Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Please cite this article in press as: Duperret et al.

, Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

Original Article

Synergy of Immune Checkpoint Blockade


with a Novel Synthetic Consensus
DNA Vaccine Targeting TERT
Elizabeth K. Duperret,1 Megan C. Wise,2,3 Aspen Trautz,1 Daniel O. Villarreal,3 Bernadette Ferraro,2 Jewell Walters,2
Jian Yan,2 Amir Khan,2 Emma Masteller,2 Laurent Humeau,2 and David B. Weiner1
1Vaccine Center, The Wistar Institute, Philadelphia, PA 19104, USA; 2Inovio Pharmaceuticals, Inc., Plymouth Meeting, PA 19462, USA; 3University of Pennsylvania,
Philadelphia, PA 19104, USA

Immune checkpoint blockade antibodies are setting a new ing these molecules have been shown to augment the effector activity
standard of care for cancer patients. It is therefore important of tumor-specific T cells and additionally inhibit regulatory T cell
to assess any new immune-based therapies in the context of (Treg) activity and reduce tumor burden in preclinical models and/or
immune checkpoint blockade. Here, we evaluate the impact in clinical trials as mono-therapies.3–5
of combining a synthetic consensus TERT DNA vaccine that
has improved capacity to break tolerance with immune check- In particular, blockade of the immune inhibitory checkpoints PD-1
point inhibitors. We observed that blockade of CTLA-4 or, to or CTLA-4 has shown promising results in the clinic for dozens of
a lesser extent, PD-1 synergized with TERT vaccine, gener- tumor types, and PD-1 blockade has become a standard of care for
ating more robust anti-tumor activity compared to check- melanoma and non-small-cell lung cancer.6–8 However, response
point alone or vaccine alone. Despite this anti-tumor synergy, rates to these mono-therapies are relatively low (33.7% response
none of these immune checkpoint therapies showed improve- for pembrolizumab [a-PD-1] and 11.9% response for ipilimumab
ment in TERT antigen-specific immune responses in tumor- [a-CTLA-4] in melanoma patients), leaving room for improve-
bearing mice. aCTLA-4 therapy enhanced the frequency of ment.9 The lack of response for the majority of such patients may
T-bet+/CD44+ effector CD8+ T cells within the tumor and be due to a lack of pre-existing tumor-associated T cell responses.
decreased the frequency of regulatory T cells within the tu- Strategies to combine PD-1 or CTLA-4 blockade with therapies
mor, but not in peripheral blood. CTLA-4 blockade syner- that prime T cells, such as radiation or irradiated cell-based vac-
gized more than Treg depletion with TERT DNA vaccine, cines, have shown improvements in pre-clinical models or in clin-
suggesting that the effect of CTLA-4 blockade is more likely ical trials.5,10–17 These approaches, however, may rely on T cell
due to the expansion of effector T cells in the tumor rather responses to neo-antigens within the tumor.18,19 Enhanced T cell
than a reduction in the frequency of Tregs. These results sug- priming by vaccines may therefore be required to break tolerance
gest that immune checkpoint inhibitors function to alter the to self-antigens for patients with poor response to immune check-
immune regulatory environment to synergize with DNA vac- point blockade. However, the mechanistic interactions between
cines, rather than boosting antigen-specific responses at the immune checkpoint blockade antibodies and vaccines are poorly
site of vaccination. understood.

INTRODUCTION Therapeutic peptide or DNA vaccination represents a more targeted


The magnitude of an adaptive immune response to a foreign antigen approach for directing T cells toward specific, less variable tumor-
is determined not only by the strength of interaction between the ma- associated antigens (TAAs). In mouse models, peptide vaccines
jor histocompatibility complex (MHC) and the T cell receptor (TCR) have been shown to synergize with immune checkpoint blockade.20,21
but also by co-stimulation at the immunological synapse.1 Without However, peptide vaccines are histocompatibility leukocyte antigen
co-stimulation, T cells will fail to initiate an effective immune (HLA) restricted and therefore cannot be used for all patients. Unlike
response. Additional co-stimulatory molecules or immune check- peptide vaccines, synthetic DNA (synDNA) vaccines are not HLA
point molecules exist to control the amplitude of T cell activation restricted, are robustly presented on both MHC class I and MHC
to prevent autoimmune responses. These immune checkpoints
(CTLA-4, PD-1, LAG3, and TIM3) are often necessary for initiation
Received 26 October 2017; accepted 14 November 2017;
of an immune response; however, as antigen persists, these check-
https://doi.org/10.1016/j.ymthe.2017.11.010.
points ultimately serve to dampen the T cell effector function against
Correspondence: David B. Weiner, The Wistar Institute, 3601 Spruce St., Phila-
the foreign antigen.2 CTLA-4, PD-1, LAG3, and TIM3 are all ex- delphia, PA 19104, USA.
pressed on T cells and limit T cell effector activity. Antibodies block- E-mail: dweiner@wistar.org

Molecular Therapy Vol. 26 No 2 February 2018 ª 2017 The American Society of Gene and Cell Therapy. 1
Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

Molecular Therapy

Figure 1. Delivery of aCTLA-4 or aPD-1 Post-first Vaccination Synergizes with TERT DNA Vaccine above Checkpoint Alone in Generating Anti-tumor
Immune Response
(A) Experimental setup. Mice were implanted with TC-1 tumor cells on day 0 and then immunized four times at 1-week intervals starting 7 days after tumor implant. Mice were
given antibodies (200 mg per mouse) every 3 days starting 1 day after the first immunization. Antibody delivery was continued until 1 week after the final vaccination. (B, D, F,
and H) Tumor volume measurements over time for naive mice, mTERT vaccine-treated mice, or mice treated with mTERT vaccine plus aCTLA-4 (B), aPD-1 (D), or a
combination of aCTLA-4 and aPD-1 (F). (C, E, and G) Mouse survival over time for naive mice, mTERT vaccine-treated mice, or mice treated with mTERT vaccine plus
aCTLA-4 (C), aPD-1 (D), or a combination of aCTLA-4 and aPD-1 (G). For (B)–(G), n = 12–13 mice per group. (H) Tumor volume measurements over time for naive mice

(legend continued on next page)

2 Molecular Therapy Vol. 26 No 2 February 2018


Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

www.moleculartherapy.org

class II, and can be designed using consensus sequences in order to effector T cells in the tumor microenvironment rather than boost-
break tolerance.22 ing vaccine responses in the periphery.

