Abstract

Though outcomes for pediatric cancer patients have significantly improved over the past several decades, too many children still experience poor outcomes and survivors suffer lifelong, debilitating late effects after conventional chemotherapy, radiation, and surgical treatment. Consequently, there has been a renewed focus on developing novel targeted therapies to improve survival outcomes. Cancer vaccines are a promising type of immunotherapy that leverage the immune system to mediate targeted, tumor-specific killing through recognition of tumor antigens, thereby minimizing off-target toxicity. As such, cancer vaccines are orthogonal to conventional cancer treatments and can therefore be used alone or in combination with other therapeutic modalities to maximize efficacy. To date, cancer vaccination has remained largely understudied in the pediatric population. In this review, we discuss the different types of tumor antigens and vaccine technologies (dendritic cells, peptides, nucleic acids, and viral vectors) evaluated in clinical trials, with a focus on those used in children. We conclude with perspectives on how advances in combination therapies, tumor antigen (eg, neoantigen) selection, and vaccine platform optimization can be translated into clinical practice to improve outcomes for children with cancer.

Cancer continues to be a major cause of morbidity and mortality in children and is the second leading cause of death before adolescence.1 The prognosis of pediatric cancer has markedly improved in recent years, with the average overall survival (OS) of pediatric cancers rising from 58% in 1975 to 83% in 2014.2 Therapies for pediatric cancer, however, often cause significant toxicity that leads to lifelong disability.3,4 Additionally, many children will develop aggressive malignancies refractory to maximal medical and surgical management and ultimately experience dismal outcomes.5 Central nervous system (CNS) malignancies are among the most aggressive of pediatric cancers and are notoriously difficult to manage.6,7 Decades of research into tumor biology and clinical studies evaluating chemotherapy, radiation, and surgical resection-based approaches have had limited success in improving survival outcomes.6,8 The current standard of care for pediatric high-grade gliomas (HGGs), which represent approximately 10% of all pediatric CNS tumors, comprises surgical resection followed by concomitant radiotherapy and chemotherapy (ie, temozolomide) with a median OS of 10–18 months.9–11 Highly targeted agents with more favorable toxicity profiles and improved therapeutic efficacy are urgently needed to improve the quality of life and long-term outcomes of children with cancer.12

Immunotherapy harnesses the ability of the immune system to combat infection and neoplasia and has emerged as a promising treatment modality for many pediatric and adult malignancies.12,13 Cancer immunotherapy can be broadly defined as any therapy that leverages autologous or engineered immune cells to mediate tumor killing and, as such, encompasses a variety of treatments with diverse compositions and mechanisms of action.14,15 Examples of successful immunotherapies include antibodies that block immunosuppressive pathways, such as pembrolizumab, a PD-1 immune checkpoint inhibitor that has gained approval for treating a myriad of solid tumors; talimogene laherparepvec, an oncolytic virus approved for treating melanoma; and, more recently, tisagenlecleucel—a chimeric antigen receptor (CAR) T-cell therapy approved for treating pediatric acute lymphoblastic leukemia.16–18

Our understanding of tumor immunology has grown in tandem with technical improvements in vaccine development, leading to a renewed focus on vaccination as cancer therapeutic.15,19–21 Prophylactic vaccination against microorganisms (eg, smallpox, polio, and influenza) is a highly effective method of preventing life-threatening infections.22 Vaccination relies on exposing patients to the target microorganism or a structural fragment (ie, antigen) to generate an immune response that protects against future infection.23 Similarly, therapeutic cancer vaccines can induce immune cells, particularly cytotoxic CD8+ T cells, to become activated, expanded, and licensed to mediate tumor-specific killing through recognition of tumor antigens.24 As tumor cells demonstrate much greater homology with healthy tissue than infectious microorganisms, the crux of successful cancer vaccine construction lies in safeguarding healthy tissue while inducing targeted immunity against tumor antigens preferentially expressed by tumor cells.20,21

Tumor antigen selection is critical to the success of any cancer vaccine. The tumor antigen should be highly expressed and have a high affinity for binding major histocompatibility complex (MHC) molecules, which are key factors for ensuring that the antigen is adequately presented to enable immune cell recognition and killing.25,26 Additionally, the length and number of antigens must be considered to maximize the breadth of both CD8+ and CD4+ T-cell responses, which each have unique requirements for antigen recognition: CD8+ T cells recognize peptides of 8–11 amino acids in length presented in MHC-I, whereas CD4+ T cells recognize peptides of 12–15 amino acids in length bound to MHC-II.27,28 Several different classes of tumor antigens exist that can meet these criteria. Here we discuss the unique advantages and challenges associated with each class of tumor antigens.

Tumor-associated antigens (TAAs) are native proteins that are quantitatively overexpressed by tumor cells.20,29 The advantage of TAAs is that they tend to be upregulated (ie, highly expressed) by certain tumor types and are thus conserved targets for vaccination.29 Though the majority of vaccine studies have evaluated TAAs, this class of antigen faces a number of limitations. A key challenge is that TAA-reactive T cells can be removed by central tolerance, leaving only T cells with low affinity for tumor antigen recognition.30 Additionally, there is a possible risk of damage to healthy tissue; although off-target toxicity has not yet been reported in vaccine studies, CAR T cells targeting certain TAAs have resulted in severe, dose-limiting toxicities.31,32

In contrast to TAAs, tumor-specific antigens (TSAs)—also known as neoantigens, are non-autologous proteins (ie, mutated proteins) arising from tumoral genetic instability.33 TSAs provide the advantages that they are not expressed by healthy cells and are not subject to central tolerance. As compared with TAAs, TSAs generate more targeted and higher affinity T cells, thus providing potentially more effective treatment with a lower risk of autoimmunity.34 While several hotspot mutations have been identified leading to commonly occurring TSAs for certain tumor types35,36 as well as shared frameshift mutations occurring in microsatellite unstable tumors,37 most TSAs are highly variant between individuals.38,39 Thus, off-the-shelf approaches for targeting TSAs may not be practical for most patients and will instead require patient-specific identification and selection of TSAs for use in personalized cancer vaccines.40,41 Fortunately, with the advent and increasing availability of high-throughput molecular and genomic profiling, TSAs have become practicable targets for cancer vaccination.42,43

The feasibility and safety of TSA-directed vaccination have been demonstrated in adult melanoma trials in which a TSA-directed vaccine alone or in conjunction with checkpoint blockade therapy yielded a robust clinical response.44 Importantly, TSA-directed vaccination has also shown some efficacy against glioblastoma (GBM), despite this tumor’s relatively low mutation load and immune-privileged environment.45,46 Thus, these early trials provide compelling support for the use of TSA-directed vaccination in the pediatric cancer population, particularly for treating CNS malignancies.

In this review, we discuss the different classes of cancer vaccines and antigen targets while emphasizing their application in pediatric oncology. Vaccine classes are divided according to their composition and include cellular47 (eg, dendritic cells48), peptide,49 nucleic acid50 (DNA or RNA), and viral vector-based.15,51 Each has unique advantages and drawbacks as summarized in Table 1. As the majority of vaccination studies have been performed in adults, the utility and efficacy of this approach for pediatric malignancies, which can differ markedly from their adult counterparts in terms of molecular characteristics, histology, mutational burden, and neoantigen profile, has yet to be determined.52–54 We provide an overview of past and current challenges faced in vaccine development and conclude by highlighting emerging technologies that overcome historic challenges and therefore have greater promise for treating childhood malignancies.

Table 1.

A Focused Comparison of the Different Classes of Cancer Vaccinations

Vaccination ClassAdvantagesDisadvantages
Dendritic cell (DC)• Clinical efficacy established (eg, Sipleucel-T)
• Greater control over DC activation and phenotype through ex vivo manipulation
• High cost, labor-intensive manufacturing to process patient samples ex vivo
Peptide • Synthetic and rapidly manufacturable using automated equipment
• Most modular, enabling immune programming through adjuvant selection
• Low toxicity, low risk for biocontamination
• Weakly immunogenic unless adequately formulated with immunostimulant (adjuvant) in nanoparticles
• Limited number of antigens compared with nucleic acid and viral approaches
Nucleic acid (DNA/RNA) • Rapid manufacturing using primarily automated equipment
• RNA has inherent innate immune (adjuvant) activity, shown to lead to robust T-cell responses
• DNA weakly induces T-cell immunity
• T-cell responses with RNA have been variable and depend on delivery platform and route of injection
Viral vectors • Most potent vaccines for inducing T-cell immunity
• Viruses with large genomes can accommodate many antigens and other encoded therapies
• Anti-vector immunity limits the number of injections that can be given to patients
• Use of cell-based expression systems leads to higher costs, potential biocontaminants
Vaccination ClassAdvantagesDisadvantages
Dendritic cell (DC)• Clinical efficacy established (eg, Sipleucel-T)
• Greater control over DC activation and phenotype through ex vivo manipulation
• High cost, labor-intensive manufacturing to process patient samples ex vivo
Peptide • Synthetic and rapidly manufacturable using automated equipment
• Most modular, enabling immune programming through adjuvant selection
• Low toxicity, low risk for biocontamination
• Weakly immunogenic unless adequately formulated with immunostimulant (adjuvant) in nanoparticles
• Limited number of antigens compared with nucleic acid and viral approaches
Nucleic acid (DNA/RNA) • Rapid manufacturing using primarily automated equipment
• RNA has inherent innate immune (adjuvant) activity, shown to lead to robust T-cell responses
• DNA weakly induces T-cell immunity
• T-cell responses with RNA have been variable and depend on delivery platform and route of injection
Viral vectors • Most potent vaccines for inducing T-cell immunity
• Viruses with large genomes can accommodate many antigens and other encoded therapies
• Anti-vector immunity limits the number of injections that can be given to patients
• Use of cell-based expression systems leads to higher costs, potential biocontaminants
Table 1.

A Focused Comparison of the Different Classes of Cancer Vaccinations

Vaccination ClassAdvantagesDisadvantages
Dendritic cell (DC)• Clinical efficacy established (eg, Sipleucel-T)
• Greater control over DC activation and phenotype through ex vivo manipulation
• High cost, labor-intensive manufacturing to process patient samples ex vivo
Peptide • Synthetic and rapidly manufacturable using automated equipment
• Most modular, enabling immune programming through adjuvant selection
• Low toxicity, low risk for biocontamination
• Weakly immunogenic unless adequately formulated with immunostimulant (adjuvant) in nanoparticles
• Limited number of antigens compared with nucleic acid and viral approaches
Nucleic acid (DNA/RNA) • Rapid manufacturing using primarily automated equipment
• RNA has inherent innate immune (adjuvant) activity, shown to lead to robust T-cell responses
• DNA weakly induces T-cell immunity
• T-cell responses with RNA have been variable and depend on delivery platform and route of injection
Viral vectors • Most potent vaccines for inducing T-cell immunity
• Viruses with large genomes can accommodate many antigens and other encoded therapies
• Anti-vector immunity limits the number of injections that can be given to patients
• Use of cell-based expression systems leads to higher costs, potential biocontaminants
Vaccination ClassAdvantagesDisadvantages
Dendritic cell (DC)• Clinical efficacy established (eg, Sipleucel-T)
• Greater control over DC activation and phenotype through ex vivo manipulation
• High cost, labor-intensive manufacturing to process patient samples ex vivo
Peptide • Synthetic and rapidly manufacturable using automated equipment
• Most modular, enabling immune programming through adjuvant selection
• Low toxicity, low risk for biocontamination
• Weakly immunogenic unless adequately formulated with immunostimulant (adjuvant) in nanoparticles
• Limited number of antigens compared with nucleic acid and viral approaches
Nucleic acid (DNA/RNA) • Rapid manufacturing using primarily automated equipment
• RNA has inherent innate immune (adjuvant) activity, shown to lead to robust T-cell responses
• DNA weakly induces T-cell immunity
• T-cell responses with RNA have been variable and depend on delivery platform and route of injection
Viral vectors • Most potent vaccines for inducing T-cell immunity
• Viruses with large genomes can accommodate many antigens and other encoded therapies
• Anti-vector immunity limits the number of injections that can be given to patients
• Use of cell-based expression systems leads to higher costs, potential biocontaminants

