Cancer vaccines: the next immunotherapy frontier


Abstract

After several decades, therapeutic cancer vaccines now show signs of efficacy and potential to help patients resistant to other standard-of-care immunotherapies, but they have yet to realize their full potential and expand the oncologic armamentarium. Here, we classify cancer vaccines by what is known of the included antigens, which tumors express those antigens and where the antigens colocalize with antigen-presenting cells, thus delineating predefined vaccines (shared or personalized) and anonymous vaccines (ex vivo or in situ). To expedite clinical development, we highlight the need for accurate immune monitoring of early trials to acknowledge failures and advance the most promising vaccines.

Similar content being viewed by others

Therapeutic cancer vaccines

Article 27 April 2021

Therapeutic cancer vaccines: advancements, challenges, and prospects

Article Open access13 December 2023

Turning the corner on therapeutic cancer vaccines

Article Open access08 February 2019

Main

Vaccines aiming to prevent infectious diseases are among the greatest medical advances of the 20th century, but the concepts underlying vaccination extend beyond prevention. Therapeutic vaccines designed to treat infections have moved into late-stage clinical trials with promising results1, made possible by a burgeoning understanding of fundamental immunology that has enabled more potent vaccine formulation. Treating established malignancy with vaccines traces back to William Coley’s injection of tumors with killed Streptococcus and Serratia in the 1910s2 and Lloyd Old’s similar approach with Bacillus Calmette–Guérin (BCG) in the 1950s3.

Despite some recent examples of vaccines that induced systemic regression of large tumors4,5 and prolonged survival6, small clinical trial sizes, marginal survival benefits and resource-intense approaches have held the field back from greater success and stirred well-justified skepticism. This is akin to the history of existing successful cancer immunotherapies, which have sparked new hope for patients with solid and hematologic malignancies despite repeated setbacks. For instance, numerous monoclonal antibody trials failed to show reproducible efficacy for nearly 20 years before the eventual success of rituximab in 1997 (ref. 7); anti-programmed cell death protein 1 (PD-1) antibody data lacked clinical efficacy for years before the first nivolumab data were published8; and many years of ineffective chimeric antigen receptor T cell (CAR T cell) clinical data prefaced their eventual success9. We propose that cancer vaccines are analogously poised for eventual success, given that they may currently show limited clinical progress but display clear rationale and compelling preclinical data for further development. Here we review this evidence and extrapolate a straightforward trajectory to the near future in which vaccines are likely to become standard anti-cancer therapies.

The success of other immunotherapies has drawn focus away from cancer vaccines, despite their distinct benefits. Although CAR T cells can be effective for cancers with identifiable tumor-specific surface antigens, vaccines have the potential to additionally target the broader set of intracellular antigens. Whereas checkpoint blockade can treat subsets of ‘inflamed’ cancers, infiltrated by previously primed tumor-reactive T cells, cancer vaccines have the potential to newly prime tumor-reactive T cells. Concurrent progress in easier-to-use therapies has also diminished vaccine enthusiasm. For example, when the sipuleucel-T vaccine was approved with a small survival benefit, enzalutamide (an oral therapy) demonstrated greater survival benefit in higher-risk patients10. Similarly, the glycoprotein 100 (gp100) vaccine given with inpatient high-dose interleukin (IL)-2 demonstrated improved survival the same year that ipilimumab (an outpatient therapy) was approved, demonstrating a more significant survival benefit that was not enhanced by co-administration with the gp100 vaccine11. Along the same lines, an idiotype vaccine trial demonstrating progression-free survival (PFS) benefit in combination with an aggressive chemotherapy regimen was supplanted by a gentler, more effective chemotherapy regimen12,13.

The history of cancer vaccines has been the subject of excellent reviews14, most of which have focused on the physical structure of the antigen being introduced: whole tumor, tumor cells, protein, peptides (long or short), RNA or DNA (directly or virally); and the adjuvants with which antigen is introduced: carrier protein, cells (for example, dendritic cells (DCs)), proteins (for example, CD40 ligand (CD40L)) or chemicals (for example, oil–water emulsions and Toll-like receptor (TLR) agonists). Here, we classify current cancer vaccines differently, based on (1) what is known of a tumor’s specific immunogenic antigen, (2) which patients’ tumors express those antigens and (3) how the antigens become colocalized with professional antigen-presenting cells (APCs). Vaccines can incorporate either predefined (known) or anonymous (unknown) antigens (Fig. 1a). The former includes either predefined shared antigens (expressed in many patient tumors) or predefined personalized antigens (exclusively determined for each patient). Anonymous antigen vaccines can be colocalized with APCs either ex vivo (in a laboratory) or in situ (at the tumor site; Fig. 1a).

figure 1
Fig. 1: Cancer vaccine types.

We consider two types of tumor-specific antigens (TSAs), including viral antigens and neo-epitopes resulting from non-synonymous somatic mutations, and two types of tumor-associated antigens (TAAs), including tissue-specific antigens and development-specific antigens (Table 1). All the vaccines discussed might mobilize T cell responses against both TSAs and TAAs, except for predefined personalized antigen vaccines, which generally use TSAs. In this latter case, it is possible that hotspot mutations in cancer-related genes could be present in the tumors of different patients sharing human leukocyte antigen (HLA) molecules15.Table 1 TSAs and TAAs classified into four groups with examples

Full size table

The uptake of tumor antigens by APCs is a critical event16 (Fig. 1b). A majority of TAAs are intracellular and thereby difficult to target with humoral responses or derived therapies such as monoclonal antibodies, CAR T cells or bispecific T cell engagers. Although intracellular TAAs can be detected by TAA-specific T cells through HLA molecules on tumor cells, deficits in tumoral costimulatory molecules generally yield T cell anergy or exhaustion. Therefore, APCs, particularly DCs, are essential for anti-tumor T cell priming. The cDC1 (type 1 conventional DC) subset (or Batf3-dependent CD103+XCR1+CD141+Clec9A+ DCs) is specifically capable of cross-presentation: taking up exogenous antigens and presenting them on HLA-I to CD8+ T cells4,17,18. Therefore, by activating tumor antigen-loaded DCs, cancer vaccines may induce immune responses against a large array of intracellular antigens. From this perspective, the different vaccine types differ merely by methods of colocalizing tumor antigens with cross-presenting DCs (Fig. 1b).

Predefined antigens

Predefined antigens can be further classified by the frequency of expression across patient cohorts. Shared antigens are those expressed in a sufficient proportion of patients such that vaccinologists can target these patient groups (frequently within patient subsets of tumor types) using standard testing. Shared antigen vaccines can thus target both TSAs and TAAs. As examples, the neo-epitope TSA epidermal growth factor receptor variant III (EGFRvIII) is expressed in ~25% of EGFR-overexpressing glioblastomas (GBMs)19 and the viral TSA human papilloma virus E6 and E7 proteins (HPV E6 and E7) are expressed in ~60% of oropharyngeal cancers and nearly all cervical cancers20, whereas the TAA Wilms’ tumor protein (WT1) is overexpressed in most acute myeloid leukemias (AMLs), breast cancers and Wilms’ tumors21. Shared antigen vaccines are distinguished from personalized antigen vaccines in that the former can be assessed with standard testing such as cytology, immunohistochemistry and flow cytometry. Predefined, shared antigen vaccines have been the primary focus of preclinical and clinical research since the 1990s and have provided foundational lessons.

Personalized antigens are unique to the vaccinated patient. Personalized antigen vaccines have developed alongside the modern era of high-throughput gene sequencing and generally consist of TSA neo-epitopes that, in contrast to the shared TSA EGFRvIII or Kirsten rat sarcoma virus (KRAS)G12D, are not sufficiently common to target a large group of patients. This approach allows the immune system to target tumors lacking known shared antigens but also places a burden on the vaccinologist to iteratively determine the optimally immunogenic epitopes. Immunogenic epitopes must bind with sufficient avidity to both the peptide groove of an HLA molecule and to the complementarity-determining regions of a reactive T cell receptor (TCR). Peptide–HLA (and, to a lesser degree, TCR) avidities can be modeled and estimated in silico for an individual patient’s tumor mutanome, although these algorithms are still improving. Such approaches also pose a logistical burden of biopsying tumors for exome and RNA sequencing or for proteomic analysis of peptides actually presented by patient HLA class I molecules22. These techniques also require time and resources inherent in vaccine design and subsequent personalized neo-epitope pool production. The same tumoral genomic, transcriptomic and proteomic steps are required for shared antigen vaccine approaches by employing public datasets (for example, the Cancer Genome Atlas) compiled from prior patients’ biopsies (Fig. 1b).

Predefined shared antigen vaccines

Shared antigen vaccines can be used as ‘off-the-shelf’ therapies, which are less resource intense and time consuming than personalized vaccines. Here we highlight a selection of optimal shared antigens ranked by their cumulative clinical and immunologic data in early trials23 with substantial immunologic or clinical achievements (Table 2).Table 2 Selected predefined shared antigen cancer vaccine trials and outcomes

Full size table

TSAs are uniquely found in tumor cells and often drive oncogenesis; one such subtype is viral antigens. Epstein–Barr virus encodes multiple antigens including latent membrane proteins (LMP1 and LMP2), which can be expressed in nasopharyngeal carcinoma, natural killer (NK)–T cell lymphoma and other tumors24. Preclinical LMP1 vaccine studies25 and successful adoptive T cell-transfer clinical studies26 have inspired clinical vaccine trials. Nonetheless, autologous DCs expressing LMP1 and LMP2 did not elicit antigen-specific T cells in patients with nasopharyngeal carcinoma27. More recently, a modified vaccinia Ankara (MVA) virus expressing an Epstein–Barr nuclear antigen (EBNA)–LMP2 fusion protein showed boosting of CD4+ and CD8+ T cell responses28, prompting a larger follow-up study (NCT01800071). Similarly, HPV E6 and E7 are viral TSAs that sequester tumor protein 53 (p53) and Rb proteins, promoting proliferation and tumorigenesis in squamous epithelia. Synthetic long peptide (SLP) vaccine (ISA101) elicited T cell responses and tumor regressions in a majority of patients with vulvar intraepithelial neoplasia29, prompting a study combining ISA101 with anti-PD-1 therapy that demonstrated clinical responses higher than those from either therapy alone, even in programmed cell death ligand 1 (PD-L1) tumors30. Both E6/E7-plasmid (VGX-3100)31 and E6/E7/Fms-like tyrosine kinase 3 ligand (Flt3L)-plasmid (GX-188E) vaccines32 induced T cell responses associated with clinical efficacy, and a randomized phase II trial using an E6/E7/IL-2 MVA vector vaccine induced superior efficacy in high-grade cervical intraepithelial neoplasia33. LCMVi vectors expressing E7 have also demonstrated potent induction of E7-specific T cells. These studies suggest that, with optimal (for example, viral) antigens, therapeutic vaccination can induce clinical remission in low-burden tumors and that DC mobilization might improve this.

Overexpressed mutant self proteins are another subclass of TSAs. EGFRvIII is a constitutively active, somatically mutated EGFR variant, commonly expressed in GBM and non-small cell lung cancer (NSCLC). Promising early results in anti-EGFRvIII CAR T cell-treated patients with GBM provide validation of this target34. A phase II trial of an EGFRvIII 14-mer peptide vaccine (Rindopepimut) given with granulocyte–monocyte colony-stimulating factor (GM-CSF) and temozolomide elicited humoral immune responses35, although a phase III trial failed to show clinical benefit despite significant humoral responses36. A randomized phase II trial of its combination with bevacizumab demonstrated greater humoral responses and an overall survival (OS) benefit as a secondary, underpowered endpoint37. These data suggest that anti-tumor humoral responses may be insufficient and that vaccine success may depend on choosing optimal combination therapies.

