v
Search
Advanced Search

Publications > Journals > Cancer Screening and Prevention > Article Full Text

  • OPEN ACCESS

Leveraging Immunogenic Cell Death in Pancreatic Cancer

  • Cong He1,2,#,
  • Yan Luo1,2,#,
  • Xin-Long Wang1,2,
  • Can Zhang1,2 and
  • Bei Sun1,2,* 
 Author information
Cancer Screening and Prevention   2023;2(2):108-116

doi: 10.14218/CSP.2023.00015

Abstract

Pancreatic cancer remains the most malignant type of cancer. Although immunotherapy has shown good efficacy in most solid tumors, it has a poor response in pancreatic cancer. The difficulty in applying immunotherapy to pancreatic cancer lies in the fact that it is an immunologically “cold” tumor. The dense and poorly vascularized microenvironment of pancreatic cancer is not conducive to the infiltration of immune cells, making it difficult to trigger an immune response. Immunogenic cell death is a specific type of cell death that can initiate an adaptive immune response and turn a “cold” tumor into a “hot” one. In this review, we discuss the basis of immunogenic cell death and its inducers in pancreatic ductal adenocarcinoma. The key point is that the manipulation of adjuvant immunogenic cell death inducers can usually synergize with other immunotherapies. This synergetic effect allows for the combination therapy to fully utilize the immune potential of each component, overcome the immune-suppressed microenvironment of pancreatic cancer, and ultimately activate immune responses that result in tumor regression. Clinical trials of therapy based on this strategy may offer hope to patients with pancreatic cancer.

Keywords

Pancreatic cancer, Immunogenic cell death, Damage-associated molecular patterns, Immunology, Immunotherapy

Introduction

Pancreatic ductal adenocarcinoma (PDAC) remains the most malignant type of cancer with a 5-year survival rate of only 11%.1 It is projected that PDAC will become the second leading cause of mortality in the United States by 2030.2 Radical surgery and adjuvant chemotherapy can gain long-term survival, but most patients have already lost the opportunity for surgery when they are first diagnosed.3,4 For most patients with locally advanced or metastatic disease, the standard treatment involves a combination of cytotoxic chemotherapy and supportive care. Unfortunately, such combination therapies come at the cost of significant side effects, while overall response rates are limited.5,6 Therefore, it is important to explore new therapeutic approaches to effectively treat PDAC.

Immunotherapy (i.e. immune checkpoint inhibitors) has achieved great success for an impressive breadth of solid tumors; however, PDAC is an exceptional case due to its distinctive histopathological features.7,8 The tumor mutational burden of pancreatic cancer is generally lower compared to other types of cancer, which in turn leads to minor antigenicity.9 The unique characteristics of the tumor microenvironment (TME) in PDAC can pose challenges for effective immune response. The dense extracellular matrix and poorly formed vascular system can limit the infiltration of immune cells. Even if a small number of immune cells do manage to infiltrate, they may not be able to function properly due to the local environment. These factors contribute to the “cold” or “immune-excluded” landscape of the TME, which is not capable of initiating a strong immune response on its own.10 It is important to continue exploring innovative approaches to address these challenges and develop effective treatments for pancreatic cancer.

Immunogenic cell death (ICD) is a variant of programmed cell death that emits damage-associated molecular patterns (DAMPs) and other signals, thereby activating the immune reaction. Mechanically, ICD provides plenty of corpses which can be identified as TNA/TSA to compensate for the low immunogenicity of pancreatic cancer. On the other hand, danger signals emitted by ICD can mobilize members of the immune system and thereby remodulate the so-called “immune-excluded” TME of pancreatic cancer to an immune-reactive state, ultimately initiating the antitumor immune response and leading to tumor regression. Indeed, part of traditional PDAC therapy has been demonstrated to exert antitumor effects through ICD. Understanding ICD also leads to the exploitation of considerable novel agents for the immunotherapy of PDAC. Hence, inducing immunogenic cell death appears to be a feasible strategy and may shed light on immunochemotherapy for PDAC.11,12

We searched MEDLINE and PubMed databases for relevant articles and reviews published from January 1, 2005, to April 31, 2023, with combinations of the following keywords: (“immunogenic cell death” OR “ICD”) AND (“PDAC” OR “pancreatic ductal adenocarcinoma” OR “pancreatic” OR “pancreas”). We also considered the reference lists of all included articles and of previous related reviews.

Paradigm of ICD

In the past decades, researchers have classified cell death into two different types: apoptosis is thought to be physiological and immuno-tolerogenic, whereas necrosis is pathological and immunogenic. The notion of ICD was first proposed by Casares et al. in 2005, identifying ICD as a variant of apoptosis that can generate cancer vaccines and stimulate antineoplastic immune responses in vivo.13 With deeper investigation, ICD has been recognized as an independent form of programmed cell death. In 2018, the Nomenclature Committee on Cell Death defined ICD as ‘a functionally peculiar form of RCD that is sufficient to activate an adaptive immune response specific for endogenous (cellular) or exogenous (viral) antigens expressed by dying cells’,14 emphasizing the immunogenicity of ICD.

A range of factors can trigger the initiation of ICD, such as viral infection, stress, therapeutic drugs (e.g. oxaliplatin, irinotecan), specific forms of radiation therapy, and some physical therapies. Certain inducers may trigger endoplasmic reticulum stress (ERS), which can raise the production of reactive oxygen species (ROS),15 while others may initially cause DNA or chromatin damage, followed by ERS. ERS subsequently generates host-derived, immune-activating molecules, such as DAMPs.16 DAMPs act as a signal of danger that can be detected by pattern recognition receptors (PRRs) present on the surface of innate immune cells. They cooperate with the new antigen presented or exposed from the cell corpse and ultimately cause an adaptive immune response. Eventually, the matured dendritic cells (DCs) recruit effective T cells, leading to long-term immunologic memory (Fig. 1).17 Indeed, the actual event of ICD can vary widely from the ideal pattern. However, the basic principles apply to any situation.