Our laboratory has pioneered efforts to develop therapeutic DNA RESULTS


vaccines for patients with human papillomavirus (HPV)-associated CTLA-4 and PD-1 Blockade Synergizes with mTERT DNA
cervical intraepithelial neoplasia (CIN2/3).23,24 We incorporated Vaccine in Generating Anti-tumor Immunity
important improvements to the DNA vaccine platform, such as For these studies, we utilized a synthetic consensus (SynCon) mouse
plasmid optimization and use of in vivo electroporation, which has TERT DNA vaccine. This vaccine administered alone exhibits a
directly led to enhanced CD4+ and CD8+ T cell responses.24–26 These robust capacity to generate an immune response against matched,
improvements led to the first DNA vaccine to demonstrate potent im- SynCon peptides, while maintaining the capacity to break tolerance
mune responses and anti-neoplastic activity in a human clinical to native mouse TERT peptides in C57BL/6 mice (Figures S1A and
trial.23,24 Specifically, a synthetic DNA vaccine targeting HPV-16 S1B). For the remaining experiments, we focused on antigen-specific
and HPV-18 E6 and E7 proteins (VGX-3100) showed clinical efficacy immune responses targeting native mouse TERT peptides. In addi-
for therapeutic treatment of cervical intraepithelial neoplasia 2/3 tion, this SynCon mouse TERT DNA vaccine is capable of slowing
(CIN2/3) in women in a double-blind, placebo-controlled phase IIb tumor growth and prolonging survival in a proportion of the mice
clinical trial.23 in a TC-1 mouse tumor challenge model (Figures S2A and S2B).

Next, we focused on developing a novel DNA vaccine platform to We first examined whether aCTLA-4 or aPD-1 therapy would
help break tolerance to non-viral tumor-associated antigens, such improve anti-tumor activity in a therapeutic tumor challenge setting.
as TERT and WT-1.27,28 This novel platform is based on the concept We implanted mice with TC-1 tumors and initiated immunizations
that the introduction of xenogeneic antigens into mice can induce after palpable tumors formed 1 week following implantation (Fig-
autoimmunity.29,30 We developed a synthetic consensus (SynCon) ure 1A). Mice were immunized at 1-week intervals for a total of
approach in which gene sequences from various species are compared four immunizations. Checkpoint treatment began 1 day following
to identify a consensus sequence (with approximately 95% homology the first immunization and continued until 1 week following the final
to the native gene sequence), an approach that has enhanced the ca- immunization (Figure 1A).
pacity to break tolerance, yet maintains T cell killing against native
MHC class I-presented sequences. We reported that this synthetic We observed that both a-CTLA-4 and a-PD-1 therapies synergized
consensus vaccine design strategy is superior to native DNA-vac- with mTERT DNA immunization above vaccine therapy alone in a
cine-encoded antigens for the WT-1 antigen.28 therapeutic TC-1 tumor challenge model when immune checkpoints
were delivered 1 day post-first immunization (Figures 1B–1G). Both
Here, we sought to further improve synthetic consensus DNA vac- a-CTLA-4 and a-PD-1 therapy alone had no initial impact in slowing
cine immune responses through combination therapy with im- tumor growth and no significant impact on mouse survival (Figures
mune checkpoint blockade. We utilized a SynCon mouse TERT 1H and 1I). SynCon mTERT immunization alone slightly slowed
DNA vaccine with enhanced capacity to break tolerance in combi- tumor growth and improved survival compared to naive mice. How-
nation with antibodies that block the immune checkpoint mole- ever, a-CTLA-4 and a-PD-1 in combination with SynCon mTERT
cules CTLA-4 or PD-1. We chose to test this combination therapy robustly slowed tumor growth and significantly improved survival
in the TC-1 tumor model, which does not respond to immune compared to DNA alone or naive mice (Figures 1B–1E). Synergy
checkpoint blockade alone. We observe that blockade of was greater for a-CTLA-4, in particular for survival, compared to
CTLA-4, PD-1, or a combination of the two effectively synergized a-PD-1 (Figures 1B–1E).
with SynCon TERT DNA vaccine to slow tumor growth in mice.
However, peripheral and systemic immune responses did not Combination therapy of aCTLA-4 and a-PD-1 in both animal
necessarily correspond with anti-tumor activity of immune therapy models and in the clinic has shown improved anti-tumor responses
combinations. These studies highlight the potential application of compared to treatment with each checkpoint alone.33–35 In particular,
these DNA vaccine/immune checkpoint blockade combinations combination therapy with aCTLA-4 and a-PD-1 was reported to
in patients that respond poorly to immune checkpoint blockade improve the anti-tumor immunity of cell-based vaccines.5,17 We
alone and allow for better rational design of combination thera- therefore examined whether a triple-combination therapy with
pies. Furthermore, these results suggest that this synergistic effect aCTLA-4, a-PD-1, and SynCon mTERT could further improve ther-
is due to the effect of immune checkpoint blockade on expanding apeutic anti-tumor activity. We observed that combination therapy of

unvaccinated mice treated with aPD-1, aCTLA-4, or both aPD-1 and aCTLA-4. (I) Mouse survival over time for naive mice or unvaccinated mice treated with aPD-1,
aCTLA-4, or both aPD-1 and aCTLA-4. Mice were euthanized if they appeared sick or if the tumor diameter exceeded 1.5 cm. For (H) and (I), n = 9 mice per groiup. Each
experiment was repeated at least once to verify results. Significance for tumor volume measurements was determined by two-way ANOVA followed by Tukey’s HSD test.
Significance for mouse survival was determined by the Gehan-Breslow-Wilcoxon test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. Error bars indicate ± SEM.

Molecular Therapy Vol. 26 No 2 February 2018 3


Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

Molecular Therapy

Figure 2. Antigen-Specific Intracellular Cytokine Production upon TERT DNA Vaccination and Immune Checkpoint Blockade in Tumor-Bearing Mice
(A) Experimental setup. Mice were implanted with TC-1 tumor cells on day 0 and then immunized on days 7 and 14. Antibody delivery was started 1 day after the first
immunization and continued every 3 days. Mice were sacrificed on day 21 for immune cell analysis. (B–F) Intracellular cytokine staining of CD8 or CD4 T cells after stimulation
with native mouse TERT peptides for 5 hr. CD4 T cells were stained for IFN-g (B) or TNF-a (C). CD8 T cells were stained for IFN-g (D), TNF-a (E), or IFN-g/CD107a/T-bet (F).
Significance was determined by two-way ANOVA followed by Tukey’s HSD test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. n = 7–10 mice per group. A representative
of three independent experiments is shown. Error bars indicate ± SEM.

