Skip to main content
Cell Death and Differentiation logoLink to Cell Death and Differentiation
. 2013 Jul 5;21(1):39–49. doi: 10.1038/cdd.2013.84

Multimodal immunogenic cancer cell death as a consequence of anticancer cytotoxic treatments

H Inoue 1,2,3, K Tani 1,3,*
PMCID: PMC3857623  PMID: 23832118

Abstract

Apoptotic cell death generally characterized by a morphologically homogenous entity has been considered to be essentially non-immunogenic. However, apoptotic cancer cell death, also known as type 1 programmed cell death (PCD), was recently found to be immunogenic after treatment with several chemotherapeutic agents and oncolytic viruses through the emission of various danger-associated molecular patterns (DAMPs). Extensive studies have revealed that two different types of immunogenic cell death (ICD) inducers, recently classified by their distinct actions in endoplasmic reticulum (ER) stress, can reinitiate immune responses suppressed by the tumor microenvironment. Indeed, recent clinical studies have shown that several immunotherapeutic modalities including therapeutic cancer vaccines and oncolytic viruses, but not conventional chemotherapies, culminate in beneficial outcomes, probably because of their different mechanisms of ICD induction. Furthermore, interests in PCD of cancer cells have shifted from its classical form to novel forms involving autophagic cell death (ACD), programmed necrotic cell death (necroptosis), and pyroptosis, some of which entail immunogenicity after anticancer treatments. In this review, we provide a brief outline of the well-characterized DAMPs such as calreticulin (CRT) exposure, high-mobility group protein B1 (HMGB1), and adenosine triphosphate (ATP) release, which are induced by the morphologically distinct types of cell death. In the latter part, our review focuses on how emerging oncolytic viruses induce different forms of cell death and the combinations of oncolytic virotherapies with further immunomodulation by cyclophosphamide and other immunotherapeutic modalities foster dendritic cell (DC)-mediated induction of antitumor immunity. Accordingly, it is increasingly important to fully understand how and which ICD inducers cause multimodal ICD, which should aid the design of reasonably multifaceted anticancer modalities to maximize ICD-triggered antitumor immunity and eliminate residual or metastasized tumors while sparing autoimmune diseases.

Keywords: ICD, DAMPs, apoptosis, necroptosis, immunotherapeutic anticancer agents, oncolytic virotherapy

Facts

  • Accelerated progresses and discoveries in the field of oncology, immunology, and virology have made it possible to translate several emerging immunostimulatory strategies to treat malignant cancers towards promising clinical benefits.

  • Profound understanding of the process of immunogenic cell death (ICD) induction by different ICD inducers such as certain chemotherapeutic agents and oncolytic viruses has highlighted the importance of immunological antitumor effects and proposed novel anticancer therapies.

  • The execution of different types of programmed cell death (PCD), including apoptosis, autophagy, necroptosis, and pyroptosis, which are driven by a plethora of stimuli, was recently found to be regulated by orchestrated interactions among them, and importantly, some of these types of PCD exhibit an ICD property.

  • Tumors and cancer cells treated with certain chemotherapeutic agents and oncolytic viruses can undergo ICD and release tumor-associated antigens (TAAs) accompanied by diverse danger-associated molecular patterns (DAMPs) and inflammatory cytokines to restore the tumor microenvironment and incite TAA-specific antitumor immunity.

Open Questions

  • What are the recent advances in the development of anticancer immunotherapeutic modalities in clinical settings?

  • In response to diversified ICD inducers, how are DAMPs such as CRT, high-mobility group protein B1 (HMGB1), and ATP expressed by or released from the dying cancer cells?

  • How do the diverse types of PCD differentially induce ICD to mount an efficient antitumor immunity?

  • What are the prerequisites for an ideal ICD inducer to obtain an optimum level of ICD for long-lasting antitumor effects?

  • It is vital to understand the molecular mechanisms of how ICD inducers, for example, infection with oncolytic viruses and resultant DAMPs, affect the host immune system. Can manipulation of ICD induction and/or combined strategies synergize with current or emerging oncolytic virotherapies?

The concept of immunogenic cell death (ICD) has recently been introduced to describe dying cancer cells that release endogenous danger molecules, the so-called damage-associated molecular patterns (DAMPs), after the exposure to certain cytotoxic agents to be recognized by antigen-presenting cells (APCs) such as dendritic cells (DCs) followed by formation of T-cell-mediated adaptive immunity.1 Although it has long been considered that apoptotic cell death is tolerogenic, DAMPs have also been found to be released from cells undergoing apoptosis, providing a promising anticancer efficacy.2, 3, 4 Therefore, comprehension of ICD induction gradually increases its significance, particularly in the field of cancer immunotherapy.

Overall prognosis of advanced cancer patients still remains dismal, thus making it imminent for oncologists to develop novel anticancer strategies. Recently, sipuleucel-T (Provenge, Dendreon, Seattle, WA, USA), indicated for patients with metastatic castration-resistant prostate cancer, received FDA's approval as the first therapeutic cancer vaccine.5 In addition, extensive Phase II clinical trials have demonstrated that the oncolytic herpes simplex virus talimogene laherparepvec (T-Vec, Amgen Inc., Thousand Oaks, CA, USA)6 and vaccinia virus JX-547 (Pexa-Vec, Jennerex Biotherapeutics, Inc., San Francisco, CA, USA),7 both of which carry the gene encoding the immunostimulatory cytokine granulocyte–macrophage colony-stimulating factor (GM-CSF), hold great promise for the treatment of advanced cancer patients. Furthermore, cytotoxic T-cell responses directed against oncolytic virus-infected cancer cells have been identified as an essential factor in the process of destruction of cancer.8 Moreover, proinflammatory cytokines generated in the virus-infected cancer cells can restore the immunosuppressive tumor microenvironment.9, 10, 11 Thus, oncolytic viruses are recently viewed as anticancer immunotherapeutic agents. These backgrounds make it imperative to update the molecular pathways and/or cellular constituents that regulate ICD.

Here, we review the progress of research on ICD, emphasizing how apoptotic, autophagic, and necroptotic cell death, called type 1, 2, and 3 PCD, respectively, are induced by various ICD inducers to achieve successful antitumor immunity. These multiple modes can be categorized by describing initiating events, intermediated changes, terminal cellular events, and their immunological responses, which are summarized in Table 1. In the later section, we outline the characteristics of anticancer agents and oncolytic viruses and how they induce diversified forms of cell death and interact with host's immune system.

Table 1. Comparison of multiple forms of programmed cell death and necrosis.

  Apoptosis (type 1 PCD) Autophagic cell death (type 2 PCD) Necroptosis (type 3 PCD) Pyroptosis Necrosis
Mode of cell death Programmed Programmed Programmed Programmed Accidental
Initiators TNF-α, FasL, or TRAIL, infectious pathogens Nutrient deprivation, HDAC inhibitors, hypoxia, infectious pathogens TNF-α, FasL, or TRAIL, microbial infections Ischemic injury DAMPs, microbial infections Toxins, infections, inflammation, trauma
Intermediate signalings Mitochondrial pathway Caspase-3, -6, -7-dependent Caspase-independent autophagosome formation Lysosomal protease TNF receptor signaling JNK activation Caspase-independent RIP1/RIP3 necrosome Nod-like receptors Caspase 1-dependent pyroptosome Inflammasome -
Terminal cellular events Non-lytic cell shrinkage DNA fragmentation apoptotic bodies Non-lytic autophagic bodies Non-lytic, loss of plasma membrane, swollen cellular organelles Lytic, rapid loss of plasma membrane, cell swelling, pore formation Lytic, plasma membrane rupture, leak of content
Inflammation Non-inflammatory Non-inflammatory Proinflammatory Proinflammatory Proinflammatory
Immunogenicity + + ++ ++ +++
DAMPs released Ecto-CRT HMGB1 and ATP release HMGB1 and ATP release Long genomic DNA IL-6 HMGB1 and ATP release IL-1α, IL-1β, IL-6, IL-18, and TNF-α chemokines HMGB1 and ATP release IL-1α, IL-33 mRNA, and genomic DNA
Eat-me signals Ecto-CRT LPC secretion PS exposure LPC secretion PS exposure PS exposure PS exposure

Abbreviations: TNF-α, tumor necrosis factor- α; FasL, Fas ligand; TRAIL, TNF-related apoptosis-inducing ligand; HDAC, histone deacetylase; IL-1α, interleukin-1α ; IL-1β, interleukin-1β; IL-6, interleukin-6; IL-18, interleukin-18; IL-33, interleukin-33; ICD, immunogenic cell death; LPC, lysophosphatidylcholine; PS, phosphatidylserine; JNK, c-Jun N-terminal kinase.