The molecular mechanism of ICD.
Fig. 1  The molecular mechanism of ICD.

Dying cancer cells succumbing to ICD emit a series of DAMPs in a temporal and spatial expression pattern. TNA/TAA released from dying cells is engulfed by immature DCs. Thereafter, TNAs/TAAs processing proceeds in mature DCs and are presented to CD8+ T cells, generating CTLs for tumor-specific immune responses. These processes ultimately lead to the eradication of tumor cells and allow for long-term immunological memory. ATP, adenosine triphosphate; CALR, calreticulin; CTL, cytotoxic T lymphocyte; DAMPs, damage-associated molecular patterns; DC, dendritic cell; HMGB1, high-mobility group box 1; ICD, immunogenic cell death; MHC, major histocompatibility complex; P2RY2, purinergic P2Y receptor 2; TAA, tumor-associated antigen; TLR4, toll-like receptor 4; TNA, tumor neoantigens.

Prerequisites of ICD: antigenicity and adjuvanticity

Immunogenic cell death requires antigenicity and adjuvanticity.18 The premise for mature T cells to recognize antigens is that they must be correctly presented through MHC-II molecules and different from those normally presented on the MHC-I. Tumor cells naturally harbor tumor-associated antigens (TAA) and tumor neoantigens (TNA) that are different from normal peptides.19,20 The release or presentation of TAA and TNA when the structure of dying cells collapses (e.g. treated with certain chemotherapies) provides the antigenicity required for ICD. However, the complexity and arbitrary nature of peptide chain structures leads to complexity in antigenicity. In contrast, adjuvanticity is completely defined by DAMPs, greatly simplifying its molecular characterization.21 Early release of ATP during ICD binds to P2RY2 (purinergic P2Y receptor 2) on the surface of DCs or macrophages, emitting a “find me” signal, followed by flipping of calreticulin (CALR) to the cell surface, emitting an “eat me” signal. In addition, high-mobility group box 1 (HMGB1) ultimately promotes DC maturation and antigen presentation. The temporal and spatial release of these DAMPs is orchestrated, ultimately leading to the recruitment of effector T cells.22 Memory T cells are subsequently produced and rapidly respond upon the second activation. Compared to complex peptide chain molecules, the adjuvanticity characterized by these clear immune small molecules is more manipulable and druggable.16

DAMPs of ICD

Molecules that are released by cells in response to various stresses are known as DAMPs. These molecules act as signals to activate the immune system, which then targets and eliminates damaged cells. DAMPs are recognized by innate immune cells through a variety of PRRs, such as Toll-like receptors (TLRs) and NOD-like receptors (NLRs). This recognition leads to the activation of downstream signaling pathways, including the NF-κB and MAPK pathways, which trigger the immune response. The three canonical DAMPs are CALR, extracellular ATP, and HMGB1, and they are thought to be biological markers for ICD.23 As research deepens, more and more DAMPs are gradually being discovered. This has improved our understanding of ICD and provided more targets for manipulating ICD.

CALR

During the early stages of ICD, CALR, an endoplasmic reticulum (ER) chaperone, is translocated from the lumen of the ER to the plasma membrane. This exposure of CALR acts as an “eat-me” signal, aiding in the engulfment of dying cells or their remnants by DCs or their precursors. This process provides these cells with a plentiful supply of antigenic material.24,25 The process of CALR translocation involves a series of events. Firstly, eIF2α is phosphorylated, leading to the inhibition of protein translation. Secondly, caspase 8 is activated, which triggers the cleavage of BCAP31. This results in the aggregation of pro-apoptotic Bcl-2 family members, BAX and BAK1, at the outer mitochondrial membrane. Lastly, the anterograde transport of CALR to the Golgi apparatus is facilitated by VAMP1 and SNAP25.26 CALR usually translocates to the cell surface along with ERp57, which is a member of the protein disulfide isomerase family A. This occurs in most cases of ICD.27

CALR functions by binding to LDL-receptor-related protein 1, also known as CD91, which is the PRR expressed by antigen-presenting cells such as DCs and serves as the main ER chaperone sensor.28,29 A mechanism that relies on BTK is involved in CD91 ligation-induced engulfment of cellular corpses.30,31 The ability of dying cancer cells to establish protective immunity is dependent on the key role of CALR in the immunogenicity of RCD. This can be disrupted through RNAi-mediated knockdown, CRISPR/Cas9-mediated deletion, or neutralizing antibody-mediated blockade of CALR. Conversely, exogenous recombinant CALR can enable the other variant of RCD to provoke protective immunity.30,32 Recently, there have been studies demonstrating that CALR can also enhance innate immunity by improving interleukin (IL)15 trans-presentation to natural killer (NK) cells. This broadens the understanding of CALR’s immunogenicity.33,34

HMGB1

HMGB1 is a protein that is abundant in the nucleus and does not belong to the histone family. During ICD in dying cells, HMGB1 is released. This release happens when both the nuclear lamina and the plasma membrane become permeable.35,36 The precise molecular mechanisms still require further clarification. HMGB1 exerts strong pro-inflammatory effects through its interaction with TLR2, TLR4, and AGER (also known as RAGE), leading to its activation.37 The binding of HGMB1 to TLR4 and RAGE receptors on DCs promotes antigen presentation and induces DC maturation.38,39 Preclinical in vivo models have shown that when cancer cells are deprived of HMGB1 or when the host is deficient in TLR4, cell death is not considered immunogenic according.40,41 It is worth mentioning that the immunogenic characteristics of HMGB1 rely on its redox condition. Studies have demonstrated that the immunostimulatory function of HMGB1 is limited by the caspase-induced generation of ROS, which leads to the oxidation of HMGB1.42 It is possible that the bidirectionality of HMGB1 could contribute to the observed paradoxical effect of HMGB1 on nucleic acid-mediated antitumor activity in mice. However, further research is needed to fully understand the mechanisms behind this phenomenon.43

ATP

ATP is released via pannexin channels in an autophagy-dependent manner through active exocytosis of ATP-containing vesicles during ICD.44,45 By binding to the purinergic receptor P2Y2 (P2RY2), extracellular ATP functions as a significant “find-me” signal for DC precursors and macrophages. This process helps in the recruitment of myeloid cells to the locations where there is active ICD.46,47 Extracellular ATP triggers pro-inflammatory effects by activating the NLRP3 inflammasome through CASP1, leading to the secretion of mature IL1B and IL18. IL1β is best known for its role in inflammation.48,49 Activation of CD8+ T cells and IL17-producing γδ T cells is the result of the effects originating from the P2X7 receptor, which is an inotropic receptor known as purinergic receptor P2X7 (P2RX7).48 The immunogenicity of cell death is dependent on events related to immune responses driven by ICD. This includes the accumulation of ATP in the microenvironment of dying cancer cells and the presence of P2RX7 or P2RY2 in the myeloid compartment of the host.

