Abstract
The interplay between tumours and the immune system has long been known to involve complex interactions between tumour cells, immune cells and the tumour microenvironment. The progress of checkpoint inhibitors in the clinic in the last decade has highlighted again the role of the immune system in the fight against cancer. Numerous efforts have been undertaken to develop ways of stimulating the cellular immune response to eradicate tumours. These interventions include the identification of appropriate tumour antigens as targets for therapy. In this review, we summarize progress in selection of target tumour antigen. Targeting self antigens has the problem of thymic deletion of high-affinity T-cell responses leaving a diminished repertoire of low-affinity T cells that fail to kill tumour cells. Thymic regulation appears to be less stringent for differentiation of cancer–testis antigens, as many tumour rejection antigens fall into this category. More recently, targeting neo-epitopes or post-translational modifications such as a phosphorylation or stress-induced citrullination has shown great promise in preclinical studies. Of particular interest is that the responses can be mediated by both CD4 and CD8 T cells. Previous vaccines have targeted CD8 T-cell responses but more recently, the central role of CD4 T cells in orchestrating inflammation within tumours and also differentiating into potent killer cells has been recognized. The design of vaccines to induce such immune responses is discussed herein. Liposomally encoded ribonucleic acid (RNA), targeted deoxyribonucleic acid (DNA) or long peptides linked to toll-like receptor (TLR) adjuvants are the most promising new vaccine approaches. These exciting new approaches suggest that the ‘Holy Grail’ of a simple nontoxic cancer vaccine may be on the horizon. A major hurdle in tumour therapy is also to overcome the suppressive tumour environment. We address current progress in combination therapies and suggest that these are likely to show the most promise for the future.
Keywords: checkpoint inhibitors, cytotoxic CD4 T cells, neo-epitopes, stress-induced post-translational modifications, T-cell avidity
Introduction
Prophylactic vaccines, stimulating protective antibody responses, have been very effective at preventing infectious disease and cancers induced by the human papilloma virus (HPV) and associated with the hepatitis B virus (HBV). In contrast, vaccines administered after disease onset to induce a therapeutic cellular immune response have yet to show significant efficacy. This is in part due to the disease process that induces an immunosuppressive environment. Induction of cellular immunity requires uptake of antigens by antigen-presenting cells (APCs) such as dendritic cells (DCs). These cells migrate to the local draining lymph node, process antigens and present peptides on major histocompatibility complex-1 (MHC-I) and MHC-II molecules. Typically, peptides presented on MHC-II stimulate the T-cell receptors (TCRs) of CD4 T cells that secrete a variety of cytokines to amplify the immune response. CD4 T cells are the first cells to migrate to the site of infection, or tumour site. They release cytokines such as interferon gamma (IFNγ) and tumour necrosis factor alpha (TNFα) which can directly induce apoptosis of target cells and also upregulate MHC to enhance T-cell killing. Furthermore, they induce an inflammatory cascade to promote the extravasation of other immune effector cells such as CD8 T cells whose TCRs recognize peptide presented on MHC-I. Both CD4 and CD8 T cells not only respond to TCR stimulation and are influenced by cytokines, but also a wide variety of costimulatory molecules that either amplify or repress TCR signalling. These include toll-like receptor (TLR) ligands (reviewed in Sasai and Yamamoto1) and costimulatory molecules such as OX40, ICOS and CD28, as well as negative regulatory mechanisms acting through CTLA4 and PD-1 pathways (summarized in Table 1).2
Table 1.
Coreceptor | Activatory/ inhibitory |
Expressed on | Ligand | Ligand expression |
---|---|---|---|---|
CD4 | Both | T cells, macrophages, monocytes | MHC-II | DC, some T cells, B cells, macrophages, monocytes |
CD8 | Both | T cells, DC subset | MHC-I | Most cells |
CD244 (2B4) | Both | Macrophages, monocytes, basophils, NK cells, some T cells | CD48 | Majority of leukocytes |
CD28 | Activatory | T cells | CD80 (B7-1), CD86 (B7-2) | DC, activated T cells, B cells, macrophages, monocytes |
CD27 | Activatory | T cells, B-cell subset and NK-cell subset | CD70 | Activated DC, B cells and T cells |
ICOS (CD278) | Activatory | Activated T cells, Th2 cells, NKT cells | ICOS-L (CD275/B7-H2) | DC, B cells, endothelial cells, macrophages, monocytes |
CD30 | Activatory | Activated T cells, B cells and NK cells | CD153 | B cells, activated T cells |
OX40 (CD134) | Activatory (inhibitory for Tregs) | Activated DC, B cells, T cells and monocytes and macrophages | OX40 ligand (CD252) | Activated T cells, Treg, NK cells, NKT cells and neutrophils |
4-1BB (CD137) | Activatory | Macrophage, monocytes, DCs, NK cells and activated T cells | 4-1BBL (CD137 ligand) | DC, B cells, macrophages, monocytes |
CD40L (CD154) | Activatory | Platelets, activated T and B cells | CD40 | DC, B cells, macrophages, monocytes, endothelial cells |
CD270 | Activatory | DC, B cells, T cells, NK cells, macrophages, monocytes | CD258, CD272, CD160 | DC, activated T and B cells, NK cells, monocytes, macrophages |
GITR (CD357) | Activatory | NK cells, activated T cells, Tregs | GITR ligand | DC, B cells, macrophages, monocytes, endothelial cells |
CTLA4 (CD152) | Inhibitory | B cells, activated T cells | CD80 (B7-1), CD86 (B7-2) | DC, activated T cells, B cells, macrophages, monocytes |
CD160 | Inhibitory | T cells, NK cells, NKT cells | CD270, MHC-I | DC, T cells, B cells, NK cells, macrophages, monocytes |
CD223 (LAG3) | Inhibitory | B cells, NK cells, activated T cells | MHC-II | DC, T cells, B cells, macrophages, monocytes |
CD272 (BTLA) | Inhibitory | Activated T cells, B cells, DCs, Th1 cells | CD270 | DC, T cells, B cells, NK cells, macrophages, monocytes |
CD274 (PD-L1/B7-H1) | Inhibitory | DC, T cells, B cells, NK cells, macrophages, monocytes | CD80 (B7-1) | DC, activated T cells, B cells, macrophages, monocytes |
PD-1 (CD279) | Inhibitory | Activated B cells, DCs and T cells, monocytes, macrophages | CD274 (PD-L2/B7-H1) CD273 (PD-L2/B7-H2) |
DC, T cells, B cells, NK cells, macrophages, monocytes |
TIM3 | Inhibitory | DC, B cells, T cells, monocytes, macrophages, NK cells | Galectin 9 | T cells, Tregs |
DC, dendritic cell; GITR, glucocorticoid-induced tumour necrosis factor receptor; MHC, major histocompatibility complex; NK, natural killer (cell); NKT, natural killer T (cell); Th, T-helper cell; Treg, regulatory T cell.
In the early stages of tumourigenesis, tumours express a range of stress-related molecules that activate DCs and the immune system. This process is termed ‘immunosurveillance’ and can result in tumour rejection. Tumours that rapidly acquire mutations are very heterogeneous, which allows selection of clonotypes that are resistant to immune attack. This process, known as ‘immune editing’, results in increasingly immunosuppressive tumour environments and rapid tumour growth. It also results in tumours with a wide spectrum of tumour-infiltrating lymphocytes from very low levels known as ‘cold’ or ‘immune desert’ tumours to very high levels termed ‘hot’ or ‘inflamed’ tumours.3 This is particularly evident in colorectal tumours where the density and type of immune infiltrate (immunoscore) is a better prognostic indicator than tumour, node and metastasis staging.4 Some tumours, particularly pancreatic tumours have a third phenotype known as ‘immune excluded’, where a physical barrier prevents infiltration.3
The clinical benefit of first-generation cancer vaccines has been limited by the choice of antigen, suboptimal design of the vaccines and by the immunosuppressive tumour environment. Second-generation vaccines and combinatorial studies may overcome some of these limitations.
