2,203
Views
0
CrossRef citations to date
0
Altmetric
Immunotherapy - Cancer

Unlocking cancer vaccine potential: What are the key factors?

, , , , , , , , & show all
Article: 2331486 | Received 08 Jan 2024, Accepted 13 Mar 2024, Published online: 02 Apr 2024

ABSTRACT

Cancer is a global health challenge, with changing demographics and lifestyle factors producing an increasing burden worldwide. Screening advancements are enabling earlier diagnoses, but current cancer immunotherapies only induce remission in a small proportion of patients and come at a high cost. Cancer vaccines may offer a solution to these challenges, but they have been mired by poor results in past decades. Greater understanding of tumor biology, coupled with the success of vaccine technologies during the COVID-19 pandemic, has reinvigorated cancer vaccine development. With the first signs of efficacy being reported, cancer vaccines may be beginning to fulfill their potential. Solid tumors, however, present different hurdles than infectious diseases. Combining insights from previous cancer vaccine clinical development and contemporary knowledge of tumor immunology, we ask: who are the ‘right’ patients, what are the ‘right’ targets, and which are the ‘right’ modalities to maximize the chances of cancer vaccine success?

This article is part of the following collections:
Cancer Vaccines

Introduction

Cancer is a major contributor to human mortality and morbidity, responsible for in one in six deaths worldwide.Citation1,Citation2 Its burden continues to increase due to shifts in demographic trends and the influence of globalization on several risk factors, such as aging populations, tobacco and alcohol use, physical activity, and environmental pollution.Citation1,Citation3 Technological advances, such as circulating tumor DNA (ctDNA) assays and low-dose computed tomography (LDCT) scans, are enabling population-level screening programs to be established for progressively lower costs. With the growing impact and accessibility of screening programs globally, there is a general trend toward earlier stage diagnoses across a range of solid tumor types.Citation4–6 Concurrently, increasingly complex cancer therapies – including antibody-drug conjugates, bioengineered cell therapies, and immune checkpoint inhibitors (ICIs) – have improved patient outcomes and survival significantly, but only for small proportions of patients.Citation7 Moreover, the high market price of these drugs has contributed to a snowballing in the cost of cancer care and, as a result, many are inaccessible to lower resourced settings.Citation8–10 The culmination of these trends in oncology – a growing global burden of cancer, a shift to earlier stage diagnosis, and the increasing cost of advanced therapeutics – highlights the need, specifically, for accessible treatments that are suitable for administration in the earlier stages of a cancer’s development.

Cancer vaccines offer a potential solution to the problems facing the field of oncology in the coming decade. These therapies benefit from economies of scale, simple administration, and may be more effective in earlier disease settings than later.Citation11,Citation12 Traditional vaccines aim to prevent or ameliorate infectious diseases through priming immune responses to exogenous antigens associated with given pathogens before a natural infection occurs. The induced adaptive immune response consists of humoral immunity (mediated predominantly by antibodies and memory B lymphocytes), and cellular immunity (typically carried out by CD4+ and CD8+ T lymphocytes). Similarly, cancer vaccines seek to induce an anti-tumor immunity using an appropriate delivery vector to prime responses to cancer antigens presented on the surface of tumor cells.Citation13,Citation14

Cancer antigens are most often variations of endogenous peptides (or their levels of expression) caused by alterations in the DNA of cancer cells during carcinogenesis. The immunogenic epitopes produced are broadly classified as Tumor-associated antigens (TAAs), Tumor-specific antigens (TSAs), and viral antigens.Citation15–17 TAAs are self-antigens that are encoded within an individual’s germline DNA, which have an abnormal level of expression on tumor cells relative to comparable normal cells. Examples of TAAs include the cancer testis antigens, prostate-specific antigen (PSA), and human epidermal growth factor receptor 2 (HER2).Citation16 TSAs are, in contrast, highly tumor-specific and are typically more immunogenic due to their complete absence in normal cells. Neoantigens are the best characterized type of TSA, which result from mutations that cause a nonsynonymous change in translated peptides. However, TSAs can emerge through a number of other less understood routes, such as post-translational modifications (e.g. glycosylation or phosphorylation) and from the translation of non-coding genomic regions.Citation18,Citation19 Viral antigens, unlike TAAs and TSAs, are completely foreign to the body and introduced to human cells via infection. These immunogenic targets can be clinically relevant cancer antigens in tumors resulting from oncogenic viruses – such as human papillomavirus (HPV).Citation20–22

A range of platforms have been explored over the decades of cancer vaccine development. Early attempts that directly administered naked, tumor-associated peptides to patients have given way to precision tumor antigen identification platforms, with the targets encoded in modalities such as bespoke mRNA and genetically modified viral constructs.Citation23,Citation24 Yet, to date, only one interventional cancer vaccine has been licensed for use: Sipuleucel-T, a Dendritic Cell (DC) vaccine created from autologous, peripheral-blood mononuclear cells (PBMCs) pulsed ex vivo with a recombinant fusion protein comprising prostatic acid phosphatase (PAP) fused to granulocyte – macrophage colony-stimulating factor (GM-CSF). Despite its approval in 2010 for use in castration-resistant prostate cancer, Sipuleucel-T was quickly superseded in common practice by hormone blockade agents that benefited from greater overall survival (OS), and a less complex and costly manufacturing process.Citation25,Citation26 However, the advances of vaccine technology made during the COVID-19 pandemic have since reinvigorated exploration of their use within oncology.Citation21 As of December 2023, there were 258 active, interventional studies of therapeutic cancer vaccines in solid tumor indications registered on ClinicalTrials.gov. While peptide and cellular modalities account for the majority of active studies, there are an increasing number of studies using mRNA and viral vectors following their efficacy in the pandemic and rapid regulatory approvals.Citation27

The type of immune response required to have a clinically significant impact on solid tumors is different to that needed in acute infections. For certain viral infections, for example, strong humoral responses are sufficient to ameliorate clinical symptoms upon later infection, even in the absence of significant cellular immunity.Citation28 The importance of humoral immunity, particularly the induction of neutralizing antibodies, was evidenced during COVID-19 vaccination where neutralization levels were associated with protection from symptomatic infection.Citation29 However, tumor targeting is instead largely dependent on T cell responses, and for cancer vaccines to be effective, there is a need for a high magnitude expansion of specific, cytotoxic T cell populations that can infiltrate and destroy tumors. It is also important that these proliferating, effector T cells do not enter an exhausted state – either as a result of persistent antigen stimulation or the immunosuppressive effects of the tumor microenvironment (TME).Citation30,Citation31 Ideally, this would be followed by a corresponding memory T cell kinetic, able to prevent clinical recurrence for several years.Citation32,Citation33 In contrast to natural infection – which tends to be intrinsically pro-inflammatory (e.g. via display of pathogen-associated molecular patterns (PAMPs)) – cancer can acquire immune evasive attributes quickly, making a durable immune memory crucial in the longer term to ensure rapid expansion and cell killing to prevent recurrent disease becoming established.Citation34,Citation35 While aspects of humoral responses have been associated with positive outcomes in the treatment of solid tumors, biomarkers of cellular immunity strongly correlate with positive prognosis.Citation36–38 Similarly, in observations of patients that have gone into long-term remission, significant proportions of tumor-reactive T cells may be found in the circulating volume many years after tumor regression.Citation39–42

Lack of understanding as to what constitutes the ‘right’ patients, targets, and modalities has hindered the progress of cancer vaccine development.Citation27,Citation43,Citation44 As discussed in detail elsewhere, improved knowledge of tumor biology and lessons learned from the development of other cancer immunotherapies have provided insights that can be applied to cancer vaccines.Citation12,Citation19 While these are transferable to selecting the right patients and targets, uncertainties around optimal modality choice remain (). We discuss all of these key considerations in detail below, with particular focus on their practicality, and ability to generate the required immune response profile to effectively target solid tumors.

Figure 1. Challenges facing cancer vaccine development.

Figure 1. Challenges facing cancer vaccine development.

Patient factors

While the general principle of a cancer ‘vaccine’ is seemingly well suited for their intended use, the vast majority of therapeutic cancer vaccine studies over the past decade have reported disappointing clinical outcomes. Early clinical studies of cancer vaccines tended to focus on patients with treatment-refractory disease. This has been standard practice for many first-in-human oncology studies but, as a consequence, these tend to recruit patients who are less physiologically fit with more advanced disease.Citation45,Citation46 As tumor burden increases, cancer can induce systemic immunosuppressive effects (e.g. increased production of neutrophils, eosinophils, and monocytes with reductions in dendritic cell, B cell and T cell populations from hemopoietic cells) as well as nurturing a localized, immune-evasive TME.Citation47,Citation48 The TME matures to a state of hypoxia, reduced pH, increased hydrostatic pressure, and erratic neo-angiogenic vasculature, which is also able to recruit immunosuppressive bystander cells.Citation49–51 These effects may reduce the ability of cancer vaccines to elicit a robust immune response.Citation20,Citation47 However, in the last 12 months alone, we have begun to see some promising results for mRNA-based cancer vaccines in melanoma and pancreatic cancer.Citation11,Citation52 Unlike many previous cancer vaccine studies that have focused on later disease settings, both of these studies have involved the adjuvant administration of treatment subsequent to curative-intent surgery. Further evidence to support the use of cancer vaccines in earlier disease settings includes HOOKIPA Pharma’s recent positive results using an arenavirus in combination with pembrolizumab in the first-line treatment of metastatic Head and Neck Squamous Cell Carcinoma (HNSCC).Citation53 These suggest that less established solid tumors may be more susceptible to the adaptive immune response generated by vaccines. Moreover, even with the ‘perfect’ cancer vaccine, primary T cell expansion in response to an antigen takes time (typically peaking around day 14) – not an insignificant length of time for patients with advanced, poor prognosis tumors.Citation54 This reframes the possible utility of cancer vaccines, and is supported by the growing understanding of immune-evasive mechanisms that solid tumors exploit as they become more advanced and able to cultivate an immune-evasive phenotype and TME.Citation11,Citation52 Exploring the window of opportunity offered in the adjuvant setting, in particular, is a strategy being adopted by a range of key players in this space during 2023 – including BioNTech, Merck, Genentech, Gritstone Bio, and Evaxion.Citation55