However, the immunogenicity of cell death can be reduced if ATP fails to accumulate or if P2RX7 or P2RY2 are absent. CD39 and CD73, which are the ectoenzymes that degrade ATP, can also exert immunosuppressive effects when overexpressed in malignant tissues.50–52 In breast cancer patients who receive anthracycline-based chemotherapy, loss-of-function polymorphisms in P2RX7 have been linked to unfavorable clinical outcomes.53 It is notable that the release of ATP during cell death is similar to the release of HGMB1 and can also occur during non-immunogenic forms of cell death. However, in vitro studies have shown that normalizing extracellular ATP levels to the percentage of dead cells or to the ATP plateau that can be achieved with a detergent can help distinguish between immunogenic and non-immunogenic forms of cell death.

Other DAMPs of ICD

Interferon (IFN)-1 is a typical DAMP molecule. Type I IFN production is a result of cells undergoing ICD, which is triggered by the detection of endogenous RNA and DNA.53–55 The immunogenicity of ICD is significantly reduced through the enzymatic degradation of extracellular nucleic acids, in line with this idea.43,56 The former is recognized by the endosomal TLR3,5,57 whereas the latter binds with cytosolic cyclic GMP-AMP synthase (cGAS), which subsequently initiates the cGAS-STING signaling pathway.58–60 The importance of cGAS-STING signaling in the immunogenicity of anthracyclines and radiation therapy has been demonstrated through studies on mice. Defects in key components of the cGAS-STING signaling pathway or cotreatment with IFNAR1-blocking antibodies have been shown to decrease therapeutic efficacy.61,62 The broad immunostimulatory effects of IFN-1 are due to its ability to interact with immune cells expressing IFNAR1.63 It has been reported that IFN-I can boost the cytotoxic capabilities of CD8+ T cells and NK cells,64 as well as facilitate cross-priming through DCs.65,66 In addition, macrophages can be stimulated to secrete pro-inflammatory mediators by type I IFN, while also inhibiting the immunosuppressive functions of regulatory T cells expressing CD4+CD25+FOXP3+.67 Malignant cells have been found to express a range of IFN-stimulated genes (ISGs) upon activation by IFN-I. This includes the expression of C-X-C motif chemokine ligand 10 (CXCL10), which acts as a chemoattractant for T cells.5

When cells undergo ICD they release various biomolecules that can trigger a pro-inflammatory response when they interact with DCs. One such biomolecule is heat shock protein, which can bind to CD91 and promote the maturation of DCs and uptake of dying cells, as previously discussed in the research on CALR.68 ANXA1 is a regulatory protein that plays a crucial role in maintaining homeostasis. It facilitates the uptake of tumor antigens by the TME when it is released by dying cells. This function is essential for the effective response to anthracycline-based chemotherapy in vivo, as malignant cells lacking ANXA1 exhibit limited sensitivity to this treatment.69 In a recent study, histones were identified as DAMPs in PDAC that are targeted by diaciclib.70 The supernatant of dinaciclib-treated KPC cell culture directly induced an increase in the expression of CD80 or CD86 on the DC cell surface, and this effect could be blocked by histone-neutralizing antibodies rather than HMGB1 antibodies.

Inducing ICD in PDAC

ICD can be induced in various manners, mainly including three categories in PDAC: 1) therapeutic drugs: including chemotherapeutics, targeted drugs, and immunomodulatory drugs; 2) biological molecules: oncolytic viruses, oncolytic peptides,71 and some biological immune regulatory factors; 3) physical therapy: stereotactic body radiotherapy (SBRT)72 and photodynamic therapy (PDT) (Table 1).73 Most of these inducers have shown profound effects on the tumor immune microenvironment in preclinical PDAC tumor models and have been confirmed to have a killing effect on tumors in various ways. There has been a growing trend in recent years towards the use of combination therapy regimens, which involve not only the combination of different ICD inducers, but also the use of ICD inducers in conjunction with other medications.74,75 The development of nanocarrier technology has enabled the use of multidrug combinations and precise pharmacokinetic control. In fact, many of the combination therapies mentioned below have used nanocarrier platforms. Combination strategies often offer more promising prospects compared to monotherapy. This leads to the following insight: in the immuno-incompetent microenvironment of pancreatic cancer, coordinated activation of multiple adaptive immune nodes and the accumulation of potential immune driving forces may be required to ultimately surpass the critical threshold to reach the trigger point (Fig. 2).