By comparison, TAAs are not exclusively but preferentially found in tumor tissue and may constitute abnormally expressed or overexpressed proteins. This broad class can be divided into development-specific (that is, oncofetal, cancer-testis), tissue type-specific or tumor-enriched proteins.

WT1 is a development-specific transcription factor that contributes to oncogenesis23. Initial trials of short (nine-mer) WT1 peptide vaccines yielded immune and clinical responses38, followed by vaccination with an altered ‘heteroclitic’ WT1 peptide with greater HLA affinity (Galinpepimut-S) that induced T cell responses in a majority of patients with AML39 and prompted an ongoing phase III trial (NCT04229979). Increasing vaccine-site DCs using GM-CSF40 or by injecting ex vivo peptide-loading DCs41 yielded greater immune efficacy, suggesting that antigen–DC colocalization may be important for enhancing clinical efficacy.

New York-esophageal cancer 1 (NY-ESO-1) is a cancer-testis antigen with restricted expression in embryonic, gonadal and cancer cells and has poorly understood function. It is highly expressed in synovial sarcomas and heterogeneously expressed in melanoma, ovarian and esophageal cancers42. Remarkably, despite patients’ frequent spontaneous anti-NY-ESO-1 immune responses, more than 20 vaccine trials have ended overall unsuccessfully, as reviewed elsewhere42. Failure may be attributable both to suboptimal vaccine design and heterogeneous tumoral antigen expression as suggested by the impressive efficacy of targeting in synovial sarcoma, a rare tumor with homogeneous antigen expression43. Seeking to improve the immunogenicity over protein-based vaccines, long peptide vaccination has been tested, yielding frequent CD4+ T cell responses but only rare CD8+ T cell responses. Attempts to increase DC antigen presentation and CD8+ T cell responses by co-administration of NY-ESO-1 with a TLR9 agonist still elicited only rare CD8+ cell responses44. Impressively, a protein conjugate of a DC-targeting (anti-DEC-205) monoclonal antibody conjugated to NY-ESO-1 (CDX-1401) combined with TLR agonists induced CD8+ T cell responses in most patients alongside tumor regression45, highlighting the need for sufficient DCs to benefit from this approach. Indeed, a randomized study of CDX-1401 with or without DC-mobilizing recombinant Flt3L46 demonstrated approximately threefold increases (86% versus 29%) in CD8+ cell responses with Flt3L. Although the study was not powered for clinical recurrence differences, it strongly suggests that effective CD8+ cell priming requires potent DC mobilization, antigen loading and activation.

Melanoma-associated antigen 3 (MAGE-A3) is a cancer-testis antigen with anti-apoptotic function preferentially expressed in melanoma, NSCLC and myeloma. The TLR4-agonist-adjuvant (AS02B) MAGE-A3 protein vaccine induced humoral anti-tumor responses but no apparent clinical benefit in a small randomized study47; however, a randomized phase II trial adding a TLR9 agonist (AS15) to the same vaccine showed greater humoral and CD4+ T cell responses with greater clinical responses and prolonged survival48. Surprisingly, large follow-up trials randomizing more than 6,000 patients did not show clinical benefit49,50. One explanation for this failure may be that MAGE-A3 is heterogeneously expressed51; thus, targeting single, heterogeneous antigens likely promotes antigen escape. To address this point, a multivalent MAGE-A3–CEA–HER2–p53 vaccine (Tedopi) improved survival in subset analysis of a randomized study of patients with NSCLC, although prospective validation is needed52. Similarly, a multivalent melanoma vaccine that includes MAGE-A3, melan A, gp100 and tyrosinase (seviprotimut-L) yielded improved outcomes for a subset of younger patients in a large randomized trial53. Most recently, an early-phase trial of prime–boost adenovirus (ChAdOx1)/MVA vaccine targeting MAGE-A3 and NY-ESO-1 for patients with lung cancer was initiated in collaboration with the Ludwig Institute in early 2022 (NCT04908111).

Human epidermal growth factor receptor 2 (HER2/Neu) is an EGFR family member kinase overexpressed in ~30% of breast cancers and smaller proportions of gastrointestinal and ovarian tumors that can be targeted by anti-HER2 monoclonal antibody. A single-epitope, HLA-I-restricted nine-mer peptide vaccine (nelipepimut-S) that induced transient CD8+ T cell responses failed to show clinical benefit54, and, similarly, a single-epitope HLA-II-restricted 15-mer peptide (AE37) induced CD4+ T cell responses but had no clinical benefit55. By contrast, a multi-epitope, combination HLA-I- and HLA-II-binding HER2 peptide vaccine induced durable (>1 year) CD8+ T cell responses in patients56, suggesting that optimal immune responses occur with priming of both CD4+ and CD8+ T cells and that targeting multiple antigenic epitopes is preferable. These lessons may be applicable in earlier-phase HER2 vaccines using pulsed DCs and alphavirus vectors showing promising preliminary immune and clinical results57.

gp100 is enriched in melanosomes and melanoma, and its target validity was demonstrated when gp100-redirecting T cell therapy induced survival prolongation58. Early trials of a heteroclitic gp100 peptide vaccine with high-dose IL-2 induced tumor-reactive T cells in most patients and a 42% overall response rate (ORR), much higher than that with IL-2 alone59. Following this result, a phase III trial of IL-2 with or without vaccine increased ORR (16% versus 6%) and survival benefit (18 versus 11 months)60, although enthusiasm was tempered by high-grade IL-2-associated toxicity and deaths. Moreover, a randomized trial failed to show benefit of the gp100 vaccine alone or with ipilimumab11. As the ORR of gp100 peptide vaccine monotherapy is <2%, these data suggest that even proven antigen targets require potent T cell priming, such as that provided by IL-2.

Prostatic acid phosphatase (PAP) is expressed on prostate epithelia and increases proportionately with cancer progression but is also expressed in other tissues61. After several smaller trials, a phase III trial of sipuleucel-T, an autologous GM-CSF-stimulated monocyte mixture pulsed with PAP, demonstrated a 4-month survival benefit versus unpulsed APC vaccination6. This promising Food and Drug Administration-approved proof of principle has had minimal clinical impact likely due to lack of clear immune or objective responses, expense and impracticalities of personalized therapy and concurrent development of easier, more effective alternatives. Addressing these shortcomings, an off-the-shelf DNA PAP vaccine has demonstrated PAP-specific T cells in a greater proportion of patients and demonstrated objective responses by positron emission tomography imaging62 and is now being tested in combination with PD-1 blockade (NCT03600350). The prolonged survival demonstrated by vaccines can be obfuscated by the obstacles of vaccinating against single, imperfectly specific antigens and the benefits of off-the-shelf over personalized therapies.

p53 is altered in half of cancers and frequently lost in tumors but also deleteriously mutated and overexpressed. Given the complexity of targeting personalized mutations63,64, small trials of wild-type (WT) p53 have included viral vector-encoded65, DC-based66 and long peptide pool vaccines67 and combination with checkpoint inhibition68, demonstrating anti-p53 T cell responses in most patients yet few clinical remissions. Conversely, a study in patients with colorectal cancer vaccinated with mutant p53 demonstrated greater T cell responses to mutant peptides versus the corresponding WT peptides, further suggesting the tolerogenicity of self peptides69. Another frequently enriched TAA, indoleamine 2,3-dioxygenase 1 (IDO), has been targeted by small-molecule inhibitors and used as a peptide vaccine70. These studies provided rationale for a trial combining IDO and/or PD-L1 vaccination with PD-1 blockade, showing peptide-specific T cells and a 42% complete response (CR), significantly higher than anti-PD-1 therapy alone71. In sum, these data suggest that inducing T cells against self proteins, even those overexpressed in tumors, requires an elevated immune response for greatest efficacy.

Predefined shared vaccines targeting well-characterized tumor antigens present a method for widespread administration constrained by heterogeneous expression, insufficient immunogenicity or suboptimal partner therapies. The more promising approaches attempt to address these shortcomings (for example, Tedopi, seviprotimut-L, MELITAC).

Predefined personalized antigen vaccines

Unlike shared antigens that exist in many individuals, personalized antigens are unique to one patient and are most commonly neo-epitope TSAs (Fig. 1a). Targeting personalized antigens allows for exquisite specificity and unleashes T cells that circumvent thymic negative selection and, in combination with checkpoint blockade, mounts widespread T cell reactivity in responding patients72. Advances in next-generation sequencing and incorporation of additional immune-stimulating factors (for example, DC recruitment and activation, myeloid suppression, CD4+ cell help) render the entire production effort of this approach more feasible and effective. As such, designing personalized antigen vaccines includes variations of DNA and RNA extraction from tumor and germline tissue for exome and RNA sequencing as well as HLA typing (Fig. 1b). Somatic mutations are selected that are present in the tumor and absent in the germline, have low ‘false discovery rate’ and cause non-synonymous protein changes. Potentially immunogenic neo-epitopes are selected from among somatic mutations by in silico prediction of their binding to that patients’ HLA alleles using approaches similar to the NetMHC algorithm73. Highly expressed neo-epitopes are prioritized by assessment of tumoral RNA-sequencing data, from which generally up to 20 neo-epitopes are selected and good manufacturing practice (GMP)-grade neo-epitope peptides, RNA or viral vectors are produced. Neo-epitope vaccines can be given with adjuvants to optimize APC uptake (for example, liposomes) or APC activation (for example, pattern-recognition receptor (PRR) agonists) to aid their immunogenicity. While these approaches are time consuming and resource intense, increased sequencing bandwidth and new algorithms including machine learning algorithms for epitope prediction make these therapies continually more promising. Here, we highlight several approaches (Table 3).Table 3 Predefined personalized antigen cancer vaccine trials and outcomes

Full size table

An early personalized vaccine using a synthetic RNA vaccine encoding ten neo-epitope candidate targets elicited mostly CD4+ and some CD8+ neo-epitope-specific T cell responses and anecdotal objective responses in patients with metastatic melanoma72. These poly-specific responses could be enhanced by PD-1 blockade or abrogated by tumor cell HLA class I presentation loss and likely contributed to a significant reduction in longitudinal metastatic events. Similarly, an academic trial delivering 13–20 long peptides of predicted neo-epitopes (NEO-PV-01) induced more CD4+ than CD8+ cell responses specific for mutated peptide74. A larger study combining neo-epitope vaccine with anti-PD-1 in 60 patients with melanoma, NSCLC and bladder cancer also noted neoantigen-specific T cell responses and clinical responses possibly higher than those expected with anti-PD-1 therapy alone75.

