You are on page 1of 16

Biomaterials 282 (2022) 121433

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Emerging photodynamic nanotherapeutics for inducing immunogenic cell


death and potentiating cancer immunotherapy
Shenwu Zhang, Jing Wang, Zhiqiang Kong, Xinxin Sun, Zhonggui He, Bingjun Sun **,
Cong Luo ***, Jin Sun *
Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China

A R T I C L E I N F O A B S T R A C T

Keywords: Immunotherapy has emerged as a promising cancer treatment modality. Despite the rapid progress in cancer
Photodynamic therapy immunotherapy, the therapeutic efficiency and clinical translation of immunotherapy are not as satisfactory as
Nanotherapeutics expected, especially for the patients with immune-cold tumors. Immunogenic cell death (ICD) represents a
Immunogenic cell death
particular form of tumor cell death accompanied by the production of tumor-specific antigens, which facilitates
Synergistic induction
Cancer immunotherapy
the infiltration of effector T cells and potentiates immune response in solid tumors. Thus, ICD contributes to
stimulating immune-cold tumors to immune-hot ones. Increasing evidence shows that photodynamic therapy
(PDT) is able to effectively induce ICD. Recently, a variety of photodynamic nanotherapeutics have been
developed to induce ICD and to potentiate cancer immunotherapy. Herein, this review outlines the recent ad­
vances in the field at the intersection of PDT, nanotechnology and ICD, including PDT-induced ICD, PDT-based
synergistic induction of ICD, and multimodal immunotherapy in basis of PDT-induced ICD. Finally, the prospects
and challenges of these photodynamic nanotherapeutics in ICD induction-based cancer immunotherapy are
discussed.

1. Introduction immune response, resulting in unsatisfactory immunotherapeutic effi­


cacy [16–18]. Therefore, how to promote the transformation of
Malignant tumors remain a severe threat to human health [1]. In immune-cold tumors to immune-hot ones could be of crucial signifi­
recent years, immunotherapy has shown promising application pros­ cance for efficient immunotherapy.
pects in clinical practice [2–9]. Compared with traditional treatment In recent decades, it has always been believed that immune re­
strategies, the emerging immunotherapy not only suppresses the growth sponses could only be triggered by exogenous substances. But in late
of primary tumors, but also effectively prevents the metastasis and 1990s, studies showed that endogenous entities could also evoke im­
relapse of tumor by harnessing the inherent immune system of patients mune responses under certain conditions [19]. In 2005, Kepp et al.
[10,11]. Typical immunotherapy strategies mainly include nonspecific firstly demonstrated that tumor cells exposed to doxorubicin (Dox) were
immunostimulation, immune checkpoint-blockade (ICB) therapy, tumor capable of acting as vaccine and activating host immunity during
vaccine, adoptive cellular immunotherapy (ACI) [12,13]. Among them, apoptosis, and named this form of cell death as immunogenic cell death
immune checkpoint programmed death 1 (PD-1)/programmed (ICD) [20,21]. Since the concept of ICD was first proposed in 2005, host
death-ligand 1 (PD-L1) pathway blockade has been discovered as a po­ immunity has received considerable attention in cancer treatment [22].
tential immunotherapy modality [14,15]. Notably, PD-1/PD-L1 The emergence of ICD and ICD-based synergistic therapeutics has
blockade immunotherapy heavily depends on the full infiltration of bridged the gap between traditional and new treatment modalities [23].
activated T lymphocytes into tumor tissues. Nevertheless, a considerable Particularly, ICD induced by traditional therapies has emerged as a
proportion of the clinical patients with immune-cold tumors exhibit low promising strategy to convert immune-cold tumors to immune-hot ones

* Corresponding author.
** Corresponding author. Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016,
China.
*** Corresponding author.
E-mail addresses: sunbingjun_spy@sina.com (B. Sun), luocong@syphu.edu.cn (C. Luo), sunjin@syphu.edu.cn (J. Sun).

https://doi.org/10.1016/j.biomaterials.2022.121433
Received 14 October 2021; Received in revised form 21 January 2022; Accepted 17 February 2022
Available online 19 February 2022
0142-9612/© 2022 Elsevier Ltd. All rights reserved.
S. Zhang et al. Biomaterials 282 (2022) 121433