Table 1

Emerging ICD inducers and their combination in PDAC

AgentMechanismRef.
Silicasome-carried platinum or OXAIncrease the release of CALR and HMGB177
Irinotecan + PDL1Neutralize acidic pH values, trigger autophagy, enhance the release perforin and granzyme B78
Irinotecan + 3M-052Mediating dendritic cell activation by activate toll-like receptor (TLR) 7/879
DiaciclibInhabit CDK1/2/5,release the histone H3/4 as DAMPs70
ACUPA/TPP-I3AInduce the release of DAMPs, upregulate CD80 and CD86 expression on DCs90
OV OBP-702Enhance secretion of extracellular ATP and HMGB1 by inducing p53-mediated apoptosis and autophagy80
Oncolytic peptide LTX-315Inhibit PD-L1 expression and enhanced lymphocyte infiltration81
TT856-1313Promote the lymph node–like structures close contact with pancreatic tumors, enhance CD4 T cells infiltration82
oHSV + CD40LIncrease the infiltration of DC and CD8 T cells and decrease the infiltration of Treg cells83
Radiotherapy/SBRTLocal release of cytokines and danger signals by dying cells, alter cytokine secretion by surviving radioresistant cells84,86
SBRT + R848Activate Toll-like receptor 7/8, increase tumor antigen-specific CD8+ T cells, decrease regulatory T cells, and enhance antigen-presenting cells maturation, as well as increase interferon gamma, granzyme B, and CCL585
SBRT + anti-CD40Promote DC maturation by activating CD40, augment T cell priming, induce memory CD8 T cell responses72
PTT + nanoplatform IMQ@IONs/ICGThermal ablation of the tumor, induce infiltration of CD8 T cells73,87
IREDirect killing effect by IRE, promotes M1 macrophage polarization88
IRE + anti-PD1Direct killing effect by IRE, activates dendritic cells, and alleviates stroma-induced immunosuppression without depleting tumor-restraining collagen89
Targeting multiple nodes of immune response for restoring or reinforcing ICD in PDAC.
Fig. 2  Targeting multiple nodes of immune response for restoring or reinforcing ICD in PDAC.

ANXA1, annexin A1; ATP, adenosine triphosphate; CALR, calreticulin; CTL, cytotoxic T lymphocyte; DC, dendritic cell; HMGB1, high-mobility group box 1; OV, oncolytic virus; PD-L1, programmed death-ligand 1; PRR, pattern recognition receptor; PTT, photothermal therapy; SBRT, stereotactic body radiotherapy; TLR, toll-like receptor; TMB, tumor mutational burden.

Therapeutic drugs

The current first-line chemotherapy regimen for PDAC involves a quadruple regimen called mFOLFIRINOX, which has shown significant extension of survival in clinical trials. Oxaliplatin has been identified as a classic ICD inducer.76 Using mesoporous silica nanoparticles as a platform to carry oxaliplatin plus an indoleamine 2,3-dioxygenase (IDO) inhibitor indoleamine 2,3-dioxygenase (IND), oxaliplatin can target dead tumor cell corpses, as well as simultaneously inhibit the IDO pathway to facilitate ICD, with smaller doses and less bone marrow toxicity.77 The same strategy is also used for irinotecan (IRIN)-encapsulation, in which silicon can deliver IRIN to the lysosomes of cancer cells, neutralize their acidic pH values, and then trigger autophagy inhibition and ER stress response, ultimately leading to “type II” ICD and excessive expression of programmed death-ligand 1 (PD-L1), while enhancing the expression of PD-L1 and antagonizing the response of PD-1.78 The team then added 3M-052, a TLR7/8 agonist, into the IRIN silicon capsule, which further strengthened the immunogenic cell death response induced by IRIN by mediating DC activation, reducing adverse pharmacokinetics and off-target systemic inflammatory effects.79

In addition to exploring the potential of traditional chemotherapy drugs for inducing ICD, some small molecule drugs that can potentially lead to ICD have also been screened and shown to have strong multiple immune effects. Diaciclib, a CDK1/2/5 inhibitor, not only overwhelms adaptive immune resistance mediated by IFN in PDAC, but also induces ICD through histone release.70 Ingenol-3-mebutate (I3A), the main active ingredient in the sap of the Euphorbia plant, can be used as an alternative immune adjuvant to upregulate CD80 and CD86 expression on DCs. Recently, a small-sized sequential receptor nanocarrier has further developed the immunogenic potential of I3A, inducing ICD and reshaping the microenvironment in PDAC.

Biological molecules

Oncolytic viruses/peptides are a promising tumor immunotherapy that uses the concept of “tumor vaccines” to destroy cancer cells. These agents trigger an antitumor immune response by releasing high-quality vaccine-like neoantigens, ultimately leading to tumor-targeted cell cytotoxic T lymphocyte-dependent immune attack. Oncolytic viruses and peptides that have been tested in multiple PDAC models include OBP-702, a p53-expressing telomerase-specific oncolytic adenovirus that significantly enhances ICD through p53-mediated cell apoptosis and autophagy;80 LTX-315, an oncolytic peptide derived from bovine lactoferrin that induces antitumor immunity by targeting the ATP11B-PD-L1 axis;81 and the use of attenuated Listeria monocytogenes to selectively deliver immunogenic tetanus toxin protein (TT856-1313).82 Insights into tumor immunity have also led to the development of other immune-positive biomolecules aimed at enhancing immune responses by increasing adjuvanticity rather than immunogenicity. For example, adding CD40L to oHSV in oncolytic viruses can significantly promote of DC maturation, followed by more ICD markers, stronger antigen presentation, and more CD8+ T cell infiltration, ultimately leading to tumor regression.83

Physical therapy

Although PDAC is considered insensitive to radiotherapy, the development of new radiotherapy techniques is challenging this notion. In theory, the therapeutic effect of radiotherapy is to kill cancer cells that are irradiated, as well as reshape the TME, ultimately initiating an immune response targeting the tumor.84 However, in the immunologically “cold” TME of PDAC, simple radiotherapy alone does not seem to be enough to overcome this barrier. To further enhance immune reactivity, the use of TLR 7/8 ligand R848hu85 or anti-CD4072 is employed to allow immunogenic cell death during SBRT, with the key point being the exogenous provision of additional “danger signals” that initiate DAMPs and downstream ICD. A recent study analyzed tumor specimens from patients receiving SBRT using mIHC, RNA-seq, and TCR-seq, further promoting the clinical efficacy of SBRT combined with immunotherapy in PDAC.86

Photothermal therapy (PTT) is a medical treatment that uses thermal energy to generate heat in targeted tissues or cells. Absorption of photon energy produces significant thermal damage in target cells, especially tumor cells that are sensitive to heat-induced damage, followed by IRS and immunogenic cell death. PTT can supply enough antigenicity, but additional adjuvants can still be added to enhance its immunogenic effect in PDAC.73,87 In addition, the oxygen consumption and microvascular damage driven by PDT will further exacerbate hypoxia and lactate formation. A recent study used a nanodrug delivery platform to reverse hypoxia and downstream lactate metabolism by carrying other precursor drugs, eliminating this side effect. Another non-thermal ablation technique that can induce ICD, irreversible electroporation (IRE), differs from PTT in that it not only kills pancreatic cancer cells to provide immunogenicity but also promotes M1 macrophage polarization88 or reprograms the matrix89 to provide adjuvanticity, making it more self-contained.