Vaccination Types

Dendritic Cell Vaccines

Dendritic cells (DCs) are “professional” antigen-presenting cells (APCs) that are specialized for processing and presenting antigens for priming CD4+ and CD8+ T cells and producing cytokines that drive expansion and differentiation of T-cell responses.55 Thus, DCs act as a bridge between innate and adaptive immunity.20,56,57 For use as cellular vaccines, autologous DCs are isolated via apheresis, matured using immunostimulatory agents (eg, filgrastim), and loaded with an antigen before reinjection into the patient via the intradermal, intravenous, intranodal, or subcutaneous route.58,59 The advantage of isolating and manipulating DCs ex vivo is that antigen loading and activation can be performed under controlled conditions. Advances in gene-editing technologies including viral transduction, RNA interference, and CRISPR/Cas9 have greatly expanded scientists’ ability to engineer DCs to optimally perform a host of antitumoral functions.60 In addition, DC vaccines are often paired with adjuvants, such as imiquimod, interleukin-2, or KLH, which may be provided in the DC cultures or concomitantly delivered to increase the magnitude and duration of the antitumor response.61 Limitations of DC-based vaccines include the labor-intensive and costly cell isolation/enrichment and ex vivo stimulation process62, potentially weak responses due to insufficient cell numbers or inadequate cell activation or phenotype; and/or T-cell inactivation from an immunosuppressive tumor environment.63 Despite these challenges, it is notable that the first cancer vaccine to receive Food and Drug Administration approval is the DC vaccine sipuleucel-T (Provenge) for prostate cancer.64

Based on data from phase I/II trials, DC-based vaccination appears to be well tolerated with minimal toxicity in children with a diverse spectrum of malignancies and pretreatment conditions.57,65–67Table 2 provides a summary of DC-based vaccine trials in children. Of note, most of these trials used autologous whole tumor lysate as antigenic material. Despite the non-randomized format of early-phase safety studies, several pediatric trials of DC-based vaccines have reported improvement in clinical outcomes, albeit transient, which directly correlated with the degree of cytotoxic T lymphocyte (CTL) response as compared to historical controls. However, it should be noted that all patients eventually progressed on therapy. A phase II trial investigating an autologous tumor lysate-pulsed DC vaccine in children with metastatic or relapsed sarcomas demonstrated a 12% increase in OS in the DC-based vaccine group compared to children receiving standard of care chemotherapy. Five-year OS of patients in the Ewing sarcoma/rhabdomyosarcoma subgroup was an unprecedented 77%68 compared with 30–50% as per historical controls.69–71 Infusion of autologous DCs and CTLs has also been shown to eradicate the minimal residual disease in children with acute myeloid leukemia following chemotherapy.72 A study evaluating an autologous tumor lysate-pulsed DC vaccine in 45 children with relapsed malignant brain tumors demonstrated efficacy.57 Median OS for relapsed HGG, GBM, and anaplastic astrocytoma patients was 13.5, 12.2, and 18.4 months, respectively, with the authors noting that HGG and atypical teratoid–rhabdoid tumor appeared to respond more favorably as compared to medulloblastoma/primitive neuro-ectodermal tumor and ependymoma. A follow-up phase IIb trial is currently ongoing (EudraCT 2009-018228-14). Benitez-Ribas recently performed a phase I study of a DC vaccine pulsed with autologous tumor cell-lines lysate as the antigen administered intradermally with KLH as the adjuvant, which is used as a source of CD4+ T-cell (helper) epitopes to augment the response, in 9 patients with newly diagnosed diffuse midline glioma (DMG) (formerly termed “diffuse intrinsic pontine glioma” or “DIPG”), a highly aggressive and universally fatal subgroup of pediatric HGG.73 A specific antitumor response was observed in 8 patients as identified by immunologic studies in peripheral blood mononuclear cells. Cerebrospinal fluid (CSF) analyses showed anti-DMG specific T lymphocytes in 2 patients. Though the authors have not yet reported clinical outcomes data, the vaccine was well tolerated and no dose-limiting toxicities were observed.

Table 2.

Trials of DC-Based Vaccines in Pediatric Patients

NCTStudy PhaseTumor TypeVaccine compositionOutcomesReference
AntigenAdjuvant
CNS
n/aIRecurrent brain tumorsAutologous RNA-pulsed DCsNonePR (1/7), SD (2/7)Caruso et al., 200465
n/aIRelapsed malignant glioma Autologous tumor peptide-pulsed DCsNoneRD patients (6): PR (1), SD (1)Rutkowski et al., 200474
CR patients (6): CCR 3 years (2)
n/aNot specifiedRecurrent malignant brain tumorsAutologous whole tumor lysate-pulsed DCsImiquimodOS:Ardon et al., 201057
HGG: 13.5 m GBM: 12.2 m AA: 18.4
NCT00107185INewly diagnosed or recurrent HGGAutologous whole tumor lysate-pulsed DCsNonePR (1/3), SD (2/3)Lasky et al., 201375
NCT02840123INew diagnosed DIPGAutologous DCs pulsed with allogeneic tumor lysateNoneNo data Benitez-Ribas et al., 201873
Solid tumor
NCT00405327IIRelapsed solid tumorsAutologous tumor peptide-pulsed DCsKLHPR (1/15), SD (5/15)Geiger et al., 200166
Recurrent alveolar rhabdomyosarcoma and Ewing sarcomaAutologous tumor peptide (breakpoint region of fusion protein)-pulsed DCsIL-2PD (15/15)Dagher et al., 200276
n/aNot specifiedAdvanced solid extra-cranial tumorsAutologous tumor lysate-pulsed DCsKLHSC-treated patients (14):Dohnal et al., 200777
- CR patients (5): CCR (4), SD (1)
- PR patient (1): PD (1)
- PD patients (8): MR (1), SD (1)
IN-treated patients (8):
- CR patients (4): CCR (3), PD (1)
- PD patients (4): PD (4
NCT00001566IIMetastatic or recurrent Ewing sarcoma and alveolar rhabdomyosarcomaAutologous tumor peptide (translocation breakpoint)-pulsed DCsIL-2CR (17/30), PR (11/30), PD (2/30)Mackall et al., 200878
n/aNot specifiedRefractory Ewing sarcoma, synovial sarcoma, neuroblastomaAutologous tumor lysate-pulsed DCsKLHCR (1/5), SD -> PD (2/5), PD (1/5)Suminoe et al., 200979
n/aIRelapsed osteosarcomaAutologous tumor lysate-pulsed DCsKLH, IL-2No evidence of tumor regressionHimoudi et al., 201280
NCT01241162IRelapsed/refractory solid tumors neuroblastoma and sarcomaAutologous tumor peptide (MAGE-A1, MAGE-A3, and NY-ESO-1 derived)-pulsed DCsImiquimodCR (1/10), SD (1/10)Krishnadas et al., 201581
NCT00923351I/IIMetastatic and relapsed high-risk sarcomasAutologous tumor lysate-pulsed DCsKLH, IL-7OS:Merchant et al., 201668
-Overall 51%
-ES/RMS: 63%
Hematological
n/aNot specifiedAMLAutologous DCs and cytokine-induced killer cellsIL-2CR (20/22), PD (2/22)Bai et al., 201572
n/aCase reportRelapsed ALLAllogeneic WT-1-pulsed DCsOK-432Relapse 14 months following treatmentSaito et al., 201567
NCT00923910I/IIPost-HSCT relapsed ALL, AML, HLAllogeneic WT-1-pulsed DCsKLHPD (5/5)Shah et al., 201682
NCTStudy PhaseTumor TypeVaccine compositionOutcomesReference
AntigenAdjuvant
CNS
n/aIRecurrent brain tumorsAutologous RNA-pulsed DCsNonePR (1/7), SD (2/7)Caruso et al., 200465
n/aIRelapsed malignant glioma Autologous tumor peptide-pulsed DCsNoneRD patients (6): PR (1), SD (1)Rutkowski et al., 200474
CR patients (6): CCR 3 years (2)
n/aNot specifiedRecurrent malignant brain tumorsAutologous whole tumor lysate-pulsed DCsImiquimodOS:Ardon et al., 201057
HGG: 13.5 m GBM: 12.2 m AA: 18.4
NCT00107185INewly diagnosed or recurrent HGGAutologous whole tumor lysate-pulsed DCsNonePR (1/3), SD (2/3)Lasky et al., 201375
NCT02840123INew diagnosed DIPGAutologous DCs pulsed with allogeneic tumor lysateNoneNo data Benitez-Ribas et al., 201873
Solid tumor
NCT00405327IIRelapsed solid tumorsAutologous tumor peptide-pulsed DCsKLHPR (1/15), SD (5/15)Geiger et al., 200166
Recurrent alveolar rhabdomyosarcoma and Ewing sarcomaAutologous tumor peptide (breakpoint region of fusion protein)-pulsed DCsIL-2PD (15/15)Dagher et al., 200276
n/aNot specifiedAdvanced solid extra-cranial tumorsAutologous tumor lysate-pulsed DCsKLHSC-treated patients (14):Dohnal et al., 200777
- CR patients (5): CCR (4), SD (1)
- PR patient (1): PD (1)
- PD patients (8): MR (1), SD (1)
IN-treated patients (8):
- CR patients (4): CCR (3), PD (1)
- PD patients (4): PD (4
NCT00001566IIMetastatic or recurrent Ewing sarcoma and alveolar rhabdomyosarcomaAutologous tumor peptide (translocation breakpoint)-pulsed DCsIL-2CR (17/30), PR (11/30), PD (2/30)Mackall et al., 200878
n/aNot specifiedRefractory Ewing sarcoma, synovial sarcoma, neuroblastomaAutologous tumor lysate-pulsed DCsKLHCR (1/5), SD -> PD (2/5), PD (1/5)Suminoe et al., 200979
n/aIRelapsed osteosarcomaAutologous tumor lysate-pulsed DCsKLH, IL-2No evidence of tumor regressionHimoudi et al., 201280
NCT01241162IRelapsed/refractory solid tumors neuroblastoma and sarcomaAutologous tumor peptide (MAGE-A1, MAGE-A3, and NY-ESO-1 derived)-pulsed DCsImiquimodCR (1/10), SD (1/10)Krishnadas et al., 201581
NCT00923351I/IIMetastatic and relapsed high-risk sarcomasAutologous tumor lysate-pulsed DCsKLH, IL-7OS:Merchant et al., 201668
-Overall 51%
-ES/RMS: 63%
Hematological
n/aNot specifiedAMLAutologous DCs and cytokine-induced killer cellsIL-2CR (20/22), PD (2/22)Bai et al., 201572
n/aCase reportRelapsed ALLAllogeneic WT-1-pulsed DCsOK-432Relapse 14 months following treatmentSaito et al., 201567
NCT00923910I/IIPost-HSCT relapsed ALL, AML, HLAllogeneic WT-1-pulsed DCsKLHPD (5/5)Shah et al., 201682

AA, anaplastic astrocytoma; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CNS, central nervous system; CR, complete response; ES, Ewing sarcoma; GBM, glioblastoma multiforme; HGG, high-grade glioma; HL, Hodgkin’s lymphoma; HSCT, hematopoietic stem cell transplantation; IN, intranodal; KLH, Keyhole limpet hemocyanin; MR, mixed response; PD, progressive disease; PR, partial response; OS, overall survival; RD, residual disease; RMS, rhabdomyosarcoma; SC, subcutaneous; SD, stable disease; WT-1, Wilms’ tumor 1.

Table 2.