Beyond treatment setting, consideration must also be given to anti-cancer therapies that patients have received previously, as well as those administered in conjunction with cancer vaccines. Previous exposure to immunotherapies may induce a selection pressure toward clones with less immunogenic antigens and/or reduced antigen presentation, while cytotoxic agents and radiotherapy may produce immunosuppression that could hamper vaccine efficacy.Citation45,Citation56,Citation57 The early signs of clinical efficacy seen in the aforementioned studies are in protocols that combined vaccines with ICIs over a long duration.Citation55 While there is logic in suppressing checkpoint activity whilst administering a treatment designed to expand specific anti-cancer populations of T cells, the addition of ICIs is costly and not without the risk of significant toxicity.Citation58 Studies exploring the synergistic role of these interventions have suggested that ICIs help overcome immunosuppressive elements of the TME, however the impact of the TME and cancer-related systemic immunosuppression may be lessened by administering vaccines at earlier disease stages in immunocompetent patients.Citation59 Similarly, discussion around ‘hot’ and ‘cold’ tumor types (i.e. tumors that do, or do not, show infiltration by lymphocytes) that have surrounded translational analysis of ICI studies may be less relevant to cancer vaccines dosed in their earlier setting. It has also been noted that in controlled studies, cancer vaccines have demonstrated the ability to increase CD8+ T cell levels within tumors – suggesting a possible utility in cold tumors, particularly.Citation60,Citation61

Human leucocyte antigen (HLA) molecules are a crucial patient factor when considering antigen presentation, as core determinants of antigen sequences that can be recognized by an individual’s cellular immunity. The importance of these molecules in immunity has long been established in transplant medicine, but their implications on the immunogenicity of cancer antigens are yet to be fully understood.Citation13,Citation17,Citation18 Importantly, the genes encoding these molecules are diverse and vary greatly within populations. There is a historical bias in HLA research that has meant that HLA haplotypes that are more common in ethnic minorities are less understood.Citation62 If cancer vaccines are to be successful – particularly on a global scale – this must be considered, and concerted efforts be made to better characterize the impact of diverse HLA genotypes on cancer antigen presentation.

Target selection

Cancer vaccines utilize either a ‘personalized’ or ‘off-the-shelf’ model when delivering target antigens. Personalized vaccines require a sample of a patient’s tumor to produce a vaccine that delivers TSAs and TAAs specific to an individual’s cancer, while off-the-shelf platforms target TSAs, TAAs, or viral antigens that are known to be associated with a given cancer type.Citation63,Citation64 Personalized platforms typically use next-generation sequencing to compare an individual’s germline DNA to that of a tumor sample, in order to find specific mutations that could lead to TSAs/TAAs. All, or a selection of these, are then delivered to the patient in a bespoke vaccine. However, this process can be costly and require many months to manufacture.Citation65,Citation66 Another technique is to personalize vaccines using ‘anonymous’ antigens (i.e. without determining what TSAs/TAAs are present), for example by producing tumor lysate ex vivo to create a peptide vaccine. However, these still require tumor samples and are difficult to measure the immunogenicity of as the antigen payload is unknown.Citation65 Off-the-shelf vaccines use a selection of TSAs/TAAs that are associated with a particular tumor type, rather than patient-specific mutations. A major benefit of this approach is the ability to manufacture the same vaccine in bulk for many patients. Off-the-shelf methodology enables patients who have yet to undergo resection, or cannot due to co-morbidity or anatomical inaccessibility, to be treated in a semi-tailored manner while avoiding treatment delays resulting from manufacture.Citation13 This format would also allow for neoadjuvant treatment that would deliver antigens prior to surgical resection of tumors, avoiding postoperative immunosuppression – a period associated with cancer progression.Citation67 However, ‘off-the-shelf’ vaccines are reliant on a robust, preexisting understanding of cancer antigen expression and presentation within a particular cancer.Citation65 While this limitation may have held this methodology back previously, there are promising results emerging from off-the-shelf vaccine studies as understanding improves.Citation68,Citation69

The choice of target antigens must give careful consideration to intrinsic patient factors (e.g. HLA genotype) and tumor biology (e.g. peptide processing and presentation).Citation70,Citation71 While antigen discovery methods have been well reviewed elsewhere, it is important to understand the limitations of current techniques.Citation72 Broadly, the technologies and approaches to identifying these potential immunotherapeutic targets – which span point mutations, novel gene isoforms, expression of non-coding genomic regions, fusion, and frameshift peptides – have rapidly evolved. As mentioned, a key approach in cancer antigen identification is the use of next-generation sequencing to identify DNA or RNA-encoded tumor mutations and aberrant expression. Sequencing approaches coupled with machine learning models – to estimate the likelihood of translation and presentation – have been used in the majority of personalized neoantigen vaccines to date.Citation21,Citation55,Citation73 However, these have been unable to reliably predict immunogenicity and presentation of delivered targets – limiting their suitability to a larger rollout in a diverse population.Citation52 A fundamental approach gaining traction is immunopeptidomics, wherein mass spectrometry is employed to identify eluted peptides from tumor samples. Currently, immunopeptidomics is one of the only methods that can directly detect presented cancer antigens. Yet, it suffers from lower sensitivity and, alone, is unable to resolve tumor-specific peptides from those found in the normal human proteome.Citation18,Citation74 A combination of these techniques in the future may be able to improve the accuracy of target selection for cancer vaccines – as well as other cancer immunotherapies.Citation12,Citation14

Disagreement persists on the best approaches for discovering the ‘ideal’ cancer antigens, but there is consensus that these need to be robustly tumor-expressed and capable of eliciting cytotoxic T cell responses.Citation13,Citation17,Citation75 The gold standard approach is to perform ex vivo cultures of patient T cells with candidate targets (using either primed DCs or bead-based assays) with readouts via antigen-specific tetramer staining, detection of cytotoxic markers through ELISPOT, or intracellular cytokine staining. Increasingly sensitive assays for peptide-T cell recognition and stimulation have also been developed – these are of particular importance to personalized platforms wherein high throughput is needed to assess individuals’ unique sets of antigens.Citation76,Citation77

Cancer antigen presentation is required to provide an opportunity for immune cells to recognize and kill malignant cells, through an adaptive immune response. All intracellular proteins eventually undergo degradation with a subset of the resulting peptides being loaded onto HLA molecules. The subsequent complex is then trafficked to the cell surface for extracellular display.Citation78 T cells – particularly cytotoxic CD8+ cells – with corresponding T cell receptors (TCRs) are able to recognize presented TSAs and TAAs, enabling an immune response against the cancer cells. This occurs when epitopes are presented by HLA class I on the surface of cancer cells directly, or indirectly by antigen-presenting cells (APCs) that engulf and present immunogenic epitopes on HLA class II molecules. The activation of these cells via the recognition of presented TSAs/TAAs leads to T cell mediated cancer cell death. This cell death, in turn, causes an agnostic release of other cancer antigens that may be present in the tumor that can prime further adaptive immune responses. This series of steps is known as the cancer immunity cycle. This concept highlights that anti-cancer immunity can be self-reinforcing – a property that cancer vaccines may be able to exploit by initiating this cycle, even in instances where only a subset of antigens are targeted by the vaccineCitation47,Citation79 ().

Figure 2. The updated cancer immunity cycle annotated with cancer vaccine technologies and how their mechanisms interact with this cycle [adapted from Mellman et al.].Citation79 APCs, antigen-presenting cells; CTLs, cytotoxic T lymphocytes; TAA, Tumor-Associated Antigen; TSA, Tumor-Specific Antigen.

Figure 2. The updated cancer immunity cycle annotated with cancer vaccine technologies and how their mechanisms interact with this cycle [adapted from Mellman et al.].Citation79 APCs, antigen-presenting cells; CTLs, cytotoxic T lymphocytes; TAA, Tumor-Associated Antigen; TSA, Tumor-Specific Antigen.

Modality choice

The COVID-19 pandemic enabled relatively novel vaccine platforms to accelerate their clinical development to address the global threat that SARS-CoV-2 posed. This has led to billions of doses of vaccines derived from these technologies to be administered in less than 3 years, providing a robust understanding of their safety profile and enabling technical optimization of their manufacture.Citation13,Citation63 Despite differing in mechanism and construction, the two most widely deployed vaccine technologies used in the COVID-19 pandemic – mRNA and viral vectors – have demonstrated the ability to rapidly scale up regulatory-compliant manufacturing capacity, establish viable international supply chains, and have begun to benefit from economies of scale.Citation80 Whilst there are lessons to be learned from the deployment of vaccine technologies during the pandemic, there are still a number of questions that need to be asked to best support the clinical development of these therapies as they are translated into solid tumors. How generalizable are the results of COVID-19 studies to malignancies? What are the optimal characteristics of a vector delivering cancer antigens? Are these quintessentially different to those needed in infectious disease prophylaxis?