Conclusions

At present, a novel therapeutic strategy is needed for PDAC. ICD, which is known as a special form of RCD that can augment immunogenicity, is a promising entity for initiating an immune response. Here, we reviewed the basis of ICD, highlighting its adjuvanticity and utility. A large amount of research demonstrates the success of exogenous provision of DAMPs and a combined regimen targeting multiple immune checkpoints, allowing for “conditional” ICD. This can transform “cold” tumors into “hot” tumors and lead to tumor regression. Meanwhile, an increasing number of new adjuvant molecules for ICD are being discovered, which lays the foundation for exploring new druggable targets. Although there are still shortcomings in the current evaluation and testing models for ICD, new technologies such as RNA-seq and TCR-seq are emerging in trial therapies for ICD, which may bring new breakthroughs in evaluating ICD.

Abbreviations

ANXA1: 

annexin A1

ATP: 

adenosine triphosphate

CALR: 

calreticulin

cGAS: 

cyclic GMP-AMP synthase

DAMPs: 

damage-associated molecular patterns

DCs: 

dendritic cells

ER: 

endoplasmic reticulum

ERS: 

endoplasmic reticulum stress

HMGB1: 

high-mobility group box 1

ICD: 

immunogenic cell death

IDO: 

indoleamine 2,3-dioxygenase

IFN: 

interferon

IL: 

interleukin

IRE: 

irreversible electroporation.

IRIN: 

irinotecan

NK cell: 

natural killer cell

NLR: 

NOD-like receptor

OXA: 

oxaliplatin

P2RX7: 

purinergic receptor P2X7

P2RY2: 

purinergic P2Y receptor 2

PDAC: 

pancreatic ductal adenocarcinoma

PD-L1: 

programmed death-ligand 1

PDT: 

photodynamic therapy

PRR: 

pattern recognition receptor

PTT: 

photothermal therapy

RAGE: 

receptor for advanced glycation end products

ROS: 

reactive oxygen species

SBRT: 

stereotactic body radiotherapy

STING: 

signaling pathway and stimulator of interferon genes

TAA: 

tumor-associated antigen

TLR: 

toll-like receptor

TME: 

tumor microenvironment

TNA: 

tumor neoantigen

Declarations

Acknowledgement

None.

Funding

The work was supported by a grant from the National Natural Science Foundation of China (Grant no. 81871974 to BS).

Conflict of interest

One of the authors, Prof. Bei Sun has been an associate editor of Cancer Screening and Prevention since March 2022. The authors have no other conflict of interests related to this publication.

Authors’ contributions

Contributed to study concept and design (CH and BS), acquisition of the data (CZ), drafting of the manuscript (CH, XLW and YL), critical revision of the manuscript (YL), and supervision (BS).