The table gives a schematic overview of the multiple forms of cell death incuding apoptotic cell death (type 1 PCD), autophagic cell death (type 2 PCD), cell death induced by necroptosis (type 3 PCD), pyroptosis and necrosis. The extent of immunogenicity in each cell death subsection is scored as +, ++, and +++, according to the expression levels of ‘eat-me' signals and DAMPs emission. ICD in cancer can display different ‘eat-me' signals, including ecto-CRT and LPC, on the cell membrane, as well as emission of DAMPs, ATP, and HMGB1. This peculiar ecto-CRT, which facilitates engulfment of TAAs from cancer cells by DCs, can only be found on cells that succumb to immunogenic apoptosis, whereas it is not present on cells dying in an immunologically silent manner. LPC secretion, PS exposure, and ATP release require autophagy induction. Numerous exquisite expression patterns shaped by the constituents of DAMPs and the interactive status of the immune system will predominantly determine the fate of subsequent immune responses, namely, immune tolerance or antitumor immunity

Apoptotic Cell Death as ICD

From ten million to billions of cells die per day as a consequence of normal tissue turnover,12 which are vital for organisms to retain homeostasis.13, 14 Therefore, the existence of multiple modes of cell death in nature is not surprising. Apoptosis, type 1 PCD, is a specialized form of cell death, characterized by typical morphological changes, including chromatin condensation, nuclear fragmentation, and membrane blebbing (Table 1).15 Apoptosis occurs ubiquitously in normal tissues and causes ‘quiet' cell death that uses phosphatidylserine (PS) as an ‘eat-me' signal to be quickly recognized by peripheral APCs. Although apoptotic cell death has been historically considered to be non-immunogenic,16 recent studies unraveled that several antineoplastic agents, including doxorubicin,1, 17 oxaliplatin,18, 19 cisplatin,20 and irradiation,21, 22, 23 can trigger immunogenic apoptosis.2 Mechanistically, the immunogenic apoptotic bodies induced by exposure to doxorubicin are sensed by APCs through their TLR-2/TLR-9-MyD88 signaling pathways.17

DAMPs: as Effectors in ICD

The primary conceptual theory of the pattern recognition of pathogen-associated molecular patterns (PAMPs), such as viral or bacterial components, has failed to fully explain the consequence of immunogenicity. Thus, the secondary concept of DAMPs has been proposed, which could provoke an immune response.24 Released DAMPs as hallmarks of ICD consisted of adenosine triphosphate (ATP), high-mobility group protein B1 (HMBG1), and exposed molecules on the outer membrane of dying cells such as CRT (ecto-CRT), heat-shock proteins (Hsp90 and Hsp70), and endoplasmic reticulum (ER) sessile proteins.25, 2627 The excretion of DAMPs was considered to occur during necrosis under inflammatory and/or pathological conditions. However, DAMPs have recently been reported to be produced from apoptotic cancer cells treated with chemotherapy1, 18 or radiotherapy.21

ICD Inducers

ICD inducers include multiple anticancer therapeutic modalities. It has been recently proposed that they can be classified into two categories, type I or II ICD inducers, based on their distinct actions to induce ER stress leading to apoptotic cell death (Tables 2 and 3).27, 28 The majority of ICD inducers such as chemotherapeutic agents (mitoxantrone,29 anthracyclines,2, 30, 31 oxaliplatin,18, 19 and cyclophosphamide32), shikonin,33, 34 the proteasome inhibitor bortezomib,35 and 7A7 (an epidermal growth factor receptor-specific antibody),36 cardiac glycosides,37 and the histone deacetylase inhibitor (vorinostat)38 are categorized as type I ICD inducers that primarily target cytosolic proteins, plasma membranes, or nucleic proteins. They also induce ER stress via collateral effects. Bortezomib, cardiac glycosides, and shikonin effectively impede protumorigenic cytokine signaling.27 Shikonin has been found to induce type 1 or 3 PCD, which is determined by caspase-8 activation as the ‘decision-making switch'.39 On the other hand, type II ICD inducers, which preferentially target the ER, include hypericin-based photodynamic therapy (PDT)40, 41, 42 and oncolytic coxsackievirus B3 (CVB3).9 Hypericin-based PDT is an anticancer therapy that utilizes hypericin to induce reactive oxygen species (ROS) in the vicinity of the ER.43 Cancer cell infection with oncolytic viruses produce large amounts of viral proteins, which inevitably cause ER stress and ROS production to promote viral replication.44, 45 The quality and/or quantity of ER stress linking ROS triggered by ICD inducers may determine the ICD properties. Indeed, the previous finding that rigorous ROS-mediated ER stress augmented the release of DAMPs revealed an unrecognized role of RNA-dependent protein kinase (PKR)-like ER kinase (PERK) as a constituent of mitochondria-associated ER membranes to exert ROS-mediated mitochondrial apoptosis.40, 41, 46 These observations indicate the superiority of type II ICD inducers with respect to immunological antitumor efficacies. However, further investigations to elucidate the precise interconnection between the ER stress and ROS production will be required to optimize antitumor immune responses.

Table 2. Classification of type I ICD inducers determined by their major targets to provoke antitumor responses.

Anticancer agents Type of cell death induced DAMPs Major targets by ICD inducers Preclinical observations for inciting antitumor immunity
Cytotoxic agents (mitoxantrone, oxaliplatin, anthracyclines) Apoptosis, autophagic cell death, necroptosis Ecto-CRT, ERp57, HMGB1, and ATP release Nucleus (DNA or DNA-related proteins for cell mitosis) In vivo antitumor effect is mitigated by depletion of CD8+T cells. Immunogenicity requires ecto-CRT in prophylactic tumor vaccination mouse models.
Cyclophosphamide (CTX) Apoptosis Ecto-CRT, HMGB1 release Nucleus (DNA) Metronomic doses of CTX deplete Treg from bed and tumors, CTX modulates DCs to produce IL-12
Shikonin Apoptosis, necroptosis Ecto-CRT, ecto-Hsp70 Cytosol (pyruvate kinase-M2 protein) DCs incubated with shikonin increase Th1 cells but decrease Treg cells
Bortezomib Apoptosis, autophagic cell death Ecto-Hsp90 Cytosol (26S proteasome) Cytotoxicity of NK cells against bortezomib-treated cells increased
7A7 (EGFR-specific antibody) Apoptosis Ecto-CRT, ERp57, ecto-Hsp70, ectp-Hsp90 Cell surface receptor (EGFR) Contribution of CD4+ T and CD8+ T to 7A7-triggered suppression of metastasis in mice model
Cardiac glycosides Apoptosis Ecto-CRT HMGB1 and ATP release Cell surface (Na+/K+-ATPase, enzyme) Prophylactic antitumor immunity is partially dependent on CD8+ T cells accompanied with Th17 cells
UVC irradiation Apoptosis, necroptosis, necrosis Ecto-CRT and ERp57, HMGB1 and ATP release Nucleus (DNA) UVC-treated cells increase susceptibility to attack by NK cells and total splenocytes
Vorinostat (HDAC inhibitor) Apoptosis Autophagic cell death Ecto-CRT Nucleus (chromatin structure) Promote the differentiation of CD8+ T cells to memory cells

Abbreviations: Ecto-CRT, calreticulin exposure; DAMPs, damage-associated molecular patterns; ICD, immunogenic cell death, HMGB1; high-mobility group protein B1; Hsp, heat-shock protein; Treg, regulatory T cells; DCs, dendritic cells; IL-12, interleukin-12; NK, natural killer; EGFR, epidermal growth factor receptor; ATP, adenosine triphosphate; UVC, ultraviolet C

Table 3. Classification of type II ICD inducers determined by their major targets to provoke antitumor responses.

Anticancer agents Type of cell death induced DAMPs Major targets by ICD inducers Preclinical observations for inciting antitumor immunity
PDT with hypericin Apoptosis, autophagic cell death dependent on Bax/Bak, necroptosis Ecto-CRT, ecto-Hsp70, ectp-Hsp90, HMGB1, and ATP release ER (ROS generation) PDT -hypericin therapy provokes antitumor immunity in both prophylactic and therapeutic murine tumor models. Same therapy-treated tumor cells result in phenotypic maturation of DCs and robust CD4+T and CD8+T cell expansion
CVB3 Apoptosis Ecto-CRT, HMGB1 translocation, ATP release ER (ROS generation) Intratumoral CVB3 administration markedly recruited NK cells and granulocytes, both of which contribute to the antitumor effects as shown by depletion assays, macrophages, and mature DCs into tumor tissues
Ad5/3-hTERT-E1A-hCD40L: chimeric Ad5/3 capsid, an hTERT promoter and human CD40L Apoptosis Ecto-CRT, HMGB1 release, ATP release ER (ROS generation) In two syngeneic mouse models, murine CD40L induced activation of APCs, leading to increased IL-12 production in splenocytes, associated with induction of the Th1 cytokines IFN-γ, RANTES, and TNF-α. Tumors treated with Ad5/3-CMV-mCD40L displayed an enhanced presence of macrophages and cytotoxic CD8+ T cells
Edmonston strain MV Apoptosis IL-6 production, HMGB1 release ER (ROS generation) Coculture of MV-infected melanoma cells with human DCs led to both CD80 and CD86 upregulation on them. CD8+ T cells cocultured with tumor cell-loaded and MV-infected DCs degranulated CD107a to target tumor cells with functional killing activity

Abbreviations: PDT, photodynamic therapy; CVB3, coxasackievirus B3; MV, measles virus; ROS, reactive oxygen species; ER, endoplasmic reticulum; hTERT, telomerase reverse transcriptase; hCD40L, human CD40 ligand; Th1, T helper type 1; RANTES, regulated and normal T cell expressed and secreted; TNF-α, tumor necrosis factor-α IL-6, interleukin-6

Calreticulin Exposure

In response to specific chemotherapeutic agents, oncolytic viruses, and vorinostat, ecto-CRT has been found only on cells succumbing to immunogenic apoptosis.2, 9, 38 This ‘eat-me' signal promotes phagocytosis by DCs, thereby facilitating their tumor antigen presentation and incitement of TAA-specific cytotoxic T cells.2, 47 It has been shown that blockade of CRT inhibits phagocytosis of anthracycline-treated tumor cells by DCs and impairs their immunogenicity in mice.2, 47 In general, CRT exposure during ICD is an earlier process occurring within a few hours than PS externalization.48 The ecto-CRT induction capacity of ICD inducers has been shown to depend on the properties of ER stress and ROS production.2, 37, 49 Cancer cells can induce ecto-CRT followed by disturbance of the ER structure with GADD34 activation and PERK phosphorylation. It has been shown that depletion of PERK abolishes anthracycline-driven ecto-CRT and immunogenicity of cellular death (ER stress module),19 and that caspase-8 acts upstream of apoptotic proteins Bax and Bak, and subsequent cleavage of its substrate Bap31 (apoptotic module) is indispensable for ecto-CRT induction.19 Furthermore, a direct interaction between ecto-CRT and ERp57 was shown to be required for their cotranslocation to the cell surface (Figure 1).29 Unlike the release of HMGB1 and ATP, ecto-CRT could be one of the determinants that distinguishes between immunogenic and non-immunogenic cell death.47

Figure 1.