Trials of DC-Based Vaccines in Pediatric Patients

NCTStudy PhaseTumor TypeVaccine compositionOutcomesReference
AntigenAdjuvant
CNS
n/aIRecurrent brain tumorsAutologous RNA-pulsed DCsNonePR (1/7), SD (2/7)Caruso et al., 200465
n/aIRelapsed malignant glioma Autologous tumor peptide-pulsed DCsNoneRD patients (6): PR (1), SD (1)Rutkowski et al., 200474
CR patients (6): CCR 3 years (2)
n/aNot specifiedRecurrent malignant brain tumorsAutologous whole tumor lysate-pulsed DCsImiquimodOS:Ardon et al., 201057
HGG: 13.5 m GBM: 12.2 m AA: 18.4
NCT00107185INewly diagnosed or recurrent HGGAutologous whole tumor lysate-pulsed DCsNonePR (1/3), SD (2/3)Lasky et al., 201375
NCT02840123INew diagnosed DIPGAutologous DCs pulsed with allogeneic tumor lysateNoneNo data Benitez-Ribas et al., 201873
Solid tumor
NCT00405327IIRelapsed solid tumorsAutologous tumor peptide-pulsed DCsKLHPR (1/15), SD (5/15)Geiger et al., 200166
Recurrent alveolar rhabdomyosarcoma and Ewing sarcomaAutologous tumor peptide (breakpoint region of fusion protein)-pulsed DCsIL-2PD (15/15)Dagher et al., 200276
n/aNot specifiedAdvanced solid extra-cranial tumorsAutologous tumor lysate-pulsed DCsKLHSC-treated patients (14):Dohnal et al., 200777
- CR patients (5): CCR (4), SD (1)
- PR patient (1): PD (1)
- PD patients (8): MR (1), SD (1)
IN-treated patients (8):
- CR patients (4): CCR (3), PD (1)
- PD patients (4): PD (4
NCT00001566IIMetastatic or recurrent Ewing sarcoma and alveolar rhabdomyosarcomaAutologous tumor peptide (translocation breakpoint)-pulsed DCsIL-2CR (17/30), PR (11/30), PD (2/30)Mackall et al., 200878
n/aNot specifiedRefractory Ewing sarcoma, synovial sarcoma, neuroblastomaAutologous tumor lysate-pulsed DCsKLHCR (1/5), SD -> PD (2/5), PD (1/5)Suminoe et al., 200979
n/aIRelapsed osteosarcomaAutologous tumor lysate-pulsed DCsKLH, IL-2No evidence of tumor regressionHimoudi et al., 201280
NCT01241162IRelapsed/refractory solid tumors neuroblastoma and sarcomaAutologous tumor peptide (MAGE-A1, MAGE-A3, and NY-ESO-1 derived)-pulsed DCsImiquimodCR (1/10), SD (1/10)Krishnadas et al., 201581
NCT00923351I/IIMetastatic and relapsed high-risk sarcomasAutologous tumor lysate-pulsed DCsKLH, IL-7OS:Merchant et al., 201668
-Overall 51%
-ES/RMS: 63%
Hematological
n/aNot specifiedAMLAutologous DCs and cytokine-induced killer cellsIL-2CR (20/22), PD (2/22)Bai et al., 201572
n/aCase reportRelapsed ALLAllogeneic WT-1-pulsed DCsOK-432Relapse 14 months following treatmentSaito et al., 201567
NCT00923910I/IIPost-HSCT relapsed ALL, AML, HLAllogeneic WT-1-pulsed DCsKLHPD (5/5)Shah et al., 201682
NCTStudy PhaseTumor TypeVaccine compositionOutcomesReference
AntigenAdjuvant
CNS
n/aIRecurrent brain tumorsAutologous RNA-pulsed DCsNonePR (1/7), SD (2/7)Caruso et al., 200465
n/aIRelapsed malignant glioma Autologous tumor peptide-pulsed DCsNoneRD patients (6): PR (1), SD (1)Rutkowski et al., 200474
CR patients (6): CCR 3 years (2)
n/aNot specifiedRecurrent malignant brain tumorsAutologous whole tumor lysate-pulsed DCsImiquimodOS:Ardon et al., 201057
HGG: 13.5 m GBM: 12.2 m AA: 18.4
NCT00107185INewly diagnosed or recurrent HGGAutologous whole tumor lysate-pulsed DCsNonePR (1/3), SD (2/3)Lasky et al., 201375
NCT02840123INew diagnosed DIPGAutologous DCs pulsed with allogeneic tumor lysateNoneNo data Benitez-Ribas et al., 201873
Solid tumor
NCT00405327IIRelapsed solid tumorsAutologous tumor peptide-pulsed DCsKLHPR (1/15), SD (5/15)Geiger et al., 200166
Recurrent alveolar rhabdomyosarcoma and Ewing sarcomaAutologous tumor peptide (breakpoint region of fusion protein)-pulsed DCsIL-2PD (15/15)Dagher et al., 200276
n/aNot specifiedAdvanced solid extra-cranial tumorsAutologous tumor lysate-pulsed DCsKLHSC-treated patients (14):Dohnal et al., 200777
- CR patients (5): CCR (4), SD (1)
- PR patient (1): PD (1)
- PD patients (8): MR (1), SD (1)
IN-treated patients (8):
- CR patients (4): CCR (3), PD (1)
- PD patients (4): PD (4
NCT00001566IIMetastatic or recurrent Ewing sarcoma and alveolar rhabdomyosarcomaAutologous tumor peptide (translocation breakpoint)-pulsed DCsIL-2CR (17/30), PR (11/30), PD (2/30)Mackall et al., 200878
n/aNot specifiedRefractory Ewing sarcoma, synovial sarcoma, neuroblastomaAutologous tumor lysate-pulsed DCsKLHCR (1/5), SD -> PD (2/5), PD (1/5)Suminoe et al., 200979
n/aIRelapsed osteosarcomaAutologous tumor lysate-pulsed DCsKLH, IL-2No evidence of tumor regressionHimoudi et al., 201280
NCT01241162IRelapsed/refractory solid tumors neuroblastoma and sarcomaAutologous tumor peptide (MAGE-A1, MAGE-A3, and NY-ESO-1 derived)-pulsed DCsImiquimodCR (1/10), SD (1/10)Krishnadas et al., 201581
NCT00923351I/IIMetastatic and relapsed high-risk sarcomasAutologous tumor lysate-pulsed DCsKLH, IL-7OS:Merchant et al., 201668
-Overall 51%
-ES/RMS: 63%
Hematological
n/aNot specifiedAMLAutologous DCs and cytokine-induced killer cellsIL-2CR (20/22), PD (2/22)Bai et al., 201572
n/aCase reportRelapsed ALLAllogeneic WT-1-pulsed DCsOK-432Relapse 14 months following treatmentSaito et al., 201567
NCT00923910I/IIPost-HSCT relapsed ALL, AML, HLAllogeneic WT-1-pulsed DCsKLHPD (5/5)Shah et al., 201682

AA, anaplastic astrocytoma; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CNS, central nervous system; CR, complete response; ES, Ewing sarcoma; GBM, glioblastoma multiforme; HGG, high-grade glioma; HL, Hodgkin’s lymphoma; HSCT, hematopoietic stem cell transplantation; IN, intranodal; KLH, Keyhole limpet hemocyanin; MR, mixed response; PD, progressive disease; PR, partial response; OS, overall survival; RD, residual disease; RMS, rhabdomyosarcoma; SC, subcutaneous; SD, stable disease; WT-1, Wilms’ tumor 1.

In summary, DC-based vaccination appears to be well tolerated and effective for inducing T-cell immunity in pediatric cancers, though additional studies will be needed to fully understand the potential of these therapies for the pediatric population.

Peptide Vaccines

An alternative to loading DCs with antigenic material ex vivo is to vaccinate patients with peptide antigens that can be processed and presented by endogenous APCs, particularly DCs, that prime T-cell immunity in lymph nodes draining the sites of vaccination.49 Peptide vaccines generally include one or more synthetic peptides comprising tumor antigens combined with immunostimulants (“adjuvants”) that are used to enhance peptide antigen immunogenicity.83,84 The length of peptide antigens is selected to maximize the breadth of the T-cell response and depends, in part, on the antigenic target. For TSAs, peptide antigens of 25 amino acids in length are typically used wherein the middle (15th amino acid) is the mutant (ie, tumor-specific) residue.85,86 This length ensures that all 8–11 amino acid CD8+ T-cell epitopes and most 12–15 amino acid CD4+ T-cell epitopes including the mutant amino acid are represented in each sequence. A similar rationale is applied to TAAs, whereby protein antigens greater than 100 amino acids in length are produced as a pool of peptide fragments overlapping by 9–14 amino acids to ensure that most CD4+ and CD8+ T-cell epitopes are represented.87,88 Finally, to maximize the breadth of T-cell responses, peptide vaccines typically composed of up to 20 unique peptide antigens, which is largely dictated by manufacturing and cost constraints.

Peptide vaccines are advantageous in that they can be produced rapidly, at relatively low cost, entirely by synthetic processes using automated equipment.89,90 Additionally, peptide vaccines are among the most modular as they, unlike, other vaccine platforms (eg, viruses), have little to no inherent immunostimulatory properties and therefore enable the quality and magnitude of the immune response to be programmed based on the adjuvant(s) used.91–93 A potential disadvantage of peptide-based vaccines is that they permit a lower antigen payload (~20 peptide antigens each of 25–35 amino acids in length) as compared with recombinant vaccine approaches (eg, viruses and nucleic acids) that can encode multiple protein antigens that are each several hundred amino acids in length.94 However, more antigens are not necessarily better. The presence of multiple antigens can lead to competition that may diminish the response against any one specific antigen.20,34 Additionally, T-cell responses directed against a single antigen can mediate durable tumor regression.95,96 Thus, it is likely that the antigen payload of peptide-based therapeutic cancer vaccines, which typically comprise about 10–20 peptide antigens, is sufficient given appropriate antigen selection.43,97

Table 3 provides a summary of peptide-based vaccine trials in children. In the pediatric setting, several early-phase studies have investigated Wilms’ Tumor gene, WT-1, targeted peptide-based vaccines in children with solid and hematologic malignancies. In a study of 5 patients with various malignancies, Hashii et al.98 found that intradermal vaccination with a single WT-1-derived short (9 amino acid) peptide antigen formulated in a water-in-oil emulsion-based adjuvant, Montanide ISA51, induced complete remission in one patient and a period of stable disease in another patient. However, clinical benefit was limited with 4 patients experiencing disease progression and/or death during the trial. In a larger study of 26 children and young adults less than 20 years old with relapsed solid and hematologic malignancies, no patients demonstrated a clinical response.99 Limited efficacy with this approach may be due to the use of only a single short (minimal epitope) peptide antigen resulting in limited T-cell breadth, and/or use of an emulsion formulation that lacks immunostimulants (eg, Toll-like receptor agonists [TLRa]) needed for strong T-cell induction100 and may instead promote T-cell exhaustion.101

Table 3.

Trials of Peptide-Based Vaccines in Pediatric Patients

NCTStudy PhaseTumor TypeVaccine CompositionOutcomesReference
AntigenAdjuvant
CNS
NCT01130077IHigh-risk gliomasIL-13 receptor alpha 2, EphA2, survivinMontanide ISA51, poly-ICLCCR (2/24), PR (2/24), MR (1/24) SD (19/24) Pollack et al., 2014102
NCT01130077IRecurrent low-grade gliomasIL-13 receptor alpha 2, EphA2, survivinMontanide ISA51, poly-ICLCPR (4/24), MR (1/24) SD (7/24)Pollack et al., 2016103
NCT02960230IDIPG, nonpontine DMGH3.3K27MMontanide ISA51, poly-ICLCOS at 12 months DIPG 44%, nonpontine DMG 39% Mueller et al., 2020104
Solid tumor
n/aIIRhabdomyosarcoma, osteosarcoma, liposarcoma, synovial sarcomaWT-1Montanide ISA51CR (1/4), SD (1/4), PD (2/4)Hashii et al., 201098
n/aI/IIRelapsed/refractory solid tumorsWT-1Montanide ISA51CR (5/9), MR (1/9), SD (1/9), PD (2/9)Sawada et al., 201699
n/aNot specifiedSolid tumorsWT-1OK-432No dataHirabayashi et al., 2018105
n/aINeuroblastomaNY-ESO-1Montanide ISA51No dataCamisaschi et al., 2018106
n/aIRefractory solid tumorsKOC1, FOXM1, KIF20AIncomplete Freund’s adjuvantSD (4/12), MR (2/12), PD (6/12)Akazawa et al., 2019107
Hematological
n/aIIALLWT-1Montanide ISA51PD (1/1)Hashii et al., 201098
n/aIIALL, AMLWT-1Montanide ISA51CR (2/3), PD (1/3)Hashii et al., 2012108
n/aI/IIALL, AML, lymphomaWT-1Montanide ISA51CR (4/4)Sawada et al., 201699
NCTStudy PhaseTumor TypeVaccine CompositionOutcomesReference
AntigenAdjuvant
CNS
NCT01130077IHigh-risk gliomasIL-13 receptor alpha 2, EphA2, survivinMontanide ISA51, poly-ICLCCR (2/24), PR (2/24), MR (1/24) SD (19/24) Pollack et al., 2014102
NCT01130077IRecurrent low-grade gliomasIL-13 receptor alpha 2, EphA2, survivinMontanide ISA51, poly-ICLCPR (4/24), MR (1/24) SD (7/24)Pollack et al., 2016103
NCT02960230IDIPG, nonpontine DMGH3.3K27MMontanide ISA51, poly-ICLCOS at 12 months DIPG 44%, nonpontine DMG 39% Mueller et al., 2020104
Solid tumor
n/aIIRhabdomyosarcoma, osteosarcoma, liposarcoma, synovial sarcomaWT-1Montanide ISA51CR (1/4), SD (1/4), PD (2/4)Hashii et al., 201098
n/aI/IIRelapsed/refractory solid tumorsWT-1Montanide ISA51CR (5/9), MR (1/9), SD (1/9), PD (2/9)Sawada et al., 201699
n/aNot specifiedSolid tumorsWT-1OK-432No dataHirabayashi et al., 2018105
n/aINeuroblastomaNY-ESO-1Montanide ISA51No dataCamisaschi et al., 2018106
n/aIRefractory solid tumorsKOC1, FOXM1, KIF20AIncomplete Freund’s adjuvantSD (4/12), MR (2/12), PD (6/12)Akazawa et al., 2019107
Hematological
n/aIIALLWT-1Montanide ISA51PD (1/1)Hashii et al., 201098
n/aIIALL, AMLWT-1Montanide ISA51CR (2/3), PD (1/3)Hashii et al., 2012108
n/aI/IIALL, AML, lymphomaWT-1Montanide ISA51CR (4/4)Sawada et al., 201699

ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CNS, central nervous system; CR, complete response; DMG, diffuse midline gliomas; MR, mixed response; PD, progressive disease; poly-ICLC, polyinosinic–polycytidylic acid complexed with poly(lysine) and carboxymethylcellulose; PR, partial response; OS, overall survival; SD, stable disease; WT, Wilms’ tumor 1.

