specific CD8 T cells in peripheral skin-draining LN. T
cells primed to the same Ag by DC that infiltrate the spleen are
capable of tumor recognition and lysis ex vivo and infiltration of
lung metastatic-like lesions in situ, yet are incapable of controlling
s.c. growing tumors. DC-based therapies may therefore lead to
restricted distributions of memory cells in different compartments
Department of Microbiology, Carter Immunology Center, and Human Immune Therapy Center, University of Virginia Health System, Charlottesville, VA 22908
Received for publication September 16, 2005. Accepted for publication January
10, 2006.
and/or the induction of restricted tissue-homing patterns on activated effectors. In addition, there is an apparent limitation on the
magnitude of immune response that can be induced in LN. We
observed steadily improving control of tumor in lung, associated
with splenic immune response, as the number of i.v. injected DC
was increased, up to at least 106 cells. In contrast, control of s.c.
growing melanoma could not be improved by increasing the number of s.c. injected DC above 104 cells. In this study, we examined
the quantitative relationship between LN infiltration by exogenous
DC and both the magnitude of immune response and control of
tumor outgrowth. Our data demonstrate that tumor control is
linked to the overall magnitude of the primary immune response,
which is directly correlated with, and constrained by, infiltration of
both DC and Ag-specific naive T cells into individual LN. We also
demonstrate strategies for circumventing both of these limitations
that are applicable in the further optimization of immunotherapeutic approaches for tumor treatment involving exogenous DC.
Materials and Methods
Animals
Transgenic mice on the C57BL/6 background expressing a chimeric MHC
class I composed of the ␣1 and ␣2 domains of HLA-A*0201 and the ␣3
domain of H2-Dd (AAD) have been described previously (22). C57BL/6
mice were obtained from Charles River Laboratories. Animals were maintained in pathogen-free facilities, and protocols were approved by the University of Virginia Institutional Animal Care and Use Committee.
Cell lines
Murine B16-F1 melanoma transfected to stably express genes for AAD and
for G418 resistance (B16-AAD) has been described previously (4). B16-F1
expressing a cytoplasmic-restricted construct of hen OVA (B16-cOVA)
was provided by Dr. T. N. J. Bullock (University of Virginia Health System, Charlottesville, VA).
Immunization with exogenous DC
Abbreviations used in this paper: DC, dendritic cell; LN, lymph node.
CD40L-activated bone marrow-derived DC were generated as described
previously (4, 23). Activated DC were CD70highCD80highCD86high and
produced IL-12 p70 (data not shown). DC were pulsed for 3 h with the
indicated peptides in the presence of human ␤2 microglobulin, as described
previously (13). Mice received DC in 200 ␮l of saline by s.c. injection into
the scapular fold or flank, as indicated, or i.v. injection into the dorsal tail
vein. For analysis of recall responses, DC-immunized mice received injections i.v. with 107 PFU of hTyrVac, a recombinant vaccinia virus encoding human tyrosinase (24).
Copyright © 2006 by The American Association of Immunologists, Inc.
0022-1767/06/$02.00
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
This work was supported by U.S. Public Health Service Grant CA78400 (to V.H.E.).
2
Address correspondence and reprint requests to Dr. Victor H. Engelhard, Carter
Immunology Center, University of Virginia, Box 801386, Charlottesville, VA 229081386. E-mail address: [email protected]
3
Downloaded from http://www.jimmunol.org/ by guest on June 17, 2017
Primary CD8 T cell responses in lymph nodes (LN) and protective immunological tumor control are quantitatively limited
following immunization with exogenous peptide-pulsed dendritic cells (DC). This arises from two constraints. First, LN are
saturated by relatively small quantities of exogenous DC. Second, circulation of new naive T cells into DC-infiltrated LN during
the functional lifespan of the DC is negligible. Limits on DC and T cellularity in, and flux through, LN constrain the magnitude
of both primary and subsequent recall responses. Enhanced immune responses and tumor control can be achieved using maneuvers to augment LN retention of DC or availability of naive T cells to Ag-presenting DC. These data offer an increased understanding of LN function in general and provide a practical basis for improvements in tumor immunotherapy. The Journal of
Immunology, 2006, 176: 4535– 4542.
4536
LIMITED INFILTRATION OF DC AND T CELLS RESTRICTS LN IMMUNITY
DC distribution in vivo
CD40L-activated DC were labeled with CFSE (Molecular Probes) or
SNARF-1 (Molecular Probes) and injected s.c. or i.v. At the indicated
times, cells from spleen and LN were collected and stained with antiCD11c (BD Pharmingen). The number of infiltrating DC
(CD11c⫹CFSEhigh or CD11c⫹ SNARF-1high cells) was quantified by flow
cytometry.
Ex vivo analysis of Ag-specific T cells
Lymphocytes were obtained from spleen and peripheral LN by homogenization. In some experiments, CD8⫹ T cells were enriched from spleens
and LN of immunized mice using a StemSep column and reagents (StemCell). Preparations were consistently 85–95% CD8⫹ as assessed by flow
cytometry. Total lymphocytes or enriched CD8⫹ T cells were directly assessed for Ag specificity by staining with anti-CD8 mAb (eBioscience) and
either HLA-A2-tetramer folded with Tyr369Y (tyrosinase369 –377) or
gp100209M peptides (National Institutes of Health Tetramer Core Facility,
Emory University Vaccine Center, Atlanta, GA) or Kb-tetramer folded
with OVA257–264.