References

  1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin 2023;73(1):17-48 View Article PubMed/NCBI
  2. Rawla P, Sunkara T, Gaduputi V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J Oncol 2019;10(1):10-27 View Article PubMed/NCBI
  3. Tesfaye AA, Philip PA. Adjuvant treatment of surgically resectable pancreatic ductal adenocarcinoma. Clin Adv Hematol Oncol 2019;17(1):54-63 PubMed/NCBI
  4. Conroy T, Hammel P, Hebbar M, Ben Abdelghani M, Wei AC, Raoul JL, et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N Engl J Med 2018;379(25):2395-2406 View Article PubMed/NCBI
  5. Sistigu A, Yamazaki T, Vacchelli E, Chaba K, Enot DP, Adam J, et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med 2014;20(11):1301-1309 View Article PubMed/NCBI
  6. Neoptolemos JP, Palmer DH, Ghaneh P, Psarelli EE, Valle JW, Halloran CM, et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, phase 3 trial. Lancet 2017;389(10073):1011-1024 View Article PubMed/NCBI
  7. Wainberg ZA, Hochster HS, Kim EJ, George B, Kaylan A, Chiorean EG, et al. Open-label, Phase I Study of Nivolumab Combined with nab-Paclitaxel Plus Gemcitabine in Advanced Pancreatic Cancer. Clin Cancer Res 2020;26(18):4814-4822 View Article PubMed/NCBI
  8. Fan JQ, Wang MF, Chen HL, Shang D, Das JK, Song J. Current advances and outlooks in immunotherapy for pancreatic ductal adenocarcinoma. Mol Cancer 2020;19(1):32 View Article PubMed/NCBI
  9. Lawlor RT, Mattiolo P, Mafficini A, Hong SM, Piredda ML, Taormina SV, et al. Tumor Mutational Burden as a Potential Biomarker for Immunotherapy in Pancreatic Cancer: Systematic Review and Still-Open Questions. Cancers (Basel) 2021;13(13):3119 View Article PubMed/NCBI
  10. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature 2017;541(7637):321-330 View Article PubMed/NCBI
  11. Dongre A, Weinberg RA. Leveraging immunochemotherapy for treating pancreatic cancer. Cell Res 2021;31(12):1228-1229 View Article PubMed/NCBI
  12. Choi M, Shin J, Lee CE, Chung JY, Kim M, Yan X, et al. Immunogenic cell death in cancer immunotherapy. BMB Rep 2023;56(5):275-286 View Article PubMed/NCBI
  13. 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(12):1691-1701 View Article PubMed/NCBI
  14. Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 2018;25(3):486-541 View Article PubMed/NCBI
  15. Serrano-Del Valle A, Reina-Ortiz C, Benedi A, Anel A, Naval J, Marzo I. Future prospects for mitosis-targeted antitumor therapies. Biochem Pharmacol 2021;190:114655 View Article PubMed/NCBI
  16. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol 2017;17(2):97-111 View Article PubMed/NCBI
  17. Green DR, Ferguson T, Zitvogel L, Kroemer G. Immunogenic and tolerogenic cell death. Nat Rev Immunol 2009;9(5):353-363 View Article PubMed/NCBI
  18. Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. Nat Immunol 2022;23(4):487-500 View Article PubMed/NCBI
  19. Vitale I, Sistigu A, Manic G, Rudqvist NP, Trajanoski Z, Galluzzi L. Mutational and Antigenic Landscape in Tumor Progression and Cancer Immunotherapy. Trends Cell Biol 2019;29(5):396-416 View Article PubMed/NCBI
  20. Ahmed A, Tait SWG. Targeting immunogenic cell death in cancer. Mol Oncol 2020;14(12):2994-3006 View Article PubMed/NCBI
  21. Galluzzi L, Kepp O, Hett E, Kroemer G, Marincola FM. Immunogenic cell death in cancer: concept and therapeutic implications. J Transl Med 2023;21(1):162 View Article PubMed/NCBI
  22. Murao A, Aziz M, Wang H, Brenner M, Wang P. Release mechanisms of major DAMPs. Apoptosis 2021;26(3-4):152-162 View Article PubMed/NCBI
  23. Jiang M, Zeng J, Zhao L, Zhang M, Ma J, Guan X, et al. Chemotherapeutic drug-induced immunogenic cell death for nanomedicine-based cancer chemo-immunotherapy. Nanoscale 2021;13(41):17218-17235 View Article PubMed/NCBI
  24. Kasikova L, Hensler M, Truxova I, Skapa P, Laco J, Belicova L, et al. Calreticulin exposure correlates with robust adaptive antitumor immunity and favorable prognosis in ovarian carcinoma patients. J Immunother Cancer 2019;7(1):312 View Article PubMed/NCBI
  25. Poon IK, Lucas CD, Rossi AG, Ravichandran KS. Apoptotic cell clearance: basic biology and therapeutic potential. Nat Rev Immunol 2014;14(3):166-180 View Article PubMed/NCBI
  26. 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(5):578-590 View Article PubMed/NCBI
  27. 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(9):1499-1509 View Article PubMed/NCBI
  28. Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-Ullrich JE, et al. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell 2005;123(2):321-334 View Article PubMed/NCBI
  29. Gardai SJ, Xiao YQ, Dickinson M, Nick JA, Voelker DR, Greene KE, et al. By binding SIRPalpha or calreticulin/CD91, lung collectins act as dual function surveillance molecules to suppress or enhance inflammation. Cell 2003;115(1):13-23 View Article PubMed/NCBI
  30. 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(5):1062-1079 View Article PubMed/NCBI
  31. Byrne JC, Ní Gabhann J, Stacey KB, Coffey BM, McCarthy E, Thomas W, et al. Bruton’s tyrosine kinase is required for apoptotic cell uptake via regulating the phosphorylation and localization of calreticulin. J Immunol 2013;190(10):5207-5215 View Article PubMed/NCBI
  32. Garg AD, Elsen S, Krysko DV, Vandenabeele P, de Witte P, Agostinis P. Resistance to anticancer vaccination effect is controlled by a cancer cell-autonomous phenotype that disrupts immunogenic phagocytic removal. Oncotarget 2015;6(29):26841-26860 View Article PubMed/NCBI
  33. Truxova I, Kasikova L, Salek C, Hensler M, Lysak D, Holicek P, et al. Calreticulin exposure on malignant blasts correlates with improved natural killer cell-mediated cytotoxicity in acute myeloid leukemia patients. Haematologica 2020;105(7):1868-1878 View Article PubMed/NCBI