Cancer vaccines, by various means, all seek to introduce TSAs and/or TAAs to the immune system. This can be achieved by direct presentation of cancer antigens or the induction of expression via normal cell machinery (). A range of technologies have been harnessed to achieve the antigen delivery that is central to cancer vaccines; these include peptides, virus-like particles, cells (e.g. DCs), DNA, mRNA, and microbial vectors (e.g. viruses).Citation19 Despite a common objective, these modalities have been shown to provoke differing immune responses. We have begun to understand the relative strengths of different vaccine platforms in inducing humoral and cellular immunity through COVID-19 vaccines. For example, the Pfizer BNT162b2 mRNA vaccine stimulated higher magnitude B cell responses compared to viral vectors, which were superior in producing T cell responses.Citation81,Citation82 While variations in dose, and route and frequency of administration have also been shown to vary with the immune response seen, the intrinsic immunokinetics of each vaccine platform must be considered cognizant of the conditions required for an anti-tumor effect.Citation83,Citation84

Peptide vaccines

Peptide vaccines consist of sequences of amino acids that are designed to directly mimic tumor antigens of interest. Therapeutic cancer vaccines using this modality have been the longest in development; initially, these vaccines consisted of recombinant proteins and epitope-length peptides, but novel approaches like synthetic long peptides (SLPs) have become more common to improve on the stability of this type of vaccine. The use of longer peptides reduces their susceptibility to enzymatic digestion and allows for multi-epitope payloads. Human studies of these vaccines have shown them to be tolerable, however they suffer from unfavorable pharmacodynamics due to their charge and solubility.Citation13 Also, owing to a specific requirement for cross presentation of epitopes by cross-presenting dendritic cells to generate vaccine-specific CD8+ T cell responses, peptide vaccines typically induce higher proportions of CD4+ T cell vaccine-specific responses.Citation85,Citation86 While these vaccines benefit from easy synthesis and low cost, no peptide-based cancer vaccines have made it beyond Phase II trials due to a lack of efficacy.Citation87,Citation88 A recent study did, however, make note of the low-level persistence of some antigen-specific CD8+ clones in a small cohort (n = 5) of long-term survivors of hepatocellular carcinoma who had received a peptide cancer vaccine in a study (n = 60) almost a decade prior. Yet, the clinical significance is unclear due to the confounding effect of other treatments received by these patients.Citation89 Many ongoing peptide vaccine studies are now focused on exploring the use of adjuvants and immunotherapy-combinations in order to improve the poor efficacy elicited by this class of vaccine.Citation90

Virus-like particles

Virus-like particles (VLPs) are nanostructures that spontaneously assemble from viral structural proteins that lack the viral genetic material. They are hollow shells that mimic the overall structure of a virus but are incapable of causing a viral infection.Citation91,Citation92 Several VLPs – designed to display cancer-specific proteins or epitopes on their surface – have been explored as cancer vaccines in preclinical studies. Like peptide vaccines, VLP-based vaccines also rely on complicated cross-presentation mechanisms to generate a CD8+ T cell response. Similarly, VLPs tend to skew toward CD4+ rather than CD8+ T cell responses that may impact their clinical efficacy in solid tumors.Citation91,Citation93 There are currently a small number of active trials using VLPs as therapeutic cancer vaccines, but these have yet to read out any mature data.Citation93

Cellular vaccines

Cell-based vaccines use autologous immune cells from patients – typically derived from PBMCs – to deliver TSAs/TAAs. Most often these have utilized monocyte-derived DCs, taken from peripheral blood sampling or by leukapheresis. These are then exposed, ex vivo, to target antigens (e.g. via incubation with tumor lysate or by transfection) before being expanded and reinfused into a patient, wherein cells carry out APC functions to elicit an adaptive immune response. This class of vaccine has been studied for over three decades and is the technology behind Sipuleucel-T, discussed earlier. A major restriction of current cell-based vaccines is their autologous origin. Extracting and stimulating these cells is a laborious process and, for immunocompromised patients in particular, can result in low yields in the final product. Co-opting the induced APCs ability to sort through a multitude of antigens within, for example, whole cell lysate provides a means of targeting anonymous antigens while avoiding the need for costly genetic analysis.Citation26,Citation77,Citation94 However, as mentioned, this method limits the ability to understand which antigens are most immunogenic and, as a consequence, the generalizability of results.Citation13

DNA vaccines

DNA-based vaccines work via ‘reverse vaccination’ strategies, wherein the vaccine does not contain the antigen(s) itself, rather the genetic blueprint with which to produce them. Once this is transfected into the nucleus of host cells, it can then exploit native machinery to produce the desired antigenic peptide. Typically, these use DNA plasmids that encode both the target antigen and stimulatory adjuvants (e.g. cytokines, colony stimulating factor). This is an established platform used experimentally in infectious disease and cancer settings and, as such, has a well characterized and tolerable safety profile.Citation95,Citation96 These compounds enjoy high stability but, due to the intrinsic negative charge of DNA, they suffer from poor cell penetrance and immunogenicity – the latter addressed somewhat by the encoding co-stimulatory molecules within the DNA sequence, but still requiring efficient protein translation for this to take effect. This requires crossing both the cell membrane and the nuclear envelope to access the necessary cellular machinery, whereas mRNA vaccines need only cross the cell membrane. This hurdle can be improved by electroporation, however this is a time-consuming and clinically skilled task limited to accessible anatomical areas.Citation19 Even so, the optimal way to deliver a DNA vaccine is still an open question and the need for specialized delivery devices adds to the overall cost of treatment. Clinical studies of a DNA vaccine encoding PAP – the same target as Sipuleucel-T – demonstrated an improved PSA doubling time in phase I. Yet, this did not translate to improved PFS in later phases.Citation25 Chimeric DNA vaccines – encoding antigens with xenogeneic domains – seek to improve immunogenicity using extra-species elements.Citation97 However, aside from the emergency approval of ZyCov-D – an DNA vaccine against SARS-Cov-2 in India – there have been no approvals of DNA vaccines in humans. Despite this, a DNA-based cancer vaccine for use in canine melanoma has been recently approved by the FDA.Citation98

mRNA vaccines

In contrast to DNA, mRNA is very unstable. It is subject to rapid degradation at room temperature and, in vivo, free mRNA is rapidly broken down by nucleases.Citation21,Citation43 As a result, mRNA vaccines consist of mRNA molecules bound to a biochemical agent (typically lipidic) to improve stability. Both elements contribute to the overall immunogenicity of the vaccine. Native uracil nucleosides in mRNA sequences trigger pattern recognition receptors in APCs, inducing an innate immune response that results in reduced antigen presentation due to rapid clearance. Replacing these with nucleosides modified analogues to avoid triggering APCs has been demonstrated to increase antigen production.Citation99 Liposome delivery formulations – such as the lipid nanoparticles (LNPs) used in BioNTech’s COVID-19 vaccine – have been developed to reduce the intrinsic immunogenicity of free mRNA, and encourage preferential endocytosis by APCs. Lipid nanoparticles also help direct the immune response by stimulating the production of IL-6, thereby promoting T follicular helper cell differentiation, and enhancing B cell and antibody responses.Citation100 This proclivity for professional APCs, including DCs, gives mRNA vaccines access to the cytoplasmic machinery needed to be translated into peptides as well as the opportunity to be presented on APC-restricted HLA-I and II molecules. HLA-I and II can readily stimulate both CD4+ and CD8+ cells, which produces a more balanced immune response than DNA-based vaccines – albeit with a shorter duration of expression and still with a disproportionate CD4+ effect.Citation58 Solutions such as self-replicating mRNA vaccines may improve these outcomes by increasing the duration of protein expression, but further refinements are needed to bolster CD8+ activation for solid tumors.Citation101 Another challenge with mRNA vaccines in oncology is the issue of rapid dispersion at the site of injection, which – depending on anatomical location – may limit their ability to reach lymphoid tissue and circulating APCs in order to maximize response. Intravenous administration allows for systemic circulation of vaccine and has been demonstrated to improve antigen production. However, this is associated with an increased risk of toxicity – although they are still well tolerated relative to other anti-cancer treatments.Citation102 Finally, the durability of immune responses generated by mRNA vaccines remains unclear with many current approaches using high-frequency repeat dosing in order to generate continuous T cell stimulation, although aforementioned developments like amplifying and circular mRNA may reduce this requirement.Citation103

Prior to the pandemic, the initial mRNA vaccines platforms were, predominately, intended for the treatment of cancer. These included longer mRNA molecules encoding multiple TSAs/TAAs, yet many of these failed to demonstrate efficacy. This resulted in cancer vaccines losing momentum relative to other immunotherapy classes.Citation19,Citation43,Citation104,Citation105 Two mRNA studies this year have reignited the interest in cancer vaccines at large, namely, the academic-led, neoadjuvant pancreatic ductal adenocarcinoma trial at Memorial Slone Kettering (co-sponsored by BioNTech) and KEYNOTE-942 (NCT03897881), the cutaneous melanoma study from a Merck and Moderna collaboration. The former is a Phase I trial of a personalized mRNA vaccine (encoding a maximum of 20 neoantigens) that was administered to 16 patients in 8 weekly doses, starting nine-week post-surgical resection, prior to atezolizumab and mFOLFIRINOX adjuvant immunochemotherapy. While data is not yet mature, the eight patients who were considered ‘responders’ (based on evidence of T cell response on ELISpot) are recurrence-free at 18 months follow-up. Interestingly, only 11% of the predicted neoantigens produced an immunogenic response, and the resulting T cell responses were not significantly impaired by the subsequent cytotoxic chemotherapy.Citation52 The Merck study combined another personalized mRNA vaccine (encoding a maximum of 34 neoantigens) with pembrolizumab in the adjuvant setting of recently resected melanoma. This study showed a 44% decreased risk of disease recurrence or death at 18 months. In February 2023, the FDA granted a breakthrough therapy designation to mRNA-4157 plus pembrolizumab as an adjuvant treatment for patients with high-risk melanoma following a complete resection.Citation11 These are significant developments, but both studies rely on combination therapy with long courses of ICIs – adding significantly to the costs of these regimens.Citation106