in tumor immunotherapy (Fig. 1) [24–27]. After the induction of ICD,


the dying tumor cells release tumor associated antigens (TAAs),
pro-inflammatory cytokines and danger associated molecular patterns
(DAMPs) [27,28], such as heat shock proteins (HSPs) [29], calreticulin
(CRT) [30–33], high mobility group box 1 (HMGB1) [33,34], and
adenosine triphosphate (ATP) [35,36]. The released DAMPs can stim­
ulate the maturation of dendritic cells (DCs), and subsequently the
mature DCs carrying cancer-specific antigens migrate to the lymph
nodes, activating effector T cells [37]. Finally, the activated effector T
cells migrate and infiltrate into tumor tissues to eliminate tumor cells
[37].
Given that ICD is induced via apoptosis in most cases, it is usually
themed as immunogenic apoptosis. Recently, several nonapoptotic cell
death modalities have also been found to be related to antitumor im­
munity [38]. For instance, necroptosis is a modality of programmed cell
death along with the release of intracellular contents amongst DAMPs,
Fig. 2. Schematic representation of photodynamic reactions.
which evokes antitumor immunity through activation of nuclear
factor-κB (NF-κB) or priming and activation of effector T cells [39,40].
Moreover, ferroptosis has also emerged as a new cell death modality half-life of most PSs, the tumor hypoxic microenvironment, and the
induced by iron ion-mediated accumulation of lipid peroxides [41]. It insufficient accumulation of ROS in ER hampers the induction of ICD
has been found that tumor cells undergoing ferroptosis can release [48,49]. In addition, there are still some inherent limitations of PDT that
HMGB1 by autophagy [42]. In turn, the infiltration of CD8+ T cells can are not sufficient to evoke a strong immune response. Moreover,
facilitate and enhance the occurrence of tumor ferroptosis [43]. Actu­ single-modal cancer immunotherapy based on PDT-driven ICD is not
ally, immunogenic cell death refers to an umbrella term, some other cell enough to attain a satisfactory therapy outcome. Therefore, it’s neces­
death modalities such as necroptosis and ferroptosis were also found sary to improve the in vivo delivery efficiency of PSs and design com­
with immunogenic features [38]. However, the detailed mechanisms of bined therapeutic strategies for efficient PDT-driven immunotherapy.
antitumor immunity induced by these nonapoptotic cell death modal­ With the burgeoning of biomaterials engineering and nanotech­
ities still remain not entirely clear. nology, nanoparticulate drug delivery system (nano-DDS) has been
Several conventional treatment modalities have been found to cause widely investigated as an efficient platform for the delivery of anticancer
tumor cell death in an immunogenic way, such as chemotherapy, drugs [50–54]. Among them, a wide range of photodynamic nano­
radiotherapy, and photodynamic therapy (PDT). Among them, PDT, as a therapeutics have been developed for cancer treatment [55–57].
non-invasive therapeutic modality, has been widely investigated for Formulating PSs into nanocarriers could offer many advantages, such as
localized cancer therapy [44–46]. Under localized tumor-focused improvements in the physicochemical and pharmacokinetics properties
near-infrared (NIR) light irradiation, photosensitizers (PSs) rapidly of PSs, tumor targeting delivery, and enhancement of combined therapy
generate cytotoxic reactive oxygen species (ROS) (Fig. 2) and kill tumor [58]. In recent years, the emerging photodynamic nanotherapeutics for
cells [44]. Notably, ROS generated from PSs can not only promote tumor inducing ICD and potentiating cancer immunotherapy have aroused
cell death but also trigger endoplasmic reticulum (ER) pressure and growing interest in cancer therapy [59–61]. In view of the rapid
elicit CRT exposure, thus inducing ICD of tumor cells and releasing development of photodynamic nanotherapeutics in inducing ICD and
pro-inflammatory cytokines and DAMPs [47,48]. However, the short potentiating cancer immunotherapy, we summarize and outline the

Fig. 1. Schematic representation of the cycle process


of immune response in tumor cells. First, antigens and
DAMPs were released from dying tumor cells. Then,
antigens were presented by antigen-presenting cells
(APCs) to promote the maturation of DCs. Antigens
were further presented to T cells in lymph nodes,
resulting in the priming and activation of T cells.
Finally, activated effector T cells were trafficked and
infiltrated to tumor tissues, where they could recog­
nize and kill tumor cells.