Tumour antigen
Early studies concentrated on stimulating T-cell responses targeting overexpressed self antigens. The TCR repertoire is both positively and negatively selected on self antigens in the thymus therfore high-affinity T cells to strongly binding self peptides are deleted, and low-affinity T cells to weakly expressed self antigens are not positively selected. Most self epitopes will stimulate moderate avidity T-cell responses that are only triggered by high levels of expression of the cognate MHC:peptide on the tumour. As there are about one million MHC molecules on any one tumour cell, but a billion possible T-cell epitopes, any one peptide MHC complex is only presented at low density. High-avidity T cells are stimulated by low levels of cognate MHC:peptide and rapidly kill tumour cells. In contrast, moderate-avidity T cells require much higher levels of signalling to reach their activation threshold and this is rarely achieved by tumour cells. Many early vaccines targeting self antigens triggered moderate-avidity T cells, which failed to result in tumour regression. Indeed, this could be one reason for the recent failure of the PROSTVAC cancer vaccine as it targeted the self-antigen prostate-specific antigen (PSA).
To gain immune recognition, the tumour must present an epitope that has avoided thymic regulation and binds with higher affinity than the self epitopes. This could be a neo-epitope which is formed by a mutation that occurs during oncogenesis, cancer-associated viral epitopes or post-translationally modified epitopes. More occasionally, there are epitopes from self antigens that have avoided thymic deletion.
Neo-epitopes
Tumour neo-antigens are the consequences of the genetic alteration accumulated by cancer cells during the tumourigenesis process. These can include single nucleotide variants, frameshifts and insertion and deletion (INDELs) mutations. The advent of next-generation sequence has now allowed rapid identification of these mutations. Most of these mutations are specific to each individual tumour, allowing the design of personalized vaccines. Somatic nonsynonymous mutations that accumulate in cancer should avoid thymic selection and stimulate high-avidity T cells. The higher the mutation rate within the tumour, the more possibilities of mutations that are potential targets for T cells. Unfortunately, most mutations do not increase MHC binding or TCR recognition and therefore do not stimulate strong T-cell responses. Tumours such as lung, melanoma, bladder and renal cancers that have high association with environmental carcinogens have the highest mutation rates5 and are most likely to benefit from immunotherapy approaches.6 The key to the success of these neo-epitope vaccines is the identification of immunogenic epitopes. Many algorithms have been designed to predict binding to common MHC-I alleles but programmes predicting MHC-II binding or TCR recognition are very limited. New algorithms rank neo-epitopes based upon their differential immunogenicity, thereby allowing rational neo-antigen selection for clinical immunotherapies.7
Despite these limitations, several groups have now successfully treated patients with neo-epitope vaccines. Carreno and colleagues, using a neo-epitope peptide-pulsed DC vaccine enhanced both pre-existing responses and induced responses that were undetectable prior to vaccination.8 Sahin and colleagues treated 13 melanoma patients with an ribonucleic acid (RNA) polyepitope vaccine by intranodal delivery.9 All patients developed T-cell responses against multiple neo-epitopes; the majority were CD4 responses. The cumulative rate of metastatic events was significantly reduced after vaccination, resulting in sustained progression-free survival. Two out of five patients with metastatic disease experienced objective responses but one had a late relapse due to outgrowth of a β2-microglobulin-negative tumour, and a third patient had a complete response when combined with anti-programmed cell death 1 (anti-PD-1) blockade therapy. Unexpectedly, high immunogenicity and antitumour responses of individual mutations identified their preferred recognition by CD4 T cells.10 Ott and colleagues treated 6 melanoma patients with up to 20 epitopes expressed as long peptides admixed with the poly ICLC (stabilization of Polyinosinic:polycytidylic acid (poly I:C) with poly-L-lysine double-stranded RNA) agonist Hiltonol. The vaccine induced polyfunctional CD4 and CD8 responses targeting 58 (60%) and 15 (16%) of the 97 unique neoantigens respectively. Four out of six patients had no recurrences at 25 months after vaccination, while two with recurrent disease were subsequently treated with anti-PD-1 blockade and experienced complete regression with expansion of the neo-epitope-specific T cells.11 Both these trials provide strong rationale for further development of this approach in patients whose tumours have a high mutation frequency, particularly in combination with checkpoint blockade.
The disadvantage of targeting somatic mutations is their great variability both within and between tumours. The former can lead to selection of tumours that no longer express the mutation12 and the latter requires the design of a new vaccine for each patient, which is both expensive and time consuming. An alternative is to search for ‘driver’ mutations which are intimately involved in tumourigenesis and are expressed by all cells within the tumour. One such driver mutation is BRAFV600E which can also be targeted by small-molecule inhibitors13 but has yet to be shown as a target of cell-mediated immune responses. However, inhibition of the mutant BRAF has been shown to enhance T-cell responses.14 It remains to be seen if other common mutations can be identified and if they have a clinical utility, as they will only bind to selective human leukocyte antigen (HLA) molecules. A better alternative may be investigation of frameshift mutations, splice variants and INDELs.15
Although neo-epitope vaccines look promising for tumours with high mutational burden, many tumours have a low mutation frequency, meaning identification of neo-epitopes may be difficult and a different vaccine for these patients remains a high priority.
Post-translational modifications
Another attractive category of ‘foreign’ antigens, not subject to thymic deletion, derives from post-translational modifications in cancer-associated proteins. In many instances, these modifications are the result of the abnormal metabolism of cancer cells, such as abnormal phosphorylation or glycosylation. Cancer cells can present phosphorylated peptides in complex with MHC-I or MHC-II. Phosphorylation cascades are often altered in tumour cells, resulting in generation of novel phosphopeptides or an increase of phosphorylated proteins. Tumour-specific phosphopeptides have been shown to stimulate CD8 and CD4 responses and repertoires of T cells recognizing these tumour antigens have been detected in human subjects.16–18 Results of a phase I trial incorporating two such peptides are awaited.
Aberrant N-acetylglucosamine (O-GlcNAc) can correlate with augmented cancer cell proliferation, survival, invasion and metastases. Elution of MHC-I peptides from cancer cells have identified numerous O-GlcNAc-modified epitopes. These peptides can stimulate multifunctional T-cell responses to the glycosylated but not the unglycosylated peptide and may form the basis for future cancer vaccines.19
In other instances, post-translational changes are the result of the stresses to which cancer cells are often subjected. A case in point is stress-induced autophagy leading to the post-translational conversion of arginine to citrulline by peptidylarginine deiminase (PAD) enzymes. These modified epitopes are presented on MHC-II and stimulate CD4 T-cell responses.20 To this end, we have demonstrated that stress-induced citrullinated peptide epitopes induce potent, cytotoxic CD4 T-cell-mediated, antitumour responses that could constitute a completely new class of cancer treatment.21
Nutrient and oxygen deprivation, endoplasmic reticulum (ER) stress, as well as accumulated deoxyribonucleic acid (DNA) damage by rapidly dividing cancer cells creates a ‘stressed’ environment. The ability of established tumours to grow and survive depends largely on autophagic flux. However, the majority of tumours do not express MHC-II, as the immunosuppressive tumour environment inhibits inflammation. Vaccinating with citrullinated peptides induces pro-inflammatory, CD4 Th1 cells that are initially reactivated within the tumour environment by APCs, constitutively expressing MHC-II and autophagy. These reactivated CD4 T cells release IFNγ, which upregulates class MHC-II on the tumours, allowing direct recognition by the cytotoxic CD4 T cells. We have shown that citrullinated peptides stimulate CD4 T cells, which do not require CD8 responses for antitumour immunity,21,22 and leave a memory response to prevent recurrence. Although our first vaccine targets are vimentin and enolase which are expressed by many cancers, other commonly expressed cytoskeletal, glycolytic, regulatory and chaperone proteins are also citrullinated under conditions of cellular stress.23–25 In theory, combinations of a select few citrullinated peptides could be used to target all major solid cancers resulting in a step change in the approach to treating tumours. In addition to citrullination, other stress-induced post-translational modifications (siPTM) have been observed within MHC-bound peptides.26,27 Our overarching hypothesis is that these vaccines based upon siPTM of proteins will elicit new or boost pre-existing immunity that will eliminate tumours with a low T-cell infiltrate.