Viral vector vaccines

Viral vector vaccines work by reprogramming viruses – which have been attenuated or rendered replication-defective – to deliver TSAs and/or TAAs using recombinant DNA technology. Like DNA and RNA vaccines, these require in vivo protein synthesis to generate antigenic peptides to be subsequently presented. In contrast, they do not require artificial constructs (e.g. LNPs) or adjuvants to produce strong innate and adaptive immune responses. Co-evolution with viral species has resulted in the human immune system producing vigorous CD4+ and CD8+ T cell responses to viral PAMPs. Virally transduced host cells present both viral peptides and the vaccine’s encoded antigens, acting as a natural adjuvant to the payload of the vaccine.Citation19,Citation95,Citation107 Common species used in this application include Adenoviruses (Ad), Adeno-associated viruses (AAV), Arenaviruses, Chimpanzee adenovirus (ChAd), and modified Vaccinia Ankara (MVA).Citation108,Citation109 The biology behind different viral vectors and their immune activity is nuanced and has been extensively reviewed elsewhere.Citation110 However, viral biology affords the opportunity to engineer vectors that enjoy advantages over other vaccine types.Citation107 Compared to mRNA, for example, viral vaccines benefit from cost efficiency, scalability, and thermostability. These features were highlighted by the significant cost-effectiveness advantage that viral vector vaccines – such as ChAdOx1 nCov-19 and Jcovden – demonstrated during the COVID-19 pandemic.Citation111–113

The immune system has evolved to respond to viruses with strong T cell responses; natural viral infections can confer immunokinetics that can persist over many decades.Citation114 The typical kinetics of a T cell response can be divided into three phases: rapid expansion of the primary effector response, contraction of the responding population upon resolution of infection, and establishment of a long‐lived memory population. Unlike the typical T cell response with distinct phases, there are observations in adenovirus vectors of “memory inflation.”Citation115 This response – also described in other natural viral infections – is characterized by persistent, high-frequency, antigen-specific CD8+ T cells driven by the prolonged presentation of low-level antigen to the immune system.Citation116 This attribute lends viral vectors a key advantage in producing the desired duration of CD8+ activation required for effective cancer vaccines.

Recombinant viral vectors have been used to prime immune responses to a range of pathogens for over 40 years; recent highlights include prophylactic vaccines against Malaria, Ebola, and COVID 19.Citation107 Their utility in cancer vaccines has also benefited from advances in cancer immunology and they, too, are beginning to show signs of efficacy.Citation44,Citation49 The viral species that are chosen for use in viral vectors tend to, deliberately, be those that are not known to cause life-threatening complications. These may be commensal species or their zoonotic variants, the latter being more prevalent in non-human hosts that also reduces the risk of preexisting immunity.Citation107,Citation117,Citation118 The immune system may already possess a primed response against a viral vector or can quickly generate one – such as neutralizing antibodies (NAbs). The existing, or resultant, immune neutralization of the viral vector has previously caused concern regarding antigen presentation efficacy and subsequent immunogenicity. However, viral vaccines in those with preexisting immunity have been shown to be immunogenic in Ebola (Ad5-EBOV) and COVID-19 (CanSino’s Ad5-nCoV vaccine).Citation49,Citation119,Citation120 Viral vector vaccines can also be modified to circumvent anti-vector immunity, improve transgene expression in target cells, and deliver multiple antigens.Citation49,Citation121 Another strategy to avoid neutralization by the immune system is heterologous dosing, a method that consists of using the same antigen payload in different vectors; subsequent ‘booster’ doses, if required, are delivered by an alternative vector avoiding the anti-vector immune response primed by the first dose. This technique consists of two or more different viral species, or multiple vaccine modalities. This strategy has been shown to precipitate superior immune responses relative to homologous regimens of both mRNA and viral vector platforms in a number of COVID-19 studies.Citation122–124

Cancer vaccines studies using viral vectors are beginning to see positive results, including in more advanced settings. As mentioned, HOOKIPA Pharma has demonstrated success in a phase I study of its two-vector, off-the-shelf, arenavirus vaccine targeting HPV viral antigens in HPV+, locally advanced or metastatic HNSCC. In patients with refractory disease, they demonstrated a disease control rate of 80% - exceeding the historical comparison of 35% with pembrolizumab monotherapy.Citation53,Citation125 Similarly, Barinthus Biotherapeutics (previously Vaccitech) are advancing an off-the-shelf, prostate cancer vaccine into phase II. Targeting TAAs associated with prostate cancer, a prior study using an early version of their ChAd and MVA platform reported that 22% of patients with metastatic, castration-resistant prostate cancer experienced biochemical remission (i.e. decrease of PSA by >50%) following treatment.Citation126 A TSA targeting vaccine has also shown promise. An off-the-shelf Great Apes Adenovirus and MVA viral platform – ‘Nous-209’ produced by Nouscom – has been used to target 209 common frameshift antigens in colorectal and gastric cancer patients with high microsatellite-instability. In combination with pembrolizumab, this treatment has resulted in clinically significant increases in progression-free survival in patients with refractory, metastatic disease. However, this study has yet to publish mature data.Citation127 Viral vectors are also being utilized in personalized platforms. Gritstone Bio’s GRANITE is a heterologous vaccine approach using ChAd68 and a self-amplifying RNA, encoding up to 20 neoantigens. Phase I/II results in advanced metastatic solid tumors demonstrated consistent induction of CD8+ T cells against multiple neoantigens and IFNγ responses, detectable by ELISpot in 100% of the patients who received both prime and boost doses. While data is immature, there were some early signs of efficacy in patients with metastatic, microsatellite-stable colorectal cancer.Citation125,Citation128

Discussion

After taking a backseat to the meteoric rise of ICIs, it is encouraging to see the first signs of efficacy within cancer vaccines. Cancer vaccines present selected antigens to the immune system outside of the immunosuppressive TME, in order to induce a specific, adaptive immune response. This offers the potential to overcome the limitations of currently licensed immunotherapies and, simultaneously, improve their safety profile.Citation13 The lackluster results seen in early cancer vaccine studies, it seems, may be attributable to trial design and contemporary understanding of target selection and antigen presentation rather than fundamental issues with the modalities used. Despite their early failure in solid tumors, these studies provided useful insights when these platforms were repurposed during the pandemic.Citation129

There is a clear shift of focus toward the adjuvant setting for cancer vaccines. This seems like a logical choice given the discussed obstacles that come with more advanced disease settings. Earlier disease settings minimize the impact of the TME, treat patients when they are most immunocompetent, and remove the confounding factor of prior anti-cancer treatment.Citation12 This environment can be used to better study the adaptive immune responses generated in a disease-free setting. Focusing on this niche also addresses the growing need for cost-effective treatments for patients increasingly diagnosed in the early stages of cancer.Citation130,Citation131 However, cancer vaccines may well be efficacious in more advanced settings, too, when combined with other treatments – as is already suggested by a number of viral vector vaccines beginning to report positive results in more advanced settings in combination with ICIs.Citation55,Citation124

As target selection and the understanding of antigen presentation continue to advance, cancer vaccines will likely improve in efficacy and safety. While there is a current trend toward personalized vaccines, these require significant resources and time to manufacture. However, the detailed analysis required by these bespoke platforms will lead to a deeper understanding of TSA/TAA prevalence across tumor types and ethnic groups. This, in turn, may enable more effective off-the-shelf vaccines to be produced, which can benefit from economies of scale. Not dissimilar to the current direction of travel toward allogenic cellular therapies, it is pragmatic to develop platforms that can deliver effective cancer treatment in a semi-tailored manner rather than a fully bespoke one. This strategy may offer the benefits of personalized medicine without the cost and infrastructure required for fully personalized therapies.Citation132 Additionally, effective off-the-shelf vaccines would enable neoadjuvant dosing to be explored; using the window of opportunity while patients are treatment-naïve and physiologically fitter before surgery and subsequent treatment (). This is a paradigm shift that has recently been effectively exploited in ICI therapies.Citation133

Figure 3. Opportunities for ‘off the shelf’ and personalized cancer vaccines in the neoadjuvant and adjuvant settings.

Figure 3. Opportunities for ‘off the shelf’ and personalized cancer vaccines in the neoadjuvant and adjuvant settings.

Many technical questions remain in regard to the use of different cancer vaccine modalities. Seemingly simple clinical parameters in other drugs, like dose and frequency of administration, are made more difficult in this setting by the variable immunogenicity of antigens and unclear half-life of presentation, factors that may be further complicated by heterologous dosing regimens.Citation83,Citation134 However, clinical optimization of these will come as they progress toward market authorization. In regard to the route of administration, at least, intravenous appears to be the most effective across modalities.Citation123 Considering the immunological characteristics of the modalities discussed, it seems the enthusiasm surrounding mRNA and viral vectors is not unfounded. It is impressive to see signs of efficacy in the adjuvant setting – especially in pancreatic cancer – and particularly remarkable to see similar suggestions in advanced, refractory cancers treated with off-the-shelf, viral vectors.Citation13,Citation55 The latter potentially owing its success to the superior ability of viral vaccines to produce cytotoxic T cell responses.Citation49 However, these results are in the context of ICI combination therapy and whether cancer vaccines are able to elicit clinically significant responses as monotherapies is yet to be seen. Regardless, it is reassuring to see – even with long courses of ICIs – both modalities have demonstrated strong safety and tolerability profiles.Citation25,Citation124,Citation135

We are, seemingly, in the midst of a cancer vaccine renaissance. Given the limitations and cost of ICIs – as well as the stalled progress in other solid tumor immunotherapies – it is important to see signs of efficacy in cancer vaccines studies.Citation136–138 The silver lining of the global COVID-19 pandemic may well be the advancement of vaccine technology to the point where it now has the potential to address the growing burden of cancer. However, it is vital that the differences between acute viral infections and solid tumors are appreciated when interpreting these results; while vaccines producing high magnitude – but short-term – responses may be efficacious in the former, evidence suggests solid tumors require high-magnitude and long-lived, anti-tumor T cell responses. It is important, too, that we learn not only from the logistics of the platforms used in the COVID-19 vaccine roll-out but also the issues that arose around equitable access and cost-effectiveness. To make a meaningful impact on the increasing global incidence of cancer, we need to place a specific focus on developing treatments that are cost-effective and efficacious across diverse populations.Citation10,Citation139

Author contributions statement

MG and LNL were involved in the conception and design, analysis, and interpretation of the data; the drafting of the paper, revising it critically for intellectual content; and the final approval of the version to be published.