Tumor induction and measurements
Statistics
Statistical comparisons were performed by Student’s t test using SigmaStat
3.01 software (Systat); all data sets were evaluated for normality of distribution before comparison by t test. Unless noted, data are presented as
means ⫾ SD of three replicates, and one experiment is shown from three
to six independent trials.
Results
Peripheral LN have a limited capacity to retain exogenous DC,
which limits primary and secondary immunity
Using mice expressing a recombinant human HLA-A*0201 class I
MHC molecule (referred to as AAD), we previously demonstrated
injection route-specific compartmentalization of immune responses using exogenous DC pulsed with the human melanoma
peptide Ag, Tyr369Y (13). Using this system, we examined the
association between the number of exogenous DC that infiltrated
lymphoid tissue and both the size of the primary immune response
and the extent of immunologic tumor control. We first injected
AAD⫹ mice with 105-activated DC by either i.v. or s.c. routes.
After i.v. injection, exogenous DC maximally infiltrated spleen
after 4 h (Fig. 1A), although only ⬃30% of the number injected
were recovered from this organ. However, exogenous DC were not
detected in any peripheral LN over at least 24 h (Fig. 1A and data
not shown). Using graded numbers of Tyr369Y-pulsed DC, the
number of exogenous DC that infiltrated spleen increased in proportion to the number injected over a range from 102 to 106 cells
(Fig. 1B). The number of infiltrating DC correlated directly over
this entire range with the size of the primary immune response,
measured as the percentage of CD8⫹ tetramer⫹ cells (Fig. 1B).
Both of these parameters also correlated with the extent of B16AAD melanoma tumor control in lung (Fig. 1B). We have previously shown this control to be dependent on splenic immunity
(13), and it likely reflects reactivation of memory T cells after
tumor injection. These results establish a direct correlation between the number of DC injected i.v. over a three-log range, the
number of splenic-infiltrating DC, and the size of primary and
recall CD8 T cell responses in that organ.
After s.c. injection, exogenous DC rapidly infiltrated the draining LN, achieving a maximal number within 30 min, and this value
was maintained for several hours (Fig. 1C). DC injected via this
route also infiltrated lungs (Fig. 1D) and spleen (Fig. 1C), and
Induction of immune responses in peripheral LN by exogenous
DC is not limited by competition for DC access or capacity to
support T cell activation
We also evaluated whether immune responses to exogenous DC in
individual LN were further limited by T cell competition for DC or
soluble factors within the LN microenvironment. To test these possibilities, we asked whether the magnitude of one Ag-specific
CD8⫹ T cell response was diminished by a simultaneous response
in the same LN, directed against a second Ag presented by the
same exogenous DC. AAD⫹ mice were immunized s.c. with 105
DC that had been pulsed with equimolar concentrations of either
Tyr369Y or gp100209M (a modified epitope from the melanocyte
differentiation protein gp100), or both Ags together. DC pulsed
with both peptides stimulated primary responses against each
epitope that were comparable to those observed using DC pulsed
with either peptide alone (Fig. 2, A and B). In addition, the outgrowth of B16-AAD tumors was delayed in animals immunized
with Tyr369Y- and gp100209M-copulsed DC as compared with single epitope-immunized littermates (Fig. 2C), demonstrating an additive secondary response. Median survival of dual epitope-immunized mice was also significantly increased by 17.8 ⫾ 2 days (␹2
test; p ⬍ 0.005) as compared with survival following single
epitope (Tyr369Y) immunization (Fig. 2D). Thus, the stimulation
of two distinct primary responses in the same peripheral LN did
not compromise the magnitude of either, nor did it limit the development of protective antitumor immunity. Collectively, these
results suggest that the primary immune response in peripheral LN
after exogenous DC immunization is not limited either by T cell
competition for DC, or by the availability of factors within the LN
microenvironment necessary for T cell activation other than the
number of DC.
Availability of naive Ag-specific T cells for activation by
exogenous DC also limits immune responses in peripheral LN
Because there appeared to be no intrinsic limit on T cell activation
within an individual LN, we next explored whether the immune
response was limited by the availability of naive T cells during the
time the DC were functional. To do this, we took advantage of the
fact that primary immune response induced by exogenous DC in
peripheral LN is limited to those nodes draining the injection site
Downloaded from http://www.jimmunol.org/ by guest on June 17, 2017
s.c. tumors or lung metastases were established by injection of 4 ⫻ 105
B16-AAD in 200 ␮l of saline via s.c. and i.v. routes, respectively (4). All
tumor measurements were performed in a blinded manner. Tumor cells
were 100% viable by trypan blue exclusion and ⬎98% AAD⫹ by flow
cytometric analysis on the day of injection.
achieved a maximal value in spleen only after 4 h. Again, however,
DC were not observed in peripheral LN noncontiguous to the injection site drainage at any time (Fig. 1C and data not shown),
consistent with entry of excess DC into circulation via efferent
lymphatics (25). DC infiltration of draining LN was very efficient:
s.c. delivery of as few as 104 DC led to ⬃3400 cells in the LN
draining the injection site. However, injecting as many as 106 DC
did not increase this number of infiltrated cells further at any time
up to 72 h (Fig. 1E and data not shown), although it did increase
the number of cells found in the spleen (Fig. 1F). As with i.v.
injection, the number of exogenous DC in spleen was directly proportional to the number injected over the entire range of 102–106
cells (Fig. 1F). Both the primary immune response in the draining
LN and the control of a s.c. growing solid tumor, which we previously showed to be dependent upon an immune response in peripheral LN (13), reached a plateau upon injection of as few as 104
DC (Fig. 1E). In contrast, the number of Ag-specific activated
CD8⫹ T cells in spleen and control of metastatic-like lesions in
lungs correlated directly to the number of DC that infiltrated this
organ after s.c. injection (Fig. 1F). These results demonstrate that
peripheral LN have a limited capacity to retain exogenous DC,
which limits the magnitude of primary and secondary immune responses in that compartment.