Table 3.

Trials of Peptide-Based Vaccines in Pediatric Patients

NCTStudy PhaseTumor TypeVaccine CompositionOutcomesReference
AntigenAdjuvant
CNS
NCT01130077IHigh-risk gliomasIL-13 receptor alpha 2, EphA2, survivinMontanide ISA51, poly-ICLCCR (2/24), PR (2/24), MR (1/24) SD (19/24) Pollack et al., 2014102
NCT01130077IRecurrent low-grade gliomasIL-13 receptor alpha 2, EphA2, survivinMontanide ISA51, poly-ICLCPR (4/24), MR (1/24) SD (7/24)Pollack et al., 2016103
NCT02960230IDIPG, nonpontine DMGH3.3K27MMontanide ISA51, poly-ICLCOS at 12 months DIPG 44%, nonpontine DMG 39% Mueller et al., 2020104
Solid tumor
n/aIIRhabdomyosarcoma, osteosarcoma, liposarcoma, synovial sarcomaWT-1Montanide ISA51CR (1/4), SD (1/4), PD (2/4)Hashii et al., 201098
n/aI/IIRelapsed/refractory solid tumorsWT-1Montanide ISA51CR (5/9), MR (1/9), SD (1/9), PD (2/9)Sawada et al., 201699
n/aNot specifiedSolid tumorsWT-1OK-432No dataHirabayashi et al., 2018105
n/aINeuroblastomaNY-ESO-1Montanide ISA51No dataCamisaschi et al., 2018106
n/aIRefractory solid tumorsKOC1, FOXM1, KIF20AIncomplete Freund’s adjuvantSD (4/12), MR (2/12), PD (6/12)Akazawa et al., 2019107
Hematological
n/aIIALLWT-1Montanide ISA51PD (1/1)Hashii et al., 201098
n/aIIALL, AMLWT-1Montanide ISA51CR (2/3), PD (1/3)Hashii et al., 2012108
n/aI/IIALL, AML, lymphomaWT-1Montanide ISA51CR (4/4)Sawada et al., 201699
NCTStudy PhaseTumor TypeVaccine CompositionOutcomesReference
AntigenAdjuvant
CNS
NCT01130077IHigh-risk gliomasIL-13 receptor alpha 2, EphA2, survivinMontanide ISA51, poly-ICLCCR (2/24), PR (2/24), MR (1/24) SD (19/24) Pollack et al., 2014102
NCT01130077IRecurrent low-grade gliomasIL-13 receptor alpha 2, EphA2, survivinMontanide ISA51, poly-ICLCPR (4/24), MR (1/24) SD (7/24)Pollack et al., 2016103
NCT02960230IDIPG, nonpontine DMGH3.3K27MMontanide ISA51, poly-ICLCOS at 12 months DIPG 44%, nonpontine DMG 39% Mueller et al., 2020104
Solid tumor
n/aIIRhabdomyosarcoma, osteosarcoma, liposarcoma, synovial sarcomaWT-1Montanide ISA51CR (1/4), SD (1/4), PD (2/4)Hashii et al., 201098
n/aI/IIRelapsed/refractory solid tumorsWT-1Montanide ISA51CR (5/9), MR (1/9), SD (1/9), PD (2/9)Sawada et al., 201699
n/aNot specifiedSolid tumorsWT-1OK-432No dataHirabayashi et al., 2018105
n/aINeuroblastomaNY-ESO-1Montanide ISA51No dataCamisaschi et al., 2018106
n/aIRefractory solid tumorsKOC1, FOXM1, KIF20AIncomplete Freund’s adjuvantSD (4/12), MR (2/12), PD (6/12)Akazawa et al., 2019107
Hematological
n/aIIALLWT-1Montanide ISA51PD (1/1)Hashii et al., 201098
n/aIIALL, AMLWT-1Montanide ISA51CR (2/3), PD (1/3)Hashii et al., 2012108
n/aI/IIALL, AML, lymphomaWT-1Montanide ISA51CR (4/4)Sawada et al., 201699

ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CNS, central nervous system; CR, complete response; DMG, diffuse midline gliomas; MR, mixed response; PD, progressive disease; poly-ICLC, polyinosinic–polycytidylic acid complexed with poly(lysine) and carboxymethylcellulose; PR, partial response; OS, overall survival; SD, stable disease; WT, Wilms’ tumor 1.

Additional peptide vaccines targeting conserved tumor antigens have been tested in pediatric clinical studies. Kushner et al.109 performed a phase I trial in which 15 children with high-risk neuroblastoma were administered a vaccine containing the immunological adjuvant OPT-821 and the neuroblastoma-associated antigens GD2 and GD3. Patients were also given β-glucan, a biologic response modifier that enhances the antitumor response.110,111 No patients had dose-limiting toxicities and 12 patients demonstrated an antibody response against GD2 and/or GD3. Carcinoembryonic antigen glypican-3 (GPC3), a hepatic heparan sulfate proteoglycan expressed in many pediatric tumors such as hepatoblastoma, yolk-sac tumors, and Wilms’ tumors, has also been piloted as a target for vaccination.112,113 A phase I study of 18 pediatric patients with GPC3-expressing solid tumors found that vaccination with a single MHC-I matched GPC3-peptide formulated in a water-in-oil emulsion improved or maintained clinical status (CR + PR + SD) in 67% of patients,114 despite using a suboptimal emulsion formulation, as discussed above.

Early peptide vaccine studies in pediatric CNS malignancies have been encouraging. Pollack et al.102 investigated the safety and performance of a peptide vaccine targeting known glioma-associated antigens (IL-13Rα2, EphA2, and survivin) administered subcutaneously with polyinosinic–polycytidylic acid (poly[I:C]), a TLR-9 agonist, stabilized by poly(lysine) and carboxymethylcellulose (poly-ICLC) adjuvant in 26 children with newly diagnosed brainstem or non-brainstem gliomas. Five children had pseudoprogression, a transient increase in edema, and contrast enhancement secondary to a treatment-induced immune response that was followed by stabilization and/or regression.115 Patients with pseudoprogression were successfully treated with dexamethasone and had a higher median survival (19.5 months vs 10.9 months). A phase I study of cytomegalovirus (CMV)-specific peptide vaccine in patients with recurrent medulloblastoma and malignant glioma is ongoing (NCT03299309) and is of interest given the noted expression of CMV proteins in such tumors.116

Peptide vaccines are also being studied in DMG. Ochs et al.117 showed that vaccination with peptides derived from H3.3K27M (a unifying oncogenic mutation resulting in global methylation perturbation), and formulated in a water-in-oil emulsion administered subcutaneously, produced an effective, mutation-specific CD4+ and CD8+ T-cell-mediated immune response with antigen presentation on both MHC classes I and II in a humanized mouse model. Although they observed tumor regression in murine models, the experimental design consisted of DMG tumors in the flank rather than intracranially. This vaccine epitope is currently being tested in phase 1 clinical trial in combination with checkpoint inhibitors in children (NCT02960230).

While it is too early to quantify the potential of peptide-based vaccines for pediatric cancers, more advanced trials have been conducted in the adult population. Indeed, several phase III adult solid tumor trials have been conducted and have failed to show any clinical benefit.118–120 While these results have dampened the initial enthusiasm for peptide-based vaccine approaches, recent mechanistic data suggest that suboptimal formulations may account, in part, for the observed weak efficacy and that formulating peptide antigens in nanoparticles that target endogenous DCs that promote T-cell immunity may be needed to improve efficacy.121 Additionally, similarly to DC vaccines, peptide vaccines appear to perform optimally in patients with lower disease burden and when administered concomitantly with other treatment modalities like checkpoint inhibitors to allow for synergistic antitumor effects.122

In summary, peptide-based cancer vaccines can safely induce anticancer T-cell immunity in pediatric and adult populations; however, further studies are needed to understand how to optimize the composition (eg, delivery vehicle and adjuvant) and combination approaches with complementary therapeutic modalities to maximize T-cell responses and efficacy in children.

Nucleic Acid Vaccines

Nucleic acid vaccines utilize plasmid DNA or mRNA to express tumor antigens through transient transfection of muscle tissue or APCs (eg, DCs) following administration. Multiple genes can be incorporated into a single vector, and the nucleic acid can be modified to modulate expression and innate stimulation to augment the immune response.65,123,124 Similar to peptide-based vaccines, nucleic acid vaccines enable lower costs and more rapid manufacturing as compared with cellular or viral vector-based vaccines, but, unlike peptides, still require the use of costly recombinant enzymes for production.125 While early clinical studies in adult patients showed that DNA vaccines were sufficient in invoking a cellular and humoral immune response, there was little evidence of clinical benefit.126 Indeed, recent preclinical data suggest that the platform and site of nucleic acid administration are critical to the capacity of such approaches to induce T-cell immunity.127 Thus, numerous delivery approaches have been developed to improve antigen expression in APCs as a means to improve efficacy through the use of direct intra-lymph node injection, gene gun, electroporation, ultrasound, laser, liposome, microparticles, and/or nanoparticles.128–131 Among the nucleic acid vaccine approaches, RNA lipoplexes appear to be one of the most promising and have shown the greatest capacity for inducing anticancer T-cell immunity.127 Though additional optimization will be needed to balance gene expression with innate stimulation that may lead to systemic toxicity and blunting of antigen expression, early data suggest that RNA-based vaccination approaches may eventually have a great therapeutic impact in pediatric cancers.

Viral Vector Vaccines

Recombinant viral vectors, typically derived from the Poxviridae, Adenoviridae, and Rhabdoviridae families, are engineered to express tumor antigen transgenes and are among the most potent vaccine technologies for inducing T-cell immunity.132–134 Viral vectors provide the advantages that they can encode a large number of antigens that can be expressed at high levels and rendered immunogenic through intrinsic innate immunostimulatory capacity of the virus. Viral vectors can be further engineered to maximize gene expression, target specific cell populations, and/or encode multiple additional therapeutic modalities.135 Though viral vectors are easier to produce, purify, and store relative to more costly and labor-intensive cellular vaccines, like DC vaccines,136 their dependence on recombinant technologies can result in higher costs than synthetic peptide-based vaccine approaches, and antibodies generated against the vector (antivector immunity) can limit their use to a single administration.137

PROSTVAC, a well-studied poxviral-based vaccine targeting prostate-specific antigen that also contains transgenes for T-cell co-stimulatory molecule expression, showed promise in an early-phase II double-blind randomized trial in metastatic castration-resistant prostate cancer; however, results from the subsequent phase III trial failed to show any survival improvement over placebo.138,139 PROSTVAC in combination with the anti-CTLA-4 checkpoint inhibitor, ipilimumab, for metastatic castration-resistant prostate cancer has been proven safe in a phase I trial140 and is currently being assessed in a randomized phase II trial (NCT02933255).