A similar but very different approach is to create a novel multivalent vaccine by disrupting the degradation of intracellular proteins by the ubiquitin proteasome system. This also involves autophagosome products and is branded as the DRibbles vaccine, containing DRiPs (defective ribosomal products) and SLiPs (short-lived proteins), including tumour-associated antigens (TAAs). This involves blocking and stabilizing these agents and harvesting autophagosomes by membrane disruption and fractionalization to create an ‘autologous vaccine’. Preclinical work showed that the DRibbles approach was much better than a whole-cell vaccine at enhancing IFNγ expression.28 Combining this approach with anti-OX40 antibodies to provide costimulation enhanced protection in mice.29
Viral epitopes
Cancer caused by viruses and other infectious agents such as Helicobacter pylori constitute approximately 20% of cancer worldwide. In theory, none of the T cells recognizing these viruses should be subjected to thymic tolerance. Indeed, vaccines for ‘HBV’ and HPV are very effective at inducing protective antibody responses to reduce the cancer risk, if they are administered before exposure to the virus.30 Despite the fact that many of the viral vaccines are directed against viral proteins involved in malignant transformation they are less effective once the virus has established a tumour. Antigen vaccination is preferable to vaccination with the whole virus as the latter exploits immune evasion and immune suppressive mechanisms that they have developed in the course of evolution. Indeed, the expression of E6 and E7 oncogenes by HPV can drive persistence and increase chance of cancer. Expression of these oncoproteins also influences innate immunity and promotes a suppressive tumour environment (reviewed in Smola et al.31). The best responses to date have been observed in patients with premalignant diseases such as cervical intraepithelial neoplasia or vulvar intraepithelial neoplasia, with up to 50% of patients achieving a partial or complete response after vaccination targeting HPV oncoproteins.32–36 Yet this vaccine was of limited value in established cancers37 due to the same peripheral tolerance/exhaustion mechanisms which operate in persistent infections and nonviral cancers.31 Combination studies described in the checkpoint inhibitor section below section show good synergies and are resulting in more encouraging immune responses. Similar findings have been observed with a DC-based vaccine targeting the latent membrane protein 1 (LMP1) and 2 proteins from Epstein-Barr virus (EBV) in nasopharangeal carcinoma.38 Other EBV-targeted vaccines have reached phase clinical trials but have yet to prove efficacy in phase II studies.39,40 Chronic infection with HBV and hepatitis C virus (HCV) can cause hepatocellular cancer (HCC). Therapeutic vaccines targeting HBV or HCV are more complex than other viral-induced cancers, as they do not contain oncogenic proteins but induce cancer due to inflammatory events. Most vaccines try to remove the infection prior to carcinogenesis but no good candidate has as yet been identified.41
Cancer–testis antigens and differentiation antigens
As previously discussed, high-avidity T cells recognizing self antigens are frequently deleted in the thymus leaving an attenuated low-avidity repertoire. However, thymic tolerance is not always complete, as has been elegantly shown by cloning T cells from regressing cancer patients. These CD8 T cells recognize differentiation antigens such as tyrosinase-related protein 2 (TRP-2)42,43 or cancer–testis antigens.44 Therapeutic vaccination of metastatic melanoma patients with peptides encoding these antigens was followed by tumour regression in a minority of the patients. In patients who do respond to the vaccine, the antivaccine T cells probably succeed in focally reversing this tumour-mediated immunosuppression and trigger a broad activation of other antitumour T cells, which proceed to destroy the tumour.45
In order to selectively stimulate high-avidity T cells that are capable of killing tumour cells, it is necessary to stimulate with low-dose antigen presented on activated DCs.46,47 A DNA vaccine, SCIB1, incorporating HLA-A*0201 restricted epitopes from differentiation antigens glycoprotein 100 (gp100) and TRP-2 plus HLA-DR*0401 and HLA-DR7/DQ6/DR53 restricted epitopes from gp100 into the Complementarity-determining regions (CDR) regions of a human immunoglobulin G1 (IgG1) monoclonal antibody (mab) was administered to 35 melanoma patients. SCIB1 induced dose-dependent T-cell responses in 88% of patients with no serious adverse effects or dose-limiting toxicities. An 8 mg dose stimulated the strongest immune responses, and these were stronger in disease-free patients than in patients with detectable tumour. A total of 2/15 patients with measurable disease showed objective reductions in tumour burden, and 7/15 showed stable disease. All 20 fully resected patients showed a T-cell response and remained alive with a median observation time of 37 months. Clinical response was related to MHC-I/MHC-II expression on tumours prior to therapy (p = 0.007) whilst tumour recurrence was related to loss of MHC-I, loss of target antigens, a lack of a CD4 T-cell infiltrate and expression of programmed cell death ligand 1 (PD-L1).48
A similar approach was used with RNA vaccines encoding the cancer–testis antigen, NY-ESO-1 or tyrosinase complexed with liposomes and delivered intranodally, ensuring delivery to DCs. T-cell responses were induced to these antigens, although they were at lower frequency than those induced to neo-epitope vaccines administered to the same patients.9
A multipeptide melanoma vaccine containing six peptides encoding CD4 epitopes from melanocytic proteins induced high Th1 CD4 responses, as well as durable clinical responses and durable stable disease up to 7 years later in 7–12% of patients. Overall, survival for patients who received the vaccine was significant when compared with matched paired controls (5-year survival was 57% versus 16%, respectively (p < 0.001). Epitope spreading to CD8 T cells was induced in 45% of patients.49–52 Ongoing trials are testing this vaccine with checkpoint blockade and BRAF/Mitogen-activated protein kinase kinase (MEK) inhibition.
In addition, the prostate-specific antigen (PSA) has been used as a target antigen in the PROSTVAC vaccine. This has limited normal tissue expression and may induce less immune tolerance. This showed promise in preclinical testing and early clinical trial but has had limited efficacy in a recent phase III clinical study.53,54 One of the limitations of the use of whole antigen is the competition in MHC binding from all possible peptide epitopes within the same APC. Thus, leading to the preferential binding of the highest-affinity peptides, which are most likely to be those subject to immune tolerance. The delivery of this vaccine as a recombinant viral vector vaccine may also have contributed to reduced efficacy, since immune responses may be subverted away from the target antigen and induced against the viral vector.
Optimal vaccine design
Stimulating any T-cell response requires uptake, processing and presentation on activated DCs. The DCs are usually activated by adjuvants, classically targeting TLRs. The dose of vaccine is also crucial, as low-but-sufficient dose is required to select the highest-affinity TCR to give the highest-avidity CD8 T-cell response.47 In contrast, CD4 T cells require a higher dose, as binding to MHC-II is lower affinity. This is difficult to achieve with a complex vaccine that presents multiple epitopes, such as cell-based,55 protein56 or oncolytic viral vaccines.57 Low doses fail to stimulate a response, and high doses present the highest-binding epitopes to which the T cells have been deleted in the thymus. If the tumour has a high mutation frequency that generates strong MHC-binding epitopes that outcompete the self epitopes, they may stimulate a de novo T-cell response. The advantage of oncolytic viruses is that they kill a low number of tumour cells in vivo, allowing efficient presentation of low-dose antigen to activated DCs in vivo. The disadvantage is that low doses may fail to stimulate efficient CD4 responses required to reverse tumour immunosuppression and perhaps explains the much lower response rate in non-injected lesions.
One solution is to use epitope-based vaccines that focus the immune response on epitopes, such as neo-epitopes and post-translational modifications that have avoided thymic deletion. Their disadvantage is that they are HLA restricted. For personalized neo-epitope vaccines, this is less of a problem, as the vaccine is designed to match a patient’s HLA type. For broader-based vaccine, selection of common HLA types such as HLA-A2, HLA-B7, B35, B44 and HLA-DP4, are desirable. The epitopes can be encoded within nucleic acid, peptide or DC vaccines. Encoded nucleic-acid vaccines are poorly transfected and translated and thereby deliver a low dose that is ideal for CD8 responses but less efficient for CD4 responses. Combining nucleic acid vaccines with electroporation (EP) or nanoparticle/liposomal delivery is more effective. In contrast, high doses of peptide are required to elicit any immune response, which is ideal for CD4 responses but induces a high-frequency, low-avidity CD8 response. New approaches with long peptides, amphiphilic peptides and linking adjuvants to peptides are showing encouraging responses. The complexity of the DC network is just beginning to be uncovered and choosing the correct subset for ex vivo loading of tumour epitopes is being explored.
Nucleic acid vaccines
The idea of using DNA vaccines to stimulate immune responses in humans was very appealing due to their simplicity and elegance. Early studies in the 1990s showed induction of strong immunity against viral and tumour antigens in small animals.58 DNA has a number of advantages as a vaccine platform. It is the only recombinant vaccine that does not induce vector immunity, allowing repeated administration. Additionally, it is safe, rapid, simple and cost effective to manufacture. It is also relatively stable at room temperature, making cold-chain transport less critical compared with other vaccine platforms. Its flexibility is enormous, allowing combinations of multiple plasmids encoding a variety of antigens.
