key: cord-0730100-8mx2b5v0 authors: Lee, Sun Min; Kim, Paul; You, Jinsuh; Kim, Eui Ho title: Role of Damage-Associated Molecular Pattern/Cell Death Pathways in Vaccine-Induced Immunity date: 2021-11-23 journal: Viruses DOI: 10.3390/v13122340 sha: 8b847a6443597da367ca0893c767c9e9f1657610 doc_id: 730100 cord_uid: 8mx2b5v0 Immune responses induced by natural infection and vaccination are known to be initiated by the recognition of microbial patterns by cognate receptors, since microbes and most vaccine components contain pathogen-associated molecular patterns. Recent discoveries on the roles of damage-associated molecular patterns (DAMPs) and cell death in immunogenicity have improved our understanding of the mechanism underlying vaccine-induced immunity. DAMPs are usually immunologically inert, but can transform into alarming signals to activate the resting immune system in response to pathogenic infection, cellular stress and death, or tissue damage. The activation of DAMPs and cell death pathways can trigger local inflammation, occasionally mediating adaptive immunity, including antibody- and cell-mediated immune responses. Emerging evidence indicates that the components of vaccines and adjuvants induce immunogenicity via the stimulation of DAMP/cell death pathways. Furthermore, strategies for targeting this pathway to enhance immunogenicity are being investigated actively. In this review, we describe various DAMPs and focus on the roles of DAMP/cell death pathways in the context of vaccines for infectious diseases and cancer. Through the course of history, fatal viral or bacterial infections have posed a threat to human health, and vaccines against these pathogens have been continuously developed. For example, during the Spanish flu, the first pandemic caused by the influenza virus, more than 20 million cases of mortality were reported among 500 million infected individuals from 1918 to 1920. After the first isolation of human influenza virus in 1933 [1] , a live attenuated influenza vaccine (LAIV) adapted to rodents, which would cause mild symptoms in humans, was developed. However, LAIV, which was almost identical to the live virus, exhibited safety and stability issues, thereby necessitating the introduction of an inactivated influenza vaccine (IIV). Influenza virus inactivated by high temperatures or chemical agents such as formaldehyde can effectively induce antibody production [2] . In the 1960s, split and subunit vaccines, which were new inactivated formulations, were tested in children under more stringent safety criteria [3] . These types of vaccines primarily contain the viral surface antigens such as hemagglutinin and neuraminidase instead of whole viruses, as the former can induce antibody responses. Although the action of these vaccines was actively investigated in the 1970s and 1980s, they were less immunogenic than LAIV and the previous version of IIV. It has been acknowledged that attempts to reduce adverse reactions and risks may have also reduced the efficacy of different vaccines [4] . Based on these findings, in the 1990s an adjuvant was used in FLUAD, an inactivated influenza vaccine for the elderly in order to enhance vaccine efficacy. In fact, various adjuvants have been applied to vaccines, commencing with the use of aluminum-based adjuvants in the 1930s, to modulate the efficacy and safety of vaccines. Aluminum salts, the most commonly used adjuvants, have been widely used for decades in vaccines against DAMPs are molecules generated either through active secretion from live cells or through passive release following cell death, such as apoptosis, and accidental or regulated necrosis, such as pyroptosis, necroptosis, and ferroptosis [13] . The types of DAMPs differ depending on the cell death type and stage, and each DAMP has been shown to perform distinct functions and exhibit different modes of action. DNA is composed of nucleotides (structured with a phosphate group, a deoxyribose sugar, and a single nitrogen-containing base) and carries the genetic code of organisms in the cell nucleus, governing biological functions. In addition to its basic function as a source of genetic information, when it is present outside cells, nuclear or mitochondrial DNA can stimulate the immune system via diverse cell death pathways or exosome/ectosome release [13] [14] [15] . Nucleus-derived extracellular DNA is generally inactive due to the fact that the DNA sensors in cells primarily recognize CpG motifs that are suppressed in mammalian nuclear DNA but expressed in bacterial DNA [16] . However, extracellular DNA can acquire immunostimulatory properties by forming complexes with other DNA-binding proteins [15] . For example, HMGB1 enables DNA to enter cells to interact with the endosomal DNA sensor TLR9. In contrast, owing