Cancer Vaccines
Cancer vaccines represent a promising frontier in oncology, leveraging the body’s immune system to target and eradicate cancer cells [
113]. These vaccines can be designed to elicit an immune response specifically against tumor-associated antigens, providing a personalized approach to cancer treatment that avoids the complications of traditional chemo and radiotherapy. Various strategies have been employed in developing cancer vaccines, primarily focusing on three main approaches: DNA-based, mRNA-based, and peptide-based vaccines [
114,
115]. DNA vaccines utilize plasmid DNA to encode tumor antigens, inducing an immune response. mRNA vaccines, like those developed for COVID-19, involve the delivery of messenger RNA encoding cancer antigens, prompting the body to produce and present these antigens to the immune system. Peptide vaccines, conversely, directly introduce tumor-specific peptides to stimulate an immune response. Each approach offers unique advantages and challenges, which will be discussed in detail.
mRNA vaccines are emerging as prominent candidates for precision treatment of PDAC [
116,
117,
118,
119,
120,
121]. mRNA vaccines generate robust anti-tumor responses that engage innate and adaptive immune systems. Initially, the innate immune system recognizes foreign mRNA via pattern recognition receptors on antigen-presenting cells like dendritic cells [
122]. This detection, in turn, triggers a cascade of pro-inflammatory signaling pathways that enhance innate immune function. mRNA vaccines can also stimulate adaptive immunity by facilitating the processing of non-self mRNA-encoded proteins into peptides, which then present on MHC-I and are transported to the cell surface, where they activate CD8+ T cells.
Additionally, these neo-antigens can be directed through the Golgi bodies to endosomes to engage in the MHC-II presentation pathway to activate CD4+ T cells. mRNA vaccine response is amplified by the upregulation of co-stimulatory molecules (such as CD40 and CD86) on antigen-presenting cells, which enhances antigen presentation and T-cell activation. Activated antigen-presenting cells, including macrophages and dendritic cells, also present antigens to B cells, initiating an antibody response. Recent clinical trials involving an mRNA-based vaccine for pancreatic cancer have shown promising results, particularly in a study at Memorial Sloan Kettering Cancer Center [
123]. The vaccine, known as autogene cevumeran (RO7198457), includes an individualized mRNA neoantigen vaccine containing up to 20 neoantigens identified in each patient’s tumor. It led to durable and functional T-cell responses in patients with resectable pancreatic cancer and was associated with a reduced risk of disease recurrence. In this phase I trial [NCT04161755], 16 patients received R07198457 combined with the checkpoint inhibitor atezolizumab and a chemotherapy regimen. Half the participants developed robust immune responses against one or more tumor neoantigens. Importantly, these T cells were long-lasting and maintained their ability to respond to neoantigens for up to three years post-vaccination. Such findings underscore the potential of a vaccine to induce a robust immune response and contribute to delayed disease recurrence in pancreatic cancer.
Safety is paramount in precision therapy, and mRNA vaccines exhibit several key safety advantages over other vaccine platforms. For example, the production and delivery of mRNA vaccines does not involve toxic chemicals, reducing potential harm to manufacturing personnel and patients [
124,
125]. Additionally, mRNA vaccine production mitigates the risk of contamination with adventitious viruses that can be introduced during the culture of host cells, a concern associated with other vaccine platforms like viral vectors, inactivated viruses, live viruses, and subunit protein vaccines. The rapid manufacture of mRNA vaccines also reduces the window of opportunity for contaminating microorganisms during production. Unlike other therapeutic modalities, mRNA cannot integrate into the host genome. Moreover, the adjustable half-life of mRNA allows for precise control over the duration and intensity of protein expression. This approach enhances safety by allowing modulation of immune responses and potential side effects.