Plasmid DNA vaccines have intrinsic adjuvant activity, resulting in recruitment of large numbers of inflammatory cells to the site of immunization. The mechanisms underlying DNA vaccine-induced immunity are complex and have yet to be fully elucidated but are thought to involve promiscuous and discriminative DNA sensors expressed by APCs. CpG oligodeoxynucleotide motifs signal through TLR9 to promote the activation and maturation of DCs.59 Interestingly, DNA vaccine activity was still observed in TLR9 knockout mice, implicating additional endosomal and cytosolic DNA sensors that mediate adjuvant activity.60 DNA sensors such as TANK Binding Kinase 1 (TBK-1) and Stimulator of interferon genes (STING) activate TLR-independent pathways and induce type I IFN.61 More recently, the helicase DDX41 was identified as a new DNA sensor in myeloid DCs.62 In addition, Retinoic acid-induced gene I (RIG-1) can also stimulate type I IFNs by sensing cytosolic DNA in association with RNA polymerase III.63 A DNA-dependent activator of IFN regulatory factors (DAI/DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immunity.64
EP, concomitant with injection of DNA vaccines has been explored as an approach to resolve the issue of poor transfection after injection with plasmid DNA alone. Brief electric pulses induce a transient increase in the permeability of cell membranes, resulting in enhanced plasmid DNA uptake and protein expression levels.65 In addition, EP is largely thought to provide an adjuvant effect with local tissue damage and subsequent expression of pro-inflammatory cytokines.66 Use of the EP device to administer a DNA vaccine caused transient pain and, on occasion, injection-site haematoma, but was successfully given on 218 occasions, including five patients who have now each received 15–17 immunizations over a period of up to 42 months. Discomfort from the EP procedure only limited treatment to three doses in a single patient.48
An alternative method of DNA delivery is the use of cationic lipids, such as polyethylenimine (PEI) or polylysine.67–72 Polyplexes and lipoplexes consisting of highly ordered structures of DNA molecules trapped within positively charged supramolecular assemblies have clearly demonstrated their transfection efficiency in vitro.72 However, aggregation in tissue fluids, toxicity and low in vivo efficiency has thus far hampered their clinical use. More recently, cationic vectors have been combined with nonionic amphiphilic polymers. Poloxamine block copolymers have a tetrafunctional structure consisting of four polyethylene oxide/polypropylene oxide (PEO/PPO) blocks centred on an ethylenediamine moiety. Block copolymers are adsorbed on nanoparticles through binding of their hydrophobic PPO segments while their hydrophilic PEO segments extend outwardly. DNA molecules collapsed into oligomolecular complexes exhibit new structural characteristics. These negatively charged nanospheres are able to deliver reporter and therapeutic genes to skeletal and heart muscle cells in vivo.73 Clinical trials are pending.
It has been known for some time that one of the components vital for induction of efficient cell-mediated immunity is the inclusion of T-cell help. Stevenson and colleagues have developed an elegant method of providing T-cell help within DNA vaccines by fusing their CD8 epitope to a CD4 epitope from tetanus toxin (FrC-DOM).74 Upon translating this into the clinic, it was found in a prostate cancer study that 29/30 patients receiving DNA with EP exhibited enhanced antibody responses and CD4 T-cell responses to DOM and 55% demonstrated detectable CD8 responses to prostate-specific membrane antigen (PSMA).75 Anti-PSMA responses were associated with an increase in PSA doubling time.75 In a second study targeting the carcinoembryonic antigen (CEA), DOM-specific immune responses demonstrated successful vaccine delivery. A total of 50% of patients without measurable disease expanded anti-CAP-1 CD8+ T cells [specific for HLA-A*0201 restricted CAP-1 peptide from CEA (aa605-613)] compared with only 20% with advanced disease. The gastrointestinal adverse event of diarrhoea was reported by 48% of patients and linked to more frequent decreases in CEA (p < 0.001) and improved immunologic responses compared with patients without diarrhoea. In advanced-disease patients, decreases in CEA were associated with better overall survival (OS) [hazard ratio (HR) = 0.14, p = 0.017]. CAP-1 peptide was detectable on MHC-I of normal bowel mucosa and primary colorectal cancer tissue by mass spectrometry, offering a mechanistic explanation for diarrhoea through CD8+ T-cell attack. It also highlights the need to be aware of autoimmune responses when targeting self antigens.76
A novel DNA vaccine approach encodes the specific T-cell epitope within the CDR region of an antibody. The constant fragment crystallizable (Fc) region of the antibody targets the high-affinity Fc receptor CD64, which is only found on activated and not immature APCs.47,77 The DNA is administered with EP which ensures good transfection, but inclusion of T-cell epitopes within the CDRs disrupts the antibody folding and leads to production of low levels of antibody. This results in higher-affinity CD8 responses than whole DNA vaccines, peptides or peptide-pulsed DC vaccines.47 In line with the CEA-DOM study, the DNA vaccine encoding epitopes within the CDR regions of a human IgG antibody stimulated T-cell responses that were stronger in patients without tumour present at screening than in patients with detectable tumour, suggesting that tumour load may attenuate the response.48 It also suggests that previous vaccine studies in patients with tumour load may have underestimated the measurable effects due to systemic or local immune suppression. DNA vaccination may therefore be particularly effective in early-stage patients with a low tumour burden.
Several groups have investigated the use of RNA for vaccination. Due to the instability of RNA, stabilized antigen encoding RNA formulations have been used and have shown antigen specific T-cell responses albeit with low antitumour activity.78,79 This could have been related to the choice of encoding whole self antigens rather than neo-epitopes, as recent studies with these RNA vaccines delivered into the lymph node stimulated potent antitumour responses.9 Similar to DNA vaccines, the efficiency of delivery of RNA is essential to the success of the vaccine. Recent studies using RNA lipoplexes have allowed systemic delivery that targets lymph nodes in vivo, where it is efficiently taken up by DCs and self adjuvants, inducing the release of type I IFN.80 Lipid nanoparticles can protect the RNA from degradation and can be engineered to express specific ligands, enabling targeting to specific cell types (reviewed in Reichmuth et al.81).
Peptide vaccines
Therapeutic vaccines require targeting and activation of DCs to stimulate both CD4 and CD8 T-cell responses. Initial trials concentrated on short peptides encoding CD8 T-cell epitopes that without CD4 help resulted in short-lived responses. Short peptides (<15 amino acids) do not require processing by APCs and can bind directly to HLA class I molecules of all nucleated cells. Thus, most of these peptides bind to non-APCs that have no costimulatory molecules and thereby cause tolerization.82–84 If the short peptide is combined with a depot, T cells elicited by the vaccine migrate to the vaccination site rather than the tumour and appear to die there.85 In contrast, immunization with synthetic long peptides (SLP) did not tolerize but stimulated a robust CD8 T-cell response due to efficient processing within APCs.82,86–88 SLPs can also encompass CD4 epitopes and it has been shown they are even more efficient at activating CD4 responses than processing from intact proteins.89,90 Furthermore, physical linkage of peptides to TLR agonists enhances the immune responses observed in three ways: (a) efficient targeting to DCs; (b) maturation of DCs to express costimulatory molecules, and secrete cytokines and chemokines; and (c) formation of an antigen depot within DCs to allow prolonged presentation of the peptide.91–94
An alternative solution is to use amphiphilic peptides that self assemble into nanostructures. In combination with poly I:C, a TLR3 agonist, they generate larger numbers of CD8 and CD4 T cells that result in reductions in the tumour growth rate in animal models.95,96
Dendritic-cell vaccines
The only approved therapeutic autologous cell-based vaccine is sipuleucel-T (Provenge®) where peripheral blood mononuclear cells are pulsed with prostate antigens. It has shown a modest survival benefit of 3 months, but the cost and time of production have severely limited its use. Dendritic cells are the major professional APC of the immune system and have been used for many years in development of vaccine therapies. Autologous DCs made in vitro by cytokine polarization are the usual candidates for this therapeutic approach. The goals for design of DC vaccines rely upon using an efficient strategy to load tumour antigen onto the DC and optimize the best way to deliver the vaccine efficiently so it can target the lymph nodes. A plethora of studies are published in which DCs derived from blood monocytes in vitro have been loaded with peptides, antigens and tumour lysates and have shown some promise in preclinical studies (reviewed in Shang et al.97 and Markov at al.98). This involves the culture of the immature DCs that are able to catch antigen by mechanisms such as macropinocytosis, phagocytosis and receptor-mediated endocytosis with the soluble antigen. Nucleic acids have also been used as a source of antigen for DCs; however, these are not able to be directly taken up by DCs. They therefore require the combination with mechanisms to internalize the nucleic acid such as electroporation, liposome formulations and viral-vector delivery. Despite efficacy in preclinical studies, these approaches have shown similar limited benefit in clinical studies.98 It is possible that this is due to a number of different issues. The activation state of the DC will determine the immune response generated, as DCs can tolerize responses, as well as activate them. It is well known that immature DCs can lead to the induction of immune tolerance.99 The extensive culture required for generation of monocyte-derived DCs may also impact their immunogenicity. Recently, work has shifted to the use of different DC subsets derived directly from blood with minimal in vitro manipulation and the incorporation of both CD4 and CD8 epitopes, with an improvement in clinical responses.100,101
The low efficacy of DC based and other vaccine therapies in clinical studies to date will suggest that generation of an immune response does not necessarily guarantee clinical outcome. This can be in part put down to the negative effect of the tumour on the immune system.