To confirm that predicted neopeptides are present in tumoral HLA, a small study used peptide elution and mass spectrometry, followed by vaccination with neopeptide-pulsed autologous IL-12-producing DCs, demonstrating induction of polyclonal, antigen-specific T cell responses76. Other small studies accomplished vaccine-site DC activation by incorporating neoantigens with poly-inosinic-polycytidylic acid, poly-L-lysine and carboxymethylcellulose (poly-ICLC) (NeoVax), leading to diverse T cell repertoires77,78. An mRNA vaccine study (CONSORT) using a new neo-epitope-selection platform that prioritized tumor-infiltrating lymphocyte (TIL)-reactive candidates found mutation-specific T cells including those against a common mutation, KRASG12D (ref. 79). To facilitate delivery of neo-epitope RNA vaccines, packaging approaches using liposomes have also entered phase II trials (NCT03815058NCT04267237), with promising preliminary immune and clinical response data. To minimize the time to therapy of personalized vaccines, GAPVAC-101 combines non-mutated ‘shared’ antigen vaccination followed by personalized neo-epitope vaccination for patients with GBM. This strategy induced both central memory CD8+ T cell and type 1 helper T (TH1) cell responses with survival results possibly superior to those of historical controls80. Another recent study demonstrated that a preclinical lung cancer neo-epitope vaccine could potentiate checkpoint blockade therapy by improving CD8+ T cell responses to subdominant antigens and preventing their differentiation toward dysfunctional CCR6+TCF1+ TC17-like cells81. Other ongoing phase I studies are using recombinant heat-killed yeast to express neo-epitopes (YE-NEO-001, NCT03552718), engineered RNA constructs expressing patient mutanomes (IVAC mutanome, NCT02035956)72 or APC-targeted delivery of RNA via lipoprotein complex (Lipo-MERIT, NCT02410733). More recently, a prime–boost vaccine with adenovirus expressing neo-epitopes followed by a self-amplifying mRNA encoding the same antigens (GO-004 and GO-005) demonstrated neoantigen-specific CD8+ T cells in a minority of patients (NCT03639714NCT03953235; Table 3)82.

Another tumor-specific mutation, although not oncogenic per se, is the unique immunoglobulin or TCR idiotype that arises from locus gene rearrangements and somatic hypermutation, which are generally maintained in transformed cells and the resulting myelomas, lymphomas or leukemias. Progressing from preclinical studies, tumor-specific idiotypes of patients with lymphoma have been tested as vaccines83. The Favrille and Genitope phase III trials vaccinated rituximab- or chemotherapy-treated patients with lymphoma with idiotype linked to KLH administered with GM-CSF, with neither study yielding clinical benefit compared to placebo. A separate phase III trial (NCI-Biovest) using the same vaccine strategy demonstrated significant disease-free survival benefit when administered to patients in complete remission after chemotherapy, but frequent patient dropout before vaccination confounded the result’s significance. Nevertheless, the equivocal results of idiotype vaccination are likely faults of implementation rather than concept, as anti-idiotype antibody therapy is effective84. Although GM-CSF has been shown to mobilize some APC subsets, other approaches, such as Flt3L, have been shown to be significantly more effective in priming adaptive immune responses85.

Predefined personalized antigen vaccines exploit the most specific tumor mutagens identified with the best computational methods available. Challenges remain to reduce the amount of required resources to produce personalized vaccines for each individual, to avoid immune escape of heterogeneous tumors and to mount effective anti-tumor CD8+ T cell immunity.

Anonymous antigens ex vivo or in situ

Instead of being classified by their antigen identity, anonymous antigens can be classified by their method and location of APC loading. Anonymous antigen ex vivo vaccines are derived from excised tumor cells that are lysed and delivered to autologous APCs (Fig. 1b). Anonymous antigen in situ vaccines rely on endogenous APCs that are induced to uptake antigen at or near the tumor site, potentially following therapy-induced immunogenic cell death. Contrary to predefined antigen vaccines, anonymous antigen vaccines may include a larger number of antigens and even new antigen types, such as peptide fusion epitopes86 and post-transcriptionally produced epitopes87, which are technically difficult to identify and not included in most neo-epitope pipelines.

Anonymous antigen vaccines ex vivo, APC colocalized

Ex vivo antigen isolation may require extraction of tumor cells (excisional biopsy), processing raw tissue into a more antigenic form and colocalization with APCs. Injected tumor cells may be taken up and their antigens may be presented by APCs, or the tumor cells themselves may present their antigens to T cells. The defining feature of this approach is the ex vivo isolation of antigens and colocalization with APCs (Fig. 1b and Table 4).Table 4 Anonymous ex vivo engineered cancer vaccine trials and outcomes

Full size table

HSPs such as gp96, HSP70 and HSP110 have been shown to chaperone neo-epitopes for APC uptake and cross-presentation without being immunogenic themselves, and preclinical tumor-derived HSP vaccines induced anti-tumor immune responses, providing evidence for clinical development88. Large randomized trials demonstrated that vaccination with autologous tumor-derived peptide–gp96 complexes (HSPPC-96) failed to improve survival for patients with melanoma89 or renal cell carcinoma90. A subsequent study of patients with GBM receiving HSPPC-96 showed that tumoral PD-L1 expression negatively correlated with survival91, prompting a follow-up study combining HSPPC-96 with anti-PD-1 antibody (NCT03018288).

Allogeneic tumor cell-based vaccines are derived from tumor biopsies subsequently transformed into immortalized cell lines and consequently enriched for commonly mutated TAAs (for example, p53, KRAS, EGFR). Several early trials of engineered allogeneic tumor cell vaccines supported the benefit of anonymous antigen vaccines, although larger randomized trials (for example, Canvaxin, Melacine, prostate GVAX, Lucanix) have been generally unimpressive92. Immunodominance of alloantigens could be a problem in this case.

Despite numerous trials showing promising tumoral immune infiltration93, autologous tumor cells transfected to express GM-CSF (personalized GVAX) infused in patients after hematopoietic stem cell transplantation did not provide survival benefit in patients with AML94. Autologous tumor cells transfected to express GM-CSF and with anti-furin shRNA to prevent transforming growth factor (TGF)-β production (gemogenovatucel-T) demonstrated promising single-arm trial efficacy in Ewing’s sarcoma95. In a randomized phase IIb trial for patients with ovarian carcinoma, the gemogenovatucel-T cohort, despite worse performance status and greater macroscopic residual disease, still demonstrated a trend toward improved recurrence-free survival (RFS) (hazard ratio of 0.69, P = 0.078) and longer RFS and OS among patients with BRCA-WT disease (hazard ratio of 0.51, P = 0.020), suggesting the need for a dedicated study of this cohort96. A phase III trial of BCG admixed with tumor cells (OncoVAX) elicited cutaneous hypersensitivity indurations and non-significantly improved RFS and OS (P = 0.330) despite promising results in stage II colorectal cancer97. These studies prove that anticipating clinical efficacy in large trials from immune responses in small trials is not always straightforward.

Autologous tumor lysate-based approaches may be preferable to shared antigens, as suggested by a study comparing parallel cohorts of autologous GBM tumor lysate-pulsed DCs versus GBM shared antigen-pulsed DCs98. This analysis found a correlation between decreased regulatory T cell (Treg) ratios and OS, including median survivals of 34 months versus 15 months favoring the autologous approach (DCVax-L), prompting an ongoing phase III trial (NCT00045968). To assess whether autologous tumor cell-based vaccines are as effective as autologous tumor lysate-pulsed DCs, a randomized phase II trial comparing the two demonstrated median survivals of 43 versus 21 months, favoring DC vaccination (P = 0.19) in patients with melanoma99, prompting follow-up studies in GBM (NCT03400917) and ovarian carcinoma (NCT02033616). Another inspiring DC vaccine using heat-shocked, autologous lymphoma-pulsed DCs demonstrated an increase in tumor-specific T cells, which correlated with the systemic tumor regressions seen in six of the 18 treated patients100. More recently, in a pilot study of 25 patients with ovarian cancer, autologous DCs with oxidized autologous tumor cell lysate were pulsed either as monotherapy or with anti-vascular endothelial growth factor A (VEGF) monoclonal antibody and chemotherapy, inducing anti-neo-epitope and anti-tumor T cell responses associated with prolonged survival101. In sum, these data suggest that autologous tumors are better sources of antigens and that DCs are more effective antigen presenters than lymphoma cells themselves. Overall, anonymous antigen ex vivo vaccines are promising for their greater potential to present the full spectrum of tumor antigens as compared to predefined antigen vaccines and their demonstrable efficacy in inducing systemic tumor regressions100. Still, these are limited by the resource commitment of creating personalized, GMP-compliant products for each patient, which has slowed their development.

Anonymous antigen vaccines in situ, APC colocalized

Anonymous antigen in situ vaccines are conceptually similar to ex vivo vaccines and bypass developing custom, GMP-compliant therapies for each patient. Although there are many types of in situ vaccines, their effective use should induce APC recruitment and tumor antigen loading and activation such that the APC can effectively cross-prime tumor-reactive T cells. In situ vaccination combines the immunologic benefits of presenting the full spectrum of tumor antigens with the practicality of off-the-shelf approaches. Numerous types of intratumorally administered agents including viruses, PRR agonists and other immune stimulants may be effective in situ vaccines if they can induce a systemic anti-tumor immune response or a vaccinal effect. Major advances across these therapy types (Table 5) have been largely driven by an increased understanding of the APC presenting tumor antigens.Table 5 Anonymous in situ loaded cancer vaccine trials and outcomes

Full size table

Dendritic cells

Given that tumors both exclude and inactivate DCs102, studies have attempted to replenish them intratumorally by direct administration, intending their subsequent uptake and presentation of tumor antigens. Autologous DCs, matured and activated ex vivo, have been injected in this manner, increasing intratumoral cytokine levels (for example, IL-12p40, IL-8, tumor necrosis factor (TNF)) that correlate with stable disease and prolonged survival103. Alternatively, immature DCs with increased phagocytic capacity have been injected alongside rituximab and GM-CSF following low-dose radiotherapy104. Frequent T cell responses and regressions at local and distant tumors correlated with the magnitude of effector responses, demonstrating the critical role of rigorous immune monitoring. A similar trial using IFN-α-activated DCs and rituximab but omitting radiotherapy induced lymphoma-specific CD4+ and CD8+ T cell responses and regressions at untreated tumors105. These two separate trials highlight the potential of endogenous colocalization of APCs and antigen to induce systemic tumor regressions. Additionally, immature, adenoviral-infected DCs expressing CCL21 were intratumorally injected in patients with NSCLC and induced tumor-infiltrating and circulating CD8+ T cells, with an upregulation in tumoral PD-L1 expression, correlating with systemic responses106.

Flt3L

Flt3L is the primary hematopoietic progenitor growth and differentiation factor responsible for mobilizing DCs, particularly the cross-presenting subset cDC1. Thus, Flt3L administration may be a more practical approach to replenish intratumoral DCs instead of their direct injection. Indeed, localized radiotherapy with Flt3L injection led to abscopal responses in nine of 29 treated patients with NSCLC105. A phase I study in which Flt3L- and herpes simplex virus 1 (HSV1)-thymidine kinase (TK)-expressing adenoviral vectors were injected into GBM tumor cavities following resection demonstrated immune cell infiltration and prolonged survival compared to contemporary controls107. Patients with low-grade B cell lymphoma treated in a phase I–II trial with intratumoral Flt3L, poly-ICLC and low-dose radiotherapy showed initial results of memory CD8+ T cell recruitment to untreated tumor sites associated with systemic tumor regression, with some lasting months to years4. A follow-up trial combines in situ vaccination with PD-1 blockade for patients with lymphoma, breast or head–neck cancer (NCT03789097). Although progress with Flt3L has been impeded by daily administration and limitation of available clinical reagents, several easier-to-use Flt3L formulations are entering the clinic (for example, NCT04747470). These data highlight the potential of DC recruitment in situ to elicit tumor-reactive T cell responses and persistent systemic remissions.

TLR agonists

TLRs are single-pass transmembrane PRR family receptors expressed on numerous leukocyte subsets such as myeloid cells and DCs that recognize structurally conserved pathogen-associated molecular patterns. Ten human and 13 murine TLRs have been identified, each with distinct pathogen-associated molecular pattern recognition. Synthetic TLR agonists have been developed to activate several human TLRs with promise to initiate anti-tumor immune responses.