to the high content of unmethylated CpG motifs, mitochondrial DNA exhibits activity independently, stimulating PRRs [17] . The majority of DNA sensing occurs through TLR9, absent in melanoma 2 (AIM2), interferon-inducible protein 16 (IFI16), cyclic GMP-AMP synthase (cGAS), and stimulator of interferon genes (STING). When these receptors recognize perturbations in extracellular DNA homeostasis, they trigger innate immune responses. TLR9 signaling stimulates NF-κB and interferon regulatory factor (IRF) pathways [18, 19] . DNA recognition by AIM2 or IFI16 activates inflammasomes to induce the production of pro-inflammatory cytokines (IL-1β and IL-18) [20, 21] . In addition, the cytosolic DNA sensing pathway mediated by cGAS-STING induces the activation of the NF-κB pathway, leading to IL-6 and tumor necrosis factor (TNF) production [22] . The activation of NF-κB pathway also mediate the anti-apoptotic signaling in DCs by inhibiting expression of a pro-apoptotic gene, Bim [23] . Uric acid is the final product of purine metabolism. Owing to its pro-inflammatory properties, uric acid has been implicated in kidney disease [24] , Alzheimer's disease [25] , and gout [26, 27] . Several recent studies have demonstrated the characteristics of uric acid as a DAMP [28] . Uric acid can be generated during massive cell damage or cell death as well as from the breakdown of exogenous purines. While uric acid is soluble inside cells, excess uric acid in serum may undergo saturation and form monosodium urate (MSU) crystals in the extracellular matrix. In humans, only a high concentration of the crystallized form of uric acid exhibits pro-inflammatory properties; the soluble form (sUA) shows no significant pro-or anti-inflammatory activity [29] . This phenomenon is consistent with the phenomena reported in rhesus monkeys or great apes [30, 31] . In rodents, both forms stimulate proinflammatory responses [31] . Braga et al. suggested that the species-dependent difference may be due to the evolutionary deletion of the Naip1-Nlrp3 inflammasome platform, which is involved in the murine recognition of sUA. With respect to the mechanism underlying inflammatory effects mediated by uric acid crystals, it is known that uric acid crystals activate NLRP3 inflammasomes with the consequent release of IL-1β and IL-18, which may induce neutrophil influx [27] . Kono et al. demonstrated that uric acid derived from sterile cell death contributes to inflammatory responses in mice [28] . Uric acid is known to directly influence various immune cells, including macrophages and T and B lymphocytes, as shown in different studies on human peripheral blood mononuclear cells. Uric acid stimulates the pro-inflammatory activity of in vitro cultured monocyte-derived macrophages by increasing the production of inflammatory factors (TNF-α, TLR4, and CD11c) and promoting macrophage phagocytosis [32] . In addition, in isolated human T cells, urate crystals elevated the zeta chain phosphorylation of the T-cell receptor (TCR) complex, indicating the direct activation of the TCR complex [33] . Similarly, urate crystals trigger B cell receptor signal transduction and induce B cell proliferation in vitro [34] . Recently, it has been shown that high levels of uric acid increased IL-1β and IL-1Ra production in human peripheral blood mononuclear cells freshly isolated from healthy people, as examined in vitro [35] . Furthermore, whole blood DNA from hyperuricemic people had different methylation features in genes related to inflammatory cytokine signaling as compared to those from normouricemic people [35] . ATP, a nucleoside triphosphate, functions as the primary molecule for energy metabolism by hydrolyzing into ADP and phosphate in cells. However, when ATP molecules are present outside cells, they act as signaling molecules [36] . Typically, ATP is generated in the nervous system as a co-transmitter with neurotransmitters and plays particular roles in specific pathways associated with the five senses (sight, smell, taste, touch/pain, and sound), breathing, and cardiac and gastrointestinal functions. Extracellular ATP is present in the healthy tissues at negligible levels (nanomolar levels), but the level increases con- siderably in an inflammatory microenvironment [37, 38] . Under inflammatory conditions, pro-inflammatory stimuli, such as LPS, reactive oxygen species (ROS), and hypoxia lead to extracellular ATP generation. The chemical properties of ATP, including low molecular weight and negative charge, facilitate translocation [39] . The major release mechanisms include necrosis, apoptosis, and pyroptosis, and the release channels include lysosomes, exosomes, and channel pores [13] . Once generated, ATP activates purinergic P2X/P2Y receptor signaling, which affects the host defense against