The Journal of Immunology
4537
(13). Therefore, we asked whether the immune response in one
DC-infiltrated node was limited by a simultaneous immune response occurring in a noncontiguous LN, because both would
compete for naive T cells from the recirculating pool. AAD⫹ mice
were immunized with a total of 105 Tyr369Y-pulsed DC as follows:
a single i.v. injection; a single s.c. injection; equally distributed
between two noncontiguous s.c. sites; or i.v. and a single s.c. site.
Although the number of DC injected into each of the two s.c. sites
(50,000) was only half as many as injected in a single-site schema,
it was still well above the number required to saturate the draining
LN (Fig. 1E).
Injection of DC at a single s.c. site, or equally distributed between i.v. and a single s.c. site, stimulated primary responses of
comparable magnitude in the LN draining the s.c. injection site
(Fig. 3A and data not shown). These primary immune responses in
peripheral LN were only detected following s.c. and not i.v. immunization. Furthermore, they were limited to the draining LN,
and not found in LN of two noncontiguous drainages. Importantly,
injection of DC into two noncontiguous s.c. sites resulted in primary responses of comparable magnitude in the LN draining both
injection sites, and the response in each LN was also comparable
to that observed in LN of mice receiving a single s.c. injection of
DC (Fig. 3A). Collectively, these data demonstrate that the immune responses induced in two individual LN by peptide-pulsed
DC do not influence one another, and demonstrate that the total
number of cognate naive cells available for activation is far greater
Downloaded from http://www.jimmunol.org/ by guest on June 17, 2017
FIGURE 1. Magnitude of primary immune response and immunologic control of tumor directly correlates with the ability of exogenous DC to be
retained in lymphoid organs. Exogenous bone marrow-derived DC were activated by coculture with CD40L-expressing National Institutes of Health-3T3
fibroblasts, pulsed with Tyr369Y peptide, and labeled with CFSE. Mice received injections i.v. in the lateral tail vein (A and B) or s.c. in the scapular fold
(C–F) with the indicated number of DC. A, Kinetics of exogenous DC infiltration of lymphoid compartments and lungs after i.v. injection of 105 DC. Organs
were harvested at the indicated times, and single-cell suspensions were prepared, stained with anti-CD11c, and analyzed by flow cytometry for
CFSE⫹CD11c⫹ cells. F, Spleen; E, lung; , axillary LN. B, Comparative analysis of the following: total exogenous DC present in spleen 4 h after i.v.
injection; primary Tyr369Y-specific immune response in spleen, measured ex vivo as CD8⫹ tetramer⫹ cells, 7 days after i.v. immunization (optimal time;
Ref. 39); and day 21 surface metastatic-like lung lesions in mice immunized i.v. with Tyr369Y-pulsed DC and challenged 3 wk later with 4 ⫻ 105 B16-AAD
tumors i.v. C, Kinetics of exogenous DC infiltration of lymphoid compartments after s.c. injection of 105 DC. F, Axillary LN; E, spleen; , inguinal LN.
D, Kinetics of exogenous DC infiltration of lungs after s.c. injection of 105 DC; data presented are from the experiment shown in C. E, Comparative analysis
of the following: total exogenous DC present in axillary LN 1 h after the s.c. injection site; primary Tyr369Y-specific immune response in injection
site-draining LN 7 days after s.c. immunization; and day 21 s.c. tumor size in mice immunized s.c. with Tyr369Y-pulsed DC, then challenged 3 wk later
with 4 ⫻ 105 B16-AAD tumors s.c. in the same anatomic location. F, Comparative analysis of the following: total exogenous DC present in spleen 4 h
after s.c. injection of DC; primary Tyr369Y-specific immune response in spleen 7 days after s.c. immunization; and day 21 surface metastatic-like lung
lesions in mice immunized s.c. with Tyr369Y-pulsed DC and challenged 3 wk later with 4 ⫻ 105 B16-AAD tumors i.v. In all panels, data are means ⫾
SDs. For DC infiltration and Ag-specific T cell assessment, n ⫽ 3 animals per treatment. For tumor outgrowth studies, n ⫽ 5 animals per treatment.
Representative data from one of four sets of similar experiments are shown.
4538
LIMITED INFILTRATION OF DC AND T CELLS RESTRICTS LN IMMUNITY
than the number activated in any individual DC-infiltrated node.
As a corollary, this result shows that the size of the primary immune response in an individual LN is limited by entry of cognate
naive T cells into it during the time that the exogenous DC remain
functional.
We next evaluated tumor control following induction of immune responses in single or multiple LN drainages. Interestingly,
mice immunized at two s.c. sites controlled s.c. growing tumor
significantly ( p ⬍ 0.001) better than those immunized with an
equivalent number of cells delivered at a single site (Fig. 3B). In
contrast, no difference was observed in control of lung metastatic
lesions following either single site or dual site s.c. immunization,
and neither of these immunization strategies was as effective as
injection solely by the i.v. route (Fig. 3C). This is in keeping with
our previous observation that control of lung metastases is dependent upon splenic immunity and occurs in proportion to the magnitude of the primary response (13).