Nonetheless, some challenges need to be addressed with mRNA vaccines. The inherent properties of naked mRNA, such as its size, degradability, and charge, can impede efficient cellular uptake and cytoplasmic entry, except in cases like immature dendritic cells that can efficiently internalize mRNA via the macropinocytosis pathway [
126]. To enhance the effective delivery of mRNA into antigen-presenting cells, appropriate mRNA formulations (e.g., liposomes, polyplexes, polysomes, and lipoplexes) and administration routes must be judiciously selected and optimized. Once successful mRNA delivery is achieved, the
in vivo half-life of transcribed mRNA requires careful regulation, as various factors influence the pharmacodynamic and pharmacokinetic properties of mRNA-based therapeutics. Structural improvements to mRNA, such as optimizing poly(A) tails, 5' cap structures, and untranslated regions, are vital for enhancing mRNA stability and overall durability. In addition to delivery and stability considerations, immunogenicity must be a focal point in mRNA vaccine design. Emerging evidence suggests a complex interplay between mRNA and its associated immune response. For example, exogenous RNA stimulates the production of type I interferon through innate immunity pathways, but excessive production can promote the degradation of both ribosomal RNA and cellular mRNA [
127]. Strategies to mitigate immunogenicity include sequence optimization and post-transcriptional purification, which can reduce innate immune responses while preserving mRNA translation [
128].
Furthermore, enhancing the immunostimulatory properties of mRNA by incorporating adjuvants, such as TriMix (mRNA encoding CD70, CD40L, and TLR4), can augment the potency of cancer mRNA vaccines [
129]. TriMix, for instance, enhances the immunogenicity of unmodified naked mRNA, facilitating the cytotoxicity of T lymphocytes and the maturation of dendritic cells [
130]. These advancements in mRNA vaccine construction are essential for improving their efficacy in treating pancreatic cancer. Furthermore, targeting KRAS mutations, one of the most commonly present mutations in PDAC, through immunotherapies has been especially challenging. In a trial [NCT03948763] utilizing mRNA-5671 (V941), which is a tetravalent vaccine targeting KRAS G12D, G12V, G13D, and G12C (Moderna Inc.) was eventually discontinued in 2022 as it did not meet efficacy endpoints and, consequently, no further progress or updates on V941 have been announced. This issue underscores the necessity for improved vaccine platforms and combinatorial therapies that can levy the immunological response provoked to infiltrate and target antigen-expressing cancer cells.
As mentioned in the CheckPAC trial, some patients experienced improved clinical response rates associated with lowered TGF-β levels, which led to the initiation of the CheckVAC trial [NCT05721846]. CheckVAC is currently exploring the combination of a TGFβ-15 peptide vaccine with nivolumab and ipilimumab treatment. TGFβ-15 is a formulated peptide vaccine containing a TGFβ-derived peptide alongside Montanide ISA-51 as an adjuvant [
131]. Upon administration, TGFβ-15 aims to restore and enhance an immunological anti-tumor response by stimulating the host immune system to mount a cytotoxic T-lymphocyte response against TGFβ-expressing immunosuppressive cells in the TME, including TAMs, MDSCs, DCs, Tregs, and CAFs.
Another cancer vaccine that is currently being tested is the combination of TG01 Vaccine / QS-21 Stimulon with or without immune checkpoint inhibitor balstilimab as maintenance therapy following adjuvant chemotherapy in patients with resected pancreatic cancer (TESLA) trial [NCT05638698]. TG01 is an experimental vaccine designed to provoke an immune response against cancer cells by targeting the seven most prevalent codon 12 and 13 oncogenic mutations in KRAS with synthetic RAS peptides. QS-21, derived from the soap bark tree, is a vaccine adjuvant known for stimulating both humoral and cell-mediated immunity, further boosting the immune response induced by TG01.
A new cancer vaccine clinical trial [NCT05964361] focuses on enhancing the body's immune response by targeting the Wilms tumor 1 (WT1) protein. WT1 is a transcription factor that plays a crucial role in both normal development and tumorigenesis [
132]. WT1 is highly overexpressed in various malignancies, including pancreatic cancer [
133]. WT1 supports tumor progression by promoting cell proliferation, inhibiting apoptosis, and enhancing angiogenesis. This trial investigates the feasibility of developing vaccines specifically targeting the Wilms' Tumor-1 (WT1) antigen alongside a novel IL15-trans presentation mechanism on their cell surface. IL15, known for its role in supporting natural killer cell function, promoting T cell memory formation, and enhancing immune response, is expected to enhance the immunogenicity of dendritic cells towards WT1-expressing cancer cells [
134,
135].