infectious microbes or the outcomes of inflammatory/infectious disorders [40] . For example, P2Y 2 R signaling mediates the clearance of mucus during inflammatory diseases and dermal wound healing [41] . In terms of cell death, extracellular ATP generated by apoptotic cells acts as a "find-me" signal via P2Y 2 R to prompt the clearance of apoptotic cells [42] . In addition, ATP-P2Y 2 R signaling contributes to migration and cytokine/DAMP production by neutrophils [43] or eosinophils [44] . The P2Y 6 R pathway is also important in innate immunity owing to its ability to trigger phagocytosis. However, it can induce chronic inflammatory diseases, such as inflammatory bowel diseases [45] and asthma [46] . P2Y 12 R signaling affects lung function in asthma in humans through the activation of dendritic cells (DCs) that promote allergic T cell activity [47] . ATP-induced P2X 7 R signaling contributes to the host defense mechanism against pathogenic infection by promoting inflammasome activation in macrophages [48] . However, the stimulation of DCs by ATP-P2X 7 R signaling primes CD8 + T cells, CD4 + T cells, or Th17 cells to promote chronic inflammation during dermatitis, asthma, psoriasis, and inflammatory bowel disease [42] . Under the inflammatory conditions, Th1 cells can also differentiate into Th17 cells which may be pathogenic in inflammatory bowel disease [49] . The high mobility group (HMG) is a group of nuclear proteins first isolated and characterized in 1973 [50, 51] and named based on their high mobility in gel electrophoresis. This protein group includes HMGA, HMGB, and HMGN, among which HMGB1 is the most abundant. HMGB1 functions as a DNA chaperone in cells [52] and functions as a DAMP outside cells. This protein can be released through active generation by activated immune cells (e.g., macrophages or monocytes) or through passive release during cell damage/death following infection or tissue injury [53] . HMGB1 is known to be released during pyroptosis [54] , ferroptosis [55] , and late stages of apoptosis [56] , among the different types of cell death. The excessive release of HMGB1 is critically associated with inflammation during sepsis or endotoxemia in infectious diseases and sterile injuries [53, 57] . HMGB1 binds to certain PAMPs and other pro-inflammatory molecules (e.g., LPS), and this enables it to interact with diverse cell surface receptors [53] . The HMGB1 complexes with cofactors operate by binding to a wide range of receptors, including RAGE, TLR4, TLR2 [58] , TLR5 [59] , and TIM3 [60] . Among these receptors, RAGE, TLR4, and TLR2 have been confirmed as the specific receptors of HMGB1 [57, 58, 61, 62] . A recent finding has shown that RAGE and TLR4 operate in a feedback interaction network, which is important in HMGB1-induced inflammation [61] . The study suggested that RAGE promotes TLR4 trafficking on the cell surface via MAPK activation, whereas TLR4 increases RAGE expression. The downstream signals of RAGE and TLR2/TLR4 induce pro-inflammatory cytokine production through the NF-κB pathway and in a MyD88-dependent manner [62] . Most modern adjuvants, in clinical use or in the late development stage, such as MPLA, CpG DNA, poly (I:C), and imiquimod, exert adjuvanticity through PRRs. Certain adjuvants, such as alum, emulsion, and saponins, indirectly activate innate immune responses by causing cell death at the site of administration and draining lymph nodes to subsequently generate DAMPs. Thus far, adjuvants, including alum, oil-in-emulsion (MF59 and AS03), HP-β-CD, and saponin-based adjuvants (e.g., QS21), have been shown to involve DAMPs in their mode of action. Generally, DAMP-inducing adjuvants promote the activation of innate immune cells [63, 64] . In this process, typical adjuvant-related DAMPs, including ATP, uric acid, and DNA, trigger the accumulation of neutrophils, monocytes, macrophages, and eosinophils [65] . Antigens are delivered to the draining lymph nodes via innate immune cells or passive draining to induce adaptive immune responses. Here, we review the role of DAMPs in the action of licensed vaccines or adjuvants currently in use or in phase 3 trials. Several viral vaccines have been shown to trigger DAMP release or cell death signaling in the absence of an adjuvant, implying that these viruses intrinsically exhibit a DAMPinducing property in addition to the PAMP-inducing property. When a UV-inactivated influenza virus vaccine is injected without an adjuvant, subcapsular sinus macrophage (SSM) death can be detected by microscopy. This type of macrophage death is a necrosislike death associated with MyD88 signaling and Toll-like receptor 7 recognition. This vaccination can also trigger the release of IL-1α, which is involved in the inflammatory pathway, suggesting the upregulation of antigen