The enhanced control of tumor following dual site s.c. injection
of DC was intriguing, given that s.c. growing tumors cause T cell
activation only in the draining LN (K. M. Hargadon and V. H.
Engelhard, unpublished observation). We hypothesized that this
enhanced control was due to induction of a larger number of memory T cells that were distributed among all peripheral LN. Therefore, we evaluated the location of early stage recall responses ac-
tivated by secondary immunization with tyrosinase-expressing
vaccinia virus (13). Secondary immunity was not confined to the
LN in which DC priming had occurred, but was also evident in
noncontiguous LN (Fig. 3C). This occurred only after primary immunization by s.c and not i.v. injection of exogenous DC, indicating that these recall responses were dependent upon memory T
cells that had been induced in LN and not in spleen. Most importantly, primary immunization by injection of DC into two noncontiguous lymphoid drainages led to an approximate doubling of the
recall response in a third noncontiguous drainage, when compared
with the response in mice immunized at a single s.c. site. These
results suggest that, as with naive T cells, the recall response is
limited by the number of memory T cells that reside in the draining
LN. In the context of exogenous DC immunization, this number of
memory cells is proportional to the size of the preceding primary
response, which is in turn restricted by the small capacities of the
priming LN for both exogenous DC and naive Ag-specific T cells.
LN conditioning enhances DC infiltration and magnitude of
immune responses in individual LN
The previous data demonstrated that recall immunity in individual
draining LN could be enhanced by increasing the number of systemically distributed memory T cells that had been activated in
multiple drainages. The functional characteristics of these memory
Downloaded from http://www.jimmunol.org/ by guest on June 17, 2017
FIGURE 2. T cell competition
does not limit activation in individual
LN following immunization with exogenous DC. Mice were immunized
s.c. with 105 DC that had been pulsed
with either Tyr369Y, gp100209M, or
an equimolar mixture of both peptides. A, Primary (day 7) immune responses to Tyr369Y and gp100209M
in axillary LN determined by costaining with specific HLA-A2-tetramer
reagents and anti-CD8␣. Numbers in
plots indicate percentage of tetramer⫹ CD8⫹ cells in the lymphocyte forward scatter/side scatter gate.
B, Total Ag-specific CD8⫹ cells in
immunization site-draining LN, calculated by multiplying the percentages of tetramer⫹ CD8⫹ lymphocytes by the total number of
lymphocytes isolated from each LN.
Data are mean of five separate animals ⫾ SD. Data from one of three
similar experiments are shown. C,
Outgrowth of melanoma and (D)
Kaplan-Meier survival data in mice
challenged with 4 ⫻ 105 B16-AAD 21
days after immunization. F, Untreated;
E, unpulsed DC; , Tyr369Y-pulsed
DC; ƒ, gp100209-pulsed DC; f, Agcopulsed DC. Data represent 10 animals per experimental grouping.
The Journal of Immunology
4539
nitude of primary immune response in spleen was unaffected by
conditioning (Fig. 4D). Furthermore, the phenotype of injected DC
recovered from the conditioned node remained unchanged (data
not shown); in combination with equivalent function in spleen,
these observations discount the possibility that enhanced primary
responses in the draining LN result from changes in the functionality of the Ag-bearing DC introduced into the conditioned LN.
Thus, conditioning can mitigate the constraints on immune responses in individual LN imposed by limited retention of Ag-specific DC and influx of naive T cells.
Discussion
cells are expected to depend on their initial activation site, and may
limit their homing or effectiveness after reactivation. Therefore, we
were also interested in whether immunity in individual draining
LN could be enhanced locally. It was recently demonstrated that
the capacity of individual LN to attract and retain CCR7⫹ cells is
significantly enhanced following conditioning with either inflammatory cytokines or DC themselves (26). Therefore, we hypothesized that conditioning of LN by injection of unpulsed exogenous
DC before injecting Ag-expressing DC would enhance primary
responses in the draining LN and subsequent tumor control. In
animals pretreated with unpulsed DC 6 h before immunization, we
observed a significant increase in Ag-bearing DC in the draining
LN (Fig. 4A). Although there was no increase in T cell number at
this time, there was a ⬃20% increase by 12 h after injection of the
conditioning DC (Fig. 4B) and in total cellularity of the conditioned LN but not spleen or distal LN (Fig. 4C). The magnitude of
the primary immune response (Fig. 4D) and subsequent control of
s.c. growing melanoma (Fig. 4E) was also significantly enhanced
by conditioning with unpulsed activated DC. Whereas retention of
Ag-bearing DC by the pretreated LN was significantly enhanced,
the majority of the 105-injected DC would likely bypass LN regardless of conditioning; consistent with this expectation, the mag-
Downloaded from http://www.jimmunol.org/ by guest on June 17, 2017
FIGURE 3. Availability of naive Ag-specific T cells for activation by
exogenous DC limits immune responses in peripheral LN. Mice were immunized with a total of 105 Tyr369Y-pulsed DC distributed as follows: a
single i.v. injection; a single s.c. site (Drainage 1); two noncontiguous s.c.
sites (Drainages 1 and 2 for injection sites 1 and 2, respectively); or i.v. and
a single s.c. site (Drainage 1). A, Primary immune responses in draining
and nondraining LN 7 days after DC immunization by different routes. B,
Control of melanoma growing s.c. or in lung, 21 and 15 days after injection
of tumor, respectively. p values indicate statistical significance compared
with unimmunized animals (f), and bracketed p value indicates statistical
significance between indicated groups. C, Recall immune responses, 4 days
after systemic challenge with tyrosinase-expressing vaccinia virus, in LN
that drained or did not drain the primary DC immunization sites. For A and
C, data are mean of three separate animals ⫾ SD; for B, data are mean of
five separate animals ⫾ SD. Data from one of three similar experiments are
shown.