2
S. Zhang et al. Biomaterials 282 (2022) 121433

latest updates in this field from various induction strategies (Fig. 3, the accumulation of Lp-KR-CCM-A in tumor was improved by 3.3 times
Table 1). Moreover, we also make some classifications of biomaterials compared with that of Lp-A owing to the high affinity to homologous
and PSs used in the development of ICD-based photodynamic nano­ tumor cells [64]. The effective distribution of KR in tumor tissues
therapeutics (Fig. 4). Finally, we briefly discuss the design principles, enabled the generation of large amounts of ROS under laser irradiation
advantages, challenges and future prospects of ICD-based photodynamic and elicited robust ICD with the release of DAMPs. Meanwhile, the host
nanotherapeutics in clinical cancer therapy. This paper could provide immune response was activated, including the maturation of DCs and
reference for the design and construction of photodynamic nano­ the proliferation of cytotoxic T lymphocytes (CTLs). As a result, the
therapeutics to induce ICD and enhance cancer immunotherapy. activated immune system significantly inhibited the growth of primary
tumor and the lung metastasis in an ectopic 4T1 tumor-bearing mouse
2. Photodynamic nanotherapeutics for ICD induction model [64].
In addition to CCM, immune cell membranes (ICM) have also been
Recently, PDT has been found to effectively induce ICD and poten­ used to develop biomimetic nanotherapeutics, such as neutrophils, DCs
tiate the immune response [62–72]. However, there are plenty of chal­ and natural killer (NK) cells [65,67]. ICM-camouflaged NPs could not
lenges for PDT-mediated induction of ICD, such as short half-life and only increase tumor targeting efficiency but also induce the secretion of
aggregation caused quenching (ACQ) effect of most PSs, tumor hypoxia pro-inflammatory cytokines. For instance, Deng et al. recently devel­
microenvironment and inefficient accumulation of PSs in ER. Recently, oped NK cell membrane-coated poly (lactic-coglycolic acid) (PLGA) NPs
several emerging photodynamic nanotherapeutics have been developed (NK-NPs) loading with 4, 4′ , 4′′ , 4′′′ -(porphine-5, 10, 15, 20-tetrayl)
to deal with multiple stages of obstacles in the whole drug delivery tetrakis (benzoic acid) (TCPP, a photosensitizer) for cancer immuno­
process [49,65,73–75]. In this section, we will discuss the emerging therapy (Fig. 5) [65]. NK-NPs could be effectively accumulated in tumor
photodynamic nanotherapeutics for ICD induction, including targeting tissues to evoke ICD and facilitate release of DAMPs, including amount
induction of ICD with biomimetic photodynamic nanotherapeutics, CRT exposure induced, secreted ATP and HMGB1 release under NIR
remodeling unfavorable tumor microenvironment (TME) for PDT, pre­ light illumination, thus propelling the maturation of DCs and increasing
cisely targeting to ER and addressing the ACQ effect of PSs. the percentage of infiltrating T cells (CD4+ or CD8+) [65]. Moreover, NK
cell membrane was found to participate in inducing the polarization of
M1 macrophages and the secretion of cytokines, and further enhancing
2.1. Biomimetically targeting induction of ICD
the infiltration of effector T cells [65]. In the 4T1 tumor model, both
primary tumors and distant tumors were significantly inhibited after
Compared with conventional nanocarriers, cell membrane-derived
PDT treatment of NK-NPs.
nanoplatform has emerged as an effective and promising biomimetic
Moreover, nanosystems modified with hybrid cell membranes were
strategy for enhancing PDT efficiency [62–67]. Particularly, it has been
also explored as multi-functional nanoplatforms. For example, a nano­
demonstrated that nanoparticles (NPs) wrapped with or embedded in
system engineered from hybrid membranes of cancerous 4T1 cells and
cancer cell membranes (CCM) can significantly improve tumor targeting
DCs was developed for reinforcing cancer immunotherapy [66]. Briefly,
capacity, thus greatly improving the efficacy of PDT-induced ICD
TCPP was dispersed in the metal-organic frameworks (MOFs), and the
[62–66]. For instance, Han et al. designed 4T1-Fluc CCM-modified li­
MOFs were further surrounded by hybrid cytomembranes to construct
posomes loading with KillerRed (a photosensitizer, KR) for improving
unique NPs (PCN@FM) [5]. PCN@FM exhibited excellent tumor
the tumor-homing ability. Firstly, the 4T1-Fluc cancer cells transfected
self-targeting ability due to the coating of functional hybrid cytomem­
with mem-KR plasmids were extracted. Then, KR-CCM, MPLA adjuvant
branes (4T1 cells and DCs) [66]. Besides, under NIR light illumination,
(A), and helper lipids (Lp) were hybridized to prepare lipocomplex
multiple-stimulation of the released DAMPs, immune co-stimulatory
(Lp-KR-CCM-A) via thin-film hydration and serial extrusion. Notably,
molecules and highly expressed antigen on hybrid cytomembranes
could synergistically enhance the activation and the tumor infiltration of
CTLs [66]. Consequently, both the primary and abscopal tumors in
4T1-tumor-bearing mice were almost entirely eliminated [66].
Except cell membranes, some serum proteins were also used to
develop biomimetic photodynamic nanosystems. For instance, Chen
et al. designed a bioinspired nanocarrier loaded with chlorine e6 (Ce6),
which was coated by hybrid protein composed of human serum albumin
(HSA) and hemoglobin (Hb) [76]. As an endogenous protein, HSA shows
good biocompatibility and specific tumor-targeting ability. As a result,
the HAS-modified nanosystem (C@HPOC) significantly promoted the
accumulation of Ce6 in tumor tissues [76]. Under laser irradiation, Ce6
produced large amounts of ROS in killing tumor cells accompanied by
CRT exposure, ATP secretion and HMGB1 release from 4T1 cells, and
then promoted DC maturation [76]. In a bilateral metastatic
triple-negative breast cancer model, the growth of primary tumors was
significantly suppressed and the distant tumors were also obtained
effective treatment attributed to the activated host immune system.
More importantly, C@HPOC plus laser treatment against primary 4T1
tumors could extremely prevent the occurrence of lung metastasis [76].
Collectively, these results demonstrated PDT-induced ICD could elicit an
intense antitumor immune response, which accordingly contributed to
promising abscopal and anti-metastatic effect [76].

2.2. Remodeling the undesirable TME

Fig. 3. Schematic representation of emerging photodynamic nanotherapeutics As discussed above, biomimetic strategy has been found to signifi­
for inducing immunogenic cell death and potentiating cancer immunotherapy. cantly increase the specific accumulation of PSs in tumors, which

3
S. Zhang et al. Biomaterials 282 (2022) 121433

Table 1
Main strategies and characterizations of the emerging photodynamic nanotherapeutics for inducing ICD and potentiating cancer immunotherapy.
Classifications Strategies Nanoplatforms Functions Refs.