  34. Fucikova J, Truxova I, Hensler M, Becht E, Kasikova L, Moserova I, et al. Calreticulin exposure by malignant blasts correlates with robust anticancer immunity and improved clinical outcome in AML patients. Blood 2016;128(26):3113-3124 View Article PubMed/NCBI
  35. Yang H, Wang H, Chavan SS, Andersson U. High Mobility Group Box Protein 1 (HMGB1): The Prototypical Endogenous Danger Molecule. Mol Med 2015;21(Suppl 1):S6-S12 View Article PubMed/NCBI
  36. Zhu X, Messer JS, Wang Y, Lin F, Cham CM, Chang J, et al. Cytosolic HMGB1 controls the cellular autophagy/apoptosis checkpoint during inflammation. J Clin Invest 2015;125(3):1098-1110 View Article PubMed/NCBI
  37. Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol 2010;28:367-388 View Article PubMed/NCBI
  38. Tittarelli A, González FE, Pereda C, Mora G, Muñoz L, Saffie C, et al. Toll-like receptor 4 gene polymorphism influences dendritic cell in vitro function and clinical outcomes in vaccinated melanoma patients. Cancer Immunol Immunother 2012;61(11):2067-2077 View Article PubMed/NCBI
  39. Dumitriu IE, Baruah P, Valentinis B, Voll RE, Herrmann M, Nawroth PP, et al. Release of high mobility group box 1 by dendritic cells controls T cell activation via the receptor for advanced glycation end products. J Immunol 2005;174(12):7506-7515 View Article PubMed/NCBI
  40. Nayagom B, Amara I, Habiballah M, Amrouche F, Beaune P, de Waziers I. Immunogenic cell death in a combined synergic gene- and immune-therapy against cancer. Oncoimmunology 2019;8(12):e1667743 View Article PubMed/NCBI
  41. 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(9):1050-1059 View Article PubMed/NCBI
  42. 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(1):21-32 View Article PubMed/NCBI
  43. 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(9):832-842 View Article PubMed/NCBI
  44. Anderson CM, Macleod KF. Autophagy and cancer cell metabolism. Int Rev Cell Mol Biol 2019;347:145-190 View Article PubMed/NCBI
  45. Follo C, Cheng Y, Richards WG, Bueno R, Broaddus VC. Autophagy facilitates the release of immunogenic signals following chemotherapy in 3D models of mesothelioma. Mol Carcinog 2019;58(10):1754-1769 View Article PubMed/NCBI
  46. Burnstock G. Physiology and pathophysiology of purinergic neurotransmission. Physiol Rev 2007;87(2):659-797 View Article PubMed/NCBI
  47. 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(7261):282-286 View Article PubMed/NCBI
  48. 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(10):1170-1178 View Article PubMed/NCBI
  49. Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol 2019;19(8):477-489 View Article PubMed/NCBI
  50. Stagg J, Beavis PA, Divisekera U, Liu MC, Möller A, Darcy PK, et al. CD73-deficient mice are resistant to carcinogenesis. Cancer Res 2012;72(9):2190-2196 View Article PubMed/NCBI
  51. Allard B, Allard D, Buisseret L, Stagg J. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol 2020;17(10):611-629 View Article PubMed/NCBI
  52. Faraoni EY, Singh K, Chandra V, Le Roux O, Dai Y, Sahin I, et al. CD73-Dependent Adenosine Signaling through Adora2b Drives Immunosuppression in Ductal Pancreatic Cancer. Cancer Res 2023;83(7):1111-1127 View Article PubMed/NCBI
  53. Rodriguez-Ruiz ME, Buqué A, Hensler M, Chen J, Bloy N, Petroni G, et al. Apoptotic caspases inhibit abscopal responses to radiation and identify a new prognostic biomarker for breast cancer patients. Oncoimmunology 2019;8(11):e1655964 View Article PubMed/NCBI
  54. Khodarev NN. Intracellular RNA Sensing in Mammalian Cells: Role in Stress Response and Cancer Therapies. Int Rev Cell Mol Biol 2019;344:31-89 View Article PubMed/NCBI
  55. Kawasaki T, Kawai T. Discrimination Between Self and Non-Self-Nucleic Acids by the Innate Immune System. Int Rev Cell Mol Biol 2019;344:1-30 View Article PubMed/NCBI
  56. Garg AD, Vandenberk L, Fang S, Fasche T, Van Eygen S, Maes J, et al. Pathogen response-like recruitment and activation of neutrophils by sterile immunogenic dying cells drives neutrophil-mediated residual cell killing. Cell Death Differ 2017;24(5):832-843 View Article PubMed/NCBI
  57. Medler T, Patel JM, Alice A, Baird JR, Hu HM, Gough MJ. Activating the Nucleic Acid-Sensing Machinery for Anticancer Immunity. Int Rev Cell Mol Biol 2019;344:173-214 View Article PubMed/NCBI
  58. Hopfner KP, Hornung V. Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat Rev Mol Cell Biol 2020;21(9):501-521 View Article PubMed/NCBI
  59. Yamazaki T, Kirchmair A, Sato A, Buqué A, Rybstein M, Petroni G, et al. Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy. Nat Immunol 2020;21(10):1160-1171 View Article PubMed/NCBI
  60. Motwani M, Pesiridis S, Fitzgerald KA. DNA sensing by the cGAS-STING pathway in health and disease. Nat Rev Genet 2019;20(11):657-674 View Article PubMed/NCBI
  61. Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun 2017;8:15618 View Article PubMed/NCBI
  62. Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 2014;41(5):843-852 View Article PubMed/NCBI
  63. Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, Kroemer G. Type I interferons in anticancer immunity. Nat Rev Immunol 2015;15(7):405-414 View Article PubMed/NCBI
  64. Oh JH, Kim MJ, Choi SJ, Ban YH, Lee HK, Shin EC, et al. Sustained Type I Interferon Reinforces NK Cell-Mediated Cancer Immunosurveillance during Chronic Virus Infection. Cancer Immunol Res 2019;7(4):584-599 View Article PubMed/NCBI
  65. Bek S, Stritzke F, Wintges A, Nedelko T, Böhmer DFR, Fischer JC, et al. Targeting intrinsic RIG-I signaling turns melanoma cells into type I interferon-releasing cellular antitumor vaccines. Oncoimmunology 2019;8(4):e1570779 View Article PubMed/NCBI
  66. Takeda Y, Azuma M, Funami K, Shime H, Matsumoto M, Seya T. Type I Interferon-Independent Dendritic Cell Priming and Antitumor T Cell Activation Induced by a Mycoplasma fermentans Lipopeptide. Front Immunol 2018;9:496 View Article PubMed/NCBI