TLR9 is an endosomal receptor highly expressed in many murine DC subsets, primarily in human B cells and plasmacytoid DCs, but not in cross-presenting cDC1 cells. Most TLR9 agonists are hypomethylated CpG-enriched oligonucleotides, classified as either CpG-A, CpG-B or CpG-C, which induce activation and proinflammatory cytokines (for example, type I IFN) in plasmacytoid DCs, B cells or both. Despite significant IFN induction and clinical enhancement of pathogen vaccines, TLR9 agonists are poor inducers of de novo human CD8+ T cell responses compared to other PRR agonists108. Despite promising early results109, a large phase III trial reported a 9% ORR with the CpG-B tilsotolimod plus ipilimumab, similar to ipilimumab alone (NCT02644967NCT03445533); studies for other tumor types are ongoing (NCT03865082). A trial in which a virus-like particle containing a CpG-A (CMP-001) was injected into patients with anti-PD-1-refractory melanoma demonstrated systemic regression as monotherapy and a 28% ORR with pembrolizumab (NCT02680184)110. Similarly, a CpG-C (SD-101) combined with pembrolizumab in a small study demonstrated a 78% ORR in anti-PD-1-naive patients but only a 15% ORR in anti-PD-1-experienced patients111. SD-101 was also studied with radiotherapy for low-grade lymphoma, leading to systemic tumor regression in six of 29 patients112. Prior studies of the CpG-B PF-3512676 (ref. 5) reflect similar results, possibly facilitated by high tumoral TLR9 expression. Overall, these data demonstrate that, while TLR9 agonists can induce intratumoral inflammation, that alone may be insufficient. If tumor antigen presentation to CD8+ T cells is critical, these antigens may need to be cross-presented by cDC1 cells, which do not strongly express TLR9.

TLR3 is primarily expressed on DCs, particularly cDC1 cells, and recognizes double-stranded RNA. It is the only described MyD88-independent TLR and signals via TIR domain-containing adaptor-inducing IFN-β (TRIF) to activate downstream nuclear factor (NF)-κB and IFN regulatory factor 3 (IRF3), among other pathways. The widely studied TLR3 agonist poly-ICLC (Hiltonol) is a synthetic complex of poly-inosinic-polycytidylic acid, poly-L-lysine and carboxymethylcellulose that activates distinct APC subsets via TLR3 and the RIG-I-like receptor (RLR) MDA-5 (ref. 113). Anecdotal reports of T cell activation, tumoral infiltration, local tumor regressions and prolonged survival after intratumoral poly-ICLC treatment have been described for patients with liver cancer114 and head and neck cancer115. Combining intratumoral poly-ICLC injection with radiotherapy and tumor lysate-pulsed DCs induced type I IFN expression, tumor-specific T cells and stable disease in a majority of patients as well as remarkable prostate cancer abscopal tumor regressions116. As noted, durable abscopal tumor regressions were observed in patients with lymphoma treated with an in situ vaccine composed of Flt3L, radiotherapy and poly-ICLC4, prompting a follow-up study combining this approach with pembrolizumab for patients with lymphoma, breast cancer or head and neck squamous cell carcinoma (NCT03789097). Newer poly-I:C formulations are immunologically distinct from poly-ICLC; rintatolimod (poly-I:C12U) activates TLR3 but uniquely avoids MDA-5 induction of TNF-dependent cytochrome c oxidase subunit II (COX2), IDO, IL-10 and Treg cell recruitment117. Additionally, intratumoral BO-112 (a nanoplexed poly-I:C) induced preclinical anti-tumor CD8+ T cell responses and, in combination with PD-1 blockade in anti-PD-1-refractory melanoma and patients with renal cancer, induced intratumoral CD8+ T cell infiltration and systemic tumor regression118.

TLR4 is a MyD88-semi-dependent PRR that binds to bacterial lipids (for example, lipopolysaccharide) to activate inflammatory responses, linking innate and adaptive immunity. Preclinical studies showed that a TLR4-binding component of inactivated Streptococcus pyogenes (OK-432) activated DCs, and intratumoral OK-432 administration has induced local recruitment of lymphocytes in patients with gastric cancer119 and increased APC levels in patients with pancreatic cancer120. A newer TLR4 agonist (G100), which contains the synthetic lipid A analog glucopyranosyl lipid A, administered intratumorally induced T cell infiltration and expression of immune-related genes correlating with clinical responses that lasted for years in a minority of patients with Merkel cell carcinoma121. In 26 patients with lymphoma receiving intratumoral G100, systemic regressions were observed in a significant minority of patients treated with G100 alone and a majority of patients when combined with pembrolizumab122.

Studies of additional TLR agonists such as TLR7, TLR8 and STING have also been reviewed123. Progress with a similar approach, activating APCs using agonistic anti-CD40 antibodies, has been stymied by toxicities when used as systemic therapy; thus, recent trials have begun to study intratumoral approaches (NCT02379741NCT04059588NCT03892525), with early clinical results showing safety of superficial intratumoral administration and PD-L1 upregulation in injected and un-injected tumors. Combining these agents for intratumoral injections could potentiate efficacy124. The induction of systemic tumor regressions in multiple tumor types is quite promising for these in situ vaccination approaches, but one concern is that tumors might exclude and inactivate APCs that express the PRR necessary for these approaches. Thus, the greatest potential may be combination approaches that recruit the PRR-expressing APC to the tumor site concurrent with intratumoral PRR-agonist administration.

Intratumorally administered oncolytic viruses and bacteria

Whereas oncolytic viruses’ preferential replication in and cytolysis of tumor cells could yield many therapeutic mechanisms, a main focus is their potential systemic vaccinal effect after intratumoral administration. Currently, the only Food and Drug Administration-approved oncolytic virus is talimogene laherparepvec (TVEC), a modified, GM-CSF-producing HSV1 virus that has demonstrated increased survival125 and tumor regression in non-injected lesions126 and is undergoing neoadjuvant and combination trials with checkpoint blockade. Similarly, since the earliest vaccinations by Drs. Coley and Old, attenuated live bacteria have been used to drive systemic anti-tumor immune responses. BCG has been administered as intravesical and intratumoral therapy, inducing local and distant tumor regression127. Similarly, attenuated Clostridium novyi intratumoral injections have demonstrated tumor-specific T cell induction and tumor regression128 and are now being combined with PD-1 blockade (NCT03435952). This broad field has great potential for rational engineering of viruses with distinct immunostimulatory profiles and clinical achievements, which are reviewed elsewhere129.

Perspectives

Although 5 decades of research have yielded many failures, vaccines are now positioned for success for several reasons. Compared to prior decades, it is now clear that (1) T cells can treat (and, in some instances, cure) patients with cancer, as seen with CAR T cells and bispecific T cell engagers; 2) patients’ endogenous T cells can be primed against their own TAAs, correlating with tumor regression, as seen with checkpoint blockade; and 3) priming of endogenous T cells requires optimal antigen presentation (for example, cDC1 cells). Which types of TAAs are the most promising (predefined or anonymous), how cDC1 cross-presentation can be optimized and by which means cross-primed tumor-reactive T cells can be measured in vaccinated patients remain to be addressed. Predefined shared antigen vaccines have dominated the field and demonstrated survival benefits, but success has been limited to tissue-specific antigens (for example, PAP, gp100). Targeting mutated TSAs (either with predefined personalized or anonymous vaccines) is appealing, but measuring resulting immune responses will be essential to their translation into the clinic. Even if using defined antigens, combinations of more than one antigen would likely offer superior efficacy. Furthermore, immune tolerance can arise from immunoediting for tumor evasion of immune cell clearance130. The clinical success of checkpoint blockade illustrates that blocking immunosuppressive pathways can be sufficient for reversing tolerance and allowing immune-mediated cancer rejection. Therefore, immunization strategies against TAAs must also address the TAA-specific immune tolerance present in the tumor host, notably by targeting or depleting TAA-specific Treg cells131,132,133.

Measuring pharmacodynamic effects before assessing anti-cancer efficacy is the gold standard of cancer therapy development; if ineffective kinase inhibitors were brought into efficacy trials, small-molecule chemotherapeutics would be hindered by numerous failures. Similar to pathogen vaccines, such as those against coronavirus disease 2019, that require potent humoral responses before clinical efficacy trials, immunotherapies should have similar metrics. The lack of reliably measurable cancer vaccine pharmacodynamics or ‘immunodynamics’ has led to insufficiently supported approaches moving to late-phase clinical trials, followed by failures that repeatedly set the field back. Effective immune monitoring will be critical to determining whether cancer vaccines accomplish their intended immunologic effects134 and to moving only immunologically effective candidates to larger studies and appropriate patient subsets. As with pathogen vaccines, early development of cancer vaccines focused on humoral responses to assess immunologic potency, rationalized by the anti-tumor efficacy of monoclonal antibody therapy for breast cancers and lymphomas. Extrapolating findings from preclinical murine models to humans has been limited by interspecies discrepancies in murine and human immune cell subsets, such as differential TLR expression on APCs. Conversely, T cell subset phenotypes and function have significant interspecies similarity. Therefore, even though personalized antigen identification is difficult, it may be possible to identify a unified tumor-reactive T cell phenotype in murine studies that could be extrapolated to human immune monitoring. Previously, murine CD8+ T cell PD-1 expression135 predicted that human PD-1 T cell expression can be an effective monitoring parameter in patients with cancer136.

Seminal studies suggest that anti-tumor T cell responses, more than those of B cells, are critical to vaccine anti-tumor efficacy17,137. However, measuring the anti-tumor function of T cells is difficult. Most T cell immune monitoring assays have been descriptive: assessing the phenotype or clonality of broad T cell populations. There is small precedent for descriptive assessment to serve as biomarkers for therapeutic efficacy: absolute lymphocyte counts correlate with some immunotherapy clinical outcomes138 and tumor-reactive T cells are enriched among CD8+ cells expressing activation or exhaustion markers such as PD-1, TIM-3 and LAG-3 (ref. 136). With high-throughput TCR sequencing, specific T cell clones can be tracked in the blood and importantly in the tumor139, with the degree of clonality predicting clinical response to some immunotherapies140. TCR identification can even be correlated with tumor antigen identity to a certain degree141,142, although the function and reactivity of most TCR clones will be unknown.

Moving beyond T cell description to assess tumor-reactive T cell function is straightforward with predefined antigen vaccines using T cell–peptide co-cultures (for example, enzyme-linked immune absorbent spot (ELISPOT) or flow cytometric analyses), and these assays have demonstrated moderate correlations with clinical response143 and survival144. Assessment of tumor-reactive T cells responding to anonymous antigen vaccines is more challenging and has been performed using T cell–tumor cell co-cultures, which have been correlated with clinical response104, although cryopreserved, autologous tumor is infrequently accessible. In principle, candidate neoantigens from anonymous antigen vaccines can be determined using mutation identification and identifying T cell responses to these antigens, as has been shown in patients treated with checkpoint blockade145, but this may be restrictively resource intense for broad use.