SC, JK, OT, VPA, and CL were involved in the drafting of the paper, revising it critically for intellectual content; and the final approval of the version to be published.

NC, LT, and AS were involved in the drafting of the paper.

All authors agree to be accountable for all aspects of the work.

Acknowledgments

The authors would like to thank Professors Teresa Lambe and Paul Klenerman (University of Oxford) for their input and guidance on the content of this article.

Disclosure statement

MG is a shareholder in Achilles Therapeutics Ltd. and has previously received honoraria from Merck Sharp & Dohme.

LNL, SC, and JK are co-founders and shareholders of Infinitopes Ltd.

Additional information

Funding

The author(s) reported that there is no funding associated with the work featured in this article.

References

  • Chen S, Cao Z, Prettner K, Kuhn M, Yang J, Jiao L, Wang Z, Li W, Geldsetzer P, Bärnighausen T, et al. Estimates and projections of the global economic cost of 29 cancers in 204 countries and territories from 2020 to 2050. JAMA Oncol. 2023;9(4):465–13. doi:10.1001/jamaoncol.2022.7826.
  • Ward ZJ, Scott AM, Hricak H, Atun R. Global costs, health benefits, and economic benefits of scaling up treatment and imaging modalities for survival of 11 cancers: a simulation-based analysis. Lancet Oncol. 2021;22(3):341–50. doi:10.1016/S1470-2045(20)30750-6.
  • Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49. doi:10.3322/caac.21660.
  • Whitaker K. Earlier diagnosis: the importance of cancer symptoms. Lancet Oncol. 2020;21(1):6–8. doi:10.1016/S1470-2045(19)30658-8.
  • Price S, Spencer A, Zhang X, Ball S, Lyratzopoulos G, Mujica-Mota R, Stapley S, Ukoumunne OC, Hamilton W. Trends in time to cancer diagnosis around the period of changing national guidance on referral of symptomatic patients: a serial cross-sectional study using UK electronic healthcare records from 2006–17. Cancer Epidemiol. 2020;69:101805. doi:10.1016/j.canep.2020.101805.
  • Swanton C, Neal RD, Johnson PWM, Dur CC, Hamilton SA, Zhang N, Kumar H, Sasieni P. NHS-Galleri trial design: equitable study recruitment tactics for targeted population-level screening with a multi-cancer early detection (MCED) test. J Clin Orthod. 2022;40:6606–6606. doi:10.1200/JCO.2022.40.16_suppl.TPS6606.
  • Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707–23. doi:10.1016/j.cell.2017.01.017.
  • Tran G, Zafar SY. Financial toxicity and implications for cancer care in the era of molecular and immune therapies. Ann Transl Med. 2018;6(9):166. doi:10.21037/atm.2018.03.28.
  • Ventola CL. Cancer immunotherapy, Part 3: challenges and future trends. P T. 2017;42:514–21.
  • Green AK. Challenges in assessing the cost-effectiveness of cancer immunotherapy. JAMA Netw Open. 2021;4:e2034020. doi:10.1001/jamanetworkopen.2020.34020.
  • Khattak A, Weber JS, Meniawy T, Taylor MH, Ansstas G, Kim KB, McKean M, Long GV, Sullivan RJ, Faries MB, et al. Distant metastasis-free survival results from the randomized, phase 2 mRNA-4157-P201/KEYNOTE-942 trial. J Clin Oncol. 2023;41:9503–9503. suppl 17; abstr LBA9503. doi:10.1200/JCO.2023.41.17_suppl.LBA9503.
  • Esfahani K, Roudaia L, Buhlaiga N, Del Rincon SV, Papneja N, WH M Jr. A review of cancer immunotherapy: from the past, to the present, to the future. Curr Oncol. 2020;27:S87–97. doi:10.3747/co.27.5223.
  • Tagliamonte M, Cavalluzzo B, Mauriello A, Ragone C, Buonaguro FM, Tornesello ML, Buonaguro L. Molecular mimicry and cancer vaccine development. Mol Cancer. 2023;22:75. doi:10.1186/s12943-023-01776-0.
  • Lang F, Schrörs B, Löwer M, Türeci Ö, Sahin U. Identification of neoantigens for individualized therapeutic cancer vaccines. Nat Rev Drug Discov. 2022;21:261–82. doi:10.1038/s41573-021-00387-y.
  • Sahin U, Derhovanessian E, Miller M, Kloke B-P, Simon P, Löwer M, Bukur V, Tadmor AD, Luxemburger U, Schrörs B, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547(7662):222–6. doi:10.1038/nature23003.
  • Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14:135–46. doi:10.1038/nrc3670.
  • Jhunjhunwala S, Hammer C, Delamarre L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat Rev Cancer. 2021;21(5):298–312. doi:10.1038/s41568-021-00339-z.
  • Bedran G, Gasser H-C, Weke K, Wang T, Bedran D, Laird A, Battail C, Zanzotto FM, Pesquita C, Axelson H, et al. The immunopeptidome from a genomic perspective: establishing the noncanonical landscape of MHC class I–associated peptides. Cancer Immunol Res. 2023;11:747–62. doi:10.1158/2326-6066.CIR-22-0621.
  • Sellars MC, Wu CJ, Fritsch EF. Cancer vaccines: building a bridge over troubled waters. Cell. 2022;185(15):2770–88. doi:10.1016/j.cell.2022.06.035.
  • Jin K-T, Du W-L, Liu Y-Y, Lan H-R, Si J-X, Mou X-Z. Oncolytic virotherapy in solid tumors: the challenges and achievements. Cancers. 2021;13:588. doi:10.3390/cancers13040588.
  • Ladak RJ, He AJ, Huang Y-H, Ding Y. The Current landscape of mRNA vaccines against viruses and cancer-A mini review. Front Immunol. 2022;13:885371. doi:10.3389/fimmu.2022.885371.
  • Elsheikh R, Makram AM, Huy NT. Therapeutic cancer vaccines and their future implications. Vaccines (Basel). 2023;11:11. doi:10.3390/vaccines11030660.
  • Guo C, Manjili MH, Subjeck JR, Sarkar D, Fisher PB, Wang X-Y. Therapeutic cancer vaccines: past, present, and future. Adv Cancer Res. 2013;119:421–75.
  • Dobosz P, Dzieciątkowski T. The intriguing history of cancer immunotherapy. Front Immunol. 2019;10:2965. doi:10.3389/fimmu.2019.02965.
  • Rastogi I, Muralidhar A, McNeel DG. Vaccines as treatments for prostate cancer. Nat Rev Urol. 2023;20(9):544–59. doi:10.1038/s41585-023-00739-w.
  • Sutherland SIM, Ju X, Horvath LG, Clark GJ. Moving on from sipuleucel-T: new dendritic cell vaccine strategies for prostate cancer. Front Immunol. 2021;12:641307. doi:10.3389/fimmu.2021.641307.
  • Chakraborty C, Sharma AR, Bhattacharya M, Lee S-S. From COVID-19 to cancer mRNA vaccines: moving from bench to clinic in the vaccine landscape. Front Immunol. 2021;12:679344. doi:10.3389/fimmu.2021.679344.
  • Wherry EJ, Barouch DH. T cell immunity to COVID-19 vaccines. Science. 2022;377(6608):821–2. doi:10.1126/science.add2897.
  • Cromer D, Steain M, Reynaldi A, Schlub TE, Wheatley AK, Juno JA, Kent SJ, Triccas JA, Khoury DS, Davenport MP, et al. Neutralising antibody titres as predictors of protection against SARS-CoV-2 variants and the impact of boosting: a meta-analysis. Lancet Microbe. 2022;3(1):e52–61. doi:10.1016/S2666-5247(21)00267-6.
  • Gumber D, Wang LD. Improving CAR-T immunotherapy: overcoming the challenges of T cell exhaustion. EBioMedicine. 2022;77:103941. doi:10.1016/j.ebiom.2022.103941.
  • Nüssing S, Trapani JA, Parish IA. Revisiting T cell tolerance as a checkpoint target for cancer immunotherapy. Front Immunol. 2020;11:589641. doi:10.3389/fimmu.2020.589641.
  • Saxena M, van der Burg SH, Melief CJM, Bhardwaj N. Therapeutic cancer vaccines. Nat Rev Cancer. 2021;21:360–78. doi:10.1038/s41568-021-00346-0.
  • Vishweshwaraiah YL, Dokholyan NV. mRNA vaccines for cancer immunotherapy. Front Immunol. 2022;13:1029069. doi:10.3389/fimmu.2022.1029069.
  • Escamilla-Tilch M, Filio-Rodríguez G, García-Rocha R, Mancilla-Herrera I, Mitchison NA, Ruiz-Pacheco JA, Sánchez‐García FJ, Sandoval‐Borrego D, Vázquez‐Sánchez EA. The interplay between pathogen-associated and danger-associated molecular patterns: an inflammatory code in cancer? Immunol Cell Biol. 2013;91:601–10. doi:10.1038/icb.2013.58.
  • Li D, Wu M. Pattern recognition receptors in health and diseases. Signal Transduct Target Ther. 2021;6(1):291. doi:10.1038/s41392-021-00687-0.