Overcoming the inhibitory tumour environment
Vaccines are likely to be more effective against immune ‘hot’ tumours which permit high immune infiltrates. ‘Cold’ or ‘immune desert’ tumours that are low or deficient in immune infiltrates often have developed suppressive mechanisms to exclude immune cells which may not be reversed by vaccines alone. These include checkpoint blockade of T cells,102–108 myeloid suppressor cells,109,110 immunosuppressive cytokines such as transforming growth factor-β and interleukin-10,111–115 and metabolic enzymes such as indoleamine 2,3 dioxygenase.116,117 To overcome these suppressive mechanisms generated by the tumour, therapies will ultimately need to combine vaccination to stimulate the immune response with other therapies that help reverse the suppressive tumour environment.
Checkpoint inhibition
Although cognate recognition of MHC:peptide by the TCR is the primary signal for T-cell activation, other costimulatory molecules can fine tune this response either positively or negatively. Some costimulatory molecules (CD28, CD27) are present on naïve cells and are essential in T-cell activation, whereas others (4-1BB, OX40 and CD40L) are expressed during T-cell activation and modulate polarization and differentiation into memory. To regulate these responses, inhibitory receptors (cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), PD-1, LAG-3, Tim-3) are induced to return the T cells to a resting state. In cancer, due to the inability of the T cells to clear the disease, persistent antigen results in chronic activation or ‘exhaustion’ of T cells. Blocking the inhibitory receptors has been shown to reverse the poor functionality of these T cells and re-establishes tumour control.118 This approach was first proven in patients with advanced melanoma, based on the improved OS of patients treated with the anti-CTLA-4 directed mab, ipilimumab.103,119 Blocking of CTLA-4 has also shown some success with long-term clinical benefit for a proportion of refractory malignant mesothelioma patients treated with the mab, tremelimumab.120 Anti-CTLA-4 antibodies have been shown to deplete regulatory T cells (Tregs) via antibody-mediated cellular cytotoxicity in murine models121,122 and it has been claimed that the clinical activity of these antibodies is mediated at least in part by this mechanism. As Tregs can inhibit both the priming and the function of vaccine-induced T-cell responses, depletion of these cells could increase vaccine efficacy. Initially, the antitumour activity of an anti-PD-1 antibody was shown in melanoma, renal cell carcinoma and lung cancer.106 These studies have now been extended to a wide range of cancers including Hodgkin’s lymphoma, head and neck cancer, urothelial tumours and microsatellite unstable tumours using antibodies that block either PD-1 or PD-L1. Nevertheless, the clinical efficacy of PD-1 pathway inhibition as monotherapy has been limited to subsets of patients within most tumour types studied to date, with response rates of 20% or less in many cancers, including common types, such as breast, colon, and prostate cancer. There is a correlation between the number of mutations within a tumour and its ability to stimulate a T-cell response, as most mutations are not immunogenic.102 A vaccine may increase the repertoire and number of responding T cells, allowing rapid tumour infiltration. In response to this brisk immune infiltrate and especially if IFNγ is released, the tumour will upregulate PD-L1 in a process known as ‘acquired resistance’.123 This will limit the efficacy of the vaccine, but synergy with checkpoint inhibition could enable eradication of tumours. In animal models, combined therapies of vaccines and checkpoint inhibition resulted in synergistic antitumour responses.124 Indeed, combination of CTLA-4 and PD-1 blockade is more effective than either modality alone in promoting the rejection of various murine tumours secreting granulocyte–macrophage colony-stimulating factor125 or McDonough Sarcoma fms-like tyrosine kinase 3 (Flt-3)126 or other vaccines.127
In animal models, SCIB1 DNA vaccination was associated with increased infiltration of CD4 and CD8 T cells within the tumour, in addition to being associated with upregulation of PD-L1 within the tumour environment. PD-1 blockade also resulted in increased CD8 T-cell infiltration and an antitumour response with 50% of mice showing long-term survival. In line with the hypothesis that PD-1/PD-L1 signalling results in inhibition of proliferation of high-avidity T cells at the tumour site, the combination of PD-1 blockade with vaccination enhanced the number and proliferation of the CD8 T-cell infiltrate.128 This resulted in a potent antitumour response with 80% survival in an aggressive mouse melanoma model. When another DNA vaccine, SCIB2, was given in combination with Treg depletion, CTLA-4 blockade or PD-1 blockade, long-term survival from established tumours was significantly enhanced to 56%, 67% and 100%, respectively.129
Clinical trials have begun to validate this concept. The response rate of melanoma patients receiving the oncolytic virus talimogene laherparepvec in combination with ipilimumab (30%) was compared with patients receiving ipilimumab alone (18%).130 Patients treated with neo-epitope vaccines that recurred showed complete responses when they were subsequently treated with checkpoint blockade. This suggests that priming a T-cell response prepares patients to respond well to checkpoint inhibition.9 In a phase II study of autologous monocyte-derived mRNA electroporated DCs plus anti-CTLA-4 in melanoma patients, the 6-month disease-control rate was 51%, which included eight complete and seven partial responses.131 In a small trial of 22 patients with incurable HPV16+ oropharyngeal cancer, the overall response rate to combined anti-HPV SLP vaccination and anti-PD-1 checkpoint blocker (nivolumab) treatment was 36% versus 16% in published monotherapy with nivolumab.132 Many of the responses were deep (two complete responses, six partial responses) and durable, warranting a randomized phase II trial.
Chemotherapy
Preclinical evidence shows that certain chemotherapeutics can synergize very effectively with therapeutic cancer vaccines. These principles have recently been carried into the clinic. For example, patients with HPV16+ recurrent or metastatic cervical cancer lived significantly longer if they had robust T-cell responses to an SLP vaccine against the HPV16 E6/E7 antigens. Vaccination was started 2 weeks after the 2-day cycle of carboplatin and paclitaxel chemotherapy, when increased immunosuppressive myeloid cell numbers had declined to the level of healthy donors. The longer survival was not due to generally better immunocompetence because high and low immune responders to vaccination had equal T-cell competence as measured by their recall responses to unrelated common microbial antigens.133
Using a spontaneous mouse model of gastrointestinal stromal tumour, oncogenic Kit tyrosine-kinase blockade augmented endogenous T-cell responses by inhibiting tumour cell production of the immunosuppressive enzyme indoleamine 2,3-dioxygenase. It was further shown that the combination of kinase inhibition and T-cell immunotherapy synergized to enhance antitumour efficacy. These findings established a critical link between cell-autonomous oncogenic signalling and cell-nonautonomous immunosuppression. It introduced the paradigm that the antitumour effects of kinase inhibition are partially T-cell dependent, and was the proof of concept for combining targeted therapy with immunotherapy.134,135
The development of new vaccine designs and identification of novel targets for vaccination is increasing rapidly and showing promising results in many preclinical studies. A few of these are also beginning to show promise in the clinical setting. Combinations with checkpoint blockade therapies and chemotherapy will ultimately lead to enhanced effects, provided adverse side effects can be minimized. Overall, vaccine design and development has progressed rapidly and shows considerable promise for the future.
Footnotes
Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
Conflict of interest statement: Lindy G Durrant is a director of Scancell Ltd. Victoria A Brentville and Katherine Cook are employees of Scancell Ltd.
Contributor Information
Victoria A Brentville, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK.
Suha Atabani, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK.
Katherine Cook, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK.
Lindy G Durrant, Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK.