presentation and humoral response [66] . Additionally, modified vaccinia Ankara virus, a viral vector actively studied for vaccine development, has been reported to induce inflammasome activation and pyroptosis in SSMs [67] . This process subsequently promotes the robust recruitment of inflammatory cells and potentiates T cell responses. Among commercialized animal vaccines, Purevax ® was developed for protection against rabies, feline leukemia, and panleukopenia in cats. Although the specific mechanism and pathway have not been identified, a histopathological study confirmed the induction of necrosis with neutrophilic and macrophage inflammatory responses within seven days of vaccine injection in cats [68] . Alum has been shown to exert adjuvanticity through the depot effect (i.e., the slow release of antigens), and it is now accepted as a typical DAMP-inducing adjuvant ( Table 1 ). The injection of alum causes necrotic cell death at the injection site, inducing the release of DAMPs, such as host DNA [69] and uric acids. Alum-induced host DNA then triggers IgG1 production from B cells in an interferon response factor 3 (Irf3)-independent manner and stimulates Th2 cells and IgE responses through Irf3-dependent mechanisms [70] . In contrast to the common mechanism of action of several adjuvants that trigger TLR signaling transduction, alum salts are known to require NLRP3 inflammasome activation for the production of pro-inflammatory cytokines [71] . NLRP3 activation is well known to induce the production of active IL-1β and IL-18 through cleavage by caspase 1. In particular, the phagocytosis of alum, MSU, and silica crystals leads to lysosomal destabilization and rupture. Furthermore, lysosomal damage can activate NALP3 inflammasomes. [72] . In addition, the direct administration of alum to the airway stimulates immune responses in the lungs. Exposure to alum induces cell death in alveolar macrophages and stimulates the release of IL-1α as an alarmin [73] . Transient IL-1α release in the lungs may subsequently cause delayed and sustained adjuvant effects on the lungs by inducing the formation of inducible bronchus-associated lymphoid tissue, which is a lymphoid cluster showing local immune responses associated with asthma. Squalene-based emulsion adjuvants, such as MF59 and AS03, are widely used as influenza vaccine adjuvants due to their ability to induce strong immunogenic responses in humans. Clinical studies have demonstrated that these adjuvants induce robust innate immune responses required for adaptive immune system activation. Notably, ATP released from the injected muscle is known to be a crucial factor contributing to the adjuvanticity of MF59 (Table 1 ) [74] . In addition, the elevation of DNA and uric acid levels was observed in the serum of Addavax (AV)-immunized mice [8] . Upon injection, squalene-based oilin-water emulsion (SE) vaccine adjuvants stimulate an immunocompetent environment through the transient release of DAMP signals, including ATP, DNA, and uric acid. These signaling molecules bind to the receptors of antigen-presenting cells (APCs), inducing cytokine and chemokine secretion, enhanced antigen uptake, and transport to draining lymph nodes [63] . However, the mechanism of action of cell death pathways underlying immune system stimulation remains poorly understood. Findings from recent studies have suggested that the efficient phagocytosis of SE adjuvants is correlated with the loss of lymph node-resident macrophages, which induces early regulated necrosis, such as RIPK3-mediated necroptosis and pyroptosis, followed by late apoptosis, in draining lymph nodes. In particular, the role of RIPK3, a key mediator in necroptosis, in SE-adjuvanted vaccination, is crucial for CD8 + T cell responses. Therefore, the importance of the kinase activity suggests that necroptotic cell death in lymph node-resident macrophages is critical for the induction of CD8 + T cell responses and the recruitment of innate immune cells by SE adjuvants [8] . Numerous studies have highlighted the role of necroptosis in the exposure to DAMPs and the regulation of inflammatory responses [75] [76] [77] [78] . Indeed, MF59 and AV engender mixed macrophage deaths associated with a highly immuno-stimulatory microenvironment upon immunization, characterized by the increased release of cytokines, including IL-6, IL-12, and TNF, and elevation in the levels of DNA and uric acid, compared to that in alum-vaccinated groups [8] . Saponins are composed of hydrophilic glycosides and lipophilic triterpenes and hence, qualify as detergents. They can destroy cell membranes by forming complexes with membrane cholesterol. Among saponin-based adjuvants, QS-21 is a component of the adjuvant AS01, a licensed vaccine against varicella-zoster virus. It has