In this study, we explored the basis for the limitation on primary
immune responses and secondary antitumor immunity following
s.c. immunization with peptide-pulsed DC. We have demonstrated
that this limitation results from constrained infiltration of peripheral LN by exogenous DC and limited availability of Ag-specific
naive T cells to the DC during the initiation of the primary response. The limited primary response in turn determines the size of
the recall response in tumor-draining LN. To our knowledge, this
is the first demonstration of a direct relationship between exogenous DC infiltration of a lymphoid compartment and the magnitude of both primary and recall immunity. We also define maneuvers that circumvent these limitations, leading to enhanced primary
and recall immune responses and prolonged control of tumor
outgrowth.
The factors that limit the infiltration of exogenous-activated DC
into draining LN are unknown. The observation of a substantial
portion of s.c. injected DC in spleen that is directly correlated with
the number injected demonstrates that LN infiltration is not limited
by sequestration of cells at the injection site. DC enter LN exclusively via afferent lymphatics, emptying into the subcapsular sinus
before at least a portion of the cells percolate into the T cell zones
of the node (25). DC infiltration of LN may be limited by saturation of the conduits through which DC transit from the subcapsular
sinus into the T cell areas of the LN. Alternatively, the T cell area
of LN may accept only a limited number of exogenous cells before
either no more cells are admitted or some infiltrating cells
emigrate.
In contrast to the LN, we observed no limitation on DC infiltration into spleen over the range of DC numbers evaluated. The
direct relationship between the number of infiltrating exogenous
DC and the magnitude of the splenic primary response discounts
the possibility that the high capacity of the spleen reflects simply
circulating or red pulp-resident cells. Furthermore, even though
circulating mature endogenous DC are rare (27, 28), activated exogenous DC efficiently enter splenic white pulp directly from circulation after i.v. injection (D. W. Mullins, unpublished observation). However, we reason that injection of exogenous DC in
sufficient number would in fact lead to saturation of spleen. Using
total T cell counts from spleen and axillary LN as a reference, we
might expect splenic retention of exogenous DC to be ⬃35-fold
greater than LN (⬃120,000 cells). In our studies, injection of 106
DC resulted in ⬃105-infiltrating DC in spleen. Although we have
not achieved a saturation in the spleen, our data demonstrate that
this compartment is infiltrated by exogenous DC in numbers significantly greater than observed in individual peripheral LN.
Two previous studies have examined the number of DC-infiltrating popliteal LN after injection in the footpad (19, 26). They
failed to find a saturation limit on infiltration into this compartment, despite injection of up to 3-fold more DC than used in our
study. On the contrary, they observed a positive cooperative relationship in which the fraction of injected DC retained in the LN
increased as the number of injected cells was increased. However,
4540
LIMITED INFILTRATION OF DC AND T CELLS RESTRICTS LN IMMUNITY
they also observed significantly slower migration to the draining
LN: whereas we observed maximal DC infiltration into the axillary
LN within ⬃30 min after s.c. injection, maximal numbers of DC
in popliteal LN are not achieved until 2– 4 days (19, 26). Therefore, we suggest that injection into the footpad results in initial
sequestration and slow efflux of DC from this tissue, such that DC
that enter the lymphatics and LN at early times lead to conditioning and acceptance of a larger number of later immigrants. Likewise, tissue retention of DC at the site of intradermal injection may
account for the reported increase in LN infiltration by DC injected
intradermally as compared with s.c (29). Alternatively, the capacity of different LN for DC retention may be significantly different,
although we have observed infiltration limits in inguinal LN comparable to those reported for axillary LN (our unpublished observation). Therefore, without prior conditioning, infiltration of exogenous DC into peripheral LN is strictly limited, and conditioning
of draining lymphatics with mature DC significantly enhances DC
infiltration and subsequent immune responses.
We also demonstrated that restricted availability of naive T cells
in individual LN constrained immune responses. Although naive T
cells circulate widely throughout secondary lymphoid compartments (30), their residence time in individual LN has been esti-
mated to be in the range of hours to days (31, 32). Indeed, we
found that the size of the immune response in one LN was not
affected by a response against the same Ag occurring in a second.
Thus, these two populations are essentially independent of one
another, and each represents a small portion of the repertoire for
that Ag available in the individual animal. This limitation may be
further constrained by the transience of the DC themselves. The
capacity of exogenous DC to induce full T cell activation, both in
vitro and in vivo, has been shown to persist for only a few hours
following maturation (33). The DC used in our studies were activated in vitro before injection and may therefore retain the ability
to induce in situ primary immune responses for only a limited time.
Therefore, only T cells that engage Ag-presenting exogenous DC
shortly following immunization would achieve full activation.