References
- 1. Sasai M, Yamamoto M. Pathogen recognition receptors: ligands and signaling pathways by Toll-like receptors. Int Rev Immunol 2013; 32: 116–133. [DOI] [PubMed] [Google Scholar]
- 2. Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 2013; 13: 227–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature 2017; 541: 321–330. [DOI] [PubMed] [Google Scholar]
- 4. Galon J, Lugli A, Bifulco C, et al. World-Wide Immunoscore Task Force: meeting report from the “Melanoma Bridge”, Napoli, November 30th–December 3rd, 2016. J Transl Med 2017; 15: 212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013; 500: 415–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015; 348: 124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Łuksza M, Riaz N, Makarov V, et al. A neoantigen fitness model predicts tumor response to checkpoint blockade immunotherapy. Nature 2017; 551: 517–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Carreno BM, Magrini V, Becker-Hapak M, et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science 2015; 348: 803–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Sahin U, Derhovanessian E, Miller M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017; 547: 222–226. [DOI] [PubMed] [Google Scholar]
- 10. Kreiter S, Vormehr M, Van de Roemer N, et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 2015; 520: 692–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ott PA, Hu Z, Keskin DB, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017; 547: 217–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Verdegaal EM, De Miranda NF, Visser M, et al. Neoantigen landscape dynamics during human melanoma-T cell interactions. Nature 2016; 536: 91–95. [DOI] [PubMed] [Google Scholar]
- 13. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011; 364: 2507–2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Boni A, Cogdill AP, Dang P, et al. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Res 2010; 70: 5213–5219. [DOI] [PubMed] [Google Scholar]
- 15. Turajlic S, Litchfield K, Xu H, et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol 2017; 18: 1009–1021. [DOI] [PubMed] [Google Scholar]
- 16. Depontieu FR, Qian J, Zarling AL, et al. Identification of tumor-associated, MHC class II-restricted phosphopeptides as targets for immunotherapy. Proc Natl Acad Sci USA 2009; 106: 12073–12078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Zarling AL, Obeng RC, Desch AN, et al. MHC-restricted phosphopeptides from insulin receptor substrate-2 and CDC25b offer broad-based immunotherapeutic agents for cancer. Cancer Res 2014; 74: 6784–6795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Cobbold M, De La Pena H, Norris A, et al. MHC class I-associated phosphopeptides are the targets of memory-like immunity in leukemia. Sci Transl Med 2013; 5 (203): 203ra125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Malaker SA, Penny SA, Steadman LG, et al. Identification of glycopeptides as posttranslationally modified neoantigens in leukemia. Cancer Immunol Res 2017; 5: 376–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Ireland JM, Unanue ER. Autophagy in antigen-presenting cells results in presentation of citrullinated peptides to CD4 T cells. J Exp Med 2011; 208: 2625–2632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Brentville VA, Metheringham RL, Gunn B, et al. Citrullinated vimentin presented on MHC-II in tumor cells is a target for CD4+ T-cell-mediated antitumor immunity. Cancer Res 2016; 76: 548–560. [DOI] [PubMed] [Google Scholar]
- 22. Durrant LG, Metheringham RL, Brentville VA. Autophagy, citrullination and cancer. Autophagy 2016; 12: 1055–1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Rondas D, Crevecoeur I, D’Hertog W, et al. Citrullinated glucose-regulated protein 78 is an autoantigen in type 1 diabetes. Diabetes 2015; 64: 573–586. [DOI] [PubMed] [Google Scholar]
- 24. Goeb V, Thomas-L’Otellier M, Daveau R, et al. Candidate autoantigens identified by mass spectrometry in early rheumatoid arthritis are chaperones and citrullinated glycolytic enzymes. Arthritis Res Ther 2009; 11: R38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Barker CE, Ali S, O’Boyle G, et al. Transplantation and inflammation: implications for the modification of chemokine function. Immunology 2014; 143: 138–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Stamnaes J, Sollid LM. Celiac disease: autoimmunity in response to food antigen. Semin Immunol 2015; 27: 343–352. [DOI] [PubMed] [Google Scholar]
- 27. Mydel P, Wang Z, Brisslert M, et al. Carbamylation-dependent activation of T cells: a novel mechanism in the pathogenesis of autoimmune arthritis. J Immunol 2010; 184: 6882–6890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Yu G, Moudgil T, Cui Z, et al. Ubiquitinated proteins isolated from tumor cells are efficient substrates for antigen cross-presentation. J Immunother 2017; 40: 155–163. [DOI] [PubMed] [Google Scholar]
- 29. Yu G, Li Y, Cui Z, et al. Combinational immunotherapy with Allo-DRibble vaccines and anti-OX40 co-stimulation leads to generation of cross-reactive effector T cells and tumor regression. Sci Rep 2016; 6: 37558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Schiller JT, Castellsague X, Garland SM. A review of clinical trials of human papillomavirus prophylactic vaccines. Vaccine 2012; 30(Suppl. 5): F123–F138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Smola S, Trimble C, Stern PL. Human papillomavirus-driven immune deviation: challenge and novel opportunity for immunotherapy. Ther Adv Vaccines 2017; 5: 69–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Kenter GG, Welters MJ, Valentijn AR, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 2009; 361: 1838–1847. [DOI] [PubMed] [Google Scholar]
- 33. Welters MJ, Kenter GG, De Vos van Steenwijk PJ, et al. Success or failure of vaccination for HPV16-positive vulvar lesions correlates with kinetics and phenotype of induced T-cell responses. Proc Natl Acad Sci USA 2010; 107: 11895–11899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Bagarazzi ML, Yan J, Morrow MP, et al. Immunotherapy against HPV16/18 generates potent TH1 and cytotoxic cellular immune responses. Sci Transl Med 2012; 4: 155ra38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Kim TJ, Jin HT, Hur SY, et al. Clearance of persistent HPV infection and cervical lesion by therapeutic DNA vaccine in CIN3 patients. Nat Commun 2014; 5: 5317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Cory L, Chu C. ADXS-HPV: a therapeutic listeria vaccination targeting cervical cancers expressing the HPV E7 antigen. Hum Vaccin Immunother 2014; 10: 3190–3195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Van Poelgeest MI, Welters MJ, Van Esch EM, et al. HPV16 synthetic long peptide (HPV16-SLP) vaccination therapy of patients with advanced or recurrent HPV16-induced gynecological carcinoma, a phase II trial. J Transl Med 2013; 11: 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Chia WK, Wang WW, Teo M, et al. A phase II study evaluating the safety and efficacy of an adenovirus-DeltaLMP1-LMP2 transduced dendritic cell vaccine in patients with advanced metastatic nasopharyngeal carcinoma. Ann Oncol 2012; 23: 997–1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Si Y, Deng Z, Lan G, et al. The safety and immunological effects of rAd5-EBV-LMP2 vaccine in nasopharyngeal carcinoma patients: a phase I clinical trial and two-year follow-up. Chem Pharm Bull (Tokyo) 2016; 64: 1118–1123. [DOI] [PubMed] [Google Scholar]
- 40. Taylor GS, Jia H, Harrington K, et al. A recombinant modified vaccinia ankara vaccine encoding Epstein-Barr Virus (EBV) target antigens: a phase I trial in UK patients with EBV-positive cancer. Clin Cancer Res 2014; 20: 5009–5022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Kapoor R, Kottilil S. Strategies to eliminate HBV infection. Future Virol 2014; 9: 565–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Kawakami Y, Eliyahu S, Delgado CH, et al. Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection. Proc Natl Acad Sci USA 1994; 91: 6458–6462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Khong HT, Rosenberg SA. Pre-existing immunity to tyrosinase-related protein (TRP)-2, a new TRP-2 isoform, and the NY-ESO-1 melanoma antigen in a patient with a dramatic response to immunotherapy. J Immunol 2002; 168: 951–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Boon T, Van der Bruggen P. Human tumor antigens recognized by T lymphocytes. J Exp Med 1996; 183: 725–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Boon T, Cerottini JC, Van den Eynde B, et al. Tumor antigens recognized by T lymphocytes. Annu Rev Immunol 1994; 12: 337–365. [DOI] [PubMed] [Google Scholar]