  • Willsmore ZN, Harris RJ, Crescioli S, Hussein K, Kakkassery H, Thapa D, Cheung A, Chauhan J, Bax HJ, Chenoweth A, et al. B cells in patients with melanoma: implications for treatment with checkpoint inhibitor antibodies. Front Immunol. 2020;11:622442. doi:10.3389/fimmu.2020.622442.
  • Litchfield K, Reading JL, Puttick C, Thakkar K, Abbosh C, Bentham R, Watkins TBK, Rosenthal R, Biswas D, Rowan A, et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell. 2021;184(3):596–614.e14. doi:10.1016/j.cell.2021.01.002.
  • McCarthy PM, Valdera FA, Smolinsky TR, Adams AM, O’Shea AE, Thomas KK, Van Decar S, Carpenter EL, Tiwari A, Myers JW, et al. Tumor infiltrating lymphocytes as an endpoint in cancer vaccine trials. Front Immunol. 2023;14:1090533. doi:10.3389/fimmu.2023.1090533.
  • Yan Y, Cao S, Liu X, Harrington SM, Bindeman WE, Adjei AA, Jang JS, Jen J, Li Y, Chanana P, et al. CX3CR1 identifies PD-1 therapy–responsive CD8+ T cells that withstand chemotherapy during cancer chemoimmunotherapy. JCI Insight. 2018;3:3. doi:10.1172/jci.insight.97828.
  • Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, Gao P, Bandyopadhyay S, Sun H, Zhao Z, et al. Decade-long leukaemia remissions with persistence of CD4+ CAR T cells. Nature. 2022;602(7897):503–9. doi:10.1038/s41586-021-04390-6.
  • D’Angelo SP, Melchiori L, Merchant MS, Bernstein D, Glod J, Kaplan R, Grupp S, Tap WD, Chagin K, Binder GK, et al. Antitumor activity associated with prolonged persistence of adoptively transferred NY-ESO-1 c259T cells in synovial sarcoma. Cancer Discov. 2018;8(8):944–57. doi:10.1158/2159-8290.CD-17-1417.
  • Crompton JG, Sukumar M, Restifo NP. Uncoupling T-cell expansion from effector differentiation in cell-based immunotherapy. Immunol Rev. 2014;257(1):264–76. doi:10.1111/imr.12135.
  • Duan L-J, Wang Q, Zhang C, Yang D-X, Zhang X-Y. Potentialities and challenges of mRNA vaccine in cancer immunotherapy. Front Immunol. 2022;13:923647. doi:10.3389/fimmu.2022.923647.
  • Chang R, Gulley JL, Fong L. Vaccinating against cancer: getting to prime time. J Immunother Cancer. 2023;11:11. doi:10.1136/jitc-2022-006628.
  • Cuzzubbo S, Mangsbo S, Nagarajan D, Habra K, Pockley AG, McArdle SEB. Cancer vaccines: adjuvant potency, importance of age, lifestyle, and treatments. Front Immunol. 2020;11:615240. doi:10.3389/fimmu.2020.615240.
  • Liu J, Fu M, Wang M, Wan D, Wei Y, Wei X. Cancer vaccines as promising immuno-therapeutics: platforms and current progress. J Hematol Oncol. 2022;15(1):28. doi:10.1186/s13045-022-01247-x.
  • Hiam-Galvez KJ, Allen BM, Spitzer MH. Systemic immunity in cancer. Nat Rev Cancer. 2021;21(6):345–59. doi:10.1038/s41568-021-00347-z.
  • Kim SK, Cho SW. The evasion mechanisms of cancer immunity and drug intervention in the tumor microenvironment. Front Pharmacol. 2022;13:868695. doi:10.3389/fphar.2022.868695.
  • Wang S, Liang B, Wang W, Li L, Feng N, Zhao Y, Wang T, Yan F, Yang S, Xia X, et al. Viral vectored vaccines: design, development, preventive and therapeutic applications in human diseases. Signal Transduct Target Ther. 2023;8(1):149. doi:10.1038/s41392-023-01408-5.
  • Nia HT, Munn LL, Jain RK. Physical traits of cancer. Science. 2020;370:370. doi:10.1126/science.aaz0868.
  • Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12(1):31–46. doi:10.1158/2159-8290.CD-21-1059.
  • Rojas LA, Sethna Z, Soares KC, Olcese C, Pang N, Patterson E, Lihm J, Ceglia N, Guasp P, Chu A, et al. Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature. 2023;618(7963):144–50. doi:10.1038/s41586-023-06063-y.
  • Pharma H. Hookipa pharma reports first Quarter 2023 financial results and recent business highlights. Hookipa Pharma Inc n.d. [accessed 2024 Mar 2]. https://ir.hookipapharma.com/node/8826/pdf.
  • Pennock ND, White JT, Cross EW, Cheney EE, Tamburini BA, Kedl RM. T cell responses: naive to memory and everything in between. Adv Physiol Educ. 2013;37:273–83. doi:10.1152/advan.00066.2013.
  • Dolgin E. Personalized cancer vaccines pass first major clinical test. Nat Rev Drug Discov. 2023;22(8):607–9. doi:10.1038/d41573-023-00118-5.
  • Kallingal A, Olszewski M, Maciejewska N, Brankiewicz W, Baginski M. Cancer immune escape: the role of antigen presentation machinery. J Cancer Res Clin Oncol. 2023;149(10):8131–41. doi:10.1007/s00432-023-04737-8.
  • Chang S, Kohrt H, Maecker HT. Monitoring the immune competence of cancer patients to predict outcome. Cancer Immunol Immunother. 2014;63(7):713–9. doi:10.1007/s00262-014-1521-3.
  • Miao L, Zhang Y, Huang L. mRNA vaccine for cancer immunotherapy. Mol Cancer. 2021;20(1):41. doi:10.1186/s12943-021-01335-5.
  • Batista-Duharte A, Hassouneh F, Alvarez-Heredia P, Pera A, Solana R. Immune checkpoint inhibitors for vaccine improvements: Current status and new approaches. Pharmaceutics. 2022;14:14. doi:10.3390/pharmaceutics14081721.
  • Wang L, Geng H, Liu Y, Liu L, Chen Y, Wu F, Liu Z, Ling S, Wang Y, Zhou L. Hot and cold tumors: immunological features and the therapeutic strategies. MedComm. 2023;4:e343. doi:10.1002/mco2.343.
  • Paston SJ, Brentville VA, Symonds P, Durrant LG. Cancer vaccines, adjuvants, and delivery systems. Front Immunol. 2021;12:627932. doi:10.3389/fimmu.2021.627932.
  • Smithy JW, Blouin A, Diamond LC, Postow M. Ensuring equity in the era of HLA-restricted cancer therapeutics. J Immunother Cancer. 2022;10:10. doi:10.1136/jitc-2022-005600.
  • Farlow A, Torreele E, Gray G, Ruxrungtham K, Rees H, Prasad S, Gomez C, Sall A, Magalhães J, Olliaro P, et al. The future of epidemic and pandemic vaccines to serve global public health needs. Vaccines (Basel). 2023;11:11. doi:10.3390/vaccines11030690.
  • Dyer O. Covid-19: countries are learning what others paid for vaccines. BMJ. 2021;372:n281. doi:10.1136/bmj.n281.
  • Lin MJ, Svensson-Arvelund J, Lubitz GS, Marabelle A, Melero I, Brown BD, Brody JD. Cancer vaccines: the next immunotherapy frontier. Nat Cancer. 2022;3:911–26. doi:10.1038/s43018-022-00418-6.
  • Khan ST, Montroy J, Forbes N, Bastin D, Kennedy MA, Diallo J-S, Kekre N, Fergusson DA, Lalu M, Auer RC, et al. Safety and efficacy of autologous tumour cell vaccines as a cancer therapeutic to treat solid tumours and haematological malignancies: a meta-analysis protocol for two systematic reviews. BMJ Open. 2020;10(6):e034714. doi:10.1136/bmjopen-2019-034714.
  • Tang F, Tie Y, Tu C, Wei X. Surgical trauma-induced immunosuppression in cancer: recent advances and the potential therapies. Clin Transl Med. 2020;10(1):199–223. doi:10.1002/ctm2.24.
  • Sahin U, Oehm P, Derhovanessian E, Jabulowsky RA, Vormehr M, Gold M, Maurus D, Schwarck-Kokarakis D, Kuhn AN, Omokoko T, et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature. 2020;585(7823):107–12. doi:10.1038/s41586-020-2537-9.
  • Badrinath S, Dellacherie MO, Li A, Zheng S, Zhang X, Sobral M, Pyrdol JW, Smith KL, Lu Y, Haag S, et al. A vaccine targeting resistant tumours by dual T cell plus NK cell attack. Nature. 2022;606(7916):992–8. doi:10.1038/s41586-022-04772-4.
  • Kalaora S, Lee JS, Barnea E, Levy R, Greenberg P, Alon M, Yagel G, Bar Eli G, Oren R, Peri A, et al. Immunoproteasome expression is associated with better prognosis and response to checkpoint therapies in melanoma. Nat Commun. 2020;11(1):896. doi:10.1038/s41467-020-14639-9.
  • Spiliopoulou P, Janse van Rensburg HJ, Avery L, Kulasingam V, Razak A, Bedard P, Hansen A, Chruscinski A, Wang B, Kulikova M, et al. Longitudinal efficacy and toxicity of SARS-CoV-2 vaccination in cancer patients treated with immunotherapy. Cell Death Disease. 2023;14(1):49. doi:10.1038/s41419-022-05548-4.
  • Peri A, Salomon N, Wolf Y, Kreiter S, Diken M, Samuels Y. The landscape of T cell antigens for cancer immunotherapy. Nat Cancer. 2023;4(7):937–54. doi:10.1038/s43018-023-00588-x.