SynCon mTERT with both aCTLA-4 and a-PD-1 slightly improved Surprisingly, there was no significant impact of any of the checkpoint
anti-tumor activity and survival of mice. This effect was greater than antibodies or combinations on systemic type 1 antigen-specific CD8+
the combined treatment of aCTLA-4 and SynCon mTERT (Figures or CD4+ immune responses, including expression of interferon-
1F and 1G). The combination therapy alone without the DNA vaccine gamma (IFNg) and tumor necrosis factor alpha (TNF-a) in CD4+
exhibited little impact on tumor growth or on mouse survival (Figures and CD8+ T cells or IFNg/CD107a/T-bet in CD8+ T cells compared
1H and 1I). to mTERT DNA alone (Figures 2B–2F).

Impact of Immune Checkpoint Blockade on Systemic Immune Next, we examined PD-1 expression on CD4 and CD8 T cells in the
Responses in Tumor-Bearing Mice spleen and the periphery of tumor-bearing mice. In tumor-bearing
Next, we explored immune responses in tumor-bearing mice treated mice, aPD-1 had no impact on PD-1 expression in both CD4+ and
with combination vaccine and the aCTLA-4/aPD-1 immune check- CD8+ T cells in spleen and in the periphery (Figures 3A–3D). How-
point blockade antibody therapy. We implanted mice with TC-1 tu- ever, both aCTLA-4 and a combination of aCTLA-4 and aPD-1
mors, immunized mice on days 7 and 14, and sacrificed mice on day enhanced the frequency of PD-1+ CD4+ and CD8+ T cells in both
21 (Figure 2A). Checkpoint inhibitor delivery was initiated one day spleen and the periphery (Figures 3A–3D).
following the first immunization, and continued every 3 days until
day 21 (Figure 2A). We examined the antigen-specific immune re- aCTLA-4 therapy is known to impact Tregs in a tissue-dependent
sponses in the spleen and the exhaustion marker PD-1 in the periph- manner, with a more profound depletion of Tregs within the tumor
ery and in spleen. compared to other tissue sites.14,36 We therefore examined the

4 Molecular Therapy Vol. 26 No 2 February 2018


Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

www.moleculartherapy.org

Figure 3. Phenotype of Peripheral and Spleen T Cells upon TERT DNA Vaccination and Immune Checkpoint Blockade in Tumor-Bearing Mice
(A–F) Staining of CD8 or CD4 T cells from spleen or peripheral blood of mice treated with the indicated therapies in accordance with the schedule in Figure 3A. CD8+
splenocytes (A) or PBMCs (B) were stained for PD-1. CD4+ splenocytes (C) or PBMCs (D) were stained for PD-1. CD4+ splenocytes (E) or PBMCs (F) were stained for CD25/
FoxP3. Significance was determined by two-way ANOVA followed by Tukey’s HSD test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. n = 7–10 mice per group.
A representative of three independent experiments is shown. Error bars indicate ± SEM.

frequency of CD4+/CD25+/FoxP3+ Tregs in spleen and in the periph- spleen, we next examined immune responses in the tumors of these
ery upon immune checkpoint blockade therapy in combination with same mice. aCTLA-4 treatment resulted in significant depletion of
SynCon mTERT DNA vaccine (Figures 3E and 3F). We observed that intra-tumoral Tregs (Figure 4A), whereas aPD-1 therapy did not
all checkpoint therapies and combinations decreased the frequency of alter the frequency of intra-tumoral Tregs, and combination ther-
Tregs in spleen, but slightly increased the frequency of Tregs in the apy with both aCTLA-4 and aPD-1 resulted in only a modest
periphery (Figures 3E and 3F). decrease in the frequency of intra-tumoral Tregs (Figure 4A).
Similar to the results observed in spleen and in the peripheral
Impact of Immune Checkpoint Blockade on Tumor-Specific T cells, the percentage of PD-1+ tumor-infiltrating CD8+ lympho-
Immune Responses in Tumor-Bearing Mice cytes also increased upon aCTLA-4 treatment and, to a lesser
Because of the surprising finding that immune checkpoint blockade extent, combination therapy with aCTLA-4 and aPD-1 treatment
had no impact on TERT antigen-specific immune responses in (Figure 4B).

Molecular Therapy Vol. 26 No 2 February 2018 5


Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

Molecular Therapy

Figure 4. Phenotype of Tumor-Infiltrating Lymphocytes upon TERT DNA Vaccination and Immune Checkpoint Blockade
(A–F) Staining of CD8 or CD4 T cells isolated from the tumors of mice treated with the indicated therapies in accordance with the schedule in Figure 4A. mTERT-stimulated
CD3+ TILs were stained for CD4/CD25/FoxP3 (A), CD8/PD-1 (B), CD8/CD44 (C), CD8/T-bet (E), or CD8/CD44/Tbet (F). PMA-stimulated CD3+ T cells were stained for CD8/
CD44 (D). Significance was determined by two-way ANOVA followed by Tukey’s HSD test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. n = 7–10 mice per group.
A representative of three independent experiments is shown. Error bars indicate ± SEM.

Next, we examined the T cell activation markers CD44 and T-bet in CTLA-4 Blockade Synergizes More Than Treg Depletion with
CD8+ tumor-infiltrating lymphocytes (TILs). aCTLA-4 treatment mTERT DNA Vaccine
alone enhanced the frequency of CD44+ CD8+ TILs (Figure 4C). It has been proposed that the primary mechanism for aCTLA-4 ther-
This effect was more pronounced after treatment of these TILs apy is depletion of Tregs in the tumor microenvironment. In order to
with PMA (Figure 4D). Interestingly, aPD-1 and the combination test the contribution of Tregs to the syngery between SynCon TERT
of aCTLA-4 and aPD-1 therapy did not significantly impact DNA vaccine and aCTLA-4 treatment, we compared the impact of
CD44 expression in the TILs (Figures 4C and 4D). Both aCTLA-4 aCTLA-4 to aCD25, a depletion antibody that systemically depletes
and the combination of aCTLA-4 and aPD-1 therapy enhanced the Tregs.37 We verified that this aCD25 depletion antibody significantly
frequency of T-bet+ CD8+ TILs (Figure 4E). This trend was similar decreased the frequency of Tregs in the peripheral blood, in spleen,
for CD8+ T cells that co-expressed T-bet and CD44 (Figure 4F). The and in the tumor (Figures 5A–5C). Importantly, the extent of Treg
SynCon TERT DNA vaccine alone did not significantly impact depletion in the tumor was similar to that of CTLA-4 blockade (Fig-
Tregs, PD-1, or CD44 expression in TILs (Figures 4A–4D). ures 4A and 5A). In these experiments, while aCD25 in combination
Together, these results demonstrate that aCTLA-4 therapy expands with mTERT significantly improved anti-tumor responses, this effect
the effector T cell population within the tumor, likely resulting was not as strong as combination therapy of aCTLA-4 and mTERT in
in the strong anti-tumor effect when combined with TERT DNA terms of tumor growth or overall survival (Figures 5D and 5E).
vaccination. Because CD25 can also be expressed by effector T cells, we examined