also been used as an approved malaria vaccine in a limited campaign in Africa. In the draining of lymph nodes, QS-21 activates caspase-1 and MyD88 pathways in SSMs and DCs to induce IL-1b and IL-18 generation, thereby inducing enhanced antibody and T cell responses. The local release of HMGB1 has been suggested to contribute to this mechanism [79] . Matrix-M™, which comprises 40 nm nanoparticles containing saponins, cholesterol, and phospholipids, is a component of NVX-CoV2373, a Novavax COVID-19 vaccine candidate undergoing phase 3 clinical trial (Clinical trial number NCT04611802). The synthetic compound HP-β-CD is also a DAMP-inducing adjuvant (Table 1) . HPβ-CD can be used as a vaccine adjuvant when administered via subcutaneous [80] or intranasal routes [81] . After administration, HP-β-CD induces local cell death to release host DNA, which stimulates Th2 immune responses [82] . This adjuvant, along with PAMP adjuvants, enhances immunogenicity when administered with an influenza split vaccine [83] . In addition, the recently developed Janssen COVID-19 vaccine contains HPβ-CD as an adjuvant and was allowed for use under emergency use authorization by the US FDA. Recently, growing evidence has corroborated that the DAMP pathway can be strategically targeted to enhance the immunogenicity of vaccines and improve the outcomes of immunotherapy. In the application of DAMPs as adjuvants against viral infection, HSP70 was integrated with a DNA vaccine encoding an antigen for protection against HIV [84] . In draining lymph nodes of mice vaccinated with DNA encoding HIV gag and HSP70 proteins, the levels of activated DCs were significantly increased, followed by the enhancement of CD4 + and CD8 + T cell responses. The vaccine, containing HSP70, showed protective potential against HIV infection [84] . In addition, it has been suggested that HMGB1 prevents viral infections by improving protective effect of vaccines. Co-immunization with plasmid-encoded HMGB1 (pHMGB1) can enhance the vaccine efficacy for antigenencoding DNA. In HIV vaccine studies, HMGB1 expression promoted DC maturation with CCR7 upregulation and antigen presentation, followed by increased antibody and IFN-γ responses to HIV [85] . Furthermore, the HMGB1-encoding plasmid also demonstrated adjuvant-like effects in the DNA-based influenza vaccine approach. Similar to that observed in the HIV vaccination, vaccination with plasmids encoding influenza antigen and HMGB1 can induce DC maturation by upregulating CD83, CD86, and CCR7, and mounted stronger neutralizing antibody response antigen-specific CD4 + and CD8 + T cells. Therefore, HMGB1 can be targeted to enhance the immunogenicity of viral vaccines that provide protection against viral challenges [86] . In the context of tumor immunotherapy, DAMPs have been tested extensively as immuno-stimulators. ATP, one of the most typical intracellular metabolites and an extracellular danger signal released by damaged cells, has been suggested as an effective vaccine adjuvant. Upon the injection of Ova as a model antigen with ATP, DCs underwent effective activation with increased expression of surface markers such as CD80, CD86, and MHC II, followed by augmented levels of anti-Ova IgG [87] . Interestingly, ATP may sufficiently inhibit the growth of tumors when peptide antigens that are weakly immunogenic are used. ATP-adjuvanted vaccination could reduce the levels of regulatory T cells and myeloid-derived suppressor cells and increase CD4 + and CD8 + effector/memory T cells [88] . Furthermore, HSP70 in combination with DNA vaccines resulted in prolonged protective effect against tumors. In a tumor challenge test, mice immunized with the DNA vaccine, encoding HSP70 linked with HPV16 mE7 and the human CD33 peptide gene, remained tumor-free for approximately two months, in contrast to unimmunized mice, which developed tumors within two weeks. It could generate considerably higher IFN-γ-producing CD8 + T cells than that observed in the control group [89] . DAMPs can be triggered as a result of ICD pathways, and this type of cell death can also enhance vaccine efficacy through DAMP signaling pathways. ICD was initially discovered during chemotherapy and radiation therapy for cancer treatment, but it is induced in response to pathogenic infection and vaccination. ICD is a more functional term classifying various types of cell death. It indicates a form of cell death triggering not only inflammation but also adaptive immunity including antibody and T cell responses [90] . ICD has been reported to be induced during photo-based therapy as well as by oncolytic viruses and natural compounds [91] . ICD inducers can exert their anti-tumor vaccine effect by direct injection of ICD inducers or the injection of dying tumor cells (killed by the inducers) as