  • Xie N, Shen G, Gao W, Huang Z, Huang C, Fu L. Neoantigens: promising targets for cancer therapy. Sig Transduct Target Ther. 2023;8:9. doi:10.1038/s41392-022-01270-x.
  • Zhang B, Bassani-Sternberg M. Current perspectives on mass spectrometry-based immunopeptidomics: the computational angle to tumor antigen discovery. J Immunother Cancer. 2023;11:11. doi:10.1136/jitc-2023-007073.
  • Becker JP, Riemer AB. The importance of being presented: target validation by immunopeptidomics for epitope-specific immunotherapies. Front Immunol. 2022;13:883989. doi:10.3389/fimmu.2022.883989.
  • Kula T, Dezfulian MH, Wang CI, Abdelfattah NS, Hartman ZC, Wucherpfennig KW, Lyerly HK, Elledge SJ. T-Scan: a genome-wide method for the systematic discovery of T cell epitopes. Cell. 2019;178:1016–28.e13. doi:10.1016/j.cell.2019.07.009.
  • Morse MA, Gwin WR III, Mitchell DA. Vaccine therapies for cancer: then and Now. Target Oncol. 2021;16:121–52. doi:10.1007/s11523-020-00788-w.
  • Cruz-Tapias P, Castiblanco J, Anaya J-M. Major histocompatibility complex: antigen processing and presentation. El Rosario University Press; 2013.
  • Mellman I, Chen DS, Powles T, Turley SJ. The cancer-immunity cycle: indication, genotype, and immunotype. Immunity. 2023;56(10):2188–205. doi:10.1016/j.immuni.2023.09.011.
  • Reza HM, Sultana F, Bari R, Cole J, Baqui AH. Local distribution infrastructure and robust vaccine manufacturing facilities in LMICs should be prioritised to tackle ongoing and future pandemic risk. Lancet Reg Health Southeast Asia. 2023;11:100158. doi:10.1016/j.lansea.2023.100158.
  • Parry H, Bruton R, Tut G, Ali M, Stephens C, Greenwood D, Faustini S, Hughes S, Huissoon A, Meade R, et al. Immunogenicity of single vaccination with BNT162b2 or ChAdOx1 nCoV-19 at 5–6 weeks post vaccine in participants aged 80 years or older: an exploratory analysis. Lancet Healthy Longev. 2021;2:e554–60. doi:10.1016/S2666-7568(21)00169-0.
  • Ward H, Whitaker M, Flower B, Tang SN, Atchison C, Darzi A, Donnelly CA, Cann A, Diggle PJ, Ashby D, et al. Population antibody responses following COVID-19 vaccination in 212,102 individuals. Nat Commun. 2022;13(1):907. doi:10.1038/s41467-022-28527-x.
  • Palgen J-L, Feraoun Y, Dzangué-Tchoupou G, Joly C, Martinon F, Le Grand R. Optimize prime/boost vaccine strategies: trained immunity as a new player in the game. Front Immunol. 2021;12:612747. doi:10.3389/fimmu.2021.612747.
  • Wang B, Pei J, Xu S, Liu J, Yu J. Recent advances in mRNA cancer vaccines: meeting challenges and embracing opportunities. Front Immunol. 2023;14:1246682. doi:10.3389/fimmu.2023.1246682.
  • Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, Zhang W, Luoma A, Giobbie-Hurder A, Peter L, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547(7662):217–21. doi:10.1038/nature22991.
  • Keskin DB, Anandappa AJ, Sun J, Tirosh I, Mathewson ND, Li S, Oliveira G, Giobbie-Hurder A, Felt K, Gjini E, et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 2019;565(7738):234–9. doi:10.1038/s41586-018-0792-9.
  • Tang M, Cai J-H, Diao H-Y, Guo W-M, Yang X, Xing S. The progress of peptide vaccine clinical trials in gynecologic oncology. Hum Vaccin Immunother. 2022;18:2062982. doi:10.1080/21645515.2022.2062982.
  • Liu W, Tang H, Li L, Wang X, Yu Z, Li J. Peptide-based therapeutic cancer vaccine: Current trends in clinical application. Cell Prolif. 2021;54(5):e13025. doi:10.1111/cpr.13025.
  • Mizukoshi E, Nakagawa H, Tamai T, Kitahara M, Fushimi K, Nio K, Terashima T, Iida N, Arai K, Yamashita T, et al. Peptide vaccine-treated, long-term surviving cancer patients harbor self-renewing tumor-specific CD8+ T cells. Nat Commun. 2022;13(1):3123. doi:10.1038/s41467-022-30861-z.
  • Nelde A, Rammensee H-G, Walz JS. The peptide vaccine of the future. Molecular & Cellular Proteomics: MCP. 2021;20:100022. doi:10.1074/mcp.R120.002309.
  • Mohsen MO, Bachmann MF. Virus-like particle vaccinology, from bench to bedside. Cell Mol Immunol. 2022;19(9):993–1011. doi:10.1038/s41423-022-00897-8.
  • McFall-Boegeman H, Huang X. Mechanisms of cellular and humoral immunity through the lens of VLP-based vaccines. Expert Rev Vaccines. 2022;21(4):453–69. doi:10.1080/14760584.2022.2029415.
  • Ruzzi F, Semprini MS, Scalambra L, Palladini A, Angelicola S, Cappello C, Pittino OM, Nanni P, Lollini P-L. Virus-like particle (VLP) vaccines for cancer immunotherapy. Int J Mol Sci. 2023;24:12963. doi:10.3390/ijms241612963.
  • Bijker MS, van den Eeden SJF, Franken KL, Melief CJM, van der Burg SH, Offringa R. Superior induction of anti-tumor CTL immunity by extended peptide vaccines involves prolonged, DC-focused antigen presentation. Eur J Immunol. 2008;38:1033–42. doi:10.1002/eji.200737995.
  • Dailey GP, Crosby EJ, Hartman ZC. Cancer vaccine strategies using self-replicating RNA viral platforms. Cancer Gene Ther. 2023;30(6):794–802. doi:10.1038/s41417-022-00499-6.
  • Melief CJM, van Hall T, Arens R, Ossendorp F, van der Burg SH. Therapeutic cancer vaccines. J Clin Invest. 2015;125(9):3401–12. doi:10.1172/JCI80009.
  • Riccardo F, Bolli E, Macagno M, Arigoni M, Cavallo F, Quaglino E. Chimeric DNA vaccines: an effective way to overcome immune tolerance. Curr Top Microbiol Immunol. 2017;405:99–122.
  • Pandya A, Shah Y, Kothari N, Postwala H, Shah A, Parekh P, Chorawala MR. The future of cancer immunotherapy: DNA vaccines leading the way. Med Oncol. 2023;40:200. doi:10.1007/s12032-023-02060-3.
  • Bernard M-C, Bazin E, Petiot N, Lemdani K, Commandeur S, Verdelet C, Margot S, Perkov V, Ripoll M, Garinot M, et al. The impact of nucleoside base modification in mRNA vaccine is influenced by the chemistry of its lipid nanoparticle delivery system. Mol Ther Nucleic Acids. 2023;32:794–806. doi:10.1016/j.omtn.2023.05.004.
  • Alameh M-G, Tombácz I, Bettini E, Lederer K, Sittplangkoon C, Wilmore JR, Wilmore JR, Gaudette BT, Soliman OY, Pine M, et al. Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses. Immunity. 2021;54:2877–92.e7. doi:10.1016/j.immuni.2021.11.001.
  • Khoury DS, Cromer D, Reynaldi A, Schlub TE, Wheatley AK, Juno JA, Subbarao K, Kent SJ, Triccas JA, Davenport MP, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021;27(7):1205–11. doi:10.1038/s41591-021-01377-8.
  • Nitika, Wei J, Hui A-M. The delivery of mRNA vaccines for therapeutics. Life. 2022;12. doi:10.3390/life12081254.
  • Aguinam ET, Nadesalingam A, Chan A, Smith P, Paloniemi M, Cantoni D, Gronlund J, Gronlund H, Carnell GW, Castillo-Olivares J, et al. Differential T-cell and antibody responses induced by mRNA versus adenoviral vectored COVID-19 vaccines in patients with immunodeficiencies. J Allergy Clin Immunol Glob. 2023;2:100091. doi:10.1016/j.jacig.2023.100091.
  • Weide B, Carralot J-P, Reese A, Scheel B, Eigentler TK, Hoerr I, Rammensee H-G, Garbe C, Pascolo S. Results of the first phase I/II clinical vaccination trial with direct injection of mRNA. J Immunother (1991). 2008;31:180–8. doi:10.1097/CJI.0b013e31815ce501.
  • Lorentzen CL, Haanen JB, Met Ö, Svane IM. Clinical advances and ongoing trials on mRNA vaccines for cancer treatment. Lancet Oncol. 2022;23:e450–8. doi:10.1016/S1470-2045(22)00372-2.
  • Kim H, Liew D, Goodall S. Cost-effectiveness and financial risks associated with immune checkpoint inhibitor therapy. Br J Clin Pharmacol. 2020;86(9):1703–10. doi:10.1111/bcp.14337.
  • McCann N, O’Connor D, Lambe T, Pollard AJ. Viral vector vaccines. Curr Opin Immunol. 2022;77:102210. doi:10.1016/j.coi.2022.102210.
  • Larocca C, Schlom J. Viral vector-based therapeutic cancer vaccines. Cancer J. 2011;17(5):359–71. doi:10.1097/PPO.0b013e3182325e63.