6 Molecular Therapy Vol. 26 No 2 February 2018


Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

www.moleculartherapy.org

Figure 5. Delivery of aCTLA-4 in Combination with TERT DNA Vaccine Is Superior to Treg Depletion in Combination with TERT DNA Vaccine
(A–C) Staining of CD8 or CD4 T cells from the peripheral blood (A), spleen (B), or tumor (C) of mice treated with the indicated therapies in accordance with the schedule in
Figure 2. (D) Tumor volume measurements over time for mice with indicated treatment regimen. Mice were treated in accordance with the schedule shown in Figure 2A.
(E) Mouse survival over time for mice with indicated treatment regimen. Mice were euthanized if they appeared sick or if the tumor diameter exceeded 1.5 cm. Significance for
tumor volume measurements was determined by two-way ANOVA followed by Tukey’s HSD test. Significance for mouse survival was determined by the Gehan-Breslow-
Wilcoxon test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. n = 10 mice per group. Error bars indicate ± SEM.

whether the aCD25 antibody treatment affected effector and central blockade. We have previously shown strong immune responses for
memory T cells (Figures S3A–S3F). We found that, in the peripheral our SynCon DNA vaccine platform in human clinical trials for viral
blood, spleen and tumor, there was no depletion of effector CD8 antigens such as HPV and Zika virus.23,38 We have now improved
T cells (CD44+/CD62Llo) or central memory CD8 T cells (CD44+/ this technology to help break tolerance to cancer-associated self-anti-
CD62L+) with aCD25 antibody treatment (Figures S3A–S3F). These gens, such as WT-1 and TERT, which we have tested in both mice and
data suggest that the mechanism of action of aCTLA-4 extends non-human primates.27,28 Because immune checkpoint blockade is
beyond depletion of Tregs in the tumor and is likely due to the expan- becoming the standard of care in cancer patients, it is important to
sion of effector T cells within the tumor (Figure 4). understand how these novel DNA vaccine therapies can work as com-
bination therapies.
DISCUSSION
Here, we utilized a synthetic consensus DNA vaccine targeting the In this study we observed robust synergy between SynCon mTERT
tumor antigen TERT in combination with immune checkpoint DNA vaccination and aCTLA-4 immune checkpoint blockade.

Molecular Therapy Vol. 26 No 2 February 2018 7


Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

Molecular Therapy

This combination exhibited long-term effects for prolonging mouse In an independent study GVAX synergized more with a-CTLA-4
survival in a therapeutic tumor challenge model. While aPD-1 therapy compared to aPD-1 therapy in CT26 tumors; however, re-
showed some synergy with SynCon mTERT DNA immunization, sponses to GVAX in combination with each checkpoint were similar
the effect was not as profound and did not result in significant prolon- in ID8 tumors5. In each of these cell-based vaccine studies in which
gation of mouse survival. Importantly, these mice did not exhibit anti- triple-combination therapies were used (GVAX/FVAX, a-CTLA-4,
tumor immunity after delivery of immune checkpoint antibodies and a-PD-1), the triple-combination therapy was profoundly supe-
alone, suggesting that addition of a DNA vaccine to an immune rior to any of the double- or mono-therapies.5,17 Our study showed
checkpoint blockade regimen may allow non-responders to become a clear bias toward DNA vaccine synergy with a-CTLA-4 blockade
responders. Interesting future studies may include testing the efficacy over a-PD-1 blockade. Furthermore, the triple-therapy was only
of this combination therapy in tumor models that already show some slightly better than double-therapy with vaccine plus aCTLA-4, indi-
response to immune checkpoint blockade alone, to evaluate whether cating that patients receiving our vaccine may not benefit as much
increased immune potency could be achieved. from receiving both aCTLA-4 and aPD-1. This is useful information,
since the double-therapy with aCTLA-4 and aPD-1 has significantly
Only a handful of preclinical studies have examined the efficacy of more toxicity compared to mono-therapy with either aCTLA-4 or
combining DNA vaccines with immune checkpoint blockade for aPD-1 alone in humans.42
tumor therapy. Two studies showed modest improvement in anti-tu-
mor immunity by combining DNA vaccines targeting the MYB onco- Furthermore, we did not observe a significant change in the T cell
protein for colorectal cancer or the SSX2 cancer antigen for sarcoma exhaustion marker PD-1 expression on CD8 or CD4 T cells in the
with aPD-1 antibody.39,40 However, neither of these studies used elec- spleen, periphery or tumor of both tumor-bearing mice after immu-
troporation in combination with the DNA vaccine, and therefore nization with SynCon TERT DNA vaccine alone. However, in other
both vaccines exhibited minimal anti-tumor effects on their own, studies upregulation of inhibitory T cell molecules, primarily PD-1,
even when delivered only 1-2 days following tumor implantation.39,40 has been observed on antigen-specific CD8 T cells in both mice and
In the report, the DNA vaccine targeting MYB was completely inef- humans after immunization with a variety of different vaccine plat-
fective if delivered 5 days or later after tumor implantation, and forms, including an SSX DNA vaccine, HLA-A2-restricted NY-ESO
was also ineffective at reducing tumor burden altogether in BALB/c peptides formulated with Frend’s and CpG adjuvants, a recombinant
mice.39 Another study combined aCTLA-4 treatment with xenoge- lentivector vaccine expressing melanoma associated antigen epitopes,
neic DNA vaccines targeting TRP-2, gp100 or PSMA in prophylactic and a HPV E7 polypeptide vaccine.40,43–45 This lack of induction
immunization studies.41 Interestingly, this study found that prophy- of PD-1 after our potent DNA vaccine immunization may explain
lactic delivery of aCTLA-4 boosted anti-tumor immunity more when why less synergy was observed between our vaccine and aPD-1
given with the second immunization instead of the first immuniza- therapy.
tion.41 No previous studies have examined combination therapy
with DNA vaccines and aCTLA-4 blocking antibody in a therapeutic We observed an interesting increase in PD-1 expression in T cells af-
tumor challenge model. In our studies we did not observe a significant ter aCTLA-4 treatment, but not aPD-1 treatment, in tumor-bearing
boosting in antigen-specific immune responses when aCTLA-4 was mice (Figure 3). Interestingly, other studies have also shown an in-
delivered following the first or second immunization. The reasons crease in the fraction of CD8+ T cells expressing PD-1 after mono-
for this discrepancy are unclear, but may be antigen-dependent, or therapy with aCTLA-4, aPD-1, or aPD-L1.17 Upregulation of
masked by the immune potency of the electroporation enhanced syn- PD-1+ and PD-L1+ cells was also shown in aCTLA-4 therapy for hu-
thetic DNA vaccine itself. man and murine bladder cancers.33 Blockade of the PD-1/PD-L1
signaling axis can only partially reverse an exhausted T cell pheno-
While there are only a few studies using DNA vaccines in combina- type; therefore, exploring methods of further improving immune acti-
tion with aCTLA-4 or aPD-1 checkpoint antibodies, studies using vation in tumor-bearing mice remains important.46
cell-based vaccines have explored these combinations more thor-
oughly. Irradiated tumor cells expressing GM-CSF or Flt-3 ligand Some differences between aCTLA-4 and aPD-1 blockade in the
(GVAX or FVAX, respectively) have been frequently used as a context of DNA vaccines may be the result of the different effects
vaccine for combination therapy studies with immune checkpoint of aCTLA-4 and aPD-1 on Tregs. aCTLA-4 antibodies, unlike
blockade.5,14–17 Interestingly, the relative effects of aCTLA-4 aPD-1 antibodies, have demonstrated robust depletion of intra-
compared to aPD-1 therapy appear to be dependent on both the tumoral Tregs in mice.14,36 There is also some evidence for ipili-
type of vaccine and the tumor model. A study that utilized a PLG mumab-dependent antibody-dependent cell-mediated cytotoxicity
(poly[lactide-co-glycolide]) cancer vaccine, in which B16 tumor ly- (ADCC) ex vivo in patient samples; however, the extent to which clin-
sates are incorporated into PLG scaffolds, showed that aCTLA-4 ical aCTLA-4 antibodies can deplete Tregs in patient tumors remains
therapy synergized to a greater extent than aPD-1 therapy.15 How- unclear.47 In the present study, aCTLA-4 antibody treatment was su-
ever, a study comparing GVAX and FVAX with aCTLA-4 or perior to depletion of Tregs (with an aCD25 depletion antibody) in
a-PD-1 therapy showed that for FVAX, aPD-1 therapy was superior combination with SynCon mTERT DNA vaccine (Figure 5). A sepa-
to aCTLA-4 therapy, and for GVAX, the two therapies were similar.17 rate study found a similar result when comparing combination