Industry–academic collaborations such as the Cancer Vaccine Consortium think tank have re-established vaccines as promising optimal combination therapies for checkpoint blockade, given their capacity to prime T cells, but emphasize that our ability to measure anti-tumor T cell responses will be even more important than the ability of vaccines to induce tumor regression as monotherapy146. To that end, innovative immune monitoring centers have now developed assays such as MANAFEST to unite functional T cell reactivity assays (for example, against neo-epitopes) with practical descriptive assays such TCR sequencing, allowing the latter to be surveyed serially in blood or tumor to measure anti-tumor T cell responses77,147. Going forward, such assays should extend beyond neo-epitope reactivity and probe for whole-tumor cell reactivity to allow measurement of the immune response to anonymous tumor antigen vaccines. As characterization data of neo-epitope or whole-tumor-reactive T cells accumulate, it is plausible that a common signature, measurable by single-cell RNA sequencing or flow cytometry, will be able to characterize effective vaccine-induced T cells. Current insensitive and nonspecific approaches (for example, IFN-γ ELISPOT) are posed to be replaced over the next 5 years with deep immune monitoring approaches to accurately characterize cancer vaccine immune responses. With such means, small trials will be able to quickly identify the most immunologically potent cancer vaccines, thereby avoiding large trials of less immunogenic vaccines. Deep immune monitoring will guide the field on a straightforward trajectory, evaluating the most promising approaches (likely neoantigen and in situ vaccines), to successful, randomized trials and ultimately commercialization. Effective vaccines are likely to be combined with other immunostimulatory approaches including adoptive T cell therapies and to be deployed in postsurgical adjuvant settings to prevent relapses.

Decades of slow progress have provided proof of principle that cancer vaccines can indeed elicit systemic tumor regression, durable remission and improvement in OS. We stand on the shoulders of pioneers who advanced our immunologic understanding and are on the precipice of using that understanding to develop rational and effective cancer vaccines, propelling the promising field of immunotherapy to a new frontier, saving resources, time and, ultimately, patients’ lives.

Remodeling How Cancer Vaccines Are Designed


Researchers from the International Institute for Nanotechnology (IIN) at Northwestern University have combined chemistry and nanotechnology to change the structural location of adjuvants and antigens on and within a nanoscale vaccine to boost potency and performance.

The study is published in Nature Biomedical Engineering in an article titled, “Multi-Antigen Spherical Nucleic Acid Cancer Vaccines.”

“Cancer vaccines must activate multiple immune cell types to be effective against aggressive tumors,” wrote the researchers. “Here we report the impact of the structural presentation of two antigenic peptides on immune responses at the transcriptomic, cellular, and organismal levels. We used spherical nucleic acid (SNA) nanoparticles to investigate how the spatial distribution and placement of two antigen classes affect antigen processing, cytokine production, and the induction of memory.”

“The work shows that vaccine structure and not just the components is a critical factor in determining vaccine efficacy,” explained lead investigator Chad A. Mirkin, PhD, director of the IIN. “Where and how we position the antigens and adjuvant within a single architecture markedly changes how the immune system recognizes and processes it.”

Mirkin and his team studied the effect of vaccine structure in the context of seven different types of cancer to date, including triple-negative breast cancer, papillomavirus-induced cervical cancer, melanoma, colon cancer, and prostate cancer.

“A challenge with conventional vaccines is that out of that blended mish mosh, an immune cell might pick up 50 antigens and one adjuvant or one antigen and 50 adjuvants,” said study author and former Northwestern postdoctoral associate Michelle Teplensky, PhD, who is now an assistant professor at Boston University. “But there must be an optimum ratio of each that would maximize the vaccine’s effectiveness.”

The researchers turned to SNAs (spherical nucleic acids), which are the structural platform used in this new class of modular vaccines. SNAs allow scientists to pinpoint exactly how many antigens and adjuvants are being delivered to cells.

“Vaccines developed through rational vaccinology deliver the precise dose of antigen and adjuvant to every immune cell, so they are all equally primed to attack cancer cells,” said Mirkin. “If your immune cells are soldiers, a traditional vaccine leaves some unarmed; our vaccine arms them all with a powerful weapon with which to kill cancer. Which immune cell ‘soldiers’ do you want to attack your cancer cells?” Mirkin asked rhetorically.

The team developed a cancer vaccine that doubled the number of cancer antigen-specific T cells and increased the activation of these cells by 30% by reconfiguring the architecture of the vaccine to contain multiple targets to help the immune system find tumor cells.

The team investigated differences in how well two antigens were recognized by the immune system depending on their placement of the SNA structure. They also studied how the different placements affected the immune system’s ability to remember the invader, and whether the memory was long-term.

“Where and how we position the antigens and adjuvant within a single architecture markedly changes how the immune system recognizes and processes it,” Mirkin said.

The study data revealed that attaching two different antigens to an SNA comprising a shell of adjuvant was the most potent approach for a cancer vaccine structure. It led to a 30% increase in antigen-specific T-cell activation and doubled the number of proliferating T cells compared to a structure in which the same two antigens were attached to two separate SNAs.

“It is remarkable,” Mirkin said. “When altering the placement of antigens in two vaccines that are nearly identical from a compositional standpoint, the treatment benefit against tumors is dramatically changed. One vaccine is potent and useful, while the other is much less effective.”

“The collective importance of this work is that it lays the foundation for developing the most effective forms of vaccine for almost any type of cancer,” Teplensky said. “It is about redefining how we develop vaccines across the board, including ones for infectious diseases.”

“The developments made in this work provide a path forward to rethinking the design of vaccines for cancer and other diseases as a whole,” Mirkin concluded.

Cancer vaccines: past, present and future; a review article


Abstract

Immunotherapy and vaccines have revolutionized disease treatment and prevention. Vaccines against infectious diseases have been in use for several decades. In contrast, only few cancer vaccines have been approved for human use. These include preventative vaccines against infectious agents associated with cancers, and therapeutic vaccines used as immunotherapy agents to treat cancers. Challenges in developing cancer vaccines include heterogeneity within and between cancer types, screening and identification of appropriate tumour-specific antigens, and the choice of vaccine delivery platforms. Recent advances in all of these areas and the lessons learnt from COVID-19 vaccines have significantly boosted interest in cancer vaccines. Further advances in these areas are expected to facilitate development of effective novel cancer vaccines. In this review, we aim to discuss the past, the present, and the future of cancer vaccines.

Introduction

Immunization has been practiced for hundreds of years starting with the use of snake venom to protect from snakebite to the development of smallpox vaccine in 1978. However, few vaccines have been effective in reducing cancer incidences in a population and in treating cancers. The past few years have seen tremendous breakthrough in vaccine technology, including the most recent use of nucleic acid vaccines against COVID-19 infection, and the development of cancer immunotherapy. Further advances in cancer vaccine technology are predicted in the future, which are discussed in this review.

A brief overview of the immune system

A full description of the immune system is beyond the scope of this review article. However, to better understand the concepts of immunotherapy and vaccines, we provide a brief overview of the immune system and its association with cancer immunity. The immune system is an integrated system of soluble molecules, cells, tissues, and organs that is capable of recognizing an invading antigen and initiating a cascade of responses that ultimately lead to the elimination of the foreign antigen. The ability to distinguish foreign versus self-antigens is a hallmark feature of the immune system. Based on the recognition mechanisms, the effector cells involved, and the speed at which effector mechanisms are elicited, immune responses can be broadly categorized into innate immunity and adaptive immunity. Innate immunity is present at birth and provides a generalized immediate response to foreign invaders [1]. The effector cells of the innate immune system including natural killer (NK) cells, neutrophils, macrophages, etc. Cells of the innate immune system recognize invading pathogens and cells with the help of germ-line encoded pattern recognition receptors (PRR) that recognize pathogen-associated molecular patterns (PAMP), which are usually shared among many different types of pathogens [1]. Adaptive immunity, on the other hand, develops after birth and is antigen specific [2]. Antigen-specific recognition is mediated by receptors, which are generated through gene recombination, giving rise to receptor diversity and antigen-specificity [2]. Adaptive immunity is mediated by B and T lymphocytes: the latter are either CD8+ cytotoxic and CD4+ helper T cells [3]. B cells work by producing antibodies (humoral immunity) against foreign antigens aiming to block their impact on cells and tissues, while T cells recognize and eliminate diseased cells (cellular immunity) [3]. An effective immune response against an antigen involves a concerted effort by both the innate and the adaptive arms of the immune system. Soluble factors, such as cytokines and chemokines, produced by the cells of the innate immune system are essential for activation of other immune cells, including B and T cells of the adaptive immune system. As well, processing of antigens by antigen presenting cells (APC), such as dendritic cells (DC), and presentation of antigenic peptides on human leukocyte antigens (HLA), the major histocompatibility complex (MHC) in humans, are critical for induction of adaptive immunity [4]. T cells possess antigen-specific T cell receptors (TCR), which recognize antigens presented on MHC molecules. A hallmark feature of the adaptive immune system, which makes long term immunity and immunization by vaccination possible, is antigen-specific immunological memory. After a primary challenge with an antigen, the adaptive immune system can mount a better, stronger, and faster response against the same antigen upon subsequence exposures [2]. Vaccines work by exposing the immune system to an immunizing antigen, which is derived from a pathogen, in the absence of an infection. Subsequently, the adaptive immunity and immunological memory elicited against the immunizing antigen protects an immunized individual against the pathogen from which the antigen was derived. Advances in understanding how the immune system recognizes and responds against pathogens and non-self/foreign cell have made possible the development of vaccines and other immune-based therapies.

In addition to immunity against foreign pathogens, several lines of evidence have demonstrated the important role of the immune system in cancer immunosurveillance and in immunity against cancers. These include: (i) increased incidences of cancers in individuals with a compromised immune system, (ii) increased incidences of cancers in patients undergoing immunosuppressive therapy, (iii) infiltration of immune cells into the tumour microenvironment and their association with better cancer prognosis, and (iv) presence of tumour antigen-specific T cells in cancer patients [5,6,7,8]. Not only the immune system responds against and eliminates cancers, but in doing so, it also shapes the tumour cells and the tumour microenvironment in a way that makes tumours resistant to further immune assault. This process is termed tumour immunoediting [9]. Tumours are also equipped with a multifaceted immune escape mechanism to evade detection and elimination by the immune system [10]. Over the past decades, advances made in understanding cancer immunosurveillance, immunoediting, immune escape processes have resulted in the development of affective cancer immunotherapies and vaccines, and will inform future development of novel immunotherapies and novel cancer vaccines.

Cancer immunotherapy

Although it is still far from being fully conquered, cancer treatment has transformed through the years. Surgery was the only treatment option prior to the development of chemotherapy, which was used for the first time in 1942 at Yale. The remarkable history of William Coley (Bone Surgeon, New York 1890–1936) in pioneering cancer immunotherapy, long before cancer immunosurveillance theory was established, is worth mentioning. Following his observation, in the late 1890s, of possible anti-cancer effect from bacteria infection, he started treating Osteosarcoma patients with a heat killed bacteria preparation (Coley’s toxin), which occasionally resulted in tumor regression [11]. Cancer immunotherapy has evolved since the pioneering work of William Coley. In the past 20 years, we have witnessed a significant breakthrough in cancer treatment with the addition of more targeted therapies and immunotherapy [12]. Cancer cells can avoid recognition by the immune system, and the immunosuppressive nature of the tumour micro-environment renders tumour-infiltrating lymphocytes ineffective at tumour elimination in vivo [1013]. The goal of immunotherapy is to overcome immune suppression in the tumour microenvironment and use the natural ability of the immune system to attack and eliminate cancers.