Photodynamic NPs for ICD Biomimetically Targeting Cancer cell membranes-decorated Enhanced homologous targeting 62–64
Induction Induction of ICD photodynamic NPs
Immune cell membranes-decorated Enhanced homologous targeting and the 65, 67
photodynamic NPs secretion of pro-inflammatory cytokines
Hybrid cell membranes-decorated Enhanced immunogenicity and tumor-targeting 66
photodynamic NPs ability
Serum proteins-decorated Enhanced tumor-targeting ability 76
photodynamic NPs
Remodeling the Photodynamic NPs based on Enhanced penetration of photodynamic NPs 68, 69
Undesirable TME destroying tumor extracellular matrix
Upconversion-based photodynamic Stronger penetration ability of light 70-72, 77
NPs
Photodynamic NPs loaded with Catalyzing H2O2 to produce oxygen 73, 78-83
metallic oxides or catalase
Photodynamic NPs loaded with Reducing oxygen consumption 59, 84, 85
mitochondrial respiration inhibitor
Precisely Targeting ER ER-targeting photodynamic NPs Enhancing ER stress and CRT exposure 48, 49
Addressing the ACQ Effect Acid-responsive photodynamic NPs Accelerating drug release and addressing ACQ 86
effect
Redox-responsive photodynamic NPs Accelerating drug release and addressing ACQ 63, 75
effect
Dual-responsive photodynamic NPs Accelerating drug release and addressing ACQ 74, 87
effect
PDT-based Synergistic Induction Photothermal/ Photodynamic NPs loaded with PTT + PDT 92, 99
of ICD Photodynamic Synergistic organic PSs
Induction Inorganic photodynamic NPs such as PTT + PDT 48, 95-98
Au NPs, black phosphorus NPs and etc.
Chemo-photodynamic Photodynamic NPs integrated with Chemotherapy + PDT 74, 88, 90, 101
Synergistic Induction chemical drugs such doxorubicin or
oxaliplatin
Other Synergistic Induction Photodynamic NPs integrated with Radiotherapy + PDT 114, 115
Combos radiation-activatable PSs
Photodynamic NPs loaded with Sonodynamic therapy + PDT 122, 23
photo/sonosensitizers
Integrating PSs with nanovesicles Carrier-based ICD + PDT 91
enabling ICD induction
Multimodal Cancer Combination with Photodynamic NPs loaded with Enhancing the PDT-based antigen presentation 131–133
Immunotherapy in basis of Adjuvants or Vaccines adjuvants
PDT-induced ICD Photodynamic NPs loaded with Combination of vaccines and the PDT-based 134, 135
vaccines DAMPs release
Combination with Photodynamic NPs integrated with Combination of PD-1/PD-L1 blockade-based 86, 125, 130,
Checkpoint Blockade anti-PD-1/PD-L1 immunotherapy and PDT 138 139, 142
Photodynamic NPs integrated with Combination of CTLA-4 blockade-based 92, 129
anti-CTLA-4 immunotherapy and PDT
Photodynamic NPs integrated with Combination of IDO blockade-based 50, 191, 105,
IDO inhibitor immunotherapy and PDT 126, 128, 147,
148
Photodynamic NPs integrated with Combination of OX40 or TIM-3 checkpoint- 152, 157
checkpoint inhibitors or agonists based immunotherapy and PDT
Combination with Other Photodynamic NPs integrated with Combination of immunotherapy strategy based 127, 158
Therapeutics imatinib or sorafenib on suppressing the immunosuppressive cells and
PDT

contributes to the induction of ICD under laser irradiation [62–67]. Subsequently, following treatment with Ce6@liposome exhibited the
However, the extremely intricate TME still imposes multiple barriers to enhanced intratumor penetration and more significant suppression in
PDT and PDT-induced ICD, such as the compact tumor extracellular 4T1 tumor-bearing BALB/c mice [68]. Moreover, the increased tumor
matrix (ECM), limited penetration ability of light into tumor tissues and infiltration of CTLs was observed in the group of DEX-HAase plus PDT
tumor hypoxia [68–73]. It has been widely accepted that overcoming (enhanced PDT), which could be ascribed to the release of TAAs after the
these barriers within TME is of crucial importance for efficient PDT and enhanced PDT-induced ICD [68]. Therefore, the combination therapy of
PDT-induced ICD. the enhanced PDT and anti-PD-L1 combination therapy displayed a
Compact ECM has been found to be a major barrier to tumor deep more significant suppression on both primary tumors and distant tumors
penetration of nanomedicines [68–72]. Recently, a variety of nano­ in 4T1 tumor-bearing BALB/c mice under laser irradiation [68]. In
systems have been designed to improve the tumor penetration of PSs for addition to HAase, Melittin (MLT) has also been found to enhance the
more efficient PDT and PDT-induced ICD [68–72]. For instance, Wang deep penetration of tumors by disrupting prokaryotic and eukaryotic
et al. developed a hyaluronidase (HAase)-modified dextran (DEX) cell membranes, which has also been utilized to facilitate the accumu­
polymer containing a pH-responsive linker for the degradation of the lation of PSs in tumors [69]. For instance, Liu et al. developed serum
cross-linked hyaluronic acid (HA) in the ECM, thus enhancing the albumin (SA)-coated boehmite NPs loading with both Ce6 and MLT
penetration capacity of PSs and then improving PDT and ICD induction peptide via electrostatic interactions, denoted as Ce6/MLT@SAB [69].
efficiency [68]. After accumulation at tumor sites, preinjected HAase Formulating MLT into the nanosystem coated with the SA not only
was released from the DEX-HAase NPs triggered by acidic stimuli to reduced the hemolysis of MLT, but also significantly enhanced the
loosen the ECM structure through degradation of HA [68]. penetration of Ce6 in tumors [69]. Under laser irradiation, because of

4
S. Zhang et al. Biomaterials 282 (2022) 121433

Fig. 4. Brief classifications and typical examples of photodynamic nanotherapeutics to induce ICD of tumor cells according to biomaterials and PSs.