The vaccine TG4010, a modified vaccinia Ankara vector expressing MUC1 and IL-2, has been evaluated in combination with first-line chemotherapy for the treatment of patients with advanced-stage non-small-cell lung cancer and was found to improve progression-free survival at 6 months.141 Though there are no current ongoing clinical trials, there are promising preclinical data to suggest that viral-based vaccines may be effective in brain tumors. Abdelaziz et al.142 designed human cytomegalovirus (HCMV)-based vaccine expressing E6-derived peptide fused to HCMV proteins. Patient-derived GBM cells infected with these vectors efficiently stimulated E6-specific T cells. Additionally, a phase I dose escalation evaluating the safety of aglatimagene besadenovec (AdV-tk), an adenoviral vector expressing herpes virus thymidine kinase, followed by anti-herpetic prodrug in pediatric malignant glioma or recurrent ependymoma found the approach to be safe in combination with radiation therapy and temozolomide143; a subsequent phase II study is planned.

Combination Immunotherapy

Therapeutic cancer vaccines principally mediate tumor clearance through the induction of cytotoxic T cells. However, cancers can evade T-cell recognition and killing by promoting an immunosuppressive environment, including through the expression of immune checkpoint molecules (eg, PD-L1) that directly inhibit T-cell killing. To overcome the immunosuppressive environment of tumors, vaccines that induce T-cell immunity should be used in combination with complementary therapies that reverse immune suppression. Indeed, many groups are starting to explore the potential of a combinatorial approach using vaccination to enhance the efficacy of other immunotherapy-based treatment modalities including checkpoint inhibitors, antiangiogenic agents, oncolytic viruses, and radiation.19,144

One of the most promising combination immunotherapies is the use of cancer vaccines with checkpoint inhibitors (eg, anti-PD-1/PD-L1 and anti-CTLA-4). Myriad preclinical studies have shown that cancer vaccines used in combination with checkpoint inhibitors lead to significantly improved efficacy as compared with either treatment used alone,145 and now multiple clinical trials are ongoing evaluating this combination in patients.19 Among the most promising studies thus far, a phase I trial found that a GM-CSF cell-based vaccine (“GVAX”) in combination with ipilimumab, an anti-CTLA-4 antibody, enhanced the preexisting endogenous tumor-specific T-cell response compared to treatment with ipilimumab alone. Posttreatment expansion of the mesothelin-specific T-cell repertoire was associated with a significant improvement in OS in the combination arm, suggesting that the frequency of preexisting mesothelin-specific T cells are low and require a vaccine to induce larger pools of precursor T-cells.146

The use of viruses to modulate the tumor microenvironment and provide a more permissive environment for T-cell killing is another promising strategy that is gaining increasing attention. Accordingly, Koske et al.147 demonstrated that combination treatment of Vesicular Stomatitis Virus-glycoprotein (VSV-GP), a chimeric VSV pseudotyped with GP of the lymphocytic choriomeningitis virus, followed by an ovalbumin peptide-loaded DC vaccine, significantly enhanced survival over either agent alone in a murine melanoma model. The authors found that this strategy alleviated local immune suppression in the tumor microenvironment by reducing regulatory T cells, activating tumor-infiltrating lymphocytes, and increasing inflammatory cytokines.

Though most combinatorial approaches have been tested in adults, there are a number of ongoing trials in the pediatric setting. A phase I trial using GVAX in combination with nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4) for refractory neuroblastoma is ongoing (NCT04239040). Krishnadas et al. reported complete remission in a patient with relapsed stage 4 neuroblastoma after treatment with decitabine to upregulate cancer testis antigen expression, followed by DC vaccine targeting the cancer testis antigens MAGE-A1, MAGE-A3, and NY-ESO. Additional phase I studies are underway to evaluate concomitant vaccination and radiation for pediatric HGG (NCT03615404, NCT02722512, and NCT00634231).

Challenges and Emerging Technologies

While a variety of cancer vaccines have been evaluated in adult and pediatric patients, most have provided only modest efficacy. The shortcomings of prior vaccines may be in part due to weak immunogenicity of the vaccine leading to insufficient magnitude of CD8+ T-cell responses; inadequate, or loss, of expression of the tumor antigen, thereby enabling tumor cells to evade recognition; and/or, inability of vaccine-induced T cells to overcome the suppressive tumor microenvironment.15,20,148 Recent advances in tumor antigen identification and selection algorithms are enabling more reliable antigen selection, and checkpoint inhibitors have proven effective for unleashing T cells to mediate tumor-specific killing. A key challenge remains the availability of vaccine technologies for reliably inducing high-magnitude CD8+ T-cell responses that correlate with immunotherapy efficacy.15,20,148 Indeed, patients treated with a peptide-based vaccine comprising peptide neoantigens admixed with the immunostimulant polyICLC (Hiltonol) experienced limited benefit from therapy despite promising preclinical data.85 Weak efficacy may be, in part, attributed to inadequate formulation of the peptide neoantigens (see below), though limitations in neoantigen prediction and selection cannot be ruled out as their validation is not possible in the absence of clear vaccine-mediated efficacy.

Toward improving peptide-based vaccine formulations for inducing CD8+ T-cell immunity, Lynn et al.121 developed a vaccine platform based on peptide antigen–TLRa conjugates that are programmed to self-assemble into nanoparticles of an optimal size (~20 nm) for targeting lymph node resident DCs that promote T-cell immunity. Programmed self-assembly was developed to ensure formulation consistency for all possible antigens that can be generated from the human genome, thus enabling a universal approach for formulating tumor antigens. Importantly, improved formulation of tumor antigens, including neoantigens, with TLRa in self-assembling nanoparticles promoted improved uptake by and activation of APCs (eg, DCs) that was associated with enhanced immunogenicity and improved tumor clearance in 3 murine tumor models.121 They also found that peptide physical form is a key determinant of CD8+ T-cell immunogenicity.

Specifically, they showed that hydrophilic, water-soluble peptide antigens are often non-immunogenic even when combined with potent immunostimulants, such as polyICLC, but that rendering such peptide antigens particulate significantly improves immunogenicity.121 These data suggest that codelivery of peptide antigens with specific immunostimulants in nanoparticles will be key to the success of peptide-based cancer vaccines and that self-assembling nanoparticles (eg, SNP-7/8a) offer an effective platform for achieving consistent nanoparticle formulations needed for reliable induction of CD8+ T cells.

Another promising peptide-based vaccine approach for ensuring codelivery of tumor antigens and immunostimulants in nanoparticles for inducing T-cell immunity is the use of synthetic high-density lipoprotein nanodiscs, which can be coupled with immunostimulatory CpG oligonucleotides and tumor antigen peptides. Kuai et al.149 recently reported that the use of this nanocarrier technology in a murine colon adenocarcinoma model generated a robust neoantigen-specific CD8+ T-cell response resulting in complete inhibition of tumor growth; additionally, concomitant vaccination with dual PD-1 and CTLA-4 blockade led to complete tumor regression in approximately 90% of mice.

Though many next-generation vaccine technologies have been focused on solid tumors, innovation in biomaterials science has also made considerable progress in the setting of hematologic malignancies. Shah et al.150 developed a macroporous cryogel composed of cross-linked polyethylene glycol and alginate scaffolding with entrapped TLR-9a cytosine–guanosine oligodeoxynucleotide and GM-CSF immunostimulants. Prophylactic administration of this vaccine with either WT-1 antigen or tumor cell lysate in a mouse model of AML elicited a potent CTL response and prevented engraftment of malignant cells in the bone marrow. Combinatorial administration of the vaccine with standard chemotherapeutic agents eradicated established AML and generated transferable protective T-cell immunity.

In addition to the type of vaccine used, the route of administration and vaccine schedule will likely require optimization in the clinical setting to achieve maximal benefit for patients. Accordingly, while most vaccines are administered by the intramuscular or subcutaneous routes, mounting evidence suggests that vaccination by the intravenous route may be favorable for promoting T-cell-mediated immunity independent of the vaccination platform used.127,151

A final consideration is how to integrate cancer vaccines into a complex treatment regimen comprising chemotherapy, radiotherapy, and immunotherapy, which may not always have synergistic effects. Indeed, many cancer patients require corticosteroid therapy at different points throughout treatment for various reasons (eg, tumor/treatment-related edema, pain relief, appetite stimulation).152 However, there is growing evidence to suggest that the immunosuppressive nature of these drugs, due in large part to their effects on T-cell apoptosis, may reduce the efficacy of therapeutic approaches that rely on stimulating a robust anticancer immune response.153,154 Wong et al.155 found that dexamethasone treatment in adult patients with recurrent GBM profoundly decreased the efficacy of radiation and chemotherapy, leading to lower OS. Pitter et al.156 found similar results in a retrospective analysis of GBM patient cohorts, which showed dexamethasone-induced antiproliferative effects conferred protection from radiotherapy- and chemotherapy-induced genotoxic stress. In a phase I study of H3.3K27M peptide vaccination in pediatric patients with DMG and DIPG, Mueller et al.104 found a negative association between dexamethasone administration and the longitudinal expansion of vaccine-reactive CD8+ T cells. Though, while immunosuppressive chemotherapy agents can have deleterious effects on cancer vaccine efficacy, certain chemotherapy regimens, particularly those that promote immunogenic cell death or selective depletion of suppressor cells (eg, regulatory T cells or myeloid-derived suppressor cells), have been shown to enhance vaccine efficacy.157,158 Therefore, further research will be needed to fully delineate the impact and optimal regimen of chemotherapies, including corticosteroids, on cancer vaccination efficacy.

Conclusions

The ability of checkpoint inhibitors and adoptive cell therapies to mediate durable regression of certain advanced cancers provides clinical proof-of-concept that tumor antigen-specific T cells can mediate tumor clearance and improve patient outcomes. These observations have fueled a resurgence in efforts to advance therapeutic cancer vaccines for priming and/or expanding tumor antigen-specific T-cell responses in patients for use alone or in combination with other therapies. Despite their immense promise, however, therapeutic cancer vaccines have only shown modest benefit in a small cohort of primarily adult patients in early-stage trials. While many challenges remain, the emergence of improved vaccine technologies for inducing T-cell immunity, as well as refined tools for tumor antigen selection, provide optimism that next-generation therapeutic cancer vaccines may effectively overcome historic limitations. Additionally, combination immunotherapies, including vaccines combined with checkpoint inhibitors, oncolytic viruses, certain chemotherapeutics,159 and/or radiation, are emerging as effective approaches for reversing immune suppression within the tumor and augmenting vaccine efficacy.

Finally, it should be noted that vaccines and other immunotherapies that are safe but fail to demonstrate efficacy in adults with advanced cancers should not be ruled out for evaluation in pediatric populations. Adults are less responsive to vaccines due to thymic atrophy, whereas younger patients are more responsive to immunotherapies and are therefore more likely to mount an effective T-cell response to therapeutic cancer vaccines. Despite this general recognition, most novel immunotherapies are evaluated in adult populations, and treatments that fail in adults are often not advanced to testing in children. However, the small number of early-phase studies that have been conducted with pediatric patients has shown great promise in terms of safety, feasibility, and ability to generate an immunologic response. Though objective clinical response rates are low, advances in antigen design, adjuvant therapy, and combinatorial approaches may drastically change the landscape of immunotherapy in pediatric cancer.

In summary, given the tremendous potential of therapeutic cancer vaccines and their higher likelihood of success in pediatric populations, cancer vaccines and combination therapies should be rigorously investigated as potentially life-saving treatments for children with advanced malignancies refractory to conventional approaches.

Acknowledgments

The authors wish to thank Dr. Kyung-Don Kang for his critical review of the manuscript.

Funding

This research was supported by grants from the Andrew McDonough B+ Foundation, Hyundai Hope on Wheels, and the Rally Foundation for Childhood Cancer Research to G.K.F.

Conflicts of interest statement. G.M.L. and A.S.I. are listed as inventors on patents describing polymer-based vaccines. G.M.L., J.D.B., and A.S.I. are employees/consultants of Avidea Technologies, Inc., which is commercializing polymer-based drug delivery technologies for immunotherapeutic applications. J.D.B. has an equity position in Treovir LLC and is a member of the POCKiT Diagnostics Board of Scientific Advisors.

References

1.

Murphy
SL
,
Xu
J
,
Kochanek
KD
,
Arias
E
.
Mortality in the United States, 2017
.
NCHS Data Brief
.
2018
(
328
):
1
8
.

2.

Jemal
A
,
Ward
EM
,
Johnson
CJ
, et al.
Annual report to the nation on the status of cancer, 1975–2014, featuring survival
.
J Natl Cancer Inst
.
2017
;
109
(
9
):
1
22
.

3.