Cancer immunotherapy has come a long way from immune surveillance hypothesis entertained by Paul Ehrlich in 1909, to the development of various immunotherapy medications that are currently in use [14]. The scope of cancer immunotherapy grew from the use of immune stimulating cytokines like interleukin-2 (IL-2) and Interferon alpha (IFNα) in renal cell carcinoma and melanoma, to the use of immune modulating antibodies against Cytotoxic T cell Lymphoma Antigen 4 (CTLA 4) and Programmed Death 1 (PD-1) molecules. Although response rate was low (10–15%) and toxicity was significant, high dose IL-2 offered durable remission for the few responding patients [15].

Discovered by Pierre Goldstein in 1987, CTLA-4 is expressed on T cells after activation and its physiologic role is to suppress T cell responses. It is also referred to as a brake to the immune system or immune checkpoint. Following the observation of tumour shrinkage in mice treated with CTLA-4 antibodies, the monoclonal antibody to CTLA-4, Ipilimumab was developed for human use [16]. Ipilimumab was approved by the FDA in 2011 for the treatment of unresectable or metastatic melanoma. The approval was based on the phase III double blind study that showed overall response rate of 10.9% with 60% maintaining the response for at least 2 years. Immune related toxicity was seen in 60% of the patients with 7 deaths attributed to immune related toxicity [17]. Although this was a great breakthrough, the low response rate, albeit durable, and high rate of immune related toxicity (20% grade III and IV) necessitated the need for safer and more effective immunotherapy drugs. The anti-CTLA-4 monoclonal antibody therapy is still used in combination with other immunotherapy drugs in patients with metastatic melanoma [1819].

PD-1 is another immune checkpoint receptor expressed on activated T cells. Following the discovery of PD-1 and its ligand PD-L1 by Tasuku Honjo and Lieping Chen, respectively, it was shown (by Lieping Chen) that PD-L1 was upregulated in several cancers and blocking of the PD-L1/PD-1 interactions lead to tumour regression in mice [20]. This discovery led to development of monoclonal antibodies targeting PD-1/PD-L1 interactions to treat several types of human cancers, including renal cell carcinoma, melanoma, Hodgkin’s lymphoma, non-small cell lung cancer and others. For instance, Nivolumab, a PD-1 monoclonal antibody is approved by the FDA for the treatment of advanced melanoma (Dec 22, 2014), metastatic renal cell carcinoma (Nov 23, 2015), Hodgkin’s lymphoma (May 17, 2016), metastatic urothelial carcinoma (Feb 2, 2017) among others [21]. Pembrolizumab, another PD-1 monoclonal antibody is FDA approved for the treatment of advanced melanoma (Sep 4, 2014), advanced non-small cell lung cancer (Oct 2, 2015), head and neck squamous cell carcinoma (Aug 5, 2016), classical Hodgkin’s lymphoma (Mar 15, 2017), and advanced renal cell carcinoma (Aug 11, 2021) among others [22].

Another fascinating progress in utilizing the immune system to eradicate cancer is the use of chimeric antigen receptor (CAR) T cell therapy. CAR is a genetically modified receptor that is specific for a tumour-associated antigen (TAA). Patient T cells are transfected with a viral vector containing the CAR genetic code to produce CAR T cells, which are then adoptively transferred to the patient. CAR T cells are potent immune effectors that will expand and form a long-term tumour antigen-specific immunological memory. Many versions of the CAR T cell have been developed over the years to achieve a better anti-tumour activity, proliferation capacity, and improved in vivo persistence [23]. Currently, CAR T cell therapy is FDA approved for use in a variety of hematologic malignancies, including acute lymphoblastic leukemia and diffuse large B cell lymphoma [24].

Despite all these success stories, cancer immunotherapy is still not curative. Further research is needed to make cancer immunotherapy more effective and safer. Based on the tremendous achievements and the progress made so far in understanding anti-cancer immune responses, it is inevitable to anticipate growing interest to further exploit the interaction between cancer and the immune system to develop additional therapeutic agents, including cancer vaccines.

The current state of cancer vaccines

Cancer vaccines are designed with the intent of inducing an immune response against tumour antigens (Fig. 1). Despite decades of research and development, only few cancer vaccines have been approved for human use (Table 1): several other vaccines are in various phases of clinical trial (described below). The success of these cancer vaccines depends on several factors, including the type of antigens used, the tumour microenvironment, the immune landscape of the tumour, and the different vaccine formulations.

figure 1
Fig. 1

Table 1 The approved vaccines for cancer prevention and therapy

Full size table

Preventative cancer vaccines

Among the first effective vaccines in preventing cancers were those targeting viral infections associated with cancer development. Hepatitis B virus (HBV) is a leading cause of chronic liver disease that increases risk of hepatocellular carcinoma (HCC) [2526]. The vaccine for HBV has been available since the early 1980s and is recommended by World Health Organization (WHO) for infants soon after birth. Three doses of the vaccine are highly affective in providing long-lasting immunity against chronic HBV infection [25]. It was the first preventative vaccine shown to reduce incidence of HCC in vaccinated individuals [27]. Taiwan was one of the first nations to implement a nation-wide HBV vaccination program, initially given to infants born to infected mothers, and later extended to all infants in 1984. Subsequent studies showed a significant drop in HCC incidence in vaccinated Taiwanese children up to 20 years after the implementation of this vaccination program [2728]. Similar studies in Thailand, where the national neonate HBV vaccination program was implemented in the late 1980s, showed significantly lower HCC incidents in children vaccinated at birth [29]. A more impressive success story comes from the United States, where universal newborn immunization with HBV vaccine implemented in 1984 in Alaska Natives, has eliminated HCC among Alaska Native children under the age of 20 years [30]. Human papilloma virus (HPV) is a sexually transmitted virus associated with a few cancers, including cervical, oropharyngeal, anal, penile, and vulvovaginal cancers [31]. HPV vaccines are available since 2006 and is recommended as prophylactic vaccine for females and males over the age of 11 years and before the onset of sexual activity, i.e., before exposure to the virus [32]. Three HPV vaccines have been approved for prevention of HPV-related diseases in humans. HPV vaccines provide immunity against high-risk HPV types. These vaccines include the bivalent vaccine (Cervarix) against HPV types 16 and 18, the quadrivalent vaccine (Gardasil-4) against HPV types 6, 11, 16, and 18, and the nonavalent vaccine (Gardasil-9) against HPV types 6, 11, 16, 18, 31, 33, 45 52, and 58 [32]. HPV vaccines are safe, highly immunogenic in adolescence, and induce long-lasting antibody response and protection into adulthood [33]. Several phase II/III clinical trials have demonstrated high efficacy of HPV vaccines in reducing the risk of HPV-related high-grade cervical, vulvar, vaginal lesions, and genital warts in females [34,35,36,37,38]. A follow up monitoring of HPV vaccination programs in the United States and Australia targeting females aged between 11 and 26 years revealed significantly reduced HPV-related cervical lesions and abnormalities in the vaccinated compared to unvaccinated women [3639]. In the long term, high coverage HPV vaccination programs are expected to substantially reduce the rate of HPV-related cancers in both males and females.

No preventative vaccine for non-viral cancers has yet been approved for use in humans. This is partly due to the unavailability of appropriate TAA and the risk of cross-reaction with self-molecules on healthy tissues causing autoimmunity. However, a few TAAs have now been safely used in therapeutic vaccine trials without substantial autoimmune effects. Moreover, presence of autoantibodies against TAAs is associated with better prognosis in cancer patients and could even reduce cancer risk [40,41,42], suggesting that prophylactic induction of anti-TAA immune responses in the absence of cancer could potentially reduce cancer incidence. Additionally, advances in clinical imaging and diagnostic tools have improved the early detection of cancers and pre-malignant lesions, which provides an early window for the use of protective vaccines that induces anti-cancer immunity prior to its development.

Therapeutic cancer vaccines

Therapeutic cancer vaccines are used as immunotherapeutic tools to treat an active disease. Only two therapeutic vaccines have been approved in cancer immunotherapy. These include Bacillus Calmette-Guerin (BCG) vaccine for treatment of early-stage bladder cancer, and Sipuleucel-T (Provenge), a dendritic cell (DC)-based vaccine for treatment of castration-resistant prostate cancer [4344].

BCG is a non-pathogenic bacterium derived from Mycobacterium bovis, which induces a protective immune response against tuberculosis caused by Mtuberculosis. It remains the only commercially available vaccine against tuberculosis [45]. The use of BCG for treatment of high-risk non-muscle-invasive bladder cancer (NMIBC) was approved after it was shown in the mid-late 1970s that intravesical instillation of this bacterium could halt disease progression and recurrence of NMIBC [4647]. BCG vaccine has since been routinely used for the treatment of NMIBC, however, the precise immunological mechanisms of BCG therapy in NMIBC is unclear. The treatment consists of a weekly instillation of BCG into the bladder for 6 weeks after resection of the tumours. Patients can then enter a phase of maintenance treatment, which consists of weekly instillation of BCG vaccine into the bladder for 3–6 weeks every 3 months for 1–3 years [44]. BCG therapy is associated with some complications in the genitourinary tract, including cystitis, bladder ulceration, penile lesions, prostatitis, and kidney infection, as well as systemic complications, such as fever, disseminated infections, BCG sepsis, etc.[48].

Sipuleucel-T vaccine is an DC-based vaccine, which uses autologous DC to stimulate cellular immune responses mediated by T cells against prostatic acid phosphatase (PAP) in castration-resistant prostate cancer patients. DCs are antigen presenting cells that can efficiently induce antigen-specific priming and activation of T cell [4]. They express class I and class II HLA molecules and present processed antigenic peptide: HLA complexes to T cells. Sipuleucel-T vaccine is prepared by incubating patient DCs with a fusion protein, consisting of PAP linked to granulocyte macrophage colony-stimulating factor (GM-CSF), to induce DC activation, processing of PAP antigenic epitopes, and expression of antigenic peptide: HLA complexes and costimulatory molecules. Activated DCs are then reinfused into the patient, which will present antigens and activate T cell responses against PAP protein [49,50,51]. Sipuleucel-T vaccine was approved for treatment of castration-resistant prostate cancer after phase III trails showed significantly improved median survival and decreased risk of death in patients receiving the vaccine compared to the placebo-treated group, most notable in patients with a Gleason score of 7 or less [495253]. The treatment consists of three infusions of approximately 50 million autologous DCs given every two weeks. Adverse reactions were found to be mild in most patients and included flu-like symptoms, back pain, joint pain, muscle aches, headache, vomiting, constipation, diarrhea, anemia, and dizziness [43].

Cancer vaccines in clinical trials

In addition to the vaccines approved for treatment of cancers, several other cancer vaccines are in various phases of clinical trial, some of which are described here. A complete list of clinical trials involving cancer vaccines can be found at clinicaltrials.gov. Amongst these, are the whole tumour cell vaccines, which use killed tumour cells to stimulate anti-cancer immune responses. The advantage of using killed tumour cells as vaccine is that they can induce immune responses against multiple tumour antigens, which do not have to be prospectively identified. Tumour cells can also be genetically modified to secret immunomodulatory cytokines, such as GM-CSF, which can promote DC activation, and enhance antigen presentation and activation of adaptive immune responses [54]. Both autologous and allogeneic tumour cell vaccines (e.g. GVAX and Vigil vaccines) engineered to produce and release GM-CSF are under investigation and have been studied as monotherapy or in combination with other immunotherapy agents in cancers including pancreatic, prostate, ovarian and colon cancer [55,56,57,58,59,60]. A randomized trial that tested Vigil autologous GM-CSF vaccine in the frontline maintenance for stage III-IV ovarian cancer showed relapse-free survival (RFS) clinical benefit, specifically in patients without BRCA1 and BRCA2 mutations [55]. Despite, inducing immune responses in patients in several trials, whole tumour cells vaccines have so far failed to show a significant therapeutic efficacy to be considered for cancer immunotherapy. Additionally, GM-CSF can induce recruitment of myeloid suppressor cells, which could adversely affect anti-cancer immune responses in the tumour microenvironment [58]. Never the less, the relative simplicity of the procedure and the range of tumour antigens provided by whole tumour vaccines make them an attractive immunotherapeutic agent. Further trials in the future are aimed at increasing immunogenicity of whole tumour vaccines to elicit protective immunity and enhanced therapeutic efficacy against cancers.