  • Travieso T, Li J, Mahesh S, Mello JDFRE, Blasi M. The use of viral vectors in vaccine development. NPJ Vaccines. 2022;7:75. doi:10.1038/s41541-022-00503-y.
  • Ahmadi SE, Soleymani M, Shahriyary F, Amirzargar MR, Ofoghi M, Fattahi MD, Safa M. Viral vectors and extracellular vesicles: innate delivery systems utilized in CRISPR/Cas-mediated cancer therapy. Cancer Gene Ther. 2023;30:936–54. doi:10.1038/s41417-023-00597-z.
  • Yarnoff B, Bodhaine S, Cohen E, Buck PO. Time and cost of administering COVID-19 mRNA vaccines in the United States. Hum Vaccin Immunother. 2021;17:3871–5. doi:10.1080/21645515.2021.1974289.
  • Ruiz FJ, Torres-Rueda S, Pearson CAB, Bergren E, Okeke C, Procter SR, Madriz-Montero A, Jit M, Vassall A, Uzochukwu BSC, et al. What, how and who: cost-effectiveness analyses of COVID-19 vaccination to inform key policies in Nigeria. PloS Glob Public Health. 2023;3(3):e0001693. doi:10.1371/journal.pgph.0001693.
  • Light DW, Lexchin J. The costs of coronavirus vaccines and their pricing. J R Soc Med. 2021;114(11):502–4. doi:10.1177/01410768211053006.
  • Cancer. Barinthus biotherapeutics 2023. [accessed 2024 Jan 3]. https://www.barinthusbio.com/pipeline/cancer/.
  • Fakih M, Le DT, Pedersen KS, Shields AF, Shah MA, Mukherjee S, Delaite P, Faivre T, D’Alise AM, Leoni G, et al. First clinical and immunogenicity results including all subjects enrolled in a phase I study of nous-209, an off-the-shelf immunotherapy, with pembrolizumab, for the treatment of tumors with a deficiency in mismatch repair/microsatellite instability (dMMR/MSI). J Clin Oncol. 2022;40:2515. doi:10.1200/JCO.22.00506.
  • Palmer CD, Rappaport AR, Davis MJ, Hart MG, Scallan CD, Hong S-J, Gitlin L, Kraemer LD, Kounlavouth S, Yang A, et al. Individualized, heterologous chimpanzee adenovirus and self-amplifying mRNA neoantigen vaccine for advanced metastatic solid tumors: phase 1 trial interim results. Nat Med. 2022;28(8):1619–29. doi:10.1038/s41591-022-01937-6.
  • Amanna IJ, Carlson NE, Slifka MK. Duration of humoral immunity to common viral and vaccine antigens. N Engl J Med. 2007;357(19):1903–15. doi:10.1056/NEJMoa066092.
  • Bolinger B, Sims S, O’Hara G, de Lara C, Tchilian E, Firner S, Engeler D, Ludewig B, Klenerman P. A new model for CD8+ T cell memory inflation based upon a recombinant adenoviral vector. J Immunol. 2013;190:4162–74. doi:10.4049/jimmunol.1202665.
  • Provine NM, Klenerman P. Adenovirus vector and mRNA vaccines: mechanisms regulating their immunogenicity. Eur J Immunol. 2023;53(6):e2250022. doi:10.1002/eji.202250022.
  • Sutton N, San Francisco Ramos A, Beales E, Smith D, Ikram S, Galiza E, Hsia Y, Heath PT. Comparing reactogenicity of COVID-19 vaccines: a systematic review and meta-analysis. Expert Rev Vaccines. 2022;21:1301–18. doi:10.1080/14760584.2022.2098719.
  • Ayati N, Evans S, Zakavi SR, Gruenewald SM. Comparison between viral vector and mRNA based COVID-19 vaccination in prevalence and severity of regional immune reactions, and 18F-FDG PET/CT features. Asia Ocean J Nucl Med Biol. 2023;11:4–12.
  • Li J-X, Hou L-H, Meng F-Y, Wu S-P, Hu Y-M, Liang Q, Chu K, Zhang Z, Xu J-J, Tang R, et al. Immunity duration of a recombinant adenovirus type-5 vector-based Ebola vaccine and a homologous prime-boost immunisation in healthy adults in China: final report of a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Glob Health. 2017;5:e324–34. doi:10.1016/S2214-109X(16)30367-9.
  • Zhu F, Jin P, Zhu T, Wang W, Ye H, Pan H, Hou L, Li J, Wang X, Wu S, et al. Safety and immunogenicity of a recombinant adenovirus type-5–vectored coronavirus disease 2019 (COVID-19) vaccine with a homologous prime-boost regimen in healthy participants aged ≥6 years: a randomized, double-blind, placebo-controlled, phase 2b trial. Clin Infect Dis. 2022;75:e783–91. doi:10.1093/cid/ciab845.
  • Franke A-C, Hardet R, Prager L, Bentler M, Demeules M, John-Neek P, Jäschke NM, Ha TC, Hacker UT, Adriouch S, et al. Capsid-modified adeno-associated virus vectors as novel vaccine platform for cancer immunotherapy. Mol Ther Methods Clin Dev. 2023;29:238–53. doi:10.1016/j.omtm.2023.03.010.
  • Costa Clemens SA, Weckx L, Clemens R, Almeida Mendes AV, Ramos Souza A, Silveira MBV, da Guarda SNF, de Nobrega MM, de Moraes Pinto MI, Gonzalez IGS, et al. Heterologous versus homologous COVID-19 booster vaccination in previous recipients of two doses of CoronaVac COVID-19 vaccine in Brazil (RHH-001): a phase 4, non-inferiority, single blind, randomised study. Lancet. 2022;399(10324):521–9. doi:10.1016/S0140-6736(22)00094-0.
  • Ramirez-Valdez RA, Baharom F, Khalilnezhad A, Fussell SC, Hermans DJ, Schrager AM, Tobin KKS, Lynn GM, Khalilnezhad S, Ginhoux F, et al. Intravenous heterologous prime-boost vaccination activates innate and adaptive immunity to promote tumor regression. Cell Rep. 2023;42(6):112599. doi:10.1016/j.celrep.2023.112599.
  • Seclì L, Leoni G, Ruzza V, Siani L, Cotugno G, Scarselli E, D’Alise AM. Personalized cancer vaccines go viral: viral vectors in the era of personalized immunotherapy of cancer. IJMS. 2023;24:16591. doi:10.3390/ijms242316591.
  • Hookipa announces positive phase 1 data and phase 2 plans for HB-200 program for the treatment of advanced head and neck cancers at ASCO. Hookipa Pharma Inc n.d. [accessed 2024 Jan 3]. https://ir.hookipapharma.com/news-releases/news-release-details/hookipa-announces-positive-phase-1-data-and-phase-2-plans-hb-200.
  • Morris VK, Kopetz S. Don’t blame the messenger: lessons learned for cancer mRNA vaccines during the COVID-19 pandemic. Nat Rev Cancer. 2022;22(6):317–8. doi:10.1038/s41568-022-00463-4.
  • Yang C-Y, Lin Y-T, Lin L-J, Chang Y-H, Chen H-Y, Wang Y-P, Shih J-Y, Yu C-J, Yang P-C. Stage shift improves lung cancer survival: real-world evidence. J Thorac Oncol. 2023;18:47–56. doi:10.1016/j.jtho.2022.09.005.
  • Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33. doi:10.3322/caac.21708.
  • Furukawa Y, Hamano Y, Shirane S, Kinoshita S, Azusawa Y, Ando J, Nakauchi H, Ando M. Advances in allogeneic cancer cell therapy and future perspectives on “off-the-shelf” T cell therapy using iPSC technology and gene editing. Cells. 2022;11:11. doi:10.3390/cells11020269.
  • Topalian SL, Forde PM, Emens LA, Yarchoan M, Smith KN, Pardoll DM. Neoadjuvant immune checkpoint blockade: a window of opportunity to advance cancer immunotherapy. Cancer Cell. 2023;41(9):1551–66. doi:10.1016/j.ccell.2023.07.011.
  • Pilla L, Ferrone S, Maccalli C. Methods for improving the immunogenicity and efficacy of cancer vaccines. Expert Opin Biol Ther. 2018;18(7):765–84. doi:10.1080/14712598.2018.1485649.
  • Zhao J, Chen Y, Ding Z-Y, Liu J-Y. Safety and efficacy of therapeutic cancer vaccines alone or in combination with immune checkpoint inhibitors in cancer treatment. Front Pharmacol. 2019;10:1184. doi:10.3389/fphar.2019.01184.
  • Marofi F, Motavalli R, Safonov VA, Thangavelu L, Yumashev AV, Alexander M, Shomali N, Chartrand MS, Pathak Y, Jarahian M, et al. CAR T cells in solid tumors: challenges and opportunities. Stem Cell Res Ther. 2021;12(1):81. doi:10.1186/s13287-020-02128-1.
  • Bonaventura P, Shekarian T, Alcazer V, Valladeau-Guilemond J, Valsesia-Wittmann S, Amigorena S. Cold tumors: a therapeutic challenge for immunotherapy. Front Immunol. 2019;10:168. doi:10.3389/fimmu.2019.00168.
  • Yan T, Zhu L, Chen J. Current advances and challenges in CAR T-Cell therapy for solid tumors: tumor-associated antigens and the tumor microenvironment. Exp Hematol Oncol. 2023;12(1):14. doi:10.1186/s40164-023-00373-7.
  • Yamey G, Garcia P, Hassan F, Mao W, McDade KK, Pai M, Saha S, Schellekens P, Taylor A, Udayakumar K, et al. It is not too late to achieve global COVID-19 vaccine equity. BMJ. 2022;376:e070650. doi:10.1136/bmj-2022-070650.