Figure 1

Oncolytic virus (CVB3) infection-triggered cancer cell death induces innate immune cell-mediated antitumor immunity. Intratumoral CVB3 infection-activated natural killer (NK) cells and granulocytes with enhanced expression of CD107a, a cytolytic degranulation marker, have been found to contribute to substantial antitumor effects as evidenced by NK cell and granulocyte depletion assays. Upon CVB3 infection, tumor cells can partially induce ecto-CRT on human tumor cells during early apoptosis, whereas majority of other viruses subvert ICD by circumventing ecto-CRT induction, and followed by robust release of DAMPs, including ATP and HMGB1, during later stages of cell death, which facilitates maturation of DCs via binding to Toll-like receptor 4 (TLR4)/RAGE and P2 × 7R, respectively. Viral genomes and/or viral progenies also stimulate DCs for their activation. Mature DCs may then efficiently phagocytose TAAs simultaneously released from dying cells and ultimately cross-present them to CD8+ T cells with the support with CD4+ T cells to elicit substantial antitumor immunity. Although ATP secretion relies on autophagic machinery, the other forms of cancer cell death, such as autophagic cell death and necroptosis, triggered by CVB3 infection have not yet been fully investigated

HMGB1

HMGB1, one of the DAMPs, is a DNA-binding protein originally known as a nuclear non-histone chromatin-binding protein.50 Although extracellular HMGB1 had been deemed to be released mainly from the nucleus during necrosis,42 it was found to be excreted from cells undergoing late stage of apoptosis and autophagy.30, 51 HMGB1 inhibition in cancers undergoing immunogenic apoptosis impaired their ability to incite antitumor immunity in a prophylactic vaccination model.30 HMGB1 initiates potent inflammation by stimulating the production of proinflammatory cytokines52 from APCs via its binding to different surface receptors including receptor for advanced glycation end-products (RAGE), TLR2, TLR4, TLR9, and TIM3 (Figure 1).53, 54 Importantly, the binding of HMGB1 to TLR4 on APCs was required to suppress tumor development, which is consistent with clinical study showing that breast cancer patients harboring a single-nucleotide polymorphism (Asp299Gly) in the TLR4 gene undergo an early relapse after anthracycline treatment.30, 55, 56 In contrast, secreted HMGB1 could induce a protumor inflammation to facilitate tumor progression.57 In addition, HMGB1 expression is significantly associated with overall survival of patients with bladder cancer.58 As HMGB1 is an intrinsic sensor of oxidative stress,59 the immunomodulatory properties of HMGB1 might be determined by its redox status.60, 61 Indeed, reduced HMGB1 production from dying cells was shown to trigger the immunogenic DCs, whereas oxidized HMGB1 during apoptosis fails.51 As the extracellular space is usually oxidative under physiological conditions but is unpredictably variable under pathogenic conditions,62 the unstable redox status of the tumor microenvironment might account for these inconsistent findings. However, the observation that the tumor microenvironment tends to be pro-oxidative63 implies that a therapeutic approach using antioxidants to decrease ROS production would be favorable to stimulate antitumor immunity. Importantly, many anticancer agents, including chemotherapy,30 radiation,22 or oncolytic viruses,9, 64, 65 have been shown to induce HMGB1 release from cancer cells, highlighting the significance of further addressing the mechanism of how these modalities affect the redox status of HMGB1.

Adenosine Triphosphate

Extracellular ATP released from apoptotic cells is another important factor in ICD induction. ATP signaling recognized by P2Y2 receptors on phagocytes as a ‘find-me' signal enables them to migrate into inflamed sites.66 Indeed, ATP released from cancer cells treated with chemotherapeutic agents is essential for effective antitumor immune responses.67 In addition, small interfering RNA-mediated inhibition of autophagic machinery abolishes ATP release from chemotherapy-treated tumor cells and mitigates the antitumor response.68 Radiotherapy triggers ATP release from dying tumor cells through its interaction with the P2 × 7 purinergic receptor,69 possibly resulting in the activation of the NLRP3–ASC–inflammasome axis and subsequent secretion of IL-1β.70

We and others recently showed that oncolytic viruses induce secretion of extracellular ATP from human cancer cells (Figure 1).9, 65 Unlike ecto-CRT induction, the release of ATP and HMGB1 is triggered by a range of death-inducing stimuli, and is not restricted to induction in apoptotic cell death.47 Although ATP production is required for efficient vaccinia virus production71 and facilitates HIV infection through its interaction with P2Y2 receptors,72 there is little knowledge of how oncolytic viruses provoke ATP release.

Autophagic Cell Death

Autophagy physiologically has catabolic roles, particularly in cell survival.73 However, persistent autophagy causes a caspase-independent form of cell death that is, morphologically defined as autophagic cell death (ACD), termed as type 2 PCD, through lysosomal proteinase-regulated elimination of cellular organelles.74, 75 Autophagy sometimes directs itself to cellular death, either in cooperation with apoptosis or as a back-up system, and thus is deemed as a cellular program with a ‘double-faced' role.

Interestingly, the key molecules of autophagy and apoptosis pathways are intricately intertwined with shared several molecules including regulatory genes such as p53 and p19ARF.76 This crosstalk therefore can be viewed as a significant clue to understand the fate of dying cancer cells from therapeutic view points. Although ACD occurs without chromatin condensation but with massive autophagic vacuolization,77 autophagy, often disabled in cancer, has been shown to be required for induction of immunogenicity.68 First, dying cells in embryoid bodies that lack autophagy-related gene are unable to express the ‘eat-me' signals and secrete lower levels of the ‘come-get-me' signal, lysophosphatidylcholine.78 Second, autophagy deficiency hinders ATP secretion from dying cancer cells, resulting in the impairment of DC recruitment and formation of adaptive immunity responses (Table 1).68 Third, the inability of autophagy-deficient cancer cells to provoke antitumor immunity after chemotherapy can be reverted by suppression of extracellular ATP-degrading enzymes.68, 79 Therefore, immunogenicity of ACD could be mediated by subtle spatiotemporal alterations in the treated cancer cells.

Novel strategy of autophagy inhibition is therapeutically effective for eliminating apoptosis-resistant cancer cells based on the rationale that growing tumors may harness autophagy as an adaptation to resist therapeutic stresses.80, 81, 82 Hence, more efforts should be made to elucidate the intricate interaction between autophagy inhibition and resulting effects on the immunogenicity.

Necrotic Cell Death and Necroptosis

Necrotic cell death is induced by external factors such as toxins, cancer, infections, and trauma, and is morphologically characterized by cellular swelling, rupture of the plasma membrane, and loss of cytoplasmic contents.83 Understanding the immunogenicity of necrotic cell death is becoming important because it frequently induces robust inflammatory reactions to mount protective immune responses (Table 1).84, 85, 86 Although necrosis has long been viewed as non-PCD, its execution was shown to be controlled by specific signal-transduction pathways and catabolic mechanism.87, 88, 89 This alternative form of necrotic PCD, aptly termed necroptosis (type 3 PCD), is induced by tumor necrosis factor (TNF) receptor signaling that involves activation of the receptor-interacting protein (RIP) family. Upon inhibition of apoptotic pathway by the caspase inhibitor, activation of RIP1 and RIP3 kinase leads to mitochondrial instability and cell death.90, 91 Phosphorylated RIP1 and RIP3 generate a molecular complex called the necrosome, which initiates necroptosis. ROS production under necroptosis has been shown to facilitate TNF-α-induced cell death by sustaining c-Jun N-terminal kinase activation.92 Intriguingly, necroptosis can also be executed via stimulation by apoptosis-inducible ligands such as TNF-α, FasL, or TRAIL (Table 1). Notably, cytotoxic agents are shown to induce necrotic cell death in apoptosis-defective cancer cells,93 probably because necroptosis is principally induced when a cell cannot die via apoptotic pathways.94 On the other hand, conventional therapy-resistant cancer stem cells (CSCs) have a higher antiapoptotic activity than that of their counterparts.95, 96 Therefore, it would be vital to clarify the key machinery of not only the necroptosis induction in cancer cells for CSC-directed therapeutic application but also the resultant immunogenicity to modulate antitumor immunity.

Pyroptosis

Pyroptosis is a recently indentified form of PCD stimulated by microbial infections and non-infectious stimuli such as myocardial infarction and cancer. In contrast to apoptosis, pyroptosis is uniquely mediated by caspase-1 activity triggered by the formation of a cytosolic complex termed the ‘inflammasome', resulting in highly inflammatory outcomes (Table 1). Pyroptotic cells represent morphological characteristics, some of which are shared with apoptosis and necrosis.97 The function of activated caspase-1 is to cleave proteolytically the proforms of the proinflammatory cytokines, IL-1β and IL-18, to their active forms.97 Although pyroptosis has been intensively studied in the context of bacteria-infected macrophages,98 it can also be triggered in human cancer cells infected with recombinant herpes simplex virus 2 (HSV-2) (Table 4).99 Pyroptotic cancer cells induced by microbial infection have been recently shown to facilitate phagocytosis by macrophages, presumably through their PS exposure and ATP release.100 Accordingly, the caspase-1-dependent generation of proinflammmatory cytokines and other DAMPs could be essential factors to provide a suitable inflammation for ICD induction.