Anti-idiotypic (anti-id) vaccines, such as Racotumumab, have shown some therapeutic efficacy and have been approved in Cuba and Argentina for treatment of advanced or recurrent non-small cell lung cancer (NSCLC). Anti-id vaccines use monoclonal antibodies that bind to antibodies specific to tumour antigens and block their interactions. Therefore, anti-id antibodies can mimic the structure of tumour antigens and elicit antigen-specific immune responses. Anti-id vaccines can be used to elicit immune responses against carbohydrate and lipid antigens, which are less immunogenic than protein antigens. Racotumumab induced immune responses against Neu-glycolyl-containing gangliosides, sulfatides, and other antigens expressed in tumours [6162]. Preclinical trials have demonstrated a strong antimetastatic effect, and clinical data have demonstrated its safety in NSCLC, breast cancer, advanced melanoma, and a significant clinical benefit in NSCLC patients compared to best supportive care [62,63,64,65].

Immunogenicity of tumour antigens and high mutational load are essential for response to immunotherapy. Glioblastoma, for instance, has low mutational load: hence, the limited intratumoral immune cell infiltration [66]. A number of different vaccine approaches including peptide vaccines and cell-based vaccines (DC and tumour-cell vaccines) have been trialed for treatment of glioblastoma, however, results of these studies have not been satisfactory to be considered for treatment [67,68,69]. In a phase I trial, glioma actively personalized vaccine (GAPVAC) using peptide antigens with polyriboinosinic-polyribocytidylic acid-poly-L-lysine carboxymethylcellulose (poly-ICLC) and GM-CSF adjuvants showed feasibility and a favourable safety with strong immunogenicity in newly diagnosed glioblastoma [70]. In this trial, patients were treated with a vaccine derived from unmutated premanufactured antigens (APVAC1) followed by a vaccine targeting mutated neoepitopes (APVAC2). APVAC1 induced a sustained response of memory CD8+ T cells, while APVAC2 mainly induced CD4+ T helper 1 type responses against predicted neoepitopes [70].

In many of the trials noted above [56646970], cancer vaccines are tried in conjunction with approved immunotherapies or other standard of care therapies. While vaccines can induce activation of anti-cancer immune effector functions, the activated immune cells must contend with the highly immunosuppressive microenvironment encountered within some tumors. The immunosuppressive mechanisms within tumor include, barriers to immune infiltration by anti-cancer immune cells, selective recruitment of cells with immunosuppressive activity, induction of T cell death, production of enzymes and metabolites that are immunosuppressive, and hypoxia conditions in tumors, all of which can hamper anti-cancer immune responses induced by vaccines [10]. Disruption of immune suppression in tumors through the use immunotherapies in combination with vaccines is, therefore, expected to enhance therapeutic efficacy. Alternatively, induction of anti-cancer immune responses by vaccines may enhance the efficacy of immunotherapeutic agents.

Tumour antigens for cancer vaccines

One of the challenges in developing cancer vaccines has been the nature of the antigens in tumours. An ideal tumour antigen for cancer vaccines would have high expression levels specifically in tumour cells, and broad expression pattern in multiple cancers. Based on their expression profiles, tumour antigens can be of two types: (i) tumour-associated antigens (TAA), and (ii) tumour-specific antigens (TSA) [71]. TAAs are self-antigens that may be expressed in normal tissues but overexpressed in multiple types of cancers (e.g., survivin), expressed in specific tissues and cancers of those tissues (e.g., Melan-A in melanoma and melanocytes), or mainly expressed in cancer tissues and their genes are silenced in normal adult tissues (e.g., cancer-testis antigens) [72,73,74]. TAA that are highly expressed in several types of tumours but have zero or at least low expression in healthy and mature somatic cells, have the potential to be targeted for cancer vaccines and immunotherapy. A major disadvantage of using TAAs for cancer immunotherapy is that TAAs are self-proteins, and therefore, inducing an immune response against them means breaking self-tolerance against self-antigens, which carries the risk of inducing autoimmunity. TSAs, on the other hand, are mutated self-antigens, also known as neoantigens, which are specific to each tumour, and are expressed on tumour cells only. Neve-rthe-less, there still remains the possible of cross-reactivity with the native antigen, resulting in autoimmune responses against self-antigens. As well, antigen loss may occur in tumours, rendering antigen-specific immune responses against the lost antigen ineffective. Additionally, not all neoantigens are immunogenic, making their identification, screening for immunogencity, and use in the development of personalized cancer vaccines expensive and arduous (Fig. 2) [75].

figure 2
Fig. 2

The discovery of neoantigens have opened new avenues for antigen screening and selection for cancer immunotherapy and vaccines. Gene mutations that occur during carcinogenesis take place in the coding and non-coding regions of the tumour cell’s genome. When a mutation occurs in the coding region of a gene, it results in a change in the amino acid sequence of the protein it codes for, and the expression of a neoantigen in the tumour cell [76]. Neoantigens are highly specific to each individual and are different from the traditional TAA [77]. Because neoantigens are expressed only on tumour cells but not normal cells, they are recognized as non-self antigens by the immune system. Tumour neoantigens are broadly categorized into two types: (i) shared neoantigens, and (ii) personalized neoantigens. Shared neoantigens are similar in different cancer patients; thus, it can be used in a wide-spectrum therapeutic approach for patients with the same mutated gene [78]. On the other hand, personalized neoantigens are unique and differ from patient to patient, therefore, the application of neoantigens in this case should be targeted to each individual (i.e. personalized therapy) [79].

All mutations that occur in tumours: point mutations, inversion, fusion, reading-frame alterations, and insertion-deletions, can generate neoantigens [80,81,82]. However, not all neoantigens are immunogenic and certain conditions must be met for a neoantigen to induce anti-tumour immunity. These conditions include sufficient expression of the specific neoantigen, high affinity of binding to the patient HLA molecules, and efficient recognition by the patient’s T cells [81]. Indeed, neoantigens with a strong affinity towards HLA molecules, as well as CD4+ and CD8+ T cells specific to these neoantigens have been detected in cancer patients [83,84,85].

The first step in neoantigen screening is the comparison of genomic DNA sequences from the normal cells and tumour cells (Fig. 2). This step is complex as some of the tumour’s mutations occur in the non-coding region of the genome and not all mutations are non-synonymous, that is they do not change the amino acid sequences of the protein. Moreover, the identification of mutant proteins that can elicit an anti-tumour immune response can be a challenging process [86]. Current development of bioinformatics tools and algorithms have increased the reliability and accuracy of neoantigen prediction and identification [87]. Next generation sequencing (NGS) has become one of the most versatile techniques that scientists have relied on for neoantigen discovery. Additionally, mass spectrometry (MS) has been used to predict post-translational modifications of neoantigens [8889]. In addition to the tumour genome sequencing data, information about the patient’s HLA haplotype, affinity of the mutated peptides towards the patient’s HLA, peptide-HLA complex stability, the affinity between peptide-HLA complex and T cell receptor, and T cell responses against the mutated peptides are needed to ascertain the immunogenicity of neoantigens and the specificity of anti-tumour immune responses [90,91,92]. It is estimated that only 10% of non-synonymous mutations in tumour cells can generate peptides with high affinity to the patient HLA molecules, and only 1–2% of peptide-HLA complex can stimulate CD8+ T cells [93].

Despite the challenges of neoantigen screening and identification, several studies have demonstrated the potential for the use of neoantigens in eliciting anti-tumour immune responses. Castle et al. was the first to use synthetic long peptide (SLP)-based vaccine using neoantigens in mouse melanoma models. From over 50 mutations identified, two mutated antigens showed significant therapeutic effects in these mouse models [87]. In 2015, Carreno et al. reported, for the first time, a personalized DC-based vaccine to treat patients with melanoma. In this study, neoantigens were identified using an immunohistochemistry approach and then patient’s DCs loaded with the neoantigens were transfused into patient, which resulted in enhanced anti-tumour T cell responses [94]. Similarly, Sahin et al. reported the use of RNA-based personalized vaccine using neoantigens identified in a trial that included 13 melanoma patients. Eight of these patients had no further tumour development over the following 23 months [95].

Several obstacles remain in the development and widespread use of neoantigen-based personalized cancer vaccines. The long cycle of cancer genome sequencing, neoantigen identification, and verification of immunogenicity add to the high cost of personalized vaccines, making them economically unfeasible for widespread use in cancer patients [79]. Development of improved bioinformatics tools for characterization of neoantigens, a more in depth understanding of tumour immunology, and advances in vaccine development and delivery methods will prove critical for the development and use of novel neoantigen-based cancer vaccines.

Modern and emerging vaccine technology

The fundamental goal of all vaccines is to prevent diseases, and their possible devastating consequences [96]. Any specific vaccine works by eliciting an immune response against an antigen that is found in the target pathogen [97]. Once primed by vaccination, the immune system will produce antibodies, cytotoxic cells, and memory cells that can neutralize and destroy the target pathogen before it causes a serious disease [9697]. There are many current and emerging types of vaccines that use either protein-based or gene-based approaches [98].

Nucleic acid vaccines

Two exciting examples of vaccine technologies that are currently in research are the deoxyribonucleic acid (DNA)- and ribonucleic acid (RNA)-based vaccines, which are referred to as genetic vaccines, or most commonly, nucleic acid vaccines [99100]. Currently, there are no DNA vaccines approved for human use, and the first RNA-based vaccines were rapidly developed in the wake of the COVID-19 pandemic [101102]. RNA-based vaccines use a type of RNA called messenger RNA (mRNA), which carries the genetic information for a protein antigen [98103]. mRNA vaccines for COVID-19 have been approved by Health Canada, granted emergency use authorization by the FDA, and granted emergency use validation by the WHO.

Unlike traditional vaccines that use a live attenuated or inactivated pathogen, or parts of the pathogen (subunit vaccines) as the source of the antigen, the nucleic acid vaccines use the genetic code for protein antigens derived from a pathogen to elicit an immune response [104]. Nucleic acid vaccines do not contain weakened or killed versions of the pathogen, or parts of the pathogen as the source of the antigen [102]. Instead, they make use of the cellular processes for protein synthesis to produce a protein, or a peptide, that triggers an immune response against a pathogen in what’s called a gene-based approach [98101105].