Fig. 5. Schematic iIllustration of NK cell-membranes-cloaked NPs for PDT-enhanced cell-membrane immunotherapy [65]. Reprinted with the permission from (Deng
et al., 2018). Copyright (2018), American Chemical Society.

the increased penetration effect of MLT, Ce6/MLT@SAB evoked a more imiquimod (R837) were loaded between the UCNP core and the PEG
intense ICD following the CRT exposure and ATP release, and promoted shell by virtue of hydrophobic interactions [70]. Under the excitation of
the maturation of DCs and the tumor infiltration of CD4+ and CD8+ T 980 nm NIR light, two upconversion luminescence peaks (550 and 660
cells, resulting in augmented PDT-based immunotherapy in 4T1 nm) were emitted from those UCNPs. The emissive 660 nm laser could
tumor-bearing BALB/c mice [69]. excite Ce6 via the resonance energy transfer. As a result, the
In addition to the inefficient tumor penetration of nanocarriers, the UCNP–Ce6-R837 NPs-mediated PDT exhibited a more potent antitumor
limited tissue penetration of laser also significantly affects PDT-based efficacy than other control groups on mice bearing CT26 tumors [70].
ICD. Generally, the long-wavelength light has better security and Moreover, both TAAs from cell residues post-PDT and immunoadjuvant
stronger penetration ability than the short-wavelength NIR light [70–72, R837 could collaboratively elicit ICD and evoke robust antitumor
77]. Upconversion NPs (UCNPs) have been extensively investigated to response to eliminate the distant tumor [70]. In the further evaluation of
improve the problems of the low energy of long-wavelength light and immune response, the proportions of effector memory T cells of the
the poor penetration of short-wavelength light into tumor tissues to UCNP–Ce6-R837 NPs-mediated PDT were significantly elevated, per­
activate PSs and enhance PDT-based induction of ICD [70–72,77]. For forming an effective inhibition on the rechallenged CT26 tumors [70].
instance, Xu et al. reported multitasking UCNPs consisted of 20% of Yb Furthermore, polydopamine (PDA)-encapsulated core–shell UCNPs with
and 2% of Er-doped NaYF4. Then, the UCNPs were further coated by Ce6 (PDA@UCNP-PEG/Ce6 NPs) were also constructed to realize deep
amphiphilic polyethylene glycol (PEG)-grafted poly (maleic anhy­ tissue penetration of visible light via conquering the aberrant TME in
dride-alt-1-octadecene) (C18PMH-PEG). Finally, both Ce6 and solid tumors [72]. Firstly, PDA with good photothermal conversion

5
S. Zhang et al. Biomaterials 282 (2022) 121433

efficiency was used as the inner core, and then lanthanide (Ln)-doped be catalyzed by Fe3O to supply oxygen via Fenton-like reaction [79].
carbonate hydroxide layer (Gd:Yb,Er(OH)CO3) was coated onto the PDA The ameliorative hypoxic condition facilitated the generation of ROS
core to construct the PDA@Ln (OH)CO3 NPs. Finally, from TBP under NIR light irradiation. As a result, in vivo high CRT
PDA@UCNP-PEG/Ce6 NPs were obtained by loading Ce6 onto PDA@Ln expression and CTLs was measured in Fe-TBP treated tumor tissue,
(OH)CO3 NPs via the mixed stir with multiarm PEG polymers [72]. which indicated the CT26 tumor cells underwent ICD and the host im­
Under the irradiation of 980 nm laser, PDA@UCNP-PEG/Ce6 NPs mune response was successfully evoked. In a bilateral model of CT26
exhibited the high abilities of photothermal conversion and upconver­ tumors, Fe-TBP NPs significantly inhibited the growth of primary and
sion emission at 663 nm for the excitation of Ce6 molecules [72]. As a distant tumor by efficiently eliciting the PDT-based ICD and promoting T
result, the designed PDA@UCNP-PEG/Ce6 effectively eliminated the cell activation and infiltration in tumor [79]. Moreover, catalase (CAT)
primary tumor and furtherly prevented the metastasis and relapse of was also utilized to catalyze H2O2 to produce oxygen in solid tumors,
tumor through the ICD induced by the DAMPs release and the activation Zhang et al. and Meng et al. have reported related studies [82,83].
of antitumor response in a 4T1 tumor-bearing mouse model [72]. Other from catalyzing H2O2 to boost oxygen concentration, another
Moreover, hypoxia is another feature of the TME in solid tumors, alternative strategy is to reduce oxygen consumption within tumor cells.
which has also been found to severely imped PDT-based ICD. Tumor- Metformin (Met), a commonly used hypoglycemic agent, has recently
specific oxygen supply has been widely accepted as an efficient strat­ been found to effectively inhibit mitochondrial respiration [59,84,85].
egy to alleviate hypoxia [73,78–82]. Some metallic oxides, such as Mai et al. designed platelet membranes (PM) as a multifunctional
copper oxide, manganese oxide, and ferric oxide, are often used to nanoplatform to co-encapsulate IR780 and Met (PM-IR780-Met NPs)
catalyze H2O2 to produce oxygen [73,78–81]. For instance, Liang et al. [59]. Met could strikingly reduce tumor oxygen consumption through
designed a core-shell gold nanocage modified with manganese dioxide inhibiting mitochondrial respiration, thereby promoting IR780-induced
(AuNC@MnO2) as an oxygen supplier for PDT to enhance immuno­ PDT and ICD-based immunotherapy [59]. Under NIR light irradiation,
therapy (Fig. 6) [73]. When exposed to acidic stimuli at tumor sites, PM-IR780-Met NPs displayed effective PDT and DAMPs were released
MnO2 in the shell of the AuNC@MnO2 NPs reacted with H2O2 to release from dying tumor cells following with the occurrence of ICD [59].
oxygen in situ to overcome hypoxia, thus facilitating the generation of Meanwhile, the relieved tumor hypoxia could also reduce the immune
ROS and the ICD induction of 4T1 tumor cells. Subsequently, these ICD suppressive cells to further enhance the antitumor therapy. Finally,
signal molecules (CRT, ATP, HMGB1) from 4T1 tumor residues could tremendous CTLs cells were recruited and invoked, providing a prom­
promote the maturation of DCs and the activation of T cells [73]. In a ising platform for elimination of the primary tumors and prevention of
4T1 tumor xenograft BALB/c mouse model, AuNC@MnO2 NPs exhibited the lung metastasis in a 4T1 tumor bearing mouse model [59].
potent antitumor activity, ablating the primary tumors under NIR light
irradiation and preventing tumors metastasis due to the activated im­
2.3. Precisely targeting ER
mune system [73]. Moreover, Lan et al. constructed a MOF-based
nanoplatform composed of Fe3O clusters and TBP (a photosensitizer)
ER pressure has been found to be a key factor for the release of
[79]. Upon exposed to hypoxic environment, intracellular H2O2 would
DAMPs. Therefore, accurately targeting of PSs to ER is of great