Table 4. DNA oncolytic viruses and their differential properties to induce either multiple forms of cell death or antitumor immunity.

Oncolytic viruses Type of cancer cells Type of cell death induced DAMPs Possible mechanism of antitumor immunity
hTERT-Ad: CRAds regulated by human hTERT promoter Human glioma, cervical and prostate cancer Autophagy NA NA
hTERT-Ad Human lung cancer Autophagy via E2F1-miR-7-EGFR NA NA
OBP-702: p53-armed hTERT-Ad Human osteosarcoma Apoptosis Autophagy NA NA
CRAd-S-RGD: Ad5 carrying the RGD motif and survivin promoter Human glioma cells Autophagy NA NA
Ad5/3-hTERT-E1A-hCD40L: chimeric Ad5/3 capsid with hTERT promoter Murine urothelial carcinoma, melanoma Apoptosis Ecto-CRT, ATP and HMGB1 Enhanced recruitment of macrophages and CD8+ T cells
ZD55-IFN-β: Oncolytic adenovirus carrying IFN-β Human hepatoma, breast cancer Apoptosis Necroptosis NA NA
Vaccinia virus Human colon, breast, ovarian cancer Not apoptosis Possibly necrosis HMGB1 release NA
vSP: antiapoptosis genes, SPI-1- and SPI-2-deleted vaccinia virus Murine colon adenocarcinoma Apoptosis Necrosis HMGB1 release NA
HSV2: Human simplex virus 2 Human endometrial cancer Apoptosis Necrosis HMGB1 release NA
HSV-1716: a replication-restricted mutant herpes simplex virus Murine ovarian cancer NA NA Intratumoral injection induced IFN-γ, CXCL9 and CXCL10 with increase in NK and CD8+ T cells
HSV-2 mutant ΔPK: ICP10PK-deleted HSV-2 virus Human melanoma cells Apoptosis Pyroptosis NA Dominant induction of CD4+ Th1 cells

Abbreviations: hTERT, telomerase reverse transcriptase; CRAds, conditionally replicative adenoviruses; miR-7, microRNA-7, EGFR, epidermal growth factor receptor, ecto-CRT, calreticulin exposure; DAMPs, damage-associated molecular patterns; ICD, immunogenic cell death, HMGB1; high-mobility group protein B1;ATP, adenosine triphosphate; IFN-β, interferon-β; HSV, herpes simplex virus; IFN-γ, interferon-γ; CXCL9, chemokine (C–X–C motif) ligand 9; CXCL10, chemokine (C–X–C motif) ligand 10; NK, natural killer cells; NA, not assessed; Th1, T helper type 1

DAMPs Induced by Infection with Oncolytic Viruses

Because oncolytic viral infection can produce abundant PAMPs, including viral proteins and nucleic acids, followed by the release of DAMPs and the entire repertoire of TAAs from treated tumors,101 oncolytic virus-triggered ICD may be more effective for induction of antitumor immunity. As viruses have developed sophisticated machineries to evade apoptotic cell death and interfere with ER stress and autophagy responses for their survival,102, 103 ICD may have played an essential role in the everlasting war between viruses and their hosts. We and other groups have found that many oncolytic viruses can induce apoptotic cell death and/or necrosis in cancer cells,9, 104, 105, 106 supporting their immunostimulatory potential to augment antitumor efficacy (Tables 4 and 5).107, 108 CVB3 infection induces multiple DAMPs including ecto-CRT, HMGB1 translocation from nuclei, and ATP release from human lung cancer cells. Importantly, intratumoral CVB3 administration can prominently recruit cytolytic degranulation marker CD107a-mobilized NK cells and granulocytes, and mature DCs into the tumor bed (Figure 1).9, 27 As pathogenic viruses have developed their strategies to subvert ecto-CRT and circumvent ICD induction,109 it is noteworthy that CVB3 infection can induce ecto-CRT accompanied by other DAMPs.9 Furthermore, we demonstrated that both NK cells and granulocytes substantially contributed to the CVB3-mediated antitumor efficacy as evidenced by in vivo depletion assays.9

Table 5. RNA oncolytic viruses and their differential properties to induce either multiple forms of cell death or antitumor immunity.

Oncolytic viruses Type of cancer cells Type of cell death induced DAMPs Possible mechanism of antitumor immunity
Edmonston vaccine strain of MV Human melanoma NA IL-6 HMGB1 release Human DC maturation Priming an adaptive T-cell response
MV-NPL: genetically engineered MV Human renal cell carcinoma Apoptosis NA NA
MV-CEA:Edmonston vaccine MV genetically engineered to produce CEA antigen Human breast cancer Apoptosis NA NA
CVB3 Human non-small-cell lung cancer Apoptosis Preapoptotic ecto-CRT, HMGB1 translocation, ATP release Phenotypic activation of immature DCs and lytic NK cells in tumors. Deletion of NK and granulocytes abrogated the CVB3-induced in vivo antitumor immunity
NDV Human glioma Autophagy NA NA
Reovirus Human multiple myeloma Apoptosis Autophagy NA NA
Live-attenuated poliovirus Human neuroblastoma Apoptosis NA NA
M51R: M protein mutant VSV Human glioblastoma multiforme Apoptosis NA NA
Interferon-sensitive VSV (AV3 strain) Human prostate cancer Apoptosis NA NA

Abbreviations: MV, measles virus; CVB3, coxasackievirus B3; NDV, New castle disease virus; CEA, carcinoembryonic antigen; VSV, vesicular stomatitis virus

Upon intratumoral replication of oncolytic viruses, resultant alterations in tumor microenvironment may restore the compromised antitumor immunity, presumably through induction of IFNs and/or cytokines that activate NK cells and APCs.110, 111 Although tumor-infiltrating DCs were impaired at maturation by immunosuppressive IL-10, PGE2, and transforming growth factor β produced from tumor cells,112 unidentified components in the culture media from reovirus-infected cancer cells facilitated maturation of DCs.113

Recent studies delineated that oncolytic viruses such as vaccinia, measles, HSV-2, and adenovirus cause the release of HMGB1.64, 65, 114, 115, 116, 117 Although HMGB1 interacts with viral components and may modulate viral replication,117 the molecular mechanisms of how each oncolytic virus differentially produces these DAMPs remain largely elusive.

Multimodal PCD Induced by Oncolytic Viruses

We showed that approximately 20% of CVB3-mediated cytotoxicity of A549 cells resulted from apoptotic cell death.9 This induction was presumably due to the capacity of CVB3 infection to induce PKR-mediated phosphorylation of eIF2 and caspase-8-mediated activation of proapoptotic mediator, caspase-3 (Figure 1).118, 119 Other DNA and RNA oncolytic viruses have been reported to induce apoptotic cancer cell death (Tables 4 and 5). However, there are only two reports showing that virus-induced ecto-CRT was correlated with enhanced intratumoral infiltrations of immune subpopulations, which accounted for the ‘in vivo' remarkable antitumor immunity.9, 65

Several studies showed that recombinant oncolytic adenoviruses induced ACD in human malignant glioma cells,120 brain tumor stem cells,121 osteosarcoma cells,105 and lung cancer cells.122 Newcastle disease virus also triggered autophagy in glioma cells to promote its viral replication.123 Reovirus-mediated oncolysis of multiple myeloma was reported to be orchestrated via upregulation of autophagy.124 Because cancer cells are largely refractory to apoptotic inducers but vulnerable to necroptosis,39 overcoming anticancer drug resistance may be achieved by activation of necroptotic rather than apoptotic pathways, where the former might be the intrinsic ‘Achilles heel' of cancers.125 So far only recombinant adenovirus has been shown to facilitate both necroptotic and apoptotic cell death with a synergistic effect on cancer cells when combined with doxorubicin (Tables 4 and 5).126 In addition, most oncolytic viruses may induce pyroptotic cancer cell death accompanied by abundant proinflammatory cytokines and DAMPs. Accordingly, some oncolytic viruses may induce multimodal ICD, allowing them to be a plausible modality as promising agents of immunotherapy.

Strategies to Enhance the Potentials of ICD Induced by Oncolytic Viruses

Besides DAMPs, massive production of type I IFNs (IFN-α/β) upon oncolytic viral infection can be a potent immunomodulator through their indirect immunostimulatory effects on neutrophils and T cells,127, 128 as well as through their direct antiproliferative effects.129 Despite a creation of multimodal ICD by oncolytic viruses to facilitate antitumor immunity, much attention should be paid to the preferential antiviral immunity that might impede direct viral oncolysis-mediated tumor destruction. To avoid this, cyclophosphamide is shown to retard immune removal of oncolytic viruses, enhancing the persistence of viral infection.130 Another promising strategy to overcome antiviral immunity could be potentiating immune responses by gene modification of oncolytic viruses to arm them with immunostimulatory cytokines, such as GM-CSF, IL-2, IL-12, and IL-15. Indeed, the results of clinical trials of the GM-CSF gene-harboring oncolytic vaccinia virus JX-594 and the GM-CSF gene-harboring oncolytic herpes virus talimogene laherparepvec demonstrated that a clinical benefit can be accomplished by combined respective oncolytic activity with the recruitment of immune cells.6, 7, 131 The combination of adoptive T-cell therapy with oncolytic viruses is shown to elicit an increased antitumor effect.131, 132 Collectively, the design of combinatorial therapies of oncolytic viruses with immunotherapeutic modalities may hold the key to mount maximally a multifaceted attack against cancers.