immunogens, via DC maturation and release of immunomodulatory factors [92] . Typically, ICDs are induced via various mechanisms, such as direct ER stress (which causes ATP secretion and surface calreticulin exposure), ROS generation, and mitochondrial outer membrane permeabilization [91] . DAMPs released as a result of these reactions activate inflammatory pathways and promote phagocytosis [93] . Although it is easy to induce ICD using particular therapeutic compounds, not all chemotherapeutic agents can induce ICD. In fact, ICD-triggering agents are limited to doxorubicin, mitoxantrone, colchicine, and shikonin (although most chemotherapeutic agents cause immune suppression). These are known as "type I ICD inducers" [94] . Doxorubicin and mitoxantrone are categorized as anthracyclines and can induce ATP secretion and CD8 + T cell responses. Colchicine, a microtubule inhibitor, triggers the release of representative DAMP molecules, such as HSP70 and HMGB1. Similarly, shikonin induces HSP70 release and calreticulin exposure [95] . As a prototype cancer vaccine, tumor cell lysate treated with a type I ICD inducer can be utilized in order to achieve DC-mediated T cell proliferation and tumor suppression [96] . In case of cancer vaccination via photobased therapy, such as photodynamic and photothermal therapy ("type II ICD inducers"), it has been shown that ER stress, and subsequent calreticulin exposure and enhanced DC maturation, was linked to the increase of CD8 + T cell responses using CT-26 and B16 tumor mouse models [97] . Moreover, CD8 + T cells were efficiently infiltrated into tumors and became more functional while eliciting an enhanced anti-tumor effect [97] . Oncolytic viruses, another type II ICD inducer, have been suggested as therapeutic candidates that can kill tumor cells and disrupt the immunosuppressive tumor microenvironment. Coxsackievirus, adenovirus, measles virus, Semliki Forest virus, reovirus, vaccinia virus, Newcastle disease virus, and influenza virus have been reported to display oncolytic effects. Oncolytic viral vaccines can activate stronger immune responses than other types of cancer vaccines since they harbor PAMPs and also trigger ICD-DAMP pathways [91, 98] . In an experimental study utilizing vaccination of oncolytic virus-infected cancer cells, the level of ICD increased upon treatment with an oncolytic virus, and tumor growth was significantly decreased in mice [99] . During pathogenic infections, host cell receptors detect microbial patterns, such as LPS, flagellin, CpG DNA, and RNA, to induce danger signaling pathways [100] . In case of bacterial infection by Listeria monocytogenes, IFNα/β-induced cell death, such as STAT1dependent apoptosis or iNOS-dependent necrosis, was induced. When host cells are infected with HIV and HCV, the expression levels of TNF-related apoptosis-inducing ligand (TRAIL), death receptor 5 (DR5), and IFNα/β from DCs are upregulated to induce T cell and B cell apoptosis [101] . Viral infection can also increase autophagy via the elF2α kinase signaling pathway related to DAMP release [102] . Although it is clear that infectious diseases are associated with ICD, the ICD pathway has not been actively targeted for the development of vaccines against infectious diseases. As new pathogens, such as SARS-CoV-2 and variants of influenza virus, continue to emerge, the development of strategies that target ICD to induce strong immunogenicity may be necessary, alongside the introduction of novel vaccine platforms. Vaccines have advanced with technological progress, and the COVID-19 pandemic has necessitated the delivery of gene-based vaccines worldwide. Although there was a lack of comprehensive information regarding the mechanism of action of vaccines in the past, recent scientific advances have allowed us to answer this critical question. Recent discoveries have highlighted the critical roles of DAMP/cell death pathways in the induction of optimal immune response during pathogenic infections and vaccinations (Figure 1 ). The significance of DAMP/cell death pathways may indicate the potential for the development of the next generation of immunomodulatory agents for effective vaccines in the future. past, recent scientific advances have allowed us to answer this critical question. Recent discoveries have highlighted the critical roles of DAMP/cell death pathways in the induction of optimal immune response during pathogenic infections and vaccinations ( Figure 1 ). The significance of DAMP/cell death pathways may indicate the potential for the development of the next generation of immunomodulatory agents for effective vaccines in the future. Figure 1 . Schematic diagram depicting how DAMPs/cell death pathways affect immunogenicity during infection, vaccination, and anti-cancer