  67. Gangaplara A, Martens C, Dahlstrom E, Metidji A, Gokhale AS, Glass DD, et al. Type I interferon signaling attenuates regulatory T cell function in viral infection and in the tumor microenvironment. PLoS Pathog 2018;14(4):e1006985 View Article PubMed/NCBI
  68. Fucikova J, Moserova I, Urbanova L, Bezu L, Kepp O, Cremer I, et al. Prognostic and Predictive Value of DAMPs and DAMP-Associated Processes in Cancer. Front Immunol 2015;6:402 View Article PubMed/NCBI
  69. Vacchelli E, Ma Y, Baracco EE, Sistigu A, Enot DP, Pietrocola F, et al. Chemotherapy-induced antitumor immunity requires formyl peptide receptor 1. Science 2015;350(6263):972-978 View Article PubMed/NCBI
  70. Huang J, Chen P, Liu K, Liu J, Zhou B, Wu R, et al. CDK1/2/5 inhibition overcomes IFNG-mediated adaptive immune resistance in pancreatic cancer. Gut 2021;70(5):890-899 View Article PubMed/NCBI
  71. Nestvold J, Wang MY, Camilio KA, Zinöcker S, Tjelle TE, Lindberg A, et al. Oncolytic peptide LTX-315 induces an immune-mediated abscopal effect in a rat sarcoma model. Oncoimmunology 2017;6(8):e1338236 View Article PubMed/NCBI
  72. Yasmin-Karim S, Bruck PT, Moreau M, Kunjachan S, Chen GZ, Kumar R, et al. Radiation and Local Anti-CD40 Generate an Effective in situ Vaccine in Preclinical Models of Pancreatic Cancer. Front Immunol 2018;9:2030 View Article PubMed/NCBI
  73. Zhou F, Yang J, Zhang Y, Liu M, Lang ML, Li M, et al. Local Phototherapy Synergizes with Immunoadjuvant for Treatment of Pancreatic Cancer through Induced Immunogenic Tumor Vaccine. Clin Cancer Res 2018;24(21):5335-5346 View Article PubMed/NCBI
  74. Zhou W, Zhou Y, Chen X, Ning T, Chen H, Guo Q, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials 2021;268:120546 View Article PubMed/NCBI
  75. Kim D, Lee S, Na K. Immune Stimulating Antibody-Photosensitizer Conjugates via Fc-Mediated Dendritic Cell Phagocytosis and Phototriggered Immunogenic Cell Death for KRAS-Mutated Pancreatic Cancer Treatment. Small 2021;17(10):e2006650 View Article PubMed/NCBI
  76. 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(4):482-491 View Article PubMed/NCBI
  77. Liu X, Jiang J, Chang CH, Liao YP, Lodico JJ, Tang I, et al. Development of Facile and Versatile Platinum Drug Delivering Silicasome Nanocarriers for Efficient Pancreatic Cancer Chemo-Immunotherapy. Small 2021;17(14):e2005993 View Article PubMed/NCBI
  78. Liu X, Jiang J, Liao YP, Tang I, Zheng E, Qiu W, et al. Combination Chemo-Immunotherapy for Pancreatic Cancer Using the Immunogenic Effects of an Irinotecan Silicasome Nanocarrier Plus Anti-PD-1. Adv Sci (Weinh) 2021;8(6):2002147 View Article PubMed/NCBI
  79. Luo L, Wang X, Liao YP, Chang CH, Nel AE. Nanocarrier Co-formulation for Delivery of a TLR7 Agonist plus an Immunogenic Cell Death Stimulus Triggers Effective Pancreatic Cancer Chemo-immunotherapy. ACS Nano 2022;16(8):13168-13182 View Article PubMed/NCBI
  80. Araki H, Tazawa H, Kanaya N, Kajiwara Y, Yamada M, Hashimoto M, et al. Oncolytic virus-mediated p53 overexpression promotes immunogenic cell death and efficacy of PD-1 blockade in pancreatic cancer. Mol Ther Oncolytics 2022;27:3-13 View Article PubMed/NCBI
  81. Tang T, Huang X, Zhang G, Lu M, Hong Z, Wang M, et al. Oncolytic peptide LTX-315 induces anti-pancreatic cancer immunity by targeting the ATP11B-PD-L1 axis. J Immunother Cancer 2022;10(3):e004129 View Article PubMed/NCBI
  82. Selvanesan BC, Chandra D, Quispe-Tintaya W, Jahangir A, Patel A, Meena K, et al. Listeria delivers tetanus toxoid protein to pancreatic tumors and induces cancer cell death in mice. Sci Transl Med 2022;14(637):eabc1600 View Article PubMed/NCBI
  83. Wang R, Chen J, Wang W, Zhao Z, Wang H, Liu S, et al. CD40L-armed oncolytic herpes simplex virus suppresses pancreatic ductal adenocarcinoma by facilitating the tumor microenvironment favorable to cytotoxic T cell response in the syngeneic mouse model. J Immunother Cancer 2022;10(1):e003809 View Article PubMed/NCBI
  84. Rodriguez-Ruiz ME, Vitale I, Harrington KJ, Melero I, Galluzzi L. Immunological impact of cell death signaling driven by radiation on the tumor microenvironment. Nat Immunol 2020;21(2):120-134 View Article PubMed/NCBI
  85. Ye J, Mills BN, Qin SS, Garrett-Larsen J, Murphy JD, Uccello TP, et al. Toll-like receptor 7/8 agonist R848 alters the immune tumor microenvironment and enhances SBRT-induced antitumor efficacy in murine models of pancreatic cancer. J Immunother Cancer 2022;10(7):e004784 View Article PubMed/NCBI
  86. Mills BN, Qiu H, Drage MG, Chen C, Mathew JS, Garrett-Larsen J, et al. Modulation of the Human Pancreatic Ductal Adenocarcinoma Immune Microenvironment by Stereotactic Body Radiotherapy. Clin Cancer Res 2022;28(1):150-162 View Article PubMed/NCBI
  87. Wang M, Li Y, Wang M, Liu K, Hoover AR, Li M, et al. Synergistic interventional photothermal therapy and immunotherapy using an iron oxide nanoplatform for the treatment of pancreatic cancer. Acta Biomater 2022;138:453-462 View Article PubMed/NCBI
  88. He C, Sun S, Zhang Y, Xie F, Li S. The role of irreversible electroporation in promoting M1 macrophage polarization via regulating the HMGB1-RAGE-MAPK axis in pancreatic cancer. Oncoimmunology 2021;10(1):1897295 View Article PubMed/NCBI
  89. Zhao J, Wen X, Tian L, Li T, Xu C, Wen X, et al. Irreversible electroporation reverses resistance to immune checkpoint blockade in pancreatic cancer. Nat Commun 2019;10(1):899 View Article PubMed/NCBI
  90. Shen J, Sun C, Wang Z, Chu Z, Liu C, Xu X, et al. Sequential receptor-mediated mixed-charge nanomedicine to target pancreatic cancer, inducing immunogenic cell death and reshaping the tumor microenvironment. Int J Pharm 2021;601:120553 View Article PubMed/NCBI
  • Cancer Screening and Prevention
  • pISSN 2993-6314
  • eISSN 2835-3315
Back to Top

Leveraging Immunogenic Cell Death in Pancreatic Cancer

Cong He, Yan Luo, Xin-Long Wang, Can Zhang, Bei Sun
  • Reset Zoom
  • Download TIFF