Conclusions

Although mechanism of ICD induction is a very complicated process, we need to elucidate how dying cells become much more stimulatory in shaping antitumor immune responses than was ever expected. Notably, intermediate death processes, including caspase activation, mitochondrial degradation by autophagy, ROS production, and oxidative modification of DAMPs, have been found to fine-tune the balance between antitumor tolerance and immunity, providing implications in manipulation of ICD.

Four forms of PCD, apoptosis, autophagy, necroptosis, and pyroptosis, may jointly decide the fate of cells of malignant cells. However, in terms of immunogenicity, investigations of only apoptotic cell death in cancer cells have just begun. Therefore, further elucidation of determinants of respective PCD-inducing pathway and characterization of resultant ICD should aid to develop novel anticancer strategies. A recent review advocates a list of characteristics for an ideal ICD inducer,27 as follows: (1) efficiently activates apoptosis or necrosis leading to emission of multiple DAMPs and TLR agonists;133, 134 (2) irrelevant in drug-efflux pathways;135 (3) can induce ER stress;134 (4) has negligible suppressive or inhibitory effects on immune cells;136 (5) counteracts immunosuppressive responses;136, 137 and (6) directly targets not only the primary tumor but also metastases.138 No ideal ICD inducer exists, but it is important to seek for ideal combinatorial therapies that could achieve these properties. Of the currently known relevant ICD inducers, those that meet most of these properties include mitoxantrone, hypericin-PDT, and shikonin. However, diverse oncolytic viruses could be the promising ICD inducer as we gain more knowledge about the properties yet to be investigated. Evidently, they can destroy conventional therapy-resistant CSCs,139 possibly through their ability to induce distinctive PCD and/or modification to express genes that target CSC-specific signaling pathways underpinning their cell survival.140

Gaining more detailed insights into the mechanisms of ICD induction, to be perceived by the immune system, will not only ameliorate the development of promising anticancer agents or combinatorial therapies but also offer useful knowledge in various life science fields including virology, immunology, and clinical medicine.

Acknowledgments

This work was supported by grants from the Ministry of Education, Culture, Sports, Science, and Technology (17016053 and 23240133), and the Ministry of Health Labour and Welfare (23080101), Japan.

Glossary

ICD

immunogenic cell death

PCD

programmed cell death

PAMPs

pathogen-associated molecular patterns

DAMPs

danger-associated molecular patterns

ecto-CRT

calreticulin exposure

HMGB1

high-mobility group protein B1

ATP

adenosine triphosphate

PS

phosphatidylserine

Hsp

heat-shock protein

ACD

autophagic cell death

ER

endoplasmic reticulum

ROS

reactive oxygen species

APCs

antigen-presenting cells

DCs

dendritic cells

TAAs

tumor-associated antigens

GM-CSF

granulocyte–macrophage colony-stimulating factor

PDT

photodynamic therapy

CVB3

coxsackievirus B3

CSCs

cancer stem cells

The authors declare no conflict of interest.