- 46. Brentville VA, Metheringham RL, Gunn B, et al. High avidity cytotoxic T lymphocytes can be selected into the memory pool but they are exquisitely sensitive to functional impairment. PLoS One 2012; 7: e41112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Pudney VA, Metheringham RL, Gunn B, et al. DNA vaccination with T-cell epitopes encoded within Ab molecules induces high-avidity anti-tumor CD8+ T cells. Eur J Immunol 2010; 40: 899–910. [DOI] [PubMed] [Google Scholar]
- 48. Patel PM, Ottensmeier CH, Mulatero C, et al. Targeting gp100 and TRP-2 with a DNA vaccine: incorporating T cell epitopes with a human IgG1 antibody induces potent T cell responses that are associated with favourable clinical outcomein a phase I/II trial. Oncoimmunology. https://doi.org/10.1080/2162402X.2018.1433516 (accessed on 22nd Feb 2018). [DOI] [PMC free article] [PubMed]
- 49. Slingluff CL, Jr, Petroni GR, Olson W, et al. Helper T-cell responses and clinical activity of a melanoma vaccine with multiple peptides from MAGE and melanocytic differentiation antigens. J Clin Oncol 2008; 26: 4973–4980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Slingluff CL, Jr, Lee S, Zhao F, et al. A randomized phase II trial of multiepitope vaccination with melanoma peptides for cytotoxic T cells and helper T cells for patients with metastatic melanoma (E1602). Clin Cancer Res 2013; 19: 4228–4238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Hu Y, Kim H, Blackwell CM, et al. Long-term outcomes of helper peptide vaccination for metastatic melanoma. Ann Surg 2015; 262: 456–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Hu Y, Petroni GR, Olson WC, et al. Immunologic hierarchy, class II MHC promiscuity, and epitope spreading of a melanoma helper peptide vaccine. Cancer Immunol Immunother 2014; 63: 779–786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Kantoff PW, Gulley JL, Pico-Navarro C. Revised overall survival analysis of a phase II, randomized, double-blind, controlled study of PROSTVAC in men with metastatic castration-resistant prostate cancer. J Clin Oncol 2017; 35: 124–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Madan RA, Arlen PM, Mohebtash M, et al. Prostvac-VF: a vector-based vaccine targeting PSA in prostate cancer. Expert Opin Investig Drugs 2009; 18: 1001–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Faries MB, Mozzillo N, Kashani-Sabet M, et al. Long-term survival after complete surgical resection and adjuvant immunotherapy for distant melanoma metastases. Ann Surg Oncol 2017; 24: 3991–4000. [DOI] [PubMed] [Google Scholar]
- 56. Vansteenkiste JF, Cho BC, Vanakesa T, et al. Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer (MAGRIT): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2016; 17: 822–835. [DOI] [PubMed] [Google Scholar]
- 57. Andtbacka RH, Kaufman HL, Collichio F, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol 2015; 33: 2780–2788. [DOI] [PubMed] [Google Scholar]
- 58. Ulmer JB, Wahren B, Liu MA. Gene-based vaccines: recent technical and clinical advances. Trends Mol Med 2006; 12: 216–222. [DOI] [PubMed] [Google Scholar]
- 59. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol 2004; 5: 987–995. [DOI] [PubMed] [Google Scholar]
- 60. Yanai H, Ban T, Taniguchi T. High-mobility group box family of proteins: ligand and sensor for innate immunity. Trends Immunol 2012; 33: 633–640. [DOI] [PubMed] [Google Scholar]
- 61. Ishii KJ, Coban C, Kato H, et al. A toll-like receptor-independent antiviral response induced by double-stranded B-form DNA. Nat Immunol 2006; 7: 40–48. [DOI] [PubMed] [Google Scholar]
- 62. Zhang Z, Yuan B, Bao M, et al. The helicase DDX41 senses intracellular DNA mediated by the adaptor STING in dendritic cells. Nat Immunol 2011; 12: 959–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Tang CK, Pietersz GA. Intracellular detection and immune signaling pathways of DNA vaccines. Expert Rev Vaccines 2009; 8: 1161–1170. [DOI] [PubMed] [Google Scholar]
- 64. Takaoka A, Wang Z, Choi MK, et al. DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response. Nature 2007; 448: 501–505. [DOI] [PubMed] [Google Scholar]
- 65. Roos AK, Eriksson F, Timmons JA, et al. Skin electroporation: effects on transgene expression, DNA persistence and local tissue environment. PLoS One 2009; 4: e7226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Chiarella P, Massi E, De Robertis M, et al. Electroporation of skeletal muscle induces danger signal release and antigen-presenting cell recruitment independently of DNA vaccine administration. Expert Opin Biol Ther 2008; 8: 1645–1657. [DOI] [PubMed] [Google Scholar]
- 67. Felgner PL, Gadek TR, Holm M, et al. Lipofection: a highly efficient, lipid-mediated DNA-transfection procedure. Proc Natl Acad Sci USA 1987; 84: 7413–7417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Gao X, Huang L. A novel cationic liposome reagent for efficient transfection of mammalian cells. Biochem Biophys Res Commun 1991; 179: 280–285. [DOI] [PubMed] [Google Scholar]
- 69. Behr JP, Demeneix B, Loeffler JP, et al. Efficient gene transfer into mammalian primary endocrine cells with lipopolyamine-coated DNA. Proc Natl Acad Sci USA 1989; 86: 6982–6986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Boussif O, Lezoualc’h F, Zanta MA, et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci USA 1995; 92: 7297–7301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Midoux P, Mendes C, Legrand A, et al. Specific gene transfer mediated by lactosylated poly-L-lysine into hepatoma cells. Nucleic Acids Res 1993; 21: 871–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Pitard B. Supramolecular assemblies of DNA delivery systems. Somat Cell Mol Genet 2002; 27: 5–15. [DOI] [PubMed] [Google Scholar]
- 73. Cany J, Barteau B, Tran L, et al. AFP-specific immunotherapy impairs growth of autochthonous hepatocellular carcinoma in mice. J Hepatol 2011; 54: 115–121. [DOI] [PubMed] [Google Scholar]
- 74. Rice J, Buchan S, Stevenson FK. Critical components of a DNA fusion vaccine able to induce protective cytotoxic T cells against a single epitope of a tumor antigen. J Immunol 2002; 169: 3908–3913. [DOI] [PubMed] [Google Scholar]
- 75. Chudley L, McCann K, Mander A, et al. DNA fusion-gene vaccination in patients with prostate cancer induces high-frequency CD8(+) T-cell responses and increases PSA doubling time. Cancer Immunol Immunother 2012; 61: 2161–2170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. McCann KJ, Mander A, Cazaly A, et al. Targeting carcinoembryonic antigen with DNA vaccination: on-target adverse events link with immunologic and clinical outcomes. Clin Cancer Res 2016; 22: 4827–4836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Metheringham RL, Pudney VA, Gunn B, et al. Antibodies designed as effective cancer vaccines. MAbs 2009; 1: 71–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Hess PR, Boczkowski D, Nair SK, et al. Vaccination with mRNAs encoding tumor-associated antigens and granulocyte-macrophage colony-stimulating factor efficiently primes CTL responses, but is insufficient to overcome tolerance to a model tumor/self antigen. Cancer Immunol Immunother 2006; 55: 672–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Hong HS, Koch SD, Scheel B, et al. Distinct transcriptional changes in non-small cell lung cancer patients associated with multi-antigenic RNActive(R) CV9201 immunotherapy. Oncoimmunology 2016; 5: e1249560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Kranz LM, Diken M, Haas H, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 2016; 534: 396–401. [DOI] [PubMed] [Google Scholar]
- 81. Reichmuth AM, Oberli MA, Jeklenec A, et al. mRNA vaccine delivery using lipid nanoparticles. Ther Deliv 2016; 7: 319–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Bijker MS, Van den Eeden SJ, Franken KL, et al. CD8+ CTL priming by exact peptide epitopes in incomplete Freund’s adjuvant induces a vanishing CTL response, whereas long peptides induce sustained CTL reactivity. J Immunol 2007; 179: 5033–5040. [DOI] [PubMed] [Google Scholar]
- 83. Toes RE, Blom RJ, Offringa R, et al. Enhanced tumor outgrowth after peptide vaccination. Functional deletion of tumor-specific CTL induced by peptide vaccination can lead to the inability to reject tumors. J Immunol 1996; 156: 3911–3918. [PubMed] [Google Scholar]
- 84. Toes RE, Offringa R, Blom RJ, et al. Peptide vaccination can lead to enhanced tumor growth through specific T-cell tolerance induction. Proc Natl Acad Sci USA 1996; 93: 7855–7860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Hailemichael Y, Dai Z, Jaffarzad N, et al. Persistent antigen at vaccination sites induces tumor-specific CD8(+) T cell sequestration, dysfunction and deletion. Nat Med 2013; 19: 465–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Zwaveling SFerreira Mota SC Nouta J et al. . Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J Immunol 2002; 169: 350–358. [DOI] [PubMed] [Google Scholar]
- 87. Sabbatini P, Tsuji T, Ferran L, et al. Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients. Clin Cancer Res 2012; 18: 6497–6508. [DOI] [PubMed] [Google Scholar]