8 Molecular Therapy Vol. 26 No 2 February 2018


Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

www.moleculartherapy.org

therapy of GVAX or FVAX with aCD25 depletion, implying that Hyclone L-15 Leibowitz Media (ThermoFisher) and RPMI, supple-
aCTLA-4 has additional effects beyond simple depletion of Tregs.16 mented with 10% FBS and 1% penicillin/streptomycin. Tumors
This suggests that the synergy between TERT and aCTLA-4 is were minced and then transferred to a 50 mL conical filled with
most likely due to the expansion of effector CD8+ T cells within the 10 mL of digestion mix. Cells were then filtered twice through a
tumor, which express the markers CD44 and T-bet, which we 40-mm mesh filter, counted, and stained.
observed upon treatment with aCTLA-4 or aCTLA-4/aPD-1 combi-
nation therapy. ELISpot Assay
Splenocytes were stimulated in the presence of native mouse TERT
Importantly, we did not observe an increase in antigen-specific im- peptides (15-mer peptides spanning the entire native mouse protein,
mune responses in the spleen upon combination therapy with overlapping by 9 amino acids, GenScript) for 24 hr in MABTECH
TERT DNA vaccine and immune checkpoint blockade, despite the Mouse IFNg ELISpotPLUS plates. Spots were developed and counted
synergy of these therapies in inducing anti-tumor immunity. These in accordance with the manufacturer’s protocol.
data suggest that immune checkpoint blockade functions to alter
the immune microenvironment and expand effector T cells at the tu- Intracellular Cytokine Staining and Flow Cytometry
mor site rather than acting on antigen-specific cells in the periphery. 2 million splenocytes per mouse were stimulated with native mouse
It is possible that addition of immune checkpoint blockade antibodies TERT peptides for 5 hr in the presence of GolgiStop GolgiPlug (BD
may alter T cell trafficking and may drive antigen-specific immune Bioscience). Phorbol myristate acetate (PMA, BD Bioscience) and
cells to peripheral tissues in the mouse. complete media were used as positive and negative controls, respec-
tively. Cells were incubated with FITC a-mouse CD107a (clone
Taken together, these studies support further rational combination 1D4B, Biolegend) during the stimulation to stain for degranulation.
immune therapies with synthetic TERT DNA vaccination and im- After stimulation, cells were washed and stained for viability using
mune checkpoint blockade. In addition, antigen-specific immune re- LIVE/DEAD violet. Surface stain was then added, followed by fixation
sponses in the periphery may not correlate with efficacy in patients. and permeabilization using a FoxP3/transcription factor fixation/per-
meabilization kit (eBioscience). The following antibodies were used in
MATERIALS AND METHODS these studies: PECy5 aCD3 (clone 145-2C11, BD PharMingen),
DNA Plasmids BV510 aCD4 (clone RM4-5, Biolegend), BV605 aTNF-a (clone
The synthetic consensus TERT sequence was generated by using 6 MP6-XT22), PE aT-bet (clone 4B10, Biolegend), BV711 aPD-1
TERT sequences collected from animals including mouse, rat, and (clone 29F.1A12, Biolegend), APC aFoxP3 (clone FJK-16 s,
hamster. The consensus sequence was obtained after alignment of eBioscience), APCCy7 aCD8 (clone 53-6.7, Biolegend), AF700
these sequences using Clustal W. Two additional mutations were aCD44 (clone IM7, Biolegend), and APC aIFNg (clone XMG1.2,
added to abolish telomerase activity.31,32 All sequences were RNA Biolegend). All data were collected on a modified LSRII flow cytom-
and codon optimized with an immunoglobulin E (IgE) leader eter (BD Bioscience) and analyzed using FlowJo software (TreeStar).
sequence and a Kozak sequence at the N terminus and cloned into
the modified pVAX vector (Genscript). The final SynCon mouse Animal Immunization
TERT sequences shares 94.6% sequence identity with native mouse Female 6- to 8-week-old C57BL/6 mice were purchased from Jackson
TERT. Laboratory. Animal care was in accordance with the guidelines of the
NIH and with the Wistar Institute Animal Care and Use Committee
Peripheral Blood Mononuclear Cell and Splenocyte Isolation (IACUC). Mice were immunized with 25 mg of each plasmid by intra-
For splenocyte isolation, spleens from mice were collected in com- muscular injection into the tibialis anterior (TA) muscle, followed by
plete RPMI media containing 10% fetal bovine serum (FBS). Cells electroporation (EP) using the CELLECTRA-3P adaptive constant
were dissociated using a stomacher and then filtered through a current device (Inovio Pharmaceuticals). Two 0.1-amp constant
40-mm mesh filter. Red blood cells were then lysed using ACK Lysis current square-wave pulses were delivered through a triangular
buffer (LifeTechnologies). Cells were then filtered again through a three-electrode array. Each pulse was 52 ms in length with a 1-s delay
40-mm mesh filter, counted, and plated for staining. For peripheral between pulses. For immunogenicity studies, mice were given a total
blood mononuclear cell (PBMC) isolation, blood was collected in of three immunizations at 2-week intervals. For tumor challenge
4% sodium citrate to prevent clotting. Blood was then layered on studies, mice were given a total of four immunizations at 1-week
top of Histopaque 1083 (Sigma-Aldrich). Cells were spun for 1 hr, intervals.
and then cells from the buffy coat were separated and stained for
analysis. Immune Checkpoint Blockade Antibodies
The following antibodies were used for animal studies (BioXCell):
Isolation of TILs a-mouse PD-1 (clone RMP1-14), a-mouse CTLA-4 (clone 9D9), or
Tumors were dissociated using digestion mix, which consisted of aCD25 (clone PC-61.5.3). Mice were given 200 mg of each antibody,
170 mg/L collagenase I, II, and IV (ThermoFisher), 12.5 mg/L DNase delivered intraperitoneally, in accordance with the schedule in each
I (Roche), and 25 mg/L Elastase (Worthington) in a 50/50 mixture of figure.