Footnotes

Edited by M Piacentini

References

  1. Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med. 2005;202:1691–1701. doi: 10.1084/jem.20050915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:54–61. doi: 10.1038/nm1523. [DOI] [PubMed] [Google Scholar]
  3. Thompson CB. Apoptosis in the pathogenesis and treatment of disease. Science. 1995;267:1456–1462. doi: 10.1126/science.7878464. [DOI] [PubMed] [Google Scholar]
  4. Garg AD, Krysko DV, Verfaillie T, Kaczmarek A, Ferreira GB, Marysael T, et al. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO J. 2012;31:1062–1079. doi: 10.1038/emboj.2011.497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Tanimoto T, Hori A, Kami M.Sipuleucel-T immunotherapy for castration-resistant prostate cancer N Engl J Med 20103631967–1968.1966; author reply. [DOI] [PubMed] [Google Scholar]
  6. Senzer NN, Kaufman HL, Amatruda T, Nemunaitis M, Reid T, Daniels G, et al. Phase II clinical trial of a granulocyte–macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J Clin Oncol. 2009;27:5763–5771. doi: 10.1200/JCO.2009.24.3675. [DOI] [PubMed] [Google Scholar]
  7. Heo J, Reid T, Ruo L, Breitbach CJ, Rose S, Bloomston M, et al. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med. 2013;19:329–336. doi: 10.1038/nm.3089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Sobol PT, Boudreau JE, Stephenson K, Wan Y, Lichty BD, Mossman KL. Adaptive antiviral immunity is a determinant of the therapeutic success of oncolytic virotherapy. Mol Ther. 2011;19:335–344. doi: 10.1038/mt.2010.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Miyamoto S, Inoue H, Nakamura T, Yamada M, Sakamoto C, Urata Y, et al. Coxsackievirus B3 is an oncolytic virus with immunostimulatory properties that is active against lung adenocarcinoma. Cancer Res. 2012;72:2609–2621. doi: 10.1158/0008-5472.CAN-11-3185. [DOI] [PubMed] [Google Scholar]
  10. Contag CH, Sikorski R, Negrin RS, Schmidt T, Fan AC, Bachireddy P, et al. Definition of an enhanced immune cell therapy in mice that can target stem-like lymphoma cells. Cancer Res. 2010;70:9837–9845. doi: 10.1158/0008-5472.CAN-10-2650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol. 2012;30:658–670. doi: 10.1038/nbt.2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Curtin JF, Cotter TG. Apoptosis: historical perspectives. Essays Biochem. 2003;39:1–10. doi: 10.1042/bse0390001. [DOI] [PubMed] [Google Scholar]
  13. Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ 2012. 19:107–120. doi: 10.1038/cdd.2011.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Brenner C, Kroemer G. Apoptosis. Mitochondria–the death signal integrators. Science. 2000;289:1150–1151. doi: 10.1126/science.289.5482.1150. [DOI] [PubMed] [Google Scholar]
  15. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35:495–516. doi: 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chen W, Frank ME, Jin W, Wahl SM. TGF-beta released by apoptotic T cells contributes to an immunosuppressive milieu. Immunity. 2001;14:715–725. doi: 10.1016/s1074-7613(01)00147-9. [DOI] [PubMed] [Google Scholar]
  17. Krysko DV, Kaczmarek A, Krysko O, Heyndrickx L, Woznicki J, Bogaert P, et al. TLR-2 and TLR-9 are sensors of apoptosis in a mouse model of doxorubicin-induced acute inflammation. Cell Death Differ. 2011;18:1316–1325. doi: 10.1038/cdd.2011.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Tesniere A, Schlemmer F, Boige V, Kepp O, Martins I, Ghiringhelli F, et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 2010;29:482–491. doi: 10.1038/onc.2009.356. [DOI] [PubMed] [Google Scholar]
  19. Panaretakis T, Kepp O, Brockmeier U, Tesniere A, Bjorklund AC, Chapman DC, et al. Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death. EMBO J. 2009;28:578–590. doi: 10.1038/emboj.2009.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Merritt RE, Mahtabifard A, Yamada RE, Crystal RG, Korst RJ. Cisplatin augments cytotoxic T-lymphocyte-mediated antitumor immunity in poorly immunogenic murine lung cancer. J Thorac Cardiovasc Surg. 2003;126:1609–1617. doi: 10.1016/s0022-5223(03)00707-4. [DOI] [PubMed] [Google Scholar]
  21. Obeid M, Panaretakis T, Joza N, Tufi R, Tesniere A, van Endert P, et al. Calreticulin exposure is required for the immunogenicity of gamma-irradiation and UVC light-induced apoptosis. Cell Death Differ. 2007;14:1848–1850. doi: 10.1038/sj.cdd.4402201. [DOI] [PubMed] [Google Scholar]
  22. Suzuki Y, Mimura K, Yoshimoto Y, Watanabe M, Ohkubo Y, Izawa S, et al. Immunogenic tumor cell death induced by chemoradiotherapy in patients with esophageal squamous cell carcinoma. Cancer Res. 2012;72:3967–3976. doi: 10.1158/0008-5472.CAN-12-0851. [DOI] [PubMed] [Google Scholar]
  23. Rubner Y, Wunderlich R, Ruhle PF, Kulzer L, Werthmoller N, Frey B, et al. How does ionizing irradiation contribute to the induction of anti-tumor immunity. Front Oncol. 2012;2:75. doi: 10.3389/fonc.2012.00075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Matzinger P. The danger model: a renewed sense of self. Science. 2002;296:301–305. doi: 10.1126/science.1071059. [DOI] [PubMed] [Google Scholar]
  25. Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12:991–1045. doi: 10.1146/annurev.iy.12.040194.005015. [DOI] [PubMed] [Google Scholar]
  26. Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2012;31:51–72. doi: 10.1146/annurev-immunol-032712-100008. [DOI] [PubMed] [Google Scholar]
  27. Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012;12:860–875. doi: 10.1038/nrc3380. [DOI] [PubMed] [Google Scholar]
  28. Dudek AM, Garg AD, Krysko DV, De Ruysscher D, Agostinis P. Inducers of immunogenic cancer cell death. Cytokine Growth Factor Rev. 2013. [DOI] [PubMed]
  29. Panaretakis T, Joza N, Modjtahedi N, Tesniere A, Vitale I, Durchschlag M, et al. The co-translocation of ERp57 and calreticulin determines the immunogenicity of cell death. Cell Death Differ. 2008;15:1499–1509. doi: 10.1038/cdd.2008.67. [DOI] [PubMed] [Google Scholar]
  30. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–1059. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
  31. Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med. 2009;15:1170–1178. doi: 10.1038/nm.2028. [DOI] [PubMed] [Google Scholar]
  32. Schiavoni G, Sistigu A, Valentini M, Mattei F, Sestili P, Spadaro F, et al. Cyclophosphamide synergizes with type I interferons through systemic dendritic cell reactivation and induction of immunogenic tumor apoptosis. Cancer Res. 2011;71:768–778. doi: 10.1158/0008-5472.CAN-10-2788. [DOI] [PubMed] [Google Scholar]
  33. Chen J, Xie J, Jiang Z, Wang B, Wang Y, Hu X. Shikonin and its analogs inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2. Oncogene. 2011;30:4297–4306. doi: 10.1038/onc.2011.137. [DOI] [PubMed] [Google Scholar]
  34. Yang H, Zhou P, Huang H, Chen D, Ma N, Cui QC, et al. Shikonin exerts antitumor activity via proteasome inhibition and cell death induction in vitro and in vivo. Int J Cancer. 2009;124:2450–2459. doi: 10.1002/ijc.24195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Spisek R, Charalambous A, Mazumder A, Vesole DH, Jagannath S, Dhodapkar MV. Bortezomib enhances dendritic cell (DC)-mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: therapeutic implications. Blood. 2007;109:4839–4845. doi: 10.1182/blood-2006-10-054221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Garrido G, Rabasa A, Sanchez B, Lopez MV, Blanco R, Lopez A, et al. Induction of immunogenic apoptosis by blockade of epidermal growth factor receptor activation with a specific antibody. J Immunol. 2011;187:4954–4966. doi: 10.4049/jimmunol.1003477. [DOI] [PubMed] [Google Scholar]
  37. Menger L, Vacchelli E, Adjemian S, Martins I, Ma Y, Shen S, et al. Cardiac glycosides exert anticancer effects by inducing immunogenic cell death. Sci Transl Med. 2012;4:143ra199. doi: 10.1126/scitranslmed.3003807. [DOI] [PubMed] [Google Scholar]
  38. Sonnemann J, Gressmann S, Becker S, Wittig S, Schmudde M, Beck JF. The histone deacetylase inhibitor vorinostat induces calreticulin exposure in childhood brain tumour cells in vitro. Cancer Chemother Pharmacol. 2010;66:611–616. doi: 10.1007/s00280-010-1302-4. [DOI] [PubMed] [Google Scholar]
  39. Han W, Li L, Qiu S, Lu Q, Pan Q, Gu Y, et al. Shikonin circumvents cancer drug resistance by induction of a necroptotic death. Mol Cancer Ther. 2007;6:1641–1649. doi: 10.1158/1535-7163.MCT-06-0511. [DOI] [PubMed] [Google Scholar]
  40. Galluzzi L, Kepp O, Kroemer G. Enlightening the impact of immunogenic cell death in photodynamic cancer therapy. EMBO J. 2012;31:1055–1057. doi: 10.1038/emboj.2012.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Verfaillie T, Rubio N, Garg AD, Bultynck G, Rizzuto R, Decuypere JP, et al. PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ. 2012;19:1880–1891. doi: 10.1038/cdd.2012.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–195. doi: 10.1038/nature00858. [DOI] [PubMed] [Google Scholar]
  43. Karioti A, Bilia AR. Hypericins as potential leads for new therapeutics. Int J Mol Sci. 2010;11:562–594. doi: 10.3390/ijms11020562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Zhang L, Wang A. Virus-induced ER stress and the unfolded protein response. Front Plant Sci. 2012;3:293. doi: 10.3389/fpls.2012.00293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Schwarz KB. Oxidative stress during viral infection: a review. Free Radic Biol Med. 1996;21:641–649. doi: 10.1016/0891-5849(96)00131-1. [DOI] [PubMed] [Google Scholar]
  46. Garg AD, Krysko DV, Vandenabeele P, Agostinis P. Hypericin-based photodynamic therapy induces surface exposure of damage-associated molecular patterns like HSP70 and calreticulin. Cancer Immunol Immunother. 2012;61:215–221. doi: 10.1007/s00262-011-1184-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Obeid M, Tesniere A, Panaretakis T, Tufi R, Joza N, van Endert P, et al. Ecto-calreticulin in immunogenic chemotherapy. Immunol Rev. 2007;220:22–34. doi: 10.1111/j.1600-065X.2007.00567.x. [DOI] [PubMed] [Google Scholar]
  48. Martin SJ, Reutelingsperger CP, McGahon AJ, Rader JA, van Schie RC, LaFace DM, et al. Early redistribution of plasma membrane phosphatidylserine is a general feature of apoptosis regardless of the initiating stimulus: inhibition by overexpression of Bcl-2 and Abl. J Exp Med. 1995;182:1545–1556. doi: 10.1084/jem.182.5.1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, et al. Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress. Oncogene. 2011;30:1147–1158. doi: 10.1038/onc.2010.500. [DOI] [PubMed] [Google Scholar]