Given these limitations, together with the large capacity of
spleen to retain DC and support activation of tumor Ag-specific T
cells, it is reasonable to suggest that i.v. immunization with DC
should be used in lieu of other routes. However, we have previously shown that cells activated in this compartment are unable to
control the subsequent outgrowth of s.c. tumors (13). Instead, this
control is mediated by cells in peripheral LN. In this study, we
show that after a localized primary response to immunization with
Downloaded from http://www.jimmunol.org/ by guest on June 17, 2017
FIGURE 4. Immune responses in LN are enhanced by conditioning with unpulsed DC. A, Mice were pretreated by s.c. injection of saline or 105-activated
DC. Six hours later, mice were immunized with 105 SNARF-labeled, Ag-pulsed activated DC. Panels show infiltration of exogenous CD40L-activated DC
1 h after injection into control (saline) and DC-preconditioned (⫺6 h or ⫺12 h) LN. Number in panel represents the percentage of SNARF⫹ cells in the
CD11c⫹ compartment. Representative data from one of three similar experiments are shown. B, Distribution of CD4 and CD8 T cells in the nonconditioned
(saline) and DC-preconditioned (⫺6 h or ⫺12 h) LN, 1 h after injection of labeled DC. Data presented are from the experiment shown in A. C, Total
cellularity cells in the draining LN (axillary), noncontiguous LN (inguinal), and spleen 1 h after injection of labeled DC, without or with pretreatment using
saline or s.c. injected activated DC (⫺12 h). D, Mice were pretreated by s.c. injection of saline or 105-activated DC or were left untreated. Six hours later,
some mice from each pretreatment cohort received injections with saline or 105 OVA257–266-pulsed DC. Data are percentage of tetramer⫹ CD8⫹ T cells
in draining (axillary) LN or spleen 7 days after immunization. Data represent mean ⫾ SD of three separate animals in a single experiment. E, Size of s.c.
growing melanoma 21 days after immunization. Data are mean ⫾ SD for five animals in each group, and representative data from one of three similar
experiments are shown.
The Journal of Immunology
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
Acknowledgments
We thank Dr. Craig L. Slingluff, Jr. for insightful comments and critically
reading this manuscript.
Disclosures
25.
26.
The authors have no financial conflict of interest.
References
1. Zitvogel, L., J. I. Mayordomo, T. Tjandrawn, A. B. DeLeo, M. R. Clarke,
M. T. Lotze, and W. J. Storkus. 1995. Therapy of murine tumors with peptide-
27.
pulsed dendritic cells: dependence of T cells, B7 costimulation, and T helper cell
1-associated cytokines. J. Exp. Med. 183: 87–97.
Hanson, H. L., S. S. Kang, L. A. Norian, K. Matsui, L. A. O’Mara, and
P. M. Allen. 2004. CD4-directed peptide vaccination augments an antitumor response, but efficacy is limited by the number of CD8⫹ T cell precursors. J. Immunol. 172: 4215– 4224.
Paglia, P., C. Chiodoni, M. Rodolfo, and M. P. Colombo. 1996. Murine dendritic
cells loaded in vitro with soluble protein prime cytotoxic T lymphocytes against
tumor antigen in vivo. J. Exp. Med. 183: 317–322.
Mullins, D. W., T. N. Bullock, T. A. Colella, V. V. Robila, and V. H. Engelhard.
2001. Immune responses to the HLA-A*0201-restricted epitopes of tyrosinase
and glycoprotein 100 enable control of melanoma outgrowth in HLA-A*0201transgenic mice. J. Immunol. 167: 4853– 4860.
Nestle, F. O., S. Alijagic, M. Gilliet, Y. Sun, S. Grabbe, R. Dummer, G. Burg, and
D. Schadendorf. 1998. Vaccination of melanoma patients with peptide- or tumor
lysate-pulsed dendritic cells. Nat. Med. 4: 328 –332.
Thurner, B., I. Haendle, C. Roder, D. Dieckmann, P. Keikavoussi, H. Jonuleit,
A. Bender, C. Maczek, D. Schreiner, D. P. von den, et al. 1999. Vaccination with
mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced
stage IV melanoma. J. Exp. Med. 190: 1669 –1678.
Dallal, R. M., and M. T. Lotze. 2000. The dendritic cell and human cancer
vaccines. Curr. Opin. Immunol. 12: 583–588.
Banchereau, J., B. Schuler-Thurner, A. K. Palucka, and G. Schuler. 2001. Dendritic cells as vectors for therapy. Cell 106: 271–274.
Gunzer, M., and S. Grabbe. 2001. Dendritic cells in cancer immunotherapy. Crit.
Rev. Immunol. 21: 133–145.
Steinman, R. M., and M. Pope. 2002. Exploiting dendritic cells to improve vaccine efficacy. J. Clin. Invest. 109: 1519 –1526.
Figdor, C. G., I. J. de Vries, W. J. Lesterhuis, and C. J. Melief. 2004. Dendritic
cell immunotherapy: mapping the way. Nat. Med. 10: 475– 480.
Fong, L., D. Brockstedt, C. Benike, L. Wu, and E. G. Engleman. 2001. Dendritic
cells injected via different routes induce immunity in cancer patients. J. Immunol.
166: 4254 – 4259.
Mullins, D. W., S. L. Sheasley, R. M. Ream, T. N. Bullock, Y. X. Fu, and
V. H. Engelhard. 2003. Route of immunization with peptide-pulsed dendritic
cells controls the distribution of memory and effector T cells in lymphoid tissues
and determines the pattern of regional tumor control. J. Exp. Med. 198:
1023–1034.