Molecular Therapy Vol. 26 No 2 February 2018 9


Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

Molecular Therapy

Tumor Challenge Studies grant (CA174523 to D.B.W.), the Wistar National Cancer Institute
The TC-1 cell line was a gift from Dr. Yvonne Paterson from the Uni- Cancer Center (P30 CA 010815), the W.W. Smith Family Trust (to
versity of Pennsylvania. This cell line was tested for Mycoplasma D.B.W.), funding from the Basser Foundation (to D.B.W.), and a
contamination prior to freezing the cells, and the test results were grant from Inovio Pharmaceuticals (to D.B.W.).
negative. Cells were thawed and cultured for fewer than 5 passages
prior to implantation into mice. Cells were cultured in DMEM REFERENCES
(Mediatech) supplemented with 10% fetal bovine serum (FBS). Cells 1. Pardoll, D.M. (2012). The blockade of immune checkpoints in cancer immuno-
were subcutaneously implanted onto the right flank of C57BL/6 mice therapy. Nat. Rev. Cancer 12, 252–264.

(50,000 cells per mouse). 1 week following tumor implantation, 2. Riley, J.L. (2009). PD-1 signaling in primary T cells. Immunol. Rev. 229, 114–125.

mouse tumors were measured, and mice were randomized to treat- 3. Walunas, T.L., and Bluestone, J.A. (1998). CTLA-4 regulates tolerance induction and
T cell differentiation in vivo. J. Immunol. 160, 3855–3860.
ment groups so that the average tumor volume and SD was equivalent
4. Anderson, A.C. (2014). Tim-3: an emerging target in the cancer immunotherapy
between groups. Tumors were monitored twice a week with electronic
landscape. Cancer Immunol. Res. 2, 393–398.
calipers. Tumor volume was calculated using the following formula:
5. Duraiswamy, J., Kaluza, K.M., Freeman, G.J., and Coukos, G. (2013). Dual blockade
volume = (p/6)*(height)*(width2). Mice were euthanized upon any of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejec-
sign of distress or sickness or when tumor diameters exceeded tion function in tumors. Cancer Res. 73, 3591–3603.
1.5 cm, in accordance with IACUC guidelines. 6. Hodi, F.S., O’Day, S.J., McDermott, D.F., Weber, R.W., Sosman, J.A., Haanen, J.B.,
Gonzalez, R., Robert, C., Schadendorf, D., Hassel, J.C., et al. (2010). Improved survival
with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363,
Statistical Analysis
711–723.
Statistical analyses were performed using GraphPad Prism Software.
7. Postow, M.A., Callahan, M.K., and Wolchok, J.D. (2015). Immune checkpoint
For each experiment, error bars represent the mean ± the SEM. For blockade in cancer therapy. J. Clin. Oncol. 33, 1974–1982.
all experiments, statistical significance was determined by one- or 8. Reck, M., Rodríguez-Abreu, D., Robinson, A.G., Hui, R., Cs}oszi, T., Fülöp, A.,
two-way ANOVA, followed by Tukey’s post hoc honest significant Gottfried, M., Peled, N., Tafreshi, A., Cuffe, S., et al.; KEYNOTE-024 Investigators
difference (HSD) test. For mouse survival studies, significance was (2016). Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell
lung cancer. N. Engl. J. Med. 375, 1823–1833.
determined by Gehan-Breslow-Wilcoxon test.
9. Robert, C., Schachter, J., Long, G.V., Arance, A., Grob, J.J., Mortier, L., Daud, A.,
Carlino, M.S., McNeil, C., Lotem, M., et al.; KEYNOTE-006 investigators (2015).
SUPPLEMENTAL INFORMATION Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 372,
Supplemental Information includes three figures and can be found 2521–2532.
with this article online at https://doi.org/10.1016/j.ymthe.2017.11.010. 10. Jing, W., Gershan, J.A., Weber, J., Tlomak, D., McOlash, L., Sabatos-Peyton, C., and
Johnson, B.D. (2015). Combined immune checkpoint protein blockade and low dose
whole body irradiation as immunotherapy for myeloma. J. Immunother. Cancer 3, 2.
AUTHOR CONTRIBUTIONS
E.K.D, M.C.W., D.O.V., B.F., J.W., J.Y., A.K., E.M., L.H., and D.B.W. 11. Li, B., VanRoey, M., Wang, C., Chen, T.H., Korman, A., and Jooss, K. (2009). Anti-
programmed death-1 synergizes with granulocyte macrophage colony-stimulating
were involved in study conception and design. E.K.D., M.C.W., A.T., factor–secreting tumor cell immunotherapy providing therapeutic benefit to mice
D.O.V., B.F., J.W., J.Y., and A.K. were involved in the acquisition, the with established tumors. Clin. Cancer Res. 15, 1623–1634.
analysis, and the interpretation of data. E.K.D. and D.B.W. drafted 12. Tang, C., Wang, X., Soh, H., Seyedin, S., Cortez, M.A., Krishnan, S., Massarelli, E.,
the manuscript. All authors were involved in critically revising the Hong, D., Naing, A., Diab, A., et al. (2014). Combining radiation and immuno-
therapy: a new systemic therapy for solid tumors? Cancer Immunol. Res. 2, 831–838.
manuscript.
13. Ito, F., Li, Q., Shreiner, A.B., Okuyama, R., Jure-Kunkel, M.N., Teitz-Tennenbaum, S.,
and Chang, A.E. (2004). Anti-CD137 monoclonal antibody administration augments
CONFLICTS OF INTEREST the antitumor efficacy of dendritic cell-based vaccines. Cancer Res. 64, 8411–8419.
M.C.W., B.F., J.W., J.Y., A.K., E.M., and L.H. are employees of Inovio 14. Quezada, S.A., Peggs, K.S., Curran, M.A., and Allison, J.P. (2006). CTLA4 blockade
Pharmaceuticals and as such receive salary and benefits, including and GM-CSF combination immunotherapy alters the intratumor balance of effector
ownership of stock and stock options. D.B.W. discloses grant funding, and regulatory T cells. J. Clin. Invest. 116, 1935–1945.
industry collaborations, speaking honoraria, and fees for consulting. 15. Ali, O.A., Lewin, S.A., Dranoff, G., and Mooney, D.J. (2016). Vaccines combined with
His service includes serving on scientific review committees and advi- immune checkpoint antibodies promote cytotoxic T-cell activity and tumor eradica-
tion. Cancer Immunol. Res. 4, 95–100.
sory boards. Remuneration includes direct payments, stock, or stock
16. Curran, M.A., and Allison, J.P. (2009). Tumor vaccines expressing flt3 ligand syner-
options. He notes potential conflicts associated with his work with gize with ctla-4 blockade to reject preimplanted tumors. Cancer Res. 69, 7747–7755.
Pfizer, Bristol Myers Squibb, Inovio Pharmaceuticals, Touchlight,
17. Curran, M.A., Montalvo, W., Yagita, H., and Allison, J.P. (2010). PD-1 and CTLA-4
Oncosec, Merck, VGXI, and potentially others. Licensing of technol- combination blockade expands infiltrating T cells and reduces regulatory T and
ogy from his laboratory has created more than 150 jobs in the private myeloid cells within B16 melanoma tumors. Proc. Natl. Acad. Sci. USA 107, 4275–
sector of the biotechnology and pharmaceutical industry. The other 4280.