- 88. Tsuji T, Sabbatini P, Jungbluth AA, et al. Effect of montanide and poly-ICLC adjuvant on human self/tumor antigen-specific CD4+ T cells in phase I overlapping long peptide vaccine trial. Cancer Immunol Res 2013; 1: 340–350. [DOI] [PubMed] [Google Scholar]
- 89. Rosalia RA, Quakkelaar ED, Redeker A, et al. Dendritic cells process synthetic long peptides better than whole protein, improving antigen presentation and T-cell activation. Eur J Immunol 2013; 43: 2554–2565. [DOI] [PubMed] [Google Scholar]
- 90. Zhang H, Hong H, Li D, et al. Comparing pooled peptides with intact protein for accessing cross-presentation pathways for protective CD8+ and CD4+ T cells. J Biol Chem 2009; 284: 9184–9191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Khan S, Bijker MS, Weterings JJ, et al. Distinct uptake mechanisms but similar intracellular processing of two different toll-like receptor ligand-peptide conjugates in dendritic cells. J Biol Chem 2007; 282: 21145–21159. [DOI] [PubMed] [Google Scholar]
- 92. Zom GG, Khan S, Britten CM, et al. Efficient induction of antitumor immunity by synthetic toll-like receptor ligand-peptide conjugates. Cancer Immunol Res 2014; 2: 756–764. [DOI] [PubMed] [Google Scholar]
- 93. Zom GG, Filippov DV, Van der Marel GA, et al. Two in one: improving synthetic long peptide vaccines by combining antigen and adjuvant in one molecule. Oncoimmunology 2014; 3: e947892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Zom GG, Welters MJ, Loof NM, et al. TLR2 ligand-synthetic long peptide conjugates effectively stimulate tumor-draining lymph node T cells of cervical cancer patients. Oncotarget 2016; 7: 67087–67100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Cho HI, Barrios K, Lee YR, et al. BiVax: a peptide/poly-IC subunit vaccine that mimics an acute infection elicits vast and effective anti-tumor CD8 T-cell responses. Cancer Immunol Immunother 2013; 62: 787–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Kumai T, Lee S, Cho HI, et al. Optimization of peptide vaccines to induce robust antitumor CD4 T-cell responses. Cancer Immunol Res 2017; 5: 72–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Shang N, Figini M, Shangguan J, et al. Dendritic cells based immunotherapy. Am J Cancer Res 2017; 7: 2091–2102. [PMC free article] [PubMed] [Google Scholar]
- 98. Markov OV, Mironova NL, Vlassov VV, et al. Antitumor vaccines based on dendritic cells: from experiments using animal tumor models to clinical trials. Acta Naturae 2017; 9: 27–38. [PMC free article] [PubMed] [Google Scholar]
- 99. Mahnke K, Schmitt E, Bonifaz L, et al. Immature, but not inactive: the tolerogenic function of immature dendritic cells. Immunol Cell Biol 2002; 80: 477–483. [DOI] [PubMed] [Google Scholar]
- 100. Wimmers F, Schreibelt G, Skold AE, et al. Paradigm shift in dendritic cell-based immunotherapy: from in vitro generated monocyte-derived DCs to naturally circulating DC subsets. Front Immunol 2014; 5: 165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Aarntzen EH, De Vries IJ, Lesterhuis WJ, et al. Targeting CD4(+) T-helper cells improves the induction of antitumor responses in dendritic cell-based vaccination. Cancer Res 2013; 73: 19–29. [DOI] [PubMed] [Google Scholar]
- 102. Gubin MM, Zhang X, Schuster H, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014; 515: 577–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363: 711–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Page DB, Postow MA, Callahan MK, et al. Immune modulation in cancer with antibodies. Annu Rev Med 2014; 65: 185–202. [DOI] [PubMed] [Google Scholar]
- 105. Sutmuller RP, Van Duivenvoorde LM, Van Elsas A, et al. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J Exp Med 2001; 194: 823–832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366: 2443–2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J Exp Med 1999; 190: 355–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013; 369: 122–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Condamine T, Ramachandran I, Youn JI, et al. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu Rev Med 2015; 66: 97–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Damuzzo V, Pinton L, Desantis G, et al. Complexity and challenges in defining myeloid-derived suppressor cells. Cytometry B Clin Cytom 2014. doi: 10.1002/cytob.21206. [DOI] [PubMed] [Google Scholar]
- 111. Dennis KL, Blatner NR, Gounari F, et al. Current status of interleukin-10 and regulatory T-cells in cancer. Curr Opin Oncol 2013; 25: 637–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Lippitz BE. Cytokine patterns in patients with cancer: a systematic review. Lancet Oncol 2013; 14: e218–e228. [DOI] [PubMed] [Google Scholar]
- 113. Mao Y, Poschke I, Kiessling R. Tumour-induced immune suppression: role of inflammatory mediators released by myelomonocytic cells. J Intern Med 2014; 276: 154–170. [DOI] [PubMed] [Google Scholar]
- 114. Mittal SK, Roche PA. Suppression of antigen presentation by IL-10. Curr Opin Immunol 2015; 34: 22–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Oh SA, Li MO. TGF-beta: guardian of T cell function. J Immunol 2013; 191: 3973–3979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Munn DH, Mellor AL. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol 2013; 34: 137–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Platten M, Von Knebel Doeberitz N, Oezen I, et al. Cancer immunotherapy by targeting IDO1/TDO and their downstream effectors. Front Immunol 2014; 5: 673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015; 348: 56–61. [DOI] [PubMed] [Google Scholar]
- 119. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011; 364: 2517–2526. [DOI] [PubMed] [Google Scholar]
- 120. Calabro L, Maio M. Immune checkpoint blockade in malignant mesothelioma: a novel therapeutic strategy against a deadly disease? Oncoimmunology 2014; 3: e27482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Simpson TR, Li F, Montalvo-Ortiz W, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med 2013; 210: 1695–1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Bulliard Y, Jolicoeur R, Windman M, et al. Activating Fc gamma receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J Exp Med 2013; 210: 1685–1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest 2015; 125: 3384–3391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Karaki S, Anson M, Tran T, et al. Is there still room for cancer vaccines at the era of checkpoint inhibitors. Vaccines (Basel) 2016; 4: pii: E37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Duraiswamy J, Kaluza KM, Freeman GJ, et al. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res 2013; 73: 3591–3603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Curran MA, Montalvo W, Yagita H, et al. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci USA 2010; 107: 4275–4280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Binder DC, Engels B, Arina A, et al. Antigen-specific bacterial vaccine combined with anti-PD-L1 rescues dysfunctional endogenous T cells to reject long-established cancer. Cancer Immunol Res 2013; 1: 123–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Xue W, Brentville VA, Symonds P, et al. SCIB1, a huIgG1 antibody DNA vaccination, combined with PD-1 blockade induced efficient therapy of poorly immunogenic tumors. Oncotarget 2016; 7: 83088–83100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Xue W, Metheringham RL, Brentville VA, et al. SCIB2, an antibody DNA vaccine encoding NY-ESO-1 epitopes, induces potent antitumor immunity which is further enhanced by checkpoint blockade. Oncoimmunology 2016; 5: e1169353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Chesney J, Puzanov I, Collichio F, et al. Randomized, open-label phase II study evaluating the efficacy and safety of talimogene laherparepvec in combination with ipilimumab versus ipilimumab alone in patients with advanced, unresectable melanoma. J Clin Oncol 2017: JCO2017737379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Wilgenhof S, Corthals J, Heirman C, et al. Phase II study of autologous monocyte-derived mRNA electroporated dendritic cells (TriMixDC-MEL) plus ipilimumab in patients with pretreated advanced melanoma. J Clin Oncol 2016; 34: 1330–1338. [DOI] [PubMed] [Google Scholar]
- 132. Ferris RL, Blumenschein G, Jr, Fayette,, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med 2016; 375: 1856–1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Welters MJ, Van der Sluis TC, Van Meir H, et al. Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses. Sci Transl Med 2016; 8: 334ra52. [DOI] [PubMed] [Google Scholar]
- 134. Balachandran VP, Cavnar MJ, Zeng S, et al. Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido. Nat Med 2011; 17: 1094–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Cavnar MJ, Zeng S, Kim TS, et al. KIT oncogene inhibition drives intratumoral macrophage M2 polarization. J Exp Med 2013; 210: 2873–2886. [DOI] [PMC free article] [PubMed] [Google Scholar]