Austyn, J. M., J. W. Kupiec-Weglinski, D. F. Hankins, and P. J. Morris. 1988.
Migration patterns of dendritic cells in the mouse: homing to T cell-dependent
areas of spleen, and binding within marginal zone. J. Exp. Med. 167: 646 – 651.
Eggert, A. A., M. W. Schreurs, O. C. Boerman, W. J. Oyen, A. J. de Boer,
C. J. Punt, C. G. Figdor, and G. J. Adema. 1999. Biodistribution and vaccine
efficiency of murine dendritic cells are dependent on the route of administration.
Cancer Res. 59: 3340 –3345.
Ohl, L., M. Mohaupt, N. Czeloth, G. Hintzen, Z. Kiafard, J. Zwirner,
T. Blankenstein, G. Henning, and R. Forster. 2004. CCR7 governs skin dendritic
cell migration under inflammatory and steady-state conditions. Immunity 21:
279 –288.
Yoneyama, H., K. Matsuno, and K. Matsushimaa. 2005. Migration of dendritic
cells. Int. J. Hematol. 81: 204 –207.
Bousso, P., and E. Robey. 2003. Dynamics of CD8⫹ T cell priming by dendritic
cells in intact lymph nodes. Nat. Immunol. 4: 579 –585.
Lappin, M. B., J. M. Weiss, V. Delattre, B. Mai, H. Dittmar, C. Maier, K. Manke,
S. Grabbe, S. Martin, and J. C. Simon. 1999. Analysis of mouse dendritic cell
migration in vivo upon subcutaneous and intravenous injection. Immunology 98:
181–188.
Lambert, L. A., G. R. Gibson, M. Maloney, B. Durell, R. J. Noelle, and
R. J. Barth, Jr. 2001. Intranodal immunization with tumor lysate-pulsed dendritic
cells enhances protective antitumor immunity. Cancer Res. 61: 641– 646.
Dudda, J. C., J. C. Simon, and S. Martin. 2004. Dendritic cell immunization route
determines CD8⫹ T cell trafficking to inflamed skin: role for tissue microenvironment and dendritic cells in establishment of T cell-homing subsets. J. Immunol. 172: 857– 863.
Ho, S. N., R. T. Abraham, A. Nilson, B. S. Handwerger, and D. J. McKean. 1987.
Interleukin 1-mediated activation of interleukin 4 (IL-4)-producing T lymphocytes: proliferation by IL-4-independent mechanisms. J. Immunol. 139:
1532–1540.
Bullock, T. N. J., D. W. Mullins, T. A. Colella, and V. H. Engelhard. 2001.
Manipulation of avidity to improve effectiveness of adoptively transferred CD8⫹
T cells for melanoma immunotherapy in human MHC class I-transgenic mice.
J. Immunol. 167: 5824 –5831.
Overwijk, W. W., A. Tsung, K. R. Irvine, M. R. Parkhurst, T. J. Goletz, K. Tsung,
M. W. Carroll, C. Liu, B. Moss, S. A. Rosenberg, and N. P. Restifo. 1998.
gp100/pmel 17 is a murine tumor rejection antigen: induction of “self”- reactive,
tumoricidal T cells using high-affinity, altered peptide ligand. J. Exp. Med. 188:
277–286.
Randolph, G. J., V. Angeli, and M. A. Swartz. 2005. Dendritic-cell trafficking to
lymph nodes through lymphatic vessels. Nat. Rev. Immunol. 5: 617– 628.
Martı́n-Fontecha, A., S. Sebastiani, U. E. Hopken, M. Uguccioni, M. Lipp,
A. Lanzavecchia, and F. Sallusto. 2003. Regulation of dendritic cell migration to
the draining lymph node: impact on T lymphocyte traffic and priming. J. Exp.
Med. 198: 615– 621.
McKenna, H. J. 1999. Generating a T cell tumor-specific immune response in
vivo: can flt3-ligand-generated dendritic cells tip the balance? Cancer Immunol.
Immunother. 48: 281–286.
Downloaded from http://www.jimmunol.org/ by guest on June 17, 2017
exogenous DC, recall responses are distributed to multiple peripheral LN. Furthermore, the size of this distributed response is dependent on the size of primary responses occurring in one or more
LN, and independent of the size of the splenic response. Our data
suggest that these responses are based on memory cells resident in
these nodes in advance of the response. We have also noted enhanced recall responses in the LN in which initial priming occurred, suggesting either that memory cells preferentially migrate
into specific LN or that some memory cells are retained in the
original priming compartment. Data consistent with the former
possibility have been reported in an adoptive transfer model (34).
Finally, there is mounting evidence that activation of T cells in
different LN compartments leads distinct tissue-homing characteristics (35–38). Therefore, targeted delivery of Ag-bearing DC to
specific LN will be crucial for the induction of tumor Ag-specific
memory and memory/effector T cells with appropriate migratory
capabilities to enter lymphatic compartments where tumor Ag is
presented and to infiltrate tumor itself upon reactivation,
respectively.