therapy. Infection, vaccination, and anti-cancer therapies can induce cell death including apoptosis, necroptosis, pyroptosis, and ferroptosis. Certain types and stages of cell death are immunogenic cell death, releasing DAMPs (e.g., ATP, nucleic acid, uric acid, and HMGB1). The release of DAMPs may trigger antibody production or T cell responses by stimulating immune responses such as cytokine production, inflammation, and DC maturation. A virus obtained from influenza patients Antibody response of human beings following vaccination with influenza viruses History and evolution of influenza control through vaccination: From the first monovalent vaccine to universal vaccines Vaccines for pandemic influenza. The history of our current vaccines, their limitations and the require-ments to deal with a pandemic threat Vaccine adjuvants: Understanding the structure and mechanism of adjuvanticity Pathogen-associated molecular patterns on biomaterials: A paradigm for engineering new vaccines Emerging concepts in the science of vaccine adjuvants Squalene emulsion-based vaccine adjuvants stimulate CD8 T cell, but not antibody responses, through a RIPK3-dependent pathway Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity Pathogen Recognition and Inflammatory Signaling in Innate Immune Defenses Complexity of Danger: The Diverse Nature of Damage-associated Molecular Patterns Key Role of DAMP in Inflammation, Cancer, and Tissue Repair Release mechanisms of major DAMPs The Alarmin Properties of DNA and DNA-associated Nuclear Proteins The origin and properties of extracellular DNA: From PAMP to DAMP CpG Motifs in Bacterial DNA and Their Immune Effects Degraded Mitochondrial DNA is a Newly Identified Subtype of the Damage Associated Molecular Pattern (DAMP) Family and Possible Trigger of Neurodegeneration Mitochondrial DNA induces inflammation and increases TLR9/NF-kappaB expression in lung tissue Master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition re-ceptors The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses The interferon-inducible DNA-sensor protein IFI16: A key player in the antiviral response DNA sensing by the cGAS-STING pathway in health and disease Human IDO-competent, long-lived immunoregulatory dendritic cells induced by intracellular pathogen, and their fate in humanized mice Uric Acid as a Marker of Kidney Disease: Review of the Current Literature Role of Uric Acid in Alzheimer's Disease. J. Alzheimer's Dis uric acid metabolism, and crystal-induced inflammation Gout-associated uric acid crystals activate the NALP3 inflammasome Uric acid promotes an acute inflammatory response to sterile cell death in mice Crystallized but not soluble uric acid elicits pro-inflammatory response in short-term whole blood cultures from healthy men Sensing soluble uric acid by Naip1-Nlrp3 platform Uric Acid Has Direct Proinflammatory Effects on Human Macrophages by Increasing Proinflammatory Mediators and Bacterial Phagocytosis Probably via URAT1 Urate crystals directly activate the T-cell receptor complex and induce T-cell proliferation Urate crystals trigger B-cell receptor signal transduction and induce B-cell proliferation Additional file 1 of Urate-induced epigenetic modifications in myeloid cells The Double Life of ATP Extracellular ATP and P2 purinergic signalling in the tumour microenvironment Pharmacology of Adenosine Receptors: The State of the Art Purinergic signaling, DAMPs, and inflammation Nucleotide signalling during inflammation Nucleotide receptors as targets in the pharmacological enhancement of dermal wound healing Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance ATP Release Guides Neutrophil Chemotaxis via P2Y2 and A3 Receptors Activation of human eosinophils via P2 receptors: Novel findings and future perspectives P2Y(6) agonist uridine 5'-diphosphate promotes host defense against bacterial infection via monocyte chemoattractant protein-1-mediated monocytes/macrophages recruitment Purinergic Receptor Type 6 Contributes to Airway Inflammation and Remodeling in Experimental Allergic Airway Inflammation Gene-by-environment effect of house dust mite on purinergic receptor P2Y12 (P2RY12) and lung function in children with asthma ATP Release from Dying Autophagic Cells and Their Phagocytosis Are Crucial for Inflammasome Activation in Macrophages Reverse plasticity: TGF-beta and IL-6 induce Th1-to-Th17-cell transdifferentiation in the gut A New Group of Chromatin-Associated Proteins with a High Content of Acidic and Basic Amino Acids Isolation and Characterisation of Two Calf-Thymus Chromatin Non-Histone Proteins with High Contents of Acidic and Basic Amino Acids The chaperone like function of the nonhistone protein HMGB1 Molecular mechanism and therapeutic modulation of high mobility group box 1 release and action: An updated review High Mobility Group Box Protein 1 (HMGB1): The Prototypical Endogenous Danger Molecule The release and activity of HMGB1 in ferroptosis Tipping the immunostimulatory and inhibitory DAMP balance to harness immunogenic cell death Targeting Inflammation Driven by HMGB1. Front. Immunol. 