authors declare no competing financial interests. 18. Snyder, A., Makarov, V., Merghoub, T., Yuan, J., Zaretsky, J.M., Desrichard, A.,
Walsh, L.A., Postow, M.A., Wong, P., Ho, T.S., et al. (2014). Genetic basis for clinical
response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199.
ACKNOWLEDGMENTS
19. Van Allen, E.M., Miao, D., Schilling, B., Shukla, S.A., Blank, C., Zimmer, L., Sucker,
This work was supported by an NIH/NCI NRSA Individual Fellow- A., Hillen, U., Foppen, M.H.G., Goldinger, S.M., et al. (2015). Genomic correlates of
ship (CA213795 to E.K.D.), a Penn/Wistar Institute NIH SPORE response to CTLA-4 blockade in metastatic melanoma. Science 350, 207–211.

10 Molecular Therapy Vol. 26 No 2 February 2018


Please cite this article in press as: Duperret et al., Synergy of Immune Checkpoint Blockade with a Novel Synthetic Consensus DNA Vaccine Targeting
TERT, Molecular Therapy (2017), https://doi.org/10.1016/j.ymthe.2017.11.010

www.moleculartherapy.org

20. Bartkowiak, T., Singh, S., Yang, G., Galvan, G., Haria, D., Ai, M., Allison, J.P., Sastry, 34. Wolchok, J.D., Kluger, H., Callahan, M.K., Postow, M.A., Rizvi, N.A., Lesokhin, A.M.,
K.J., and Curran, M.A. (2015). Unique potential of 4-1BB agonist antibody to pro- Segal, N.H., Ariyan, C.E., Gordon, R.A., Reed, K., et al. (2013). Nivolumab plus ipi-
mote durable regression of HPV+ tumors when combined with an E6/E7 peptide vac- limumab in advanced melanoma. N. Engl. J. Med. 369, 122–133.
cine. Proc. Natl. Acad. Sci. USA 112, E5290–E5299.
35. Lussier, D.M., Johnson, J.L., Hingorani, P., and Blattman, J.N. (2015). Combination
21. Madan, R.A., Heery, C.R., and Gulley, J.L. (2012). Combination of vaccine immunotherapy with a-CTLA-4 and a-PD-L1 antibody blockade prevents immune
and immune checkpoint inhibitor is safe with encouraging clinical activity. escape and leads to complete control of metastatic osteosarcoma. J. Immunother.
OncoImmunology 1, 1167–1168. Cancer 3, 21.
22. Flingai, S., Czerwonko, M., Goodman, J., Kudchodkar, S.B., Muthumani, K., and 36. Selby, M.J., Engelhardt, J.J., Quigley, M., Henning, K.A., Chen, T., Srinivasan, M., and
Weiner, D.B. (2013). Synthetic DNA vaccines: improved vaccine potency by electro- Korman, A.J. (2013). Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor
poration and co-delivered genetic adjuvants. Front. Immunol. 4, 354. activity through reduction of intratumoral regulatory T cells. Cancer Immunol.
23. Trimble, C.L., Morrow, M.P., Kraynyak, K.A., Shen, X., Dallas, M., Yan, J., et al. Res. 1, 32–42.
(2015). Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic 37. Stephens, L.A., Gray, D., and Anderton, S.M. (2005). CD4+CD25+ regulatory T cells
DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cer- limit the risk of autoimmune disease arising from T cell receptor crossreactivity. Proc.
vical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled Natl. Acad. Sci. USA 102, 17418–17423.
phase 2b trial. Lancet 386, 2078–2088.
38. Tebas, P., Roberts, C.C., Muthumani, K., Reuschel, E.L., Kudchodkar, S.B., Zaidi, F.I.,
24. Bagarazzi, M.L., Yan, J., Morrow, M.P., Shen, X., Parker, R.L., Lee, J.C., Giffear, M., White, S., Khan, A.S., Racine, T., Choi, H., et al. (2017). Safety and immunogenicity
Pankhong, P., Khan, A.S., Broderick, K.E., et al. (2012). Immunotherapy against of an anti-Zika virus DNA vaccine - preliminary report. N. Engl. J. Med., Published
HPV16/18 generates potent TH1 and cytotoxic cellular immune responses. Sci. online October 4, 2017. https://doi.org/10.1056/NEJMoa1708120.
Transl. Med. 4, 155ra138.
39. Cross, R.S., Malaterre, J., Davenport, A.J., Carpinteri, S., Anderson, R.L., Darcy, P.K.,
25. Morrow, M.P., Tebas, P., Yan, J., Ramirez, L., Slager, A., Kraynyak, K., Diehl, M., and Ramsay, R.G. (2015). Therapeutic DNA vaccination against colorectal cancer by
Shah, D., Khan, A., Lee, J., et al. (2015). Synthetic consensus HIV-1 DNA induces targeting the MYB oncoprotein. Clin. Transl. Immunology 4, e30.
potent cellular immune responses and synthesis of granzyme B, perforin in HIV in-
fected individuals. Mol. Ther. 23, 591–601. 40. Rekoske, B.T., Smith, H.A., Olson, B.M., Maricque, B.B., and McNeel, D.G. (2015).
PD-1 or PD-L1 blockade restores antitumor efficacy following SSX2 epitope-modi-
26. Kalams, S.A., Parker, S.D., Elizaga, M., Metch, B., Edupuganti, S., Hural, J., De Rosa,