Because of the limited ability of DC to engage naive T cells in
peripheral LN, our work also offers practical insights to enhance
the use of DC as immunotherapeutic reagents. First, conditioning
of draining lymphatics by pretreatment with unpulsed, activated
DC leads to enhanced primary immune responses and antitumor
immunity. Our data are consistent with earlier work demonstrating
that conditioning enhanced the magnitude of primary responses by
increasing the number of Ag-bearing DC in the LN as well as the
subsequent flux of naive T cells (26). However, we have extended
it to demonstrate that this also leads to enhanced secondary immunity and associated tumor control. Second, tumor immunity can
be enhanced by injection of DC simultaneously into multiple noncontiguous LN. In this case, the total number of DC and T cells in
any individual LN remains unchanged. However, the total number
of LN-infiltrating DC is increased, allowing greater access to the
pool of naive T cells resident in, or circulating through, separate
LN compartments. This leads to enhanced primary in local LN, but
also a larger secondary immune response that is distributed more
systemically among other peripheral LN. Finally, tumor control
can be enhanced by immunization with DC presenting two different peptide Ag. We find no evidence of competition among T cells
within individual LN, and the total number of tumor-specific effector or memory cells is enhanced without altering the number of
infiltrating DC of T cells. Each of these maneuvers can be readily
incorporated into clinical trial protocols that use DC for tumor
immunotherapy.
In summary, our results provide strong evidence that the immune response in individual LN is limited by the availability of
both exogenous DC and Ag-specific T cells. These limitations can
be circumvented by distribution of exogenous DC into multiple
LN compartments, induction of multiple T cell specificities in each
LN compartment, and conditioning of LN to support the infiltration and/or retention of exogenous DC. These data offer an increased understanding of LN function in general and provide a
practical basis for improvements in tumor immunotherapy.
4541
4542
LIMITED INFILTRATION OF DC AND T CELLS RESTRICTS LN IMMUNITY
28. Marroquin, C. E., J. A. Westwood, R. Lapointe, A. Mixon, J. R. Wunderlich,
D. Caron, S. A. Rosenberg, and P. Hwu. 2002. Mobilization of dendritic cell
precursors in patients with cancer by flt3 ligand allows the generation of higher
yields of cultured dendritic cells. J. Immunother. 25: 278 –288.
29. Ridolfi, R., A. Riccobon, R. Galassi, G. Giorgetti, M. Petrini, L. Fiammenghi,
M. Stefanelli, L. Ridolfi, A. Moretti, G. Migliori, and G. Fiorentini. 2004. Evaluation of in vivo labelled dendritic cell migration in cancer patients. J. Transl.
Med. 2: 27.
30. Butcher, E. C., and L. J. Picker. 1996. Lymphocyte homing and homeostasis.
Science 272: 60 – 66.
31. Young, A. J. 1999. The physiology of lymphocyte migration through the single
lymph node in vivo. Semin. Immunol. 11: 73– 83.
32. Iparraguirre, A., and W. Weninger. 2003. Visualizing T cell migration in vivo.
Int. Arch. Allergy Immunol. 132: 277–293.
33. Hugues, S., L. Fetler, L. Bonifaz, J. Helft, F. Amblard, and S. Amigorena. 2004.
Distinct T cell dynamics in lymph nodes during the induction of tolerance and
immunity. Nat. Immunol. 5: 1235–1242.
34. Masopust, D., V. Vezys, E. J. Usherwood, L. S. Cauley, S. Olson, A. L. Marzo,
R. L. Ward, D. L. Woodland, and L. Lefrancois. 2004. Activated primary and
memory CD8 T cells migrate to nonlymphoid tissues regardless of site of activation or tissue of origin. J. Immunol. 172: 4875– 4882.
35. Xu, B., N. Wagner, L. N. Pham, V. Magno, Z. Shan, E. C. Butcher, and
S. A. Michie. 2003. Lymphocyte homing to bronchus-associated lymphoid tissue
(BALT) is mediated by L-selectin/PNAd, ␣4␤1 integrin/VCAM-1, and LFA-1
adhesion pathways. J. Exp. Med. 197: 1255–1267.
36. Nakajima, H., H. Sano, T. Nishimura, S. Yoshida, and I. Iwamoto. 1994. Role of
vascular cell adhesion molecule 1/very late activation antigen 4 and intercellular
adhesion molecule 1/lymphocyte function-associated antigen 1 interactions in
antigen-induced eosinophil and T cell recruitment into the tissue. J. Exp. Med.
179: 1145–1154.
37. Chin, J. E., C. A. Hatfield, G. E. Winterrowd, J. R. Brashler, S. L. Vonderfecht,
S. F. Fidler, R. L. Griffin, K. P. Kolbasa, R. F. Krzesicki, L. M. Sly, et al. 1997.
Airway recruitment of leukocytes in mice is dependent on ␣4-integrins and vascular cell adhesion molecule-1. Am. J. Physiol. 272: L219 –L229.
38. Wolber, F. M., J. L. Curtis, P. Maly, R. J. Kelly, P. Smith, T. A. Yednock,
J. B. Lowe, and L. M. Stoolman. 1998. Endothelial selectins and ␣4 integrins
regulate independent pathways of T lymphocyte recruitment in the pulmonary
immune response. J. Immunol. 161: 4396 – 4403.
39. Bullock, T. N., T. A. Colella, and V. H. Engelhard. 2000. The density of peptides
displayed by dendritic cells affects immune responses to human tyrosinase and
gp100 in HLA-A2 transgenic mice. J. Immunol. 164: 2354 –2361.
Downloaded from http://www.jimmunol.org/ by guest on June 17, 2017