  50. Garg AD, Nowis D, Golab J, Vandenabeele P, Krysko DV, Agostinis P. Immunogenic cell death, DAMPs and anticancer therapeutics: an emerging amalgamation. Biochim Biophys Acta. 2010;1805:53–71. doi: 10.1016/j.bbcan.2009.08.003. [DOI] [PubMed] [Google Scholar]
  51. Kazama H, Ricci JE, Herndon JM, Hoppe G, Green DR, Ferguson TA. Induction of immunological tolerance by apoptotic cells requires caspase-dependent oxidation of high-mobility group box-1 protein. Immunity. 2008;29:21–32. doi: 10.1016/j.immuni.2008.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Andersson U, Wang H, Palmblad K, Aveberger AC, Bloom O, Erlandsson-Harris H, et al. High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. J Exp Med. 2000;192:565–570. doi: 10.1084/jem.192.4.565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Tesniere A, Apetoh L, Ghiringhelli F, Joza N, Panaretakis T, Kepp O, et al. Immunogenic cancer cell death: a key–lock paradigm. Curr Opin Immunol. 2008;20:504–511. doi: 10.1016/j.coi.2008.05.007. [DOI] [PubMed] [Google Scholar]
  54. Chiba S, Baghdadi M, Akiba H, Yoshiyama H, Kinoshita I, Dosaka-Akita H, et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat Immunol. 2012;13:832–842. doi: 10.1038/ni.2376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Apetoh L, Ghiringhelli F, Tesniere A, Criollo A, Ortiz C, Lidereau R, et al. The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol Rev. 2007;220:47–59. doi: 10.1111/j.1600-065X.2007.00573.x. [DOI] [PubMed] [Google Scholar]
  56. Yang H, Hreggvidsdottir HS, Palmblad K, Wang H, Ochani M, Li J, et al. A critical cysteine is required for HMGB1 binding to Toll-like receptor 4 and activation of macrophage cytokine release. Proc Natl Acad Sci USA. 2010;107:11942–11947. doi: 10.1073/pnas.1003893107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Jube S, Rivera ZS, Bianchi ME, Powers A, Wang E, Pagano I, et al. Cancer cell secretion of the DAMP protein HMGB1 supports progression in malignant mesothelioma. Cancer Res. 2012;72:3290–3301. doi: 10.1158/0008-5472.CAN-11-3481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Yang GL, Zhang LH, Bo JJ, Huo XJ, Chen HG, Cao M, et al. Increased expression of HMGB1 is associated with poor prognosis in human bladder cancer. J Surg Oncol. 2012;106:57–61. doi: 10.1002/jso.23040. [DOI] [PubMed] [Google Scholar]
  59. Hoppe G, Talcott KE, Bhattacharya SK, Crabb JW, Sears JE. Molecular basis for the redox control of nuclear transport of the structural chromatin protein Hmgb1. Exp Cell Res. 2006;312:3526–3538. doi: 10.1016/j.yexcr.2006.07.020. [DOI] [PubMed] [Google Scholar]
  60. Venereau E, Casalgrandi M, Schiraldi M, Antoine DJ, Cattaneo A, De Marchis F, et al. Mutually exclusive redox forms of HMGB1 promote cell recruitment or proinflammatory cytokine release. J Exp Med. 2012;209:1519–1528. doi: 10.1084/jem.20120189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Yang H, Lundback P, Ottosson L, Erlandsson-Harris H, Venereau E, Bianchi ME, et al. Redox modification of cysteine residues regulates the cytokine activity of high mobility group box-1 (HMGB1) Mol Med. 2012;18:250–259. doi: 10.2119/molmed.2011.00389. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  62. Chaiswing L, Oberley TD. Extracellular/microenvironmental redox state. Antioxid Redox Signal. 2010;13:449–465. doi: 10.1089/ars.2009.3020. [DOI] [PubMed] [Google Scholar]
  63. Policastro LL, Ibanez IL, Notcovich C, Duran HA, Podhajcer OL. The tumor microenvironment: characterization, redox considerations, and novel approaches for reactive oxygen species-targeted gene therapy. Antioxid Redox Signal. 2012. [DOI] [PubMed]
  64. Guo ZS, Naik A, O'Malley ME, Popovic P, Demarco R, Hu Y, et al. The enhanced tumor selectivity of an oncolytic vaccinia lacking the host range and antiapoptosis genes SPI-1 and SPI-2. Cancer Res. 2005;65:9991–9998. doi: 10.1158/0008-5472.CAN-05-1630. [DOI] [PubMed] [Google Scholar]
  65. Diaconu I, Cerullo V, Hirvinen ML, Escutenaire S, Ugolini M, Pesonen SK, et al. Immune response is an important aspect of the antitumor effect produced by a CD40L-encoding oncolytic adenovirus. Cancer Res. 2012;72:2327–2338. doi: 10.1158/0008-5472.CAN-11-2975. [DOI] [PubMed] [Google Scholar]
  66. Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461:282–286. doi: 10.1038/nature08296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Martins I, Tesniere A, Kepp O, Michaud M, Schlemmer F, Senovilla L, et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle. 2009;8:3723–3728. doi: 10.4161/cc.8.22.10026. [DOI] [PubMed] [Google Scholar]
  68. Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573–1577. doi: 10.1126/science.1208347. [DOI] [PubMed] [Google Scholar]
  69. Ohshima Y, Tsukimoto M, Takenouchi T, Harada H, Suzuki A, Sato M, et al. Gamma-irradiation induces P2X(7) receptor-dependent ATP release from B16 melanoma cells. Biochim Biophys Acta. 2010;1800:40–46. doi: 10.1016/j.bbagen.2009.10.008. [DOI] [PubMed] [Google Scholar]
  70. Petrovski G, Ayna G, Majai G, Hodrea J, Benko S, Madi A, et al. Phagocytosis of cells dying through autophagy induces inflammasome activation and IL-1beta release in human macrophages. Autophagy. 2011;7:321–330. doi: 10.4161/auto.7.3.14583. [DOI] [PubMed] [Google Scholar]
  71. Chang CW, Li HC, Hsu CF, Chang CY, Lo SY. Increased ATP generation in the host cell is required for efficient vaccinia virus production. J Biomed Sci. 2009;16:80. doi: 10.1186/1423-0127-16-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Seror C, Melki MT, Subra F, Raza SQ, Bras M, Saidi H, et al. Extracellular ATP acts on P2Y2 purinergic receptors to facilitate HIV-1 infection. J Exp Med. 2011;208:1823–1834. doi: 10.1084/jem.20101805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 2007;8:741–752. doi: 10.1038/nrm2239. [DOI] [PubMed] [Google Scholar]
  74. Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res. 2007;13:7271–7279. doi: 10.1158/1078-0432.CCR-07-1595. [DOI] [PubMed] [Google Scholar]
  75. Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science. 2000;290:1717–1721. doi: 10.1126/science.290.5497.1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Eisenberg-Lerner A, Bialik S, Simon HU, Kimchi A. Life and death partners: apoptosis, autophagy and the cross-talk between them. Cell Death Differ. 2009;16:966–975. doi: 10.1038/cdd.2009.33. [DOI] [PubMed] [Google Scholar]
  77. Kroemer G, Levine B. Autophagic cell death: the story of a misnomer. Nat Rev Mol Cell Biol. 2008;9:1004–1010. doi: 10.1038/nrm2527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Qu X, Zou Z, Sun Q, Luby-Phelps K, Cheng P, Hogan RN, et al. Autophagy gene-dependent clearance of apoptotic cells during embryonic development. Cell. 2007;128:931–946. doi: 10.1016/j.cell.2006.12.044. [DOI] [PubMed] [Google Scholar]
  79. Martins I, Michaud M, Sukkurwala AQ, Adjemian S, Ma Y, Shen S, et al. Premortem autophagy determines the immunogenicity of chemotherapy-induced cancer cell death. Autophagy. 2012;8:413–415. doi: 10.4161/auto.19009. [DOI] [PubMed] [Google Scholar]
  80. Townsend KN, Hughson LR, Schlie K, Poon VI, Westerback A, Lum JJ. Autophagy inhibition in cancer therapy: metabolic considerations for antitumor immunity. Immunol Rev. 2012;249:176–194. doi: 10.1111/j.1600-065X.2012.01141.x. [DOI] [PubMed] [Google Scholar]
  81. Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, et al. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest. 2007;117:326–336. doi: 10.1172/JCI28833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Yang ZJ, Chee CE, Huang S, Sinicrope FA. The role of autophagy in cancer: therapeutic implications. Mol Cancer Ther. 2011;10:1533–1541. doi: 10.1158/1535-7163.MCT-11-0047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Proskuryakov SY, Konoplyannikov AG, Gabai VL. Necrosis: a specific form of programmed cell death. Exp Cell Res. 2003;283:1–16. doi: 10.1016/s0014-4827(02)00027-7. [DOI] [PubMed] [Google Scholar]
  84. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972;26:239–257. doi: 10.1038/bjc.1972.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Vandenabeele P, Declercq W, Van Herreweghe F, Vanden Berghe T. The role of the kinases RIP1 and RIP3 in TNF-induced necrosis. Sci Signal. 2010;3:re4. doi: 10.1126/scisignal.3115re4. [DOI] [PubMed] [Google Scholar]
  86. Wyllie AH, Kerr JF, Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol. 1980;68:251–306. doi: 10.1016/s0074-7696(08)62312-8. [DOI] [PubMed] [Google Scholar]
  87. Van Herreweghe F, Festjens N, Declercq W, Vandenabeele P. Tumor necrosis factor-mediated cell death: to break or to burst, that's the question. Cell Mol Life Sci. 2010;67:1567–1579. doi: 10.1007/s00018-010-0283-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Festjens N, Vanden Berghe T, Vandenabeele P. Necrosis, a well-orchestrated form of cell demise: signalling cascades, important mediators and concomitant immune response. Biochim Biophys Acta. 2006;1757:1371–1387. doi: 10.1016/j.bbabio.2006.06.014. [DOI] [PubMed] [Google Scholar]
  89. Golstein P, Kroemer G. Cell death by necrosis: towards a molecular definition. Trends Biochem Sci. 2007;32:37–43. doi: 10.1016/j.tibs.2006.11.001. [DOI] [PubMed] [Google Scholar]
  90. Zhang DW, Shao J, Lin J, Zhang N, Lu BJ, Lin SC, et al. RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science. 2009;325:332–336. doi: 10.1126/science.1172308. [DOI] [PubMed] [Google Scholar]
  91. Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, et al. Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol. 2000;1:489–495. doi: 10.1038/82732. [DOI] [PubMed] [Google Scholar]
  92. Kamata H, Honda S, Maeda S, Chang L, Hirata H, Karin M. Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell. 2005;120:649–661. doi: 10.1016/j.cell.2004.12.041. [DOI] [PubMed] [Google Scholar]
  93. Zong WX, Ditsworth D, Bauer DE, Wang ZQ, Thompson CB. Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev. 2004;18:1272–1282. doi: 10.1101/gad.1199904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Skulachev VP. Bioenergetic aspects of apoptosis, necrosis and mitoptosis. Apoptosis. 2006;11:473–485. doi: 10.1007/s10495-006-5881-9. [DOI] [PubMed] [Google Scholar]
  95. Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen L, Iovino F, et al. Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4. Cell Stem Cell. 2007;1:389–402. doi: 10.1016/j.stem.2007.08.001. [DOI] [PubMed] [Google Scholar]
  96. Madjd Z, Mehrjerdi AZ, Sharifi AM, Molanaei S, Shahzadi SZ, Asadi-Lari M. CD44+ cancer cells express higher levels of the anti-apoptotic protein Bcl-2 in breast tumours. Cancer Immun. 2009;9:4. [PMC free article] [PubMed] [Google Scholar]