Fig. 6. Schematic illustration of the design and function of AuNC@MnO2 (AM) as TME responsive oxygen producer for oxygen-boosted immunogenic PDT [73].
Reprinted with the permission from (Liang et al., 2018). Copyright (2018), Elsevier Ltd.

6
S. Zhang et al. Biomaterials 282 (2022) 121433

importance for the induction of ICD [48,49]. For example, Li et al. re­ amounts of carrier materials has long been a challenge for PDT [75,86].
ported indocyanine green (ICG)-coupled hollow gold NPs (FAL-IC­ The ACQ effect significantly impairs the generation and diffusion of
GHAuNS), which were decorated with ER-targeting pardaxin (FAL) ROS. Therefore, how to realize tumor-specific release of PSs from
peptides (Fig. 7) [48]. Moreover, a FLA-modified liposome containing nanocarriers is of crucial importance for PDT-based immunotherapy.
the oxygen-supplying Hb was integrated into the system to alleviate Compared to normal tissues, most tumors are characterized with highly
hypoxia [48]. The composite nanosystem induced robust ER pressure acidic and redox microenvironment [75]. Recently, several tumor
under laser irradiation. As a result, a large amount of CRT was exposed stimuli-responsive nanosystems loaded with PSs have been developed to
on cell membrane to stimulate the maturation of DCs and the prolifer­ address the ACQ problems and to promote PDT-based cancer immuno­
ation of CD8+ cells [48]. The ER-targeting nanosystem effectively acti­ therapy [63,75,86]. For instance, Wang’s group developed
vated the dormant host immune system, facilitating a potent acid-activatable cationic nanomicelles co-loaded with PSs (Pyropheo­
ICD-associated immunotherapy in a B16 tumor-bearing mouse model phorbide a, PPa) and small interfering RNA (siRNA) [86]. After the
[48]. tertiary amino groups of pH-responsive diblock copolymer fully pro­
In addition to modifying nanocarriers with ER-targeting ligands, tonated in the acidic TME, the designed micelleplexes were dissociated,
formulating PSs with ER-targeting capacity into nanocarriers represents and PPa was rapidly released from the nanosystem [86]. Upon NIR light
another strategy for PDT-based ICD induction. For instance, Deng et al. treatment at pH 6.0, the nanomicelles generated much more ROS
designed and synthesized an ER-targeted photosensitizer, named TCPP- (5.6-fold) than that at pH 7.4, resulting in more efficient ICD of B16–F10
TER, which was loaded into reduction-sensitive Ds-sP NPs through thiol- tumor cells [86]. The high expressions of heat shock protein 70 (HSP70)
disulfide exchange reaction [49]. When exposed to the high level of and NF-κB in B16–F10 tumor cells were observed in the treatment group
glutathione (GSH) in tumor cells, TCPP-TER could be quickly released of pH-responsive nanomicelles, suggesting the activation of the innate
from the Ds-sP/TCPP-TER NPs. Then, TCPP-TER was selectively accu­ and adaptive immune response [86]. As a result, the pH-responsive
mulated in ER to induce intense ER pressure by the generated abundant nanomicelles showed significant inhibition on tumor growth and
ROS [49]. Both in vitro and in vivo results demonstrated that ER-targeting metastasis by evoking antitumor immune response and recruiting tumor
PDT could elevate ICD level and promote release of DAMPs [49]. In a infiltrating CD4+ and CD8+ T cells in a B16–F10 melanoma xenograft
4T1 tumor-bearing mouse model, Ds-sP/TCPP-TER NPs-mediated PDT tumor model.
exhibited potent antitumor effect for the primary tumors [49]. In addition to acidic stimulation, high concentrations of intracellular
Furthermore, an outstanding anti-distant tumor effect was obtained in GSH and ROS were also utilized to develop redox-responsive photody­
the Ds-sP/TCPP-TER NPs-treated group [49]. Meanwhile, the augmented namic nanotherapeutics for alleviation of the ACQ of PSs [63,75]. For
secretion of cytokines and infiltration of CD8+ T cells were observed at instance, Liu et al. designed redox-activated liposomes (RAL) consisted
the tumor tissue, suggesting that the ER-targeting PDT strategy could of disulfide bond-linked phospholipid-porphyrin conjugates [75]. Upon
amplify immunotherapeutic efficiency to inhibit the growth and internalization into tumor cells, porphyrin could be rapidly released in
metastasis of tumors [49]. the presence of high concentration of intracellular GSH, thereby acti­
vating the fluorescence signal and ROS generation capacity of porphyrin
2.4. Addressing the ACQ effect [75]. Moreover, GSH depletion contributed to the sensitivity of PDT
[75]. Under NIR light irradiation, efficient PDT-induced ICD of tumor
Although a variety of nanomaterials have been used for improving cells following with CRT exposure, HMGB-1 release, and ATP secretion
the delivery efficiency of PSs, the ACQ effect of PSs trapped in large was observed, and more activated CTLs were transferred to tumor