2020, 11, 484 Involvement of Toll-like Receptors 2 and 4 in Cellular Activation by High Mobility Group Box 1 Yin, H. HMGB1 Activates Proinflammatory Signaling via TLR5 Leading to Allodynia. Cell Rep PAMPs and alarmins: All we need to know about danger Interplay between RAGE and TLR4 Regulates HMGB1-Induced Inflammation by Promoting Cell Surface Expression of RAGE and TLR4 High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal Vaccine adjuvants alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodes Vaccine priming is restricted to draining lymph nodes and controlled by adjuvant-mediated antigen uptake Kinetics of the inflammatory response following intramuscular injection of aluminum adjuvant Macrophage Death following Influenza Vaccination Initiates the Inflammatory Response that Promotes Dendritic Cell Function in the Draining Lymph Node In vivo imaging of inflammasome activation reveals a subcapsular macrophage burst response that mobilizes innate and adaptive immunity A kinetic study of histopathological changes in the subcutis of cats injected with non-adjuvanted and adjuvanted multi-component vaccines Host DNA released in response to aluminum adjuvant enhances MHC class II-mediated antigen presentation and prolongs CD4 T-cell interactions with dendritic cells DNA released from dying host cells mediates aluminum adjuvant activity Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization Inhaled Fine Particles Induce Alveolar Macrophage Death and Interleukin-1alpha Release to Promote Inducible Bronchus-Associated Lymphoid Tissue Formation The adjuvant MF59 induces ATP release from muscle that potentiates response to vaccination Pyroptosis: Host cell death and inflammation A long-awaited merger of the pathways mediating host defence and programmed cell death True grit: Programmed necrosis in antiviral host defense, inflammation, and immunogenicity Necroptosis and its role in inflammation Central Role of CD169(+) Lymph Node Resident Macrophages in the Adjuvanticity of the QS-21 Component of AS01 Hydroxypropyl-beta-cyclodextrin spikes local inflammation that induces Th2 cell and T follicular helper cell responses to the coadministered antigen Intranasal hydroxypropylbeta-cyclodextrin-adjuvanted influenza vaccine protects against sub-heterologous virus infection Modes of Action for Mucosal Vaccine Adjuvants DAMP-Inducing Adjuvant and PAMP Ad-juvants Parallelly Enhance Protective Type-2 and Type-1 Immune Responses to Influenza Split Vaccination DNA vaccines encoding membrane-bound or secreted forms of heat shock protein 70 exhibit improved potency Co-immunization with an optimized plasmid-encoded immune stimulatory interleukin, high-mobility group box 1 protein, results in enhanced interferon-γ secretion by antigen-specific CD8 T cells Molecular adjuvant HMGB1 enhances anti-influenza immunity during DNA vaccination Vaccination with Antigen Combined with alphabeta-ATP as a Vaccine Adjuvant Enhances Antigen-Specific Antibody Production via Dendritic Cell Activation Employing ATP as a New Adjuvant Promotes the Induction of Robust Antitumor Cellular Immunity by a PLGA Nanoparticle Vaccine Human HSP70 and modified HPV16 E7 fusion DNA vaccine induces enhanced specific CD8+ T cell responses and anti-tumor effects The Diversification of Cell Death and Immunity: Memento Mori Immunogenic Cell Death-Based Cancer Vaccines Immunogenic Apoptosis as a Novel Tool for Anticancer Vaccine Development Evaluation of cell surface reactive immuno-adjuvant in combination with immunogenic cell death inducing drug for in situ chemo-immunotherapy Natural modulators of the hallmarks of immunogenic cell death Natural compound inducers of immunogenic cell death Shikonin-enhanced cell immunogenicity of tumor vaccine is mediated by the differential effects of DAMP components Targeting photodynamic and photothermal therapy to the endoplasmic reticulum enhances immunogenic cancer cell death Clinical development of talimogene laherparepvec (T-VEC): A modified herpes simplex virus type-1-derived oncolytic immunotherapy Whole cell vaccination using immunogenic cell death by an oncolytic adenovirus is effective against a colorectal cancer model Immunogenic cell death in cancer and infectious disease Type I interferons in infectious disease Regulation of starvationand virus-induced autophagy by the eIF2 kinase signaling pathway Institutional Review Board Statement: Not applicable. Data Availability Statement: Not applicable. The authors declare no conflict of interest.