Another clinical trial [NCT05846516] evaluates the experimental immunotherapy KISIMA-02 for pancreatic cancer patients. The KISIMA platform is a single therapeutic vaccine that is comprised of three components: a cell-penetrating peptide (CPP) for transportation of the vaccine contents across the cell membrane, antigens that can be tailored to each indication, and a TLR peptide agonist that acts as an adjuvant [
136]. KISIMA-02 is a combination treatment for patients with KRAS G12D/G12V mutated PDAC. This regimen includes the KISIMA therapeutic protein vaccine (ATP150 or ATP152), an oncolytic viral vector (VSV-GP154), and an immune checkpoint inhibitor. This study is designed to assess the safety and tolerability of the KISIMA-02 regimen before examining its impact on delaying tumor recurrence. Participants will receive a therapeutic protein vaccine (either ATP150 or ATP152), a viral vector VSV-GP154, and an immune checkpoint inhibitor, ezabenlimab, or be in an observational group. The ATP-150 vaccine, specifically developed for KRAS G12D and G12V mutated PDAC, employs a heterologous prime-boost approach, integrating both protein and viral vector components. This vaccine is administered parenterally and is part of the KISIMA immunization platform, designed to enhance immune response through self-adjuvant mechanisms.
The pancreatic GVAX platform is another example of a novel strategy that has seen success in other cancers, with the pancreatic version being a vaccine composed of genetically modified pancreatic cancer cells [
137,
138]. These engineered pancreatic cells secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) to facilitate a strong dendritic cell-dependent immunological response. The GVAX induces the formation of tertiary lymphoid structures within the tumor, providing a localized site for immune cell activation for an effective immune response. In certain trial arms, GVAX is combined with immune checkpoint inhibitors like nivolumab and ipilimumab. Combining GVAX with these checkpoint inhibitors aims to prime the immune system against the tumor and prevent immune escape. GVAX is also commonly administered with low-dose cyclophosphamide, which selectively depletes Tregs that suppress immune responses, thus enhancing the vaccine's effectiveness [
139]. A three-arm, phase 2 clinical trial [NCT02451982] is currently underway, aiming to determine the effect of co-treatment of GVAX with CY on patient outcomes. This trial aims to compare the DFS and OS of patients in three treatment arms: Arm A (GVAX alone), Arm B (GVAX and then low-dose CY), and Arm C (low-dose CY and then GVAX). The study evaluates the effect of the sequencing and combination of GVAX and low-dose CY on patient outcomes. By comparing these three treatment strategies, this trial aims to determine whether the addition of low-dose CY before or after GVAX could enhance or impair the efficacy of the immunotherapy. Another GVAX-related phase 2 clinical trial [NCT03190265] is focused on the investigation of ORR and AEs with combination therapy of CRS-207, nivolumab, and ipilimumab, with or without the GVAX (and CY)). CRS-207 is a live-attenuated, double-deleted Listeria monocytogenes strain engineered to express the pancreatic tumor-associated antigen mesothelin [
140,
141]. By infecting antigen-presenting cells, CRS-207 helps break immune tolerance by inducing a potent innate and adaptive immune response, including activating T cells specific to mesothelin. Overall, GVAX provides a broad array of tumor antigens in the context of GM-CSF. At the same time, CRS-207 focuses on a specific antigen, mesothelin, enhancing the overall presentation of tumor antigens to the immune system. This combination can lead to a more robust activation and expansion of tumor-specific T cells, as both therapies independently activate and mature dendritic cells. CRS-207’s ability to break immune tolerance complements GVAX’s ability to stimulate an immune response, which may lead to a more effective and sustained anti-tumor immune response. Targeting multiple tumor-associated antigens between GVAX and CRS-207 would also reduce the likelihood of immune escape by tumor cells.