  97. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7:99–109. doi: 10.1038/nrmicro2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Frantz S, Ducharme A, Sawyer D, Rohde LE, Kobzik L, Fukazawa R, et al. Targeted deletion of caspase-1 reduces early mortality and left ventricular dilatation following myocardial infarction. J Mol Cell Cardiol. 2003;35:685–694. doi: 10.1016/s0022-2828(03)00113-5. [DOI] [PubMed] [Google Scholar]
  99. Colunga AG, Laing JM, Aurelian L. The HSV-2 mutant DeltaPK induces melanoma oncolysis through nonredundant death programs and associated with autophagy and pyroptosis proteins. Gene Ther. 2010;17:315–327. doi: 10.1038/gt.2009.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Wang Q, Imamura R, Motani K, Kushiyama H, Nagata S, Suda T. Pyroptotic cells externalize eat-me and release find-me signals and are efficiently engulfed by macrophages. Int Immunol. 2013;25:363–372. doi: 10.1093/intimm/dxs161. [DOI] [PubMed] [Google Scholar]
  101. Bridle BW, Stephenson KB, Boudreau JE, Koshy S, Kazdhan N, Pullenayegum E, et al. Potentiating cancer immunotherapy using an oncolytic virus. Mol Ther. 2010;18:1430–1439. doi: 10.1038/mt.2010.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Galluzzi L, Brenner C, Morselli E, Touat Z, Kroemer G. Viral control of mitochondrial apoptosis. PLoS Pathogen. 2008;4:e1000018. doi: 10.1371/journal.ppat.1000018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature. 2011;469:323–335. doi: 10.1038/nature09782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Meng X, Nakamura T, Okazaki T, Inoue H, Takahashi A, Miyamoto S, et al. Enhanced antitumor effects of an engineered measles virus Edmonston strain expressing the wild-type N, P, L genes on human renal cell carcinoma. Mol Ther. 2010;18:544–551. doi: 10.1038/mt.2009.296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Hasei J, Sasaki T, Tazawa H, Osaki S, Yamakawa Y, Kunisada T, et al. Dual programmed cell death pathways induced by p53 transactivation overcome resistance to oncolytic adenovirus in human osteosarcoma cells. Mol Cancer Ther. 2013;12:314–325. doi: 10.1158/1535-7163.MCT-12-0869. [DOI] [PubMed] [Google Scholar]
  106. Moussavi M, Fazli L, Tearle H, Guo Y, Cox M, Bell J, et al. Oncolysis of prostate cancers induced by vesicular stomatitis virus in PTEN knockout mice. Cancer Res. 2010;70:1367–1376. doi: 10.1158/0008-5472.CAN-09-2377. [DOI] [PubMed] [Google Scholar]
  107. Kirn D, Martuza RL, Zwiebel J. Replication-selective virotherapy for cancer: biological principles, risk management and future directions. Nat Med. 2001;7:781–787. doi: 10.1038/89901. [DOI] [PubMed] [Google Scholar]
  108. Prestwich RJ, Harrington KJ, Pandha HS, Vile RG, Melcher AA, Errington F. Oncolytic viruses: a novel form of immunotherapy. Expert Rev Anticancer Ther. 2008;8:1581–1588. doi: 10.1586/14737140.8.10.1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Kepp O, Senovilla L, Galluzzi L, Panaretakis T, Tesniere A, Schlemmer F, et al. Viral subversion of immunogenic cell death. Cell Cycle. 2009;8:860–869. doi: 10.4161/cc.8.6.7939. [DOI] [PubMed] [Google Scholar]
  110. Zhang Y, Chirmule N, Gao GP, Qian R, Croyle M, Joshi B, et al. Acute cytokine response to systemic adenoviral vectors in mice is mediated by dendritic cells and macrophages. Mol Ther. 2001;3 (Part 1:697–707. doi: 10.1006/mthe.2001.0329. [DOI] [PubMed] [Google Scholar]
  111. Benencia F, Courreges MC, Conejo-Garcia JR, Mohamed-Hadley A, Zhang L, Buckanovich RJ, et al. HSV oncolytic therapy upregulates interferon-inducible chemokines and recruits immune effector cells in ovarian cancer. Mol Ther. 2005;12:789–802. doi: 10.1016/j.ymthe.2005.03.026. [DOI] [PubMed] [Google Scholar]
  112. Vicari AP, Chiodoni C, Vaure C, Ait-Yahia S, Dercamp C, Matsos F, et al. Reversal of tumor-induced dendritic cell paralysis by CpG immunostimulatory oligonucleotide and anti-interleukin 10 receptor antibody. J Exp Med. 2002;196:541–549. doi: 10.1084/jem.20020732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Errington F, Steele L, Prestwich R, Harrington KJ, Pandha HS, Vidal L, et al. Reovirus activates human dendritic cells to promote innate antitumor immunity. J Immunol. 2008;180:6018–6026. doi: 10.4049/jimmunol.180.9.6018. [DOI] [PubMed] [Google Scholar]
  114. Donnelly OG, Errington-Mais F, Steele L, Hadac E, Jennings V, Scott K, et al. Measles virus causes immunogenic cell death in human melanoma. Gene Ther. 2013;20:7–15. doi: 10.1038/gt.2011.205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Huang B, Sikorski R, Kirn DH, Thorne SH. Synergistic anti-tumor effects between oncolytic vaccinia virus and paclitaxel are mediated by the IFN response and HMGB1. Gene Ther. 2011;18:164–172. doi: 10.1038/gt.2010.121. [DOI] [PubMed] [Google Scholar]
  116. Borde C, Barnay-Verdier S, Gaillard C, Hocini H, Marechal V, Gozlan J. Stepwise release of biologically active HMGB1 during HSV-2 infection. PLoS One. 2011;6:e16145. doi: 10.1371/journal.pone.0016145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Moisy D, Avilov SV, Jacob Y, Laoide BM, Ge X, Baudin F, et al. HMGB1 protein binds to influenza virus nucleoprotein and promotes viral replication. J Virol. 2012;86:9122–9133. doi: 10.1128/JVI.00789-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Zhang HM, Ye X, Su Y, Yuan J, Liu Z, Stein DA, et al. Coxsackievirus B3 infection activates the unfolded protein response and induces apoptosis through downregulation of p58IPK and activation of CHOP and SREBP1. J Virol. 2010;84:8446–8459. doi: 10.1128/JVI.01416-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Chau DH, Yuan J, Zhang H, Cheung P, Lim T, Liu Z, et al. Coxsackievirus B3 proteases 2A and 3C induce apoptotic cell death through mitochondrial injury and cleavage of eIF4GI but not DAP5/p97/NAT1. Apoptosis. 2007;12:513–524. doi: 10.1007/s10495-006-0013-0. [DOI] [PubMed] [Google Scholar]
  120. Ito H, Aoki H, Kuhnel F, Kondo Y, Kubicka S, Wirth T, et al. Autophagic cell death of malignant glioma cells induced by a conditionally replicating adenovirus. J Natl Cancer Inst. 2006;98:625–636. doi: 10.1093/jnci/djj161. [DOI] [PubMed] [Google Scholar]
  121. Jiang H, Gomez-Manzano C, Aoki H, Alonso MM, Kondo S, McCormick F, et al. Examination of the therapeutic potential of Delta-24-RGD in brain tumor stem cells: role of autophagic cell death. J Natl Cancer Inst. 2007;99:1410–1414. doi: 10.1093/jnci/djm102. [DOI] [PubMed] [Google Scholar]
  122. Tazawa H, Yano S, Yoshida R, Yamasaki Y, Sasaki T, Hashimoto Y, et al. Genetically engineered oncolytic adenovirus induces autophagic cell death through an E2F1–microRNA-7–epidermal growth factor receptor axis. Int J Cancer. 2012;131:2939–2950. doi: 10.1002/ijc.27589. [DOI] [PubMed] [Google Scholar]
  123. Meng C, Zhou Z, Jiang K, Yu S, Jia L, Wu Y, et al. Newcastle disease virus triggers autophagy in U251 glioma cells to enhance virus replication. Arch Virol. 2012;157:1011–1018. doi: 10.1007/s00705-012-1270-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Thirukkumaran CM, Shi ZQ, Luider J, Kopciuk K, Gao H, Bahlis N, et al. Reovirus modulates autophagy during oncolysis of multiple myeloma. Autophagy. 2013;9:413–414. doi: 10.4161/auto.22867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Hu X, Xuan Y. Bypassing cancer drug resistance by activating multiple death pathways–a proposal from the study of circumventing cancer drug resistance by induction of necroptosis. Cancer Lett. 2008;259:127–137. doi: 10.1016/j.canlet.2007.11.007. [DOI] [PubMed] [Google Scholar]
  126. Huang H, Xiao T, He L, Ji H, Liu XY. Interferon-beta-armed oncolytic adenovirus induces both apoptosis and necroptosis in cancer cells. Acta Biochim Biophys Sin (Shanghai) 2012;44:737–745. doi: 10.1093/abbs/gms060. [DOI] [PubMed] [Google Scholar]
  127. Jablonska J, Leschner S, Westphal K, Lienenklaus S, Weiss S. Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J Clin Invest. 2010;120:1151–1164. doi: 10.1172/JCI37223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Tough DF, Borrow P, Sprent J. Induction of bystander T cell proliferation by viruses and type I interferon in vivo. Science. 1996;272:1947–1950. doi: 10.1126/science.272.5270.1947. [DOI] [PubMed] [Google Scholar]
  129. Qin XQ, Beckham C, Brown JL, Lukashev M, Barsoum J. Human and mouse IFN-beta gene therapy exhibits different anti-tumor mechanisms in mouse models. Mol Ther. 2001;4:356–364. doi: 10.1006/mthe.2001.0464. [DOI] [PubMed] [Google Scholar]
  130. Chiocca EA. The host response to cancer virotherapy. Curr Opin Mol Ther. 2008;10:38–45. [PubMed] [Google Scholar]
  131. Melcher A, Parato K, Rooney CM, Bell JC. Thunder and lightning: immunotherapy and oncolytic viruses collide. Mol Ther. 2011;19:1008–1016. doi: 10.1038/mt.2011.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Kottke T, Diaz RM, Kaluza K, Pulido J, Galivo F, Wongthida P, et al. Use of biological therapy to enhance both virotherapy and adoptive T-cell therapy for cancer. Mol Ther. 2008;16:1910–1918. doi: 10.1038/mt.2008.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Garg AD, Krysko DV, Vandenabeele P, Agostinis P. DAMPs and PDT-mediated photo-oxidative stress: exploring the unknown. Photochem Photobiol Sci. 2011;10:670–680. doi: 10.1039/c0pp00294a. [DOI] [PubMed] [Google Scholar]
  134. Garg AD, Krysko DV, Vandenabeele P, Agostinis P. The emergence of phox-ER stress induced immunogenic apoptosis. Oncoimmunology. 2012;1:786–788. doi: 10.4161/onci.19750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Shen F, Chu S, Bence AK, Bailey B, Xue X, Erickson PA, et al. Quantitation of doxorubicin uptake, efflux, and modulation of multidrug resistance (MDR) in MDR human cancer cells. J Pharmacol Exp Ther. 2008;324:95–102. doi: 10.1124/jpet.107.127704. [DOI] [PubMed] [Google Scholar]
  136. Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012;12:298–306. doi: 10.1038/nrc3245. [DOI] [PubMed] [Google Scholar]
  137. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  139. Kanai R, Wakimoto H, Martuza RL, Rabkin SD. A novel oncolytic herpes simplex virus that synergizes with phosphoinositide 3-kinase/Akt pathway inhibitors to target glioblastoma stem cells. Clin Cancer Res. 2011;17:3686–3696. doi: 10.1158/1078-0432.CCR-10-3142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Cripe TP, Wang PY, Marcato P, Mahller YY, Lee PW. Targeting cancer-initiating cells with oncolytic viruses. Mol Ther. 2009;17:1677–1682. doi: 10.1038/mt.2009.193. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cell Death and Differentiation are provided here courtesy of Nature Publishing Group

RESOURCES