Diller
L
,
Chow
EJ
,
Gurney
JG
, et al.
Chronic disease in the childhood cancer survivor study cohort: a review of published findings
.
J Clin Oncol.
2009
;
27
(
14
):
2339
2355
.

4.

Bhattacharya
D
,
Pomeroy
SL
,
Pomeranz Krummel
DA
,
Sengupta
S
.
Epigenetics and survivorship in pediatric brain tumor patients
.
J Neurooncol.
2020
;
150
(
1
):
77
83
.

5.

Smith
MA
,
Seibel
NL
,
Altekruse
SF
, et al.
Outcomes for children and adolescents with cancer: challenges for the twenty-first century
.
J Clin Oncol.
2010
;
28
(
15
):
2625
2634
.

6.

Karajannis
M
,
Allen
JC
,
Newcomb
EW
.
Treatment of pediatric brain tumors
.
J Cell Physiol.
2008
;
217
(
3
):
584
589
.

7.

Gajjar
A
,
Pfister
SM
,
Taylor
MD
,
Gilbertson
RJ
.
Molecular insights into pediatric brain tumors have the potential to transform therapy
.
Clin Cancer Res.
2014
;
20
(
22
):
5630
5640
.

8.

Turner
CD
,
Rey-Casserly
C
,
Liptak
CC
,
Chordas
C
.
Late effects of therapy for pediatric brain tumor survivors
.
J Child Neurol.
2009
;
24
(
11
):
1455
1463
.

9.

Cohen
KJ
,
Pollack
IF
,
Zhou
T
, et al.
Temozolomide in the treatment of high-grade gliomas in children: a report from the Children’s Oncology Group
.
Neuro Oncol.
2011
;
13
(
3
):
317
323
.

10.

Jakacki
RI
,
Cohen
KJ
,
Buxton
A
, et al.
Phase 2 study of concurrent radiotherapy and temozolomide followed by temozolomide and lomustine in the treatment of children with high-grade glioma: a report of the Children’s Oncology Group ACNS0423 study
.
Neuro Oncol.
2016
;
18
(
10
):
1442
1450
.

11.

Guerra-García
P
,
Marshall
LV
,
Cockle
JV
, et al.
Challenging the indiscriminate use of temozolomide in pediatric high-grade gliomas: a review of past, current, and emerging therapies
.
Pediatr Blood Cancer.
2020
;
67
(
1
):
e28011
.

12.

Foster
JB
,
Madsen
PJ
,
Hegde
M
, et al.
Immunotherapy for pediatric brain tumors: past and present
.
Neuro Oncol.
2019
;
21
(
10
):
1226
1238
.

13.

Ring
EK
,
Markert
JM
,
Gillespie
GY
,
Friedman
GK
.
Checkpoint proteins in pediatric brain and extracranial solid tumors: opportunities for immunotherapy
.
Clin Cancer Res.
2017
;
23
(
2
):
342
350
.

14.

Zhang
H
,
Chen
J
.
Current status and future directions of cancer immunotherapy
.
J Cancer.
2018
;
9
(
10
):
1773
1781
.

15.

Wang
SS
,
Bandopadhayay
P
,
Jenkins
MR
.
Towards immunotherapy for pediatric brain tumors
.
Trends Immunol.
2019
;
40
(
8
):
748
761
.

16.

Paz-Ares
L
,
Luft
A
,
Vicente
D
, et al.
Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer
.
N Engl J Med.
2018
;
379
(
21
):
2040
2051
.

17.

Andtbacka
RH
,
Ross
M
,
Puzanov
I
, et al.
Patterns of clinical response with talimogene laherparepvec (T-VEC) in patients with melanoma treated in the OPTiM Phase III clinical trial
.
Ann Surg Oncol.
2016
;
23
(
13
):
4169
4177
.

18.

Neelapu
SS
,
Locke
FL
,
Bartlett
NL
, et al.
Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma
.
N Engl J Med.
2017
;
377
(
26
):
2531
2544
.

19.

Mougel
A
,
Terme
M
,
Tanchot
C
.
Therapeutic cancer vaccine and combinations with antiangiogenic therapies and immune checkpoint blockade
.
Front Immunol.
2019
;
10
:
467
.

20.

Sampson
JH
,
Mitchell
DA
.
Vaccination strategies for neuro-oncology
.
Neuro Oncol
.
2015
;
17
(
suppl 7
):
vii15
vii25
.

21.

Hollingsworth
RE
,
Jansen
K
.
Turning the corner on therapeutic cancer vaccines
.
NPJ Vaccines.
2019
;
4
:
7
.

22.

Plotkin
SA
.
Correlates of protection induced by vaccination
.
Clin Vaccine Immunol.
2010
;
17
(
7
):
1055
1065
.

23.

Pulendran
B
,
Ahmed
R
.
Immunological mechanisms of vaccination
.
Nat Immunol.
2011
;
12
(
6
):
509
517
.

24.

Chen
DS
,
Mellman
I
.
Oncology meets immunology: the cancer-immunity cycle
.
Immunity.
2013
;
39
(
1
):
1
10
.

25.

Abelin
JG
,
Keskin
DB
,
Sarkizova
S
, et al.
Mass spectrometry profiling of HLA-associated peptidomes in mono-allelic cells enables more accurate epitope prediction
.
Immunity.
2017
;
46
(
2
):
315
326
.

26.

McGranahan
N
,
Furness
AJ
,
Rosenthal
R
, et al.
Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade
.
Science.
2016
;
351
(
6280
):
1463
1469
.

27.

Comber
JD
,
Philip
R
.
MHC class I antigen presentation and implications for developing a new generation of therapeutic vaccines
.
Ther Adv Vaccines.
2014
;
2
(
3
):
77
89
.

28.

Lundegaard
C
,
Lund
O
,
Nielsen
M
.
Predictions versus high-throughput experiments in T-cell epitope discovery: competition or synergy?
Expert Rev Vaccines.
2012
;
11
(
1
):
43
54
.

29.

Ilyas
S
,
Yang
JC
.
Landscape of tumor antigens in T cell immunotherapy
.
J Immunol.
2015
;
195
(
11
):
5117
5122
.

30.

Pedersen
SR
,
Sørensen
MR
,
Buus
S
,
Christensen
JP
,
Thomsen
AR
.
Comparison of vaccine-induced effector CD8 T cell responses directed against self- and non-self-tumor antigens: implications for cancer immunotherapy
.
J Immunol.
2013
;
191
(
7
):
3955
3967
.

31.

Morgan
RA
,
Yang
JC
,
Kitano
M
,
Dudley
ME
,
Laurencot
CM
,
Rosenberg
SA
.
Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2
.
Mol Ther.
2010
;
18
(
4
):
843
851
.

32.

Parkhurst
MR
,
Yang
JC
,
Langan
RC
, et al.
T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis
.
Mol Ther.
2011
;
19
(
3
):
620
626
.

33.

Castle
JC
,
Kreiter
S
,
Diekmann
J
, et al.
Exploiting the mutanome for tumor vaccination
.
Cancer Res.
2012
;
72
(
5
):
1081
1091
.

34.

Zamora
AE
,
Crawford
JC
,
Thomas
PG
.
Hitting the target: how T cells detect and eliminate tumors
.
J Immunol.
2018
;
200
(
2
):
392
399
.

35.

Tran
E
,
Ahmadzadeh
M
,
Lu
YC
, et al.
Immunogenicity of somatic mutations in human gastrointestinal cancers
.
Science.
2015
;
350
(
6266
):
1387
1390
.

36.

Chang
MT
,
Asthana
S
,
Gao
SP
, et al.
Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity
.
Nat Biotechnol.
2016
;
34
(
2
):
155
163
.

37.

Roudko
V
,
Bozkus
CC
,
Orfanelli
T
, et al.
Shared immunogenic poly-epitope frameshift mutations in microsatellite unstable tumors
.
Cell
.
2020
;
183
(
6
):
1634
1649
e1617.

38.

Chen
DS
,
Mellman
I
.
Elements of cancer immunity and the cancer-immune set point
.
Nature.
2017
;
541
(
7637
):
321
330
.

39.

Ward
JP
,
Gubin
MM
,
Schreiber
RD
.
The role of neoantigens in naturally occurring and therapeutically induced immune responses to cancer
.
Adv Immunol.
2016
;
130
:
25
74
.

40.

Kreiter
S
,
Castle
JC
,
Türeci
O
,
Sahin
U
.
Targeting the tumor mutanome for personalized vaccination therapy
.
Oncoimmunology.
2012
;
1
(
5
):
768
769
.

41.

Sensi
M
,
Anichini
A
.
Unique tumor antigens: evidence for immune control of genome integrity and immunogenic targets for T cell-mediated patient-specific immunotherapy
.
Clin Cancer Res.
2006
;
12
(
17
):
5023
5032
.

42.

Sahin
U
,
Türeci
Ö
.
Personalized vaccines for cancer immunotherapy
.
Science.
2018
;
359
(
6382
):
1355
1360
.

43.

Hu
Z
,
Ott
PA
,
Wu
CJ
.
Towards personalized, tumour-specific, therapeutic vaccines for cancer
.
Nat Rev Immunol.
2018
;
18
(
3
):
168
182
.

44.

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
(
6236
):
803
808
.

45.

Keskin
DB
,
Anandappa
AJ
,
Sun
J
, et al.
Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial
.
Nature.
2019
;
565
(
7738
):
234
239
.

46.

Sampson
JH
,
Gunn
MD
,
Fecci
PE
,
Ashley
DM
.
Brain immunology and immunotherapy in brain tumours
.
Nat Rev Cancer.
2020
;
20
(
1
):
12
25
.

47.

Palucka
K
,
Banchereau
J
.
Dendritic-cell-based therapeutic cancer vaccines
.
Immunity.
2013
;
39
(
1
):
38
48
.

48.

Sabado
RL
,
Balan
S
,
Bhardwaj
N
.
Dendritic cell-based immunotherapy
.
Cell Res.
2017
;
27
(
1
):
74
95
.

49.

Melief
CJM
.
Smart delivery of vaccines
.
Nat Mater.
2018
;
17
(
6
):
482
483
.

50.

Deering
RP
,
Kommareddy
S
,
Ulmer
JB
,
Brito
LA
,
Geall
AJ
.
Nucleic acid vaccines: prospects for non-viral delivery of mRNA vaccines
.
Expert Opin Drug Deliv.
2014
;
11
(
6
):
885
899
.

51.

Draper
SJ
,
Heeney
JL
.
Viruses as vaccine vectors for infectious diseases and cancer
.
Nat Rev Microbiol.
2010
;
8
(
1
):
62
73
.

52.

Paugh
BS
,
Qu
C
,
Jones
C
, et al.
Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease
.
J Clin Oncol.
2010
;
28
(
18
):
3061
3068
.

53.

Jones
C
,
Karajannis
MA
,
Jones
DTW
, et al.
Pediatric high-grade glioma: biologically and clinically in need of new thinking
.
Neuro Oncol.
2017
;
19
(
2
):
153
161
.

54.

Gröbner
SN
,
Worst
BC
,
Weischenfeldt
J
, et al.
The landscape of genomic alterations across childhood cancers
.
Nature.
2018
;
555
(
7696
):
321
327
.

55.

Steinman
RM
.
Dendritic cells in vivo: a key target for a new vaccine science
.
Immunity.
2008
;
29
(
3
):
319
324
.

56.

Guo
C
,
Manjili
MH
,
Subjeck
JR
,
Sarkar
D
,
Fisher
PB
,
Wang
XY
.
Therapeutic cancer vaccines: past, present, and future
.
Adv Cancer Res.
2013
;
119
:
421
475
.

57.

Ardon
H
,
De Vleeschouwer
S
,
Van Calenbergh
F
, et al.
Adjuvant dendritic cell-based tumour vaccination for children with malignant brain tumours
.
Pediatr Blood Cancer.
2010
;
54
(
4
):
519
525
.

58.

Banchereau
J
,
Palucka
AK
.
Dendritic cells as therapeutic vaccines against cancer
.
Nat Rev Immunol.
2005
;
5
(
4
):
296
306
.

59.

Tuyaerts
S
,
Aerts
JL
,
Corthals
J
, et al.
Current approaches in dendritic cell generation and future implications for cancer immunotherapy
.
Cancer Immunol Immunother.
2007
;
56
(
10
):
1513
1537
.

60.

Perez
CR
,
De Palma
M
.
Engineering dendritic cell vaccines to improve cancer immunotherapy
.
Nat Commun.
2019
;
10
(
1
):
5408
.

61.