Fig. 7. The antitumor mechanism of FAL-ICG-HAuNS plus FAL-Hb-lipo. Schematic illustration of enhanced immunogenic cancer cell death and anticancer effect
induced by ER-targeting photothermal therapy (PTT)/PDT [48]. Reprinted with the permission from (Wei et al., 2019). Copyright (2019), Nature Communication
Publishing Group.

7
S. Zhang et al. Biomaterials 282 (2022) 121433

tissues, effectively eradicating the primary tumors and contralateral et al. developed PDA-encapsulated Ce6-loaded core–shell UCNPs for
tumors and preventing the occurrence of metastatic nodules in 4T1 PTT&PDT induced ICD [99]. Upon laser irradiation, the PDA core could
breast tumor-bearing mice [75]. perform an optimal photothermal effect for PTT while synchronously
Moreover, several dual-responsive nanosystems have been devel­ optimized the generation of ROS for PDT [99]. The combinational
oped for PDT-based ICD and immunotherapy [74,87]. Compared with application of PDT and PTT in vivo effectively promoted the release of
the single-responsive nanosystem, the dual-responsive NPs could release TAAs and DAMPs, triggered the maturation of DCs and robustly acti­
PSs more quickly, and sufficiently alleviate the ACQ effect and enhance vated CTLs against the metastasis and relapse of 4T1 tumors [99]. Taken
PDT efficiency [74,87]. For instance, Wang et al. fabricated together, the photothermal/photodynamic combination offers a
GSH/pH-triggered mesoporous organosilicas (MONs) loaded with Ce6, high-efficiency synergistic therapeutic modality for cancer
and was further modified with magnetic heads (M-MONs@Ce6). In immunotherapy.
comparison with the single stimulation of GSH or acid, M-MONs@Ce6
exhibited more thorough degradation and faster drug release, which 3.2. chemo-photodynamic synergistic induction
enhanced the photodynamic efficiency of Ce6 and reduced the toxicity
to normal tissues [87]. When exposed to NIR light and alternating cur­ In addition to PDT, recent evidence has revealed that some chemo­
rent magnetic field (ACMF), the nanosystems induced the greatest therapeutic drugs such as oxaliplatin (Oxa), cyclophosphamide, pacli­
amount of CRT exposure and HMGB1 release to evoke robust ICD of taxel (PTX) or Dox could also induce ICD of tumor cells and provoke the
tumor cells, due to efficient generation of ROS and magnetic hyper­ host immune response [26,88,100–104]. Therefore, the combination of
thermia. The induced ICD could further promote DCs maturation and T PDT and chemotherapy will initiate a stronger immune response than
cells activation, priming tumor-specific immune response in 4T1 PDT and chemotherapy alone [74,88,101,105]. For instance, He et al.
tumor-bearing mice [87]. Consequently, the activated immune response developed a core-shell nanoscale coordination polymer (NCP) contain­
exhibited highly effective suppression on both primary tumors and ing Oxa and photosensitizer pyropheophorbide-lipid conjugate (pyroli­
metastatic tumors in 4T1 tumor-bearing mouse models [87]. pid) [88]. In bilateral tumor models of MC38, the effective
chemo-photodynamic synergistic therapy between Oxa and PPa pro­
3. PDT-based synergistic induction of ICD voked a high level of ICD, leading to CRT exposure, antitumor vacci­
nation and an abscopal effect [88]. Recently, Huang et al. developed
Though PDT has been extensively explored to induce ICD, there are engineered macrophages (Oxa (IV)@ZnPc@M) carrying nanostructured
still some inherent limitations of PDT that are not sufficient to evoke a lipid, Oxa precursors and Zinc phthalocyanine (ZnPc, as PSs) [90].
strong immune response [88,89]. Recently, multiple combined thera­ Firstly, Zn2+ was crosslinked with the carboxyl group of Oxa prodrug to
peutic strategies have been used to elevate the efficiency of ICD by form the core, and ZnPc was simultaneously loaded in the lipid bilayer
compensating the deficiencies of a single PDT [90–92]. The synergistic through hydrophobic interaction for the construction of Oxa (IV)@ZnPc
efficacy to induce ICD can be achieved by combining PDT with (Fig. 8) [90]. Then, Oxa (IV)@ZnPc was further loaded into macro­
chemotherapy, PTT, radiotherapy and so on. phages to improve tumor-targeted ability [90]. Under NIR light irradi­
ation, drugs inside Oxa (IV)@ZnPc were released and then performed a
3.1. Photothermal/photodynamic synergistic induction combined chemo-photodynamic therapy with the second laser, which
led to successfully kill 4T1 primary tumors [90]. Meanwhile, DAMPs
Similar to PDT, non-invasive PTT can convert light into heat energy, released from killed 4T1 tumor cells could cause ICD of tumor cells and
which enables the temperature of the cells to be heated up rapidly, potentiate the antitumor immune response against primary and bone