fied DNA vaccine immunization. Cancer Immunol. Res. 3, 946–955.
S., Carter, D.K., Rybczyk, K., Frank, I., et al.; NIAID HIV Vaccine Trials Network
(2013). Safety and comparative immunogenicity of an HIV-1 DNA vaccine in com- 41. Gregor, P.D., Wolchok, J.D., Ferrone, C.R., Buchinshky, H., Guevara-Patiño, J.A.,
bination with plasmid interleukin 12 and impact of intramuscular electroporation for Perales, M.-A., Mortazavi, F., Bacich, D., Heston, W., Latouche, J.B., et al. (2004).
delivery. J. Infect. Dis. 208, 818–829. CTLA-4 blockade in combination with xenogeneic DNA vaccines enhances T-cell re-
sponses, tumor immunity and autoimmunity to self antigens in animal and cellular
27. Yan, J., Pankhong, P., Shin, T.H., Obeng-Adjei, N., Morrow, M.P., Walters, J.N.,
model systems. Vaccine 22, 1700–1708.
Khan, A.S., Sardesai, N.Y., and Weiner, D.B. (2013). Highly optimized DNA vaccine
targeting human telomerase reverse transcriptase stimulates potent antitumor immu- 42. Boutros, C., Tarhini, A., Routier, E., Lambotte, O., Ladurie, F.L., Carbonnel, F.,
nity. Cancer Immunol. Res. 1, 179–189. Izzeddine, H., Marabelle, A., Champiat, S., Berdelou, A., et al. (2016). Safety profiles
28. Walters, J.N., Ferraro, B., Duperret, E.K., Kraynyak, K.A., Chu, J., Saint-Fleur, A., of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat. Rev. Clin.
Yan, J., Levitsky, H., Khan, A.S., Sardesai, N.Y., and Weiner, D.B. (2017). A novel Oncol. 13, 473–486.
DNA vaccine platform enhances neo-antigen-like T cell responses against WT1 to 43. Sierro, S.R., Donda, A., Perret, R., Guillaume, P., Yagita, H., Levy, F., and Romero, P.
break tolerance and induce anti-tumor immunity. Mol. Ther. 25, 976–988. (2011). Combination of lentivector immunization and low-dose chemotherapy or
29. Engelhorn, M.E., Guevara-Patiño, J.A., Noffz, G., Hooper, A.T., Lou, O., Gold, J.S., PD-1/PD-L1 blocking primes self-reactive T cells and induces anti-tumor immunity.
Kappel, B.J., and Houghton, A.N. (2006). Autoimmunity and tumor immunity Eur. J. Immunol. 41, 2217–2228.
induced by immune responses to mutations in self. Nat. Med. 12, 198–206. 44. Badoual, C., Hans, S., Merillon, N., Van Ryswick, C., Ravel, P., Benhamouda, N.,
30. Guevara-Patiño, J.A., Engelhorn, M.E., Turk, M.J., Liu, C., Duan, F., Rizzuto, G., Levionnois, E., Nizard, M., Si-Mohamed, A., Besnier, N., et al. (2013). PD-1-express-
Cohen, A.D., Merghoub, T., Wolchok, J.D., and Houghton, A.N. (2006). ing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated
Optimization of a self antigen for presentation of multiple epitopes in cancer immu- head and neck cancer. Cancer Res. 73, 128–138.
nity. J. Clin. Invest. 116, 1382–1390. 45. Fourcade, J., Sun, Z., Pagliano, O., Chauvin, J.-M., Sander, C., Janjic, B., Tarhini, A.A.,
31. Weinrich, S.L., Pruzan, R., Ma, L., Ouellette, M., Tesmer, V.M., Holt, S.E., Bodnar, Tawbi, H.A., Kirkwood, J.M., Moschos, S., et al. (2014). PD-1 and Tim-3 regulate the
A.G., Lichtsteiner, S., Kim, N.W., Trager, J.B., et al. (1997). Reconstitution of human expansion of tumor antigen-specific CD8 5 T cells induced by melanoma vaccines.
telomerase with the template RNA component hTR and the catalytic protein subunit Cancer Res 74, 1045–1055.
hTRT. Nat. Genet. 17, 498–502. 46. Pauken, K.E., Sammons, M.A., Odorizzi, P.M., Manne, S., Godec, J., Khan, O., Drake,
32. Lingner, J., Hughes, T.R., Shevchenko, A., Mann, M., Lundblad, V., and Cech, T.R. A.M., Chen, Z., Sen, D.R., Kurachi, M., et al. (2016). Epigenetic stability of exhausted
(1997). Reverse transcriptase motifs in the catalytic subunit of telomerase. Science T cells limits durability of reinvigoration by PD-1 blockade. Science 354, 1160–1165.
276, 561–567. 47. Romano, E., Kusio-Kobialka, M., Foukas, P.G., Baumgaertner, P., Meyer, C.,
33. Shi, L.Z., Fu, T., Guan, B., Chen, J., Blando, J.M., Allison, J.P., Xiong, L., Subudhi, S.K., Ballabeni, P., Michielin, O., Weide, B., Romero, P., and Speiser, D.E. (2015).
Gao, J., and Sharma, P. (2016). Interdependent IL-7 and IFN-g signalling in T-cell Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by
controls tumour eradication by combined a-CTLA-4+a-PD-1 therapy. Nat. nonclassical monocytes in melanoma patients. Proc. Natl. Acad. Sci. USA 112,
Commun. 7, 12335. 6140–6145.

Molecular Therapy Vol. 26 No 2 February 2018 11

You might also like