Vermaelen
K
.
Vaccine strategies to improve anti-cancer cellular immune responses
.
Front Immunol.
2019
;
10
:
8
.

62.

Boudousquie
C
,
Boand
V
,
Lingre
E
, et al.
Development and optimization of a GMP-Compliant manufacturing process for a personalized tumor lysate dendritic cell vaccine
.
Vaccines (Basel)
.
2020
;
8
(
1
).

63.

Kalinski
P
,
Muthuswamy
R
,
Urban
J
.
Dendritic cells in cancer immunotherapy: vaccines and combination immunotherapies
.
Expert Rev Vaccines.
2013
;
12
(
3
):
285
295
.

64.

Kantoff
PW
,
Higano
CS
,
Shore
ND
, et al.
Sipuleucel-T immunotherapy for castration-resistant prostate cancer
.
N Engl J Med.
2010
;
363
(
5
):
411
422
.

65.

Caruso
DA
,
Orme
LM
,
Neale
AM
, et al.
Results of a phase 1 study utilizing monocyte-derived dendritic cells pulsed with tumor RNA in children and young adults with brain cancer
.
Neuro Oncol.
2004
;
6
(
3
):
236
246
.

66.

Geiger
JD
,
Hutchinson
RJ
,
Hohenkirk
LF
, et al.
Vaccination of pediatric solid tumor patients with tumor lysate-pulsed dendritic cells can expand specific T cells and mediate tumor regression
.
Cancer Res.
2001
;
61
(
23
):
8513
8519
.

67.

Saito
S
,
Yanagisawa
R
,
Yoshikawa
K
, et al.
Safety and tolerability of allogeneic dendritic cell vaccination with induction of Wilms tumor 1-specific T cells in a pediatric donor and pediatric patient with relapsed leukemia: a case report and review of the literature
.
Cytotherapy.
2015
;
17
(
3
):
330
335
.

68.

Merchant
MS
,
Bernstein
D
,
Amoako
M
, et al.
Adjuvant immunotherapy to improve outcome in high-risk pediatric sarcomas
.
Clin Cancer Res.
2016
;
22
(
13
):
3182
3191
.

69.

Carli
M
,
Colombatti
R
,
Oberlin
O
, et al.
European intergroup studies (MMT4-89 and MMT4-91) on childhood metastatic rhabdomyosarcoma: final results and analysis of prognostic factors
.
J Clin Oncol.
2004
;
22
(
23
):
4787
4794
.

70.

Paulussen
M
,
Ahrens
S
,
Burdach
S
, et al.
Primary metastatic (stage IV) Ewing tumor: survival analysis of 171 patients from the EICESS studies. European Intergroup Cooperative Ewing Sarcoma Studies
.
Ann Oncol.
1998
;
9
(
3
):
275
281
.

71.

Pappo
AS
,
Anderson
JR
,
Crist
WM
, et al.
Survival after relapse in children and adolescents with rhabdomyosarcoma: a report from the Intergroup Rhabdomyosarcoma Study Group
.
J Clin Oncol.
1999
;
17
(
11
):
3487
3493
.

72.

Bai
Y
,
Zheng
JE
,
Wang
N
, et al.
Effects of dendritic cell-activated and cytokine-induced killer cell therapy on 22 children with acute myeloid leukemia after chemotherapy
.
J Huazhong Univ Sci Technolog Med Sci.
2015
;
35
(
5
):
689
693
.

73.

Benitez-Ribas
D
,
Cabezón
R
,
Flórez-Grau
G
, et al.
Immune response generated with the administration of autologous dendritic cells pulsed with an allogenic tumoral cell-lines lysate in patients with newly diagnosed diffuse intrinsic pontine glioma
.
Front Oncol.
2018
;
8
:
127
.

74.

Rutkowski
S
,
De Vleeschouwer
S
,
Kaempgen
E
, et al.
Surgery and adjuvant dendritic cell-based tumour vaccination for patients with relapsed malignant glioma, a feasibility study
.
Br J Cancer.
2004
;
91
(
9
):
1656
1662
.

75.

Lasky
JL
3rd,
Panosyan
EH
,
Plant
A
, et al.
Autologous tumor lysate-pulsed dendritic cell immunotherapy for pediatric patients with newly diagnosed or recurrent high-grade gliomas
.
Anticancer Res.
2013
;
33
(
5
):
2047
2056
.

76.

Dagher
R
,
Long
LM
,
Read
EJ
, et al.
Pilot trial of tumor-specific peptide vaccination and continuous infusion interleukin-2 in patients with recurrent Ewing sarcoma and alveolar rhabdomyosarcoma: an inter-institute NIH study
.
Med Pediatr Oncol.
2002
;
38
(
3
):
158
64
.

77.

Dohnal
AM
,
Witt
V
,
Hügel
H
,
Holter
W
,
Gadner
H
,
Felzmann
T
.
Phase I study of tumor Ag-loaded IL-12 secreting semi-mature DC for the treatment of pediatric cancer
.
Cytotherapy.
2007
;
9
(
8
):
755
770
.

78.

Mackall
CL
,
Rhee
EH
,
Read
EJ
, et al.
A pilot study of consolidative immunotherapy in patients with high-risk pediatric sarcomas
.
Clin Cancer Res.
2008
;
14
(
15
):
4850
4858
.

79.

Suminoe
A
,
Matsuzaki
A
,
Hattori
H
,
Koga
Y
,
Hara
T
.
Immunotherapy with autologous dendritic cells and tumor antigens for children with refractory malignant solid tumors
.
Pediatr Transplant.
2009
;
13
(
6
):
746
753
.

80.

Himoudi
N
,
Wallace
R
,
Parsley
KL
, et al.
Lack of T-cell responses following autologous tumour lysate pulsed dendritic cell vaccination, in patients with relapsed osteosarcoma
.
Clin Transl Oncol.
2012
;
14
(
4
):
271
279
.

81.

Krishnadas
DK
,
Shusterman
S
,
Bai
F
, et al.
A phase I trial combining decitabine/dendritic cell vaccine targeting MAGE-A1, MAGE-A3 and NY-ESO-1 for children with relapsed or therapy-refractory neuroblastoma and sarcoma
.
Cancer Immunol Immunother.
2015
;
64
(
10
):
1251
1260
.

82.

Shah
NN
,
Loeb
DM
,
Khuu
H
, et al.
Induction of immune response after allogeneic Wilms’ tumor 1 dendritic cell vaccination and donor lymphocyte infusion in patients with hematologic malignancies and post-transplantation relapse
.
Biol Blood Marrow Transplant.
2016
;
22
(
12
):
2149
2154
.

83.

Bezu
L
,
Kepp
O
,
Cerrato
G
, et al.
Trial watch: peptide-based vaccines in anticancer therapy
.
Oncoimmunology.
2018
;
7
(
12
):
e1511506
.

84.

Dubensky
TW
Jr,
Reed
SG
.
Adjuvants for cancer vaccines
.
Semin Immunol.
2010
;
22
(
3
):
155
161
.

85.

Ott
PA
,
Hu
Z
,
Keskin
DB
, et al.
An immunogenic personal neoantigen vaccine for patients with melanoma
.
Nature.
2017
;
547
(
7662
):
217
221
.

86.

Yadav
M
,
Jhunjhunwala
S
,
Phung
QT
, et al.
Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing
.
Nature.
2014
;
515
(
7528
):
572
576
.

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
(
23
):
6497
6508
.

88.

Melief
CJ
,
van der Burg
SH
.
Immunotherapy of established (pre)malignant disease by synthetic long peptide vaccines
.
Nat Rev Cancer.
2008
;
8
(
5
):
351
360
.

89.

Slingluff
CL
Jr.
The present and future of peptide vaccines for cancer: single or multiple, long or short, alone or in combination?
Cancer J.
2011
;
17
(
5
):
343
350
.

90.

Calvo Tardón
M
,
Allard
M
,
Dutoit
V
,
Dietrich
PY
,
Walker
PR
.
Peptides as cancer vaccines
.
Curr Opin Pharmacol.
2019
;
47
:
20
26
.

91.

Brito
LA
,
O’Hagan
DT
.
Designing and building the next generation of improved vaccine adjuvants
.
J Control Release.
2014
;
190
:
563
579
.

92.

Lynn
GM
,
Laga
R
,
Darrah
PA
, et al.
In vivo characterization of the physicochemical properties of polymer-linked TLR agonists that enhance vaccine immunogenicity
.
Nat Biotechnol.
2015
;
33
(
11
):
1201
1210
.

93.

Lynn
GM
,
Laga
R
,
Jewell
CM
.
Induction of anti-cancer T cell immunity by in situ vaccination using systemically administered nanomedicines
.
Cancer Lett.
2019
;
459
:
192
203
.

94.

Prow
NA
,
Jimenez Martinez
R
,
Hayball
JD
,
Howley
PM
,
Suhrbier
A
.
Poxvirus-based vector systems and the potential for multi-valent and multi-pathogen vaccines
.
Expert Rev Vaccines.
2018
;
17
(
10
):
925
934
.

95.

Tran
E
,
Turcotte
S
,
Gros
A
, et al.
Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer
.
Science.
2014
;
344
(
6184
):
641
645
.

96.

Tran
E
,
Robbins
PF
,
Lu
YC
, et al.
T-Cell transfer therapy targeting mutant KRAS in cancer
.
N Engl J Med.
2016
;
375
(
23
):
2255
2262
.

97.

Schneble
E
,
Clifton
GT
,
Hale
DF
,
Peoples
GE
.
Peptide-based cancer vaccine strategies and clinical results
.
Methods Mol Biol.
2016
;
1403
:
797
817
.

98.

Hashii
Y
,
Sato
E
,
Ohta
H
,
Oka
Y
,
Sugiyama
H
,
Ozono
K
.
WT1 peptide immunotherapy for cancer in children and young adults
.
Pediatr Blood Cancer.
2010
;
55
(
2
):
352
355
.

99.

Sawada
A
,
Inoue
M
,
Kondo
O
, et al.
Feasibility of cancer immunotherapy with WT1 peptide vaccination for solid and hematological malignancies in children
.
Pediatr Blood Cancer.
2016
;
63
(
2
):
234
241
.

100.

Coffman
RL
,
Sher
A
,
Seder
RA
.
Vaccine adjuvants: putting innate immunity to work
.
Immunity.
2010
;
33
(
4
):
492
503
.

101.

Hailemichael
Y
,
Overwijk
WW
.
Peptide-based anticancer vaccines: the making and unmaking of a T-cell graveyard
.
Oncoimmunology.
2013
;
2
(
7
):
e24743
.

102.

Pollack
IF
,
Jakacki
RI
,
Butterfield
LH
, et al.
Antigen-specific immune responses and clinical outcome after vaccination with glioma-associated antigen peptides and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in children with newly diagnosed malignant brainstem and nonbrainstem gliomas
.
J Clin Oncol.
2014
;
32
(
19
):
2050
2058
.

103.

Pollack
IF
,
Jakacki
RI
,
Butterfield
LH
, et al.
Immune responses and outcome after vaccination with glioma-associated antigen peptides and poly-ICLC in a pilot study for pediatric recurrent low-grade gliomas
.
Neuro Oncol.
2016
;
18
(
8
):
1157
1168
.

104.

Mueller
S
,
Taitt
JM
,
Villanueva-Meyer
JE
, et al.
Mass cytometry detects H3.3K27M-specific vaccine responses in diffuse midline glioma
.
J Clin Invest.
2020
;
130
(
12
):
6325
6337
.

105.

Hirabayashi
K
,
Yanagisawa
R
,
Saito
S
, et al.
Feasibility and immune response of WT1 peptide vaccination in combination with OK-432 for paediatric solid tumors
.
Anticancer Res.
2018
;
38
(
4
):
2227
2234
.

106.

Camisaschi
C
,
Renne
SL
,
Beretta
V
, et al.
Immune landscape and in vivo immunogenicity of NY-ESO-1 tumor antigen in advanced neuroblastoma patients
.
BMC Cancer.
2018
;
18
(
1
):
983
.

107.

Akazawa
Y
,
Hosono
A
,
Yoshikawa
T
, et al.
Efficacy of the NCCV Cocktail-1 vaccine for refractory pediatric solid tumors: a phase I clinical trial
.
Cancer Sci.
2019
;
110
(
12
):
3650
3662
.

108.

Hashii
Y
,
Sato-Miyashita
E
,
Matsumura
R
, et al.
WT1 peptide vaccination following allogeneic stem cell transplantation in pediatric leukemic patients with high risk for relapse: successful maintenance of durable remission
.
Leukemia.
2012
;
26
(
3
):
530
532
.