A key challenge in nucleic acid vaccines is shielding them from being degraded before reaching the target site. To avoid degradation of DNA vaccines by DNases and other nucleases, nano-carriers are often used [101]. There are many types of nano-carriers that are being studied, such as: nano-carriers composed of inorganic material, lipid-based nano-carriers, protein-based nano-carriers, and polymeric nano-carriers [101]. mRNA vaccines have been found to be most effective when the mRNA is bound to lipofection formulation [98]. A critical part of mRNA vaccines is successfully transporting the mRNA into the cells [106]. Utilizing lipid nanoparticles (LNP) is the most common method of mRNA vaccine delivery. Liposome-based transfection reagents containing cationic lipids are used to protect the mRNA from degradation by RNases as it makes its way into the cell through the extracellular space [98106]. LNPs are designed with an aqueous core, where the mRNA resides, which is enclosed by a lipid bilayer shell [106]. The recent development of ionizable lipids and lipid-like materials has increased the potency of LNPs [106]. Phospholipids, cholesterol, and lipid-anchored polyethylene glycol are also commonly used for both structural and functional purposes in LNPs [106]. Another exciting prospect of LNPs is the potential to embed lipophilic compounds into the lipid bilayer, including adjuvants to increase efficacy of the mRNA vaccine [106]. In addition to LNPs, extracellular exomes, which are key mediators of intercellular communication, are also extensively studied as a method for cancer vaccine delivery. Their optimal size, biocompatibility, stability and target specific delivery makes exomes attractive nano-carriers for vaccines [107].

There are several different routes of administration for DNA vaccines, including injection (Intravenous or Intramuscular), oral, topical, and pulmonary routes [101]. The most common method of delivery for mRNA vaccines is intramuscular injection [106]. This is the route of administration used in all the seven mRNA vaccines for COVID-19 that contain lipid nanoparticles [98]. Intramuscular injections are simple to perform, requiring little personnel training, and mRNA molecules are delivered to the muscle cells for protein synthesis [106]. Other modes of mRNA vaccine delivery that have been investigated include subcutaneous, intradermal, intravenous, intranasal, intranodal, and intraperitoneal methods [106].

How do nucleic acid vaccines work?

To understand how nucleic acid vaccines work, it is crucial to understand the normal protein synthesis process at the cellular level. Proteins are essential for many cellular functions in our body. Synthesis of proteins using the genetic code in DNA occurs through 2 stages: transcription, and translation [108]. In transcription, the nucleotide sequences in DNA are copied into a complimentary sequence in an mRNA molecule. Transcription occurs in the nucleus where the DNA is stored. The mRNA molecules are then transported into the cytoplasm where translation occurs. During translation, the nucleotide sequence in the mRNA, specifying an amino acid sequence, is read by ribosomes, which are organelles in the cytoplasm. Ribosomes move along the mRNA strand and use the genetic code of the mRNA sequence to translate the codons into their corresponding amino acids, which are then assembled to form a polypeptide and further processed to form mature proteins.

Nucleic acid vaccines use the cellular process of protein synthesis to produce antigenic peptides in the cells that will trigger an antigen-specific immune response. Following a route of injection, nucleic acid molecules (DNA or RNA) must enter keratinocytes and myocytes, as well as antigen-presenting cells (APC) near the injection site, in a process called transfection [101109]. DNA vaccines supply the DNA molecule, which contains the genetic code for a protein antigen, to our cells [110]. The DNA in the vaccine is in the form of a circular plasmid, which is derived from bacterial cells [110]. This DNA enters our cells and translocates to the nucleus, where it undergoes transcription to form a mRNA molecule [101]. The mRNA molecules are then translocated back to the cytoplasm, where the genetic code is read by ribosomes to synthesize a protein. mRNA vaccines work through the same cellular processes, however, instead of a DNA molecule it directly provides a pre-synthesized mRNA for translation into an antigenic protein (Fig. 3) [104]. Cellular transfection of myocytes and APCs by the LNP-encapsulated mRNA occur through endocytosis [98]. When the lipid nanoparticle enters the cytoplasm of the cell, they are degraded to release the mRNA in the cytoplasm, where they are translated into proteins by ribosomes [105]. The antigenic proteins are process by the antigen processing and presentation machinery in the cells to generate antigenic peptides, which can be recognized as foreign antigens by B cells directly and by T cells when they are presented on MHC molecules. APCs also take up proteins released from dying myocytes, which are then processed and presented on MHC molecules to activate T cells.

figure 3
Fig. 3

Advantages of nucleic acid vaccines over traditional vaccines

The benefit of mRNA vaccines lies in the possibilities that they hold for the future of safe and effective vaccination. Live-attenuated vaccines are the most potent type of vaccine in terms of activating both humoral and cellular immune responses [106]. Subunit vaccines are successfully used to generate humoral immunity against pathogens, but they do not generate cellular immunity, and often require the use of adjuvants to boost immunogenicity of the antigens [106]. Generating cellular immunity is critical to destroying the intercellular pathogen reservoir in some chronic diseases [106]. A significant drawback of live-attenuated vaccines is their rare but concerning potential to cause disease in immunosuppressed individuals [106]. Therefore, it is usually recommended that immunosuppressed patients do not receive live-attenuated vaccines [106]. mRNA and DNA vaccines have been developed with the hope of solving both safety and efficacy issues [106]. Nucleic acid vaccines have been proven to induce both cellular and humoral immunity (like live-attenuated vaccines), while also being safe (like subunit vaccines) since they do not contain whole or parts of the pathogen [106111]. Another advantage of nucleic acid vaccines, when compared to traditional vaccines, like live-attenuated vaccines and inactivated vaccines, is their ability to be manufactured rapidly [98]. The manufacturing process of the more traditional vaccines have safety concerns and carry a high risk of contamination with live pathogens [99]. The simple and safe manufacturing processes for nucleic acid vaccines are key reasons that they have been extensively researched [99].

Nevertheless, there are still safety concerns with nucleic acid vaccines that must not be overlooked, particularly with DNA vaccines. Since, DNA resides in the nucleus of cells, the DNA molecules in the vaccine must be transported to the nucleus of the cells [102106]. To facilitate transport of DNA into the nucleus, viral vectors, such as adenoviruses, are used as vehicles for DNA delivery [112]. However, the use of such delivery vectors could have potential safety concerns collectively referred to as vector-mediated genotoxicity [112113]. mRNA vaccines do not have to cross the nuclear membrane and are translated in the cytoplasm. Hence, they do not have similar safety concern as DNA vaccines [106]. In addition, mRNA molecules stay in the cytoplasm of the cell for a relatively short period of time before it is degraded [106].

Nucleic acid vaccines for cancer

The idea of using nucleic acid vaccines for the treatment of cancer, using both DNA- and RNA-based approaches, is a radical departure from the more traditional methods of cancer treatment, and represents a new era in cancer therapy. Generally, the goal of most vaccines is to prevent someone from either contracting a pathogen, or become seriously ill from the disease it causes, hence are called prophylactic vaccines [114]. Most cancer vaccines being researched are therapeutic in nature, i.e., they are used to treat the patient after they have developed a disease [99]. This is a key difference between the goals of nucleic acid vaccines for pathogens and nucleic acid vaccines for cancers. Therapeutic nucleic acid vaccines for the treatment of cancers rely on similar principles as the preventative vaccines, i.e., to teach the immune system to identify tumours and destroy them [99]. This is done by targeting antigens that are commonly expressed on cancer cells [115]. These antigens include growth associated factors, or antigens that are unique to cancer cells due to somatic mutations (neoantigens) [115]. Both mRNA and DNA vaccines work by delivering the genetic information to cells necessary to produce tumour antigens, and then induce immune responses against these antigens [99]. These antigen-specific immune responses, in particular the cytotoxic T cells, have the potential to clear tumour cells from the body, and therefore aid in cancer treatment (Fig. 3) [115116]. In a recent study, whether anti-tumour responses mediated by immune checkpoint inhibitors (ICIs) in solid tumour could be enhanced through immunizing patients against tumour antigens using mRNA vaccines was studied [117]. In the Phase I of the trials sponsored by ModernaTX Inc., mRNA-4157 was used as monotherapy in patients with resected solid tumours and in combination with pembrolizumab in patients with unresectable solid tumors. This combination was found to be safe and it induced neoantigen specific T cell responses in the immunized patients [117]. Currently, phase II trial is in progress (NCT03739931).

Lessons learned from the COVID-19 vaccines

COVID-19 is the disease caused by the virus SARS-CoV-2[98118]. SARS-CoV-2 is an enveloped virus with a positive-sense single-stranded RNA genome, belonging to the β-coronavirus subfamily [98]. An important part of the SARS-CoV-2 virus is it’s ‘spike (S) protein, which is a relatively large surface glycoprotein containing 1300 amino acids [98105]. It is encoded by the virus’ 3,822-bp S gene [98]. The role of the S protein is to interact with the host cells by binding to angiotensin-converting enzyme 2 (ACE2), allowing the virus to fuse with the cell membrane and enter cells [119120]. COVID-19 has a profound impact on certain organs, such as the lungs, heart, and kidneys, likely due to high expression of ACE2 on epithelial cells in these organs [121].

In response to the COVID-19 pandemic, many vaccines were developed. Safe and effective vaccines against SARS-CoV-2 are viewed as essential to conquering the COVID-19 pandemic, which has had an exceptionally negative impact on physical and mental health. Currently, there are over 100 vaccine candidates for SARS-CoV-2 being studied around the world according to the WHO. The leaders in the race to develop a vaccine against SARS-CoV-2 emerged as two mRNA vaccines, made by Pfizer and BioNTech, and by Moderna. These vaccines were both developed in under a year, which is considered incredible, since the fastest prior vaccine brought to the market was made in approximately 4 years. Along with the two mRNA vaccines, some of the other fastest developed vaccines against SARS-CoV-2 use viral vector technology, such as those developed by AstraZeneca [122].

Conventional vaccines usually take years or decades to manufacture and test, and ultimately obtain approval for human use [105]. Typically, they are very expensive to produce [105]. This makes them ill-suited to combat global pandemic from novel viruses, like SARS-CoV-2. mRNA vaccines however have the advantage of relatively low cost and rapid manufacturing.

To accelerate the process of approval, there were numerous strategies employed. In the United States, the FDA granted three vaccines developed by Pfizer-BioNTech, Moderna, and Janssen an Emergency Use Authorization (EUA). According to the FDA, an EUA is granted when the known and potential benefits outweigh the known and potential risks of the vaccine [123]. Granting an EUA to these vaccines allows them to be rolled out much faster than having to wait for a full FDA approval, which takes longer to achieve. As of August 23, 2021, the FDA has given full approval to the Pfizer-BioNTech COVID-19 Vaccine [123].

A critical problem in the effort to vaccinate the world against COVID-19 comes from widespread vaccine hesitancy and/or skepticism. As of early August 2021, the US COVID-19 vaccine rollout has slowed to concerning levels, with a substantial portion of the population refusing to be vaccinated against COVID-19. This is often attributed to a rise in misinformation due to the development of social media platforms with several conspiracy theories, specifically regarding the mRNA technology. Some of the conspiracy theories include belief of impaired fertility following vaccination, fear of mRNA integrating itself into the human genome and altering our DNA, and several others related to religious and political views. While none of these conspiracy theories are merit based, they persist and circulate in populations around the world, including the United States and Canada where vaccines are widely accessible [124].

Another challenge for some mRNA vaccines is the requirement for storage and transport at ultra-low temperatures [98]. One possible avenue for combatting this challenge of mRNA vaccines is developing new types of lipid nanoparticle technologies that are more stable at ambient temperature [98]. These lessons learnt and experiences gained from developing mRNA vaccines and immunizing nations against COVID-19 are going to be valuable in developing mRNA vaccines against cancer [125].

Conclusion

Cancer Immunotherapy properly named as the “Breakthrough of the year in 2013” has revolutionized the treatment of several types of cancer. The tremendous experience with nucleic acid vaccines in the COVID-19 era, will move the file of cancer vaccine forward. A time may come when cancer vaccines will be part of the immunization history, and oncologists share the joy of a pediatrician knowing that immunized patients are protected not only against infectious pathogens but against specific types of cancers