109.

Kushner
BH
,
Cheung
IY
,
Modak
S
,
Kramer
K
,
Ragupathi
G
,
Cheung
NK
.
Phase I trial of a bivalent gangliosides vaccine in combination with β-glucan for high-risk neuroblastoma in second or later remission
.
Clin Cancer Res.
2014
;
20
(
5
):
1375
1382
.

110.

Cheung
NK
,
Modak
S
,
Vickers
A
,
Knuckles
B
.
Orally administered beta-glucans enhance anti-tumor effects of monoclonal antibodies
.
Cancer Immunol Immunother.
2002
;
51
(
10
):
557
564
.

111.

Yan
JL
,
van der Hoorn
A
,
Larkin
TJ
,
Boonzaier
NR
,
Matys
T
,
Price
SJ
.
Extent of resection of peritumoral diffusion tensor imaging-detected abnormality as a predictor of survival in adult glioblastoma patients
.
J Neurosurg.
2017
;
126
(
1
):
234
241
.

112.

Filmus
J
.
Glypicans in growth control and cancer
.
Glycobiology.
2001
;
11
(
3
):
19R
23R
.

113.

Kinoshita
Y
,
Tanaka
S
,
Souzaki
R
, et al.
Glypican 3 expression in pediatric malignant solid tumors
.
Eur J Pediatr Surg.
2015
;
25
(
1
):
138
144
.

114.

Tsuchiya
N
,
Hosono
A
,
Yoshikawa
T
, et al.
Phase I study of glypican-3-derived peptide vaccine therapy for patients with refractory pediatric solid tumors
.
Oncoimmunology.
2017
;
7
(
1
):
e1377872
.

115.

Thust
SC
,
van den Bent
MJ
,
Smits
M
.
Pseudoprogression of brain tumors
.
J Magn Reson Imaging
.
2018
;
48
(
3
):
571
589
.

116.

Cobbs
CS
,
Harkins
L
,
Samanta
M
, et al.
Human cytomegalovirus infection and expression in human malignant glioma
.
Cancer Res.
2002
;
62
(
12
):
3347
3350
.

117.

Ochs
K
,
Ott
M
,
Bunse
T
, et al.
K27M-mutant histone-3 as a novel target for glioma immunotherapy
.
Oncoimmunology.
2017
;
6
(
7
):
e1328340
.

118.

Middleton
G
,
Silcocks
P
,
Cox
T
, et al.
Gemcitabine and capecitabine with or without telomerase peptide vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer (TeloVac): an open-label, randomised, phase 3 trial
.
Lancet Oncol.
2014
;
15
(
8
):
829
840
.

119.

Mitchell
P
,
Thatcher
N
,
Socinski
MA
, et al.
Tecemotide in unresectable stage III non-small-cell lung cancer in the phase III START study: updated overall survival and biomarker analyses
.
Ann Oncol.
2015
;
26
(
6
):
1134
1142
.

120.

Rini
BI
,
Stenzl
A
,
Zdrojowy
R
, et al.
IMA901, a multipeptide cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy for advanced or metastatic renal cell carcinoma (IMPRINT): a multicentre, open-label, randomised, controlled, phase 3 trial
.
Lancet Oncol.
2016
;
17
(
11
):
1599
1611
.

121.

Lynn
GM
,
Sedlik
C
,
Baharom
F
, et al.
Peptide-TLR-7/8a conjugate vaccines chemically programmed for nanoparticle self-assembly enhance CD8 T-cell immunity to tumor antigens
.
Nat Biotechnol.
2020
;
38
(
3
):
320
332
.

122.

Crotty
E
,
Downey
K
,
Ferrerosa
L
, et al.
Considerations when treating high-grade pediatric glioma patients with immunotherapy
.
Expert Rev Neurother
.
2020
;
21
(
2
):
1
15
.

123.

Aurisicchio
L
,
Ciliberto
G
.
Genetic cancer vaccines: current status and perspectives
.
Expert Opin Biol Ther.
2012
;
12
(
8
):
1043
1058
.

124.

Sahin
U
,
Derhovanessian
E
,
Miller
M
, et al.
Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer
.
Nature.
2017
;
547
(
7662
):
222
226
.

125.

Pardi
N
,
Hogan
MJ
,
Porter
FW
,
Weissman
D
.
mRNA vaccines—a new era in vaccinology
.
Nat Rev Drug Discov.
2018
;
17
(
4
):
261
279
.

126.

Tiptiri-Kourpeti
A
,
Spyridopoulou
K
,
Pappa
A
,
Chlichlia
K
.
DNA vaccines to attack cancer: strategies for improving immunogenicity and efficacy
.
Pharmacol Ther.
2016
;
165
:
32
49
.

127.

Kranz
LM
,
Diken
M
,
Haas
H
, et al.
Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy
.
Nature.
2016
;
534
(
7607
):
396
401
.

128.

Ribas
A
,
Weber
JS
,
Chmielowski
B
, et al.
Intra-lymph node prime-boost vaccination against Melan A and tyrosinase for the treatment of metastatic melanoma: results of a phase 1 clinical trial
.
Clin Cancer Res.
2011
;
17
(
9
):
2987
2996
.

129.

Buchan
S
,
Grønevik
E
,
Mathiesen
I
,
King
CA
,
Stevenson
FK
,
Rice
J
.
Electroporation as a “prime/boost” strategy for naked DNA vaccination against a tumor antigen
.
J Immunol.
2005
;
174
(
10
):
6292
6298
.

130.

Greenland
JR
,
Letvin
NL
.
Chemical adjuvants for plasmid DNA vaccines
.
Vaccine.
2007
;
25
(
19
):
3731
3741
.

131.

Cui
Z
,
Mumper
RJ
.
Microparticles and nanoparticles as delivery systems for DNA vaccines
.
Crit Rev Ther Drug Carrier Syst.
2003
;
20
(
2–3
):
103
137
.

132.

Manikandan
C
,
Kaushik
A
,
Sen
D
.
Viral vector: potential therapeutic for glioblastoma multiforme
.
Cancer Gene Ther.
2020
;
27
(
5
):
270
279
.

133.

Chiocca
EA
,
Nassiri
F
,
Wang
J
,
Peruzzi
P
,
Zadeh
G
.
Viral and other therapies for recurrent glioblastoma: is a 24-month durable response unusual?
Neuro Oncol.
2019
;
21
(
1
):
14
25
.

134.

Pol
JG
,
Zhang
L
,
Bridle
BW
, et al.
Maraba virus as a potent oncolytic vaccine vector
.
Mol Ther.
2014
;
22
(
2
):
420
429
.

135.

Guo
ZS
,
Lu
B
,
Guo
Z
, et al.
Vaccinia virus-mediated cancer immunotherapy: cancer vaccines and oncolytics
.
J Immunother Cancer.
2019
;
7
(
1
):
6
.

136.

Larocca
C
,
Schlom
J
.
Viral vector-based therapeutic cancer vaccines
.
Cancer J.
2011
;
17
(
5
):
359
371
.

137.

Iampietro
MJ
,
Larocca
RA
,
Provine
NM
, et al.
Immunogenicity and cross-reactivity of rhesus adenoviral vectors
.
J Virol
.
2018
;
92
(
11
).

138.

Kantoff
PW
,
Schuetz
TJ
,
Blumenstein
BA
, et al.
Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer
.
J Clin Oncol.
2010
;
28
(
7
):
1099
1105
.

139.

Gulley
JL
,
Borre
M
,
Vogelzang
NJ
, et al.
Phase III trial of PROSTVAC in asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer
.
J Clin Oncol.
2019
;
37
(
13
):
1051
1061
.

140.

Madan
RA
,
Mohebtash
M
,
Arlen
PM
, et al.
Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial
.
Lancet Oncol.
2012
;
13
(
5
):
501
508
.

141.

Quoix
E
,
Ramlau
R
,
Westeel
V
, et al.
Therapeutic vaccination with TG4010 and first-line chemotherapy in advanced non-small-cell lung cancer: a controlled phase 2B trial
.
Lancet Oncol.
2011
;
12
(
12
):
1125
1133
.

142.

Abdelaziz
MO
,
Ossmann
S
,
Kaufmann
AM
, et al.
Development of a human cytomegalovirus (HCMV)-based therapeutic cancer vaccine uncovers a previously unsuspected viral block of MHC Class I antigen presentation
.
Front Immunol.
2019
;
10
:
1776
.

143.

Kieran
MW
,
Goumnerova
L
,
Manley
P
, et al.
Phase I study of gene-mediated cytotoxic immunotherapy with AdV-tk as adjuvant to surgery and radiation for pediatric malignant glioma and recurrent ependymoma
.
Neuro Oncol.
2019
;
21
(
4
):
537
546
.

144.

Totsch
SK
,
Schlappi
C
,
Kang
KD
, et al.
Oncolytic herpes simplex virus immunotherapy for brain tumors: current pitfalls and emerging strategies to overcome therapeutic resistance
.
Oncogene.
2019
;
38
(
34
):
6159
6171
.

145.

Gatti-Mays
ME
,
Redman
JM
,
Collins
JM
,
Bilusic
M
.
Cancer vaccines: enhanced immunogenic modulation through therapeutic combinations
.
Hum Vaccin Immunother.
2017
;
13
(
11
):
2561
2574
.

146.

Le
DT
,
Lutz
E
,
Uram
JN
, et al.
Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer
.
J Immunother.
2013
;
36
(
7
):
382
389
.

147.

Koske
I
,
Rössler
A
,
Pipperger
L
, et al.
Oncolytic virotherapy enhances the efficacy of a cancer vaccine by modulating the tumor microenvironment
.
Int J Cancer.
2019
;
145
(
7
):
1958
1969
.

148.

Kwok
D
,
Okada
H
.
T-Cell based therapies for overcoming neuroanatomical and immunosuppressive challenges within the glioma microenvironment
.
J Neurooncol.
2020
;
147
(
2
):
281
295
.

149.

Kuai
R
,
Ochyl
LJ
,
Bahjat
KS
,
Schwendeman
A
,
Moon
JJ
.
Designer vaccine nanodiscs for personalized cancer immunotherapy
.
Nat Mater.
2017
;
16
(
4
):
489
496
.

150.

Shah
NJ
,
Najibi
AJ
,
Shih
TY
, et al.
A biomaterial-based vaccine eliciting durable tumour-specific responses against acute myeloid leukaemia
.
Nat Biomed Eng.
2020
;
4
(
1
):
40
51
.

151.

Baharom
F
,
Ramirez-Valdez
RA
,
Tobin
KKS
, et al.
Intravenous nanoparticle vaccination generates stem-like TCF1+ neoantigen-specific CD8+ T cells
.
Nat Immunol.
2021
;
22
(
1
):
41
52
.

152.

Twycross
R
.
The risks and benefits of corticosteroids in advanced cancer
.
Drug Saf.
1994
;
11
(
3
):
163
178
.

153.

Adhikaree
J
,
Moreno-Vicente
J
,
Kaur
AP
,
Jackson
AM
,
Patel
PM
.
Resistance mechanisms and barriers to successful immunotherapy for treating glioblastoma
.
Cells
.
2020
;
9
(
2
):
263
.

154.

Herold
MJ
,
McPherson
KG
,
Reichardt
HM
.
Glucocorticoids in T cell apoptosis and function
.
Cell Mol Life Sci.
2006
;
63
(
1
):
60
72
.

155.

Wong
ET
,
Lok
E
,
Gautam
S
,
Swanson
KD
.
Dexamethasone exerts profound immunologic interference on treatment efficacy for recurrent glioblastoma
.
Br J Cancer.
2015
;
113
(
2
):
232
241
.

156.

Pitter
KL
,
Tamagno
I
,
Alikhanyan
K
, et al.
Corticosteroids compromise survival in glioblastoma
.
Brain.
2016
;
139
(
Pt 5
):
1458
1471
.

157.

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
(
334
):
334ra352
.

158.

Zitvogel
L
,
Galluzzi
L
,
Smyth
MJ
,
Kroemer
G
.
Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance
.
Immunity.
2013
;
39
(
1
):
74
88
.

159.

Pol
JG
,
Atherton
MJ
,
Stephenson
KB
, et al.
Enhanced immunotherapeutic profile of oncolytic virus-based cancer vaccination using cyclophosphamide preconditioning
.
J Immunother Cancer
.
2020
;
8
(
2
).

Author notes

These authors jointly supervised this work.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.