killing the cell in the form of thermal ablation [93,94]. Recent studies metastatic tumors in a 4T1-bearing mouse model [90].
also described that PTT could cause antigen release and activate im­ In addition to Oxa, existing researches also clarified the capability of
mune response, resulting in significant enhancement in the level of ICD Dox for ICD induction [74,101,106]. Hu’s research group designed a
via combining with PDT [95,96]. Apart from acting as a photosensitizer cascade chemo-photodynamic therapy (Ce6/Dox) with ROS-responsive
for PDT, ICG also can serve as a light absorber for PTT [92]. Wang’s lipid-polymer hybrid NPs (TKHNP-C/D) to promote ICD and tumor
group fabricated a light-triggered and antigen-capturing UCNPs-based immunotherapy [101]. Under 660 nm light irradiation, Dox was rapidly
nanoplatform (UCNP/ICG/RB-mal), which was obtained by assembling released through the degradation of the designed nanocarrier, formu­
ICG and DSPE-PEG-maleimide onto UCNPs and then loading the lating an efficient synergy with PDT to stimulate the maturation of DCs
photosensitizer rose bengal (RB) [92]. In the nanoplatform, PDT and and the infiltration of T cells [101]. Furthermore, a dual-sensitive
PTT can be simultaneously achieved to enhance the release of antigens (GSH/pH) prodrug-based nanoplatform was designed by conjugating
under the NIR laser irradiation [92]. In addition, the TAAs and DAMPs 5-aminolevulinic acid (5-ALA, as PSs) and Dox via hydrazine backbones
released from 4T1 cells could be captured in situ due to the maleimide, and disulfide bonds [74]. When exposed to the intracellular TME, the
further enhancing the uptake and presentation of tumor antigens [92]. dual-response prodrug self-assembly NPs could rapidly release 5-ALA
In vivo results demonstrated that the synergistic treatment of PDT and and Dox, and the 5-ALA was subsequently converted to protoporphy­
PTT significantly inhibited the growth of primary and distant 4T1 tu­ rin IX (PpIX) by intracellular enzyme catalysis [74]. Under NIR light
mors, contributing to a potent therapeutic effect in a highly metastatic irradiation, the generated ROS and the released Dox synchronously
4T1 mammary tumor model [92]. Moreover, due to the effective in­ induced ICD with the release of calreticulin, HMGB1, and ATP from
duction of ICD, the combination group of UCNP/ICG/RB-mal and im­ dying 4T1 cells. These released DAMPs subsequently activated DCs and
mune checkpoint blockade (ICB) successfully rejected 4T1 tumor stimulated host immune response to lead to the increased number of
rechallenge and realized long-term immune response [92]. CTLs at tumor site [74]. Ultimately, the prodrug-based chemo-­
Due to the intrinsic absorption to light, amounts of inorganic nano­ photodynamic strategy not only significantly reduced the undesirable
therapeutics such as Au NPs [48,96], CuS NPs [97], ZrC [98] or black side effects but also markedly inhibited the growth and metastasis of
phosphorus NPs [95] have been developed for PTT. Liu et al. constructed tumor in a 4T1 mouse model [74].
an ultrasmall gold nanocluster (Au25Capt18) for the combined PTT and
PDT against skin cancer [96]. Upon laser irradiation, the prepared Au 3.3. Other synergistic induction combos
NPs could generate high level of photothermal energy and ROS, eliciting
robust ICD of cutaneous squamous cell carcinoma [96]. In addition to In addition to some conventional treatments, the combination of PDT
metallic material, some polymers have also been applied for PTT in with radiotherapy or sonodynamic therapy has also been explored to
combination strategies for potentiating ICD induction. For instance, Yan synergistically induce ICD [91,107,108]. Radiotherapy involves one or

8
S. Zhang et al. Biomaterials 282 (2022) 121433

Fig. 8. The preparation and mechanism of the artificially engineered macrophages. a Schematic illustration of the preparation of Oxa (IV)@ZnPc@M. b The
mechanism of Oxa (IV)@ZnPc@M mediated chemo-photodynamic therapy to trigger robust antitumor immune responses and potentiate PD-L1 blockade immu­
notherapies for anti-primary and bone metastatic tumors [90]. Reprinted with the permission from (Wei et al., 2021). Copyright (2021), Nature Communication
Publishing Group.

more kinds of ionizing radiation such as X-rays and high-energy electron radiotherapy. On the other hand, X-ray triggered the generation of 1O2
beams to induce the DNA damage in tumor cells [109,110]. Recently, from PSs to facilitate PDT, which was known as radiodynamic therapy
there’s evidence that radiotherapy-induced DNA damage is capable of (RDT) [114]. Under the induction of X-rays, MOFs loaded with
triggering ICD and stimulating T cell proliferation [109,110]. Moreover, indoleamine-pyrrole 2, 3-dioxygenase (IDO) inhibitor (IDOi@nMOFs)
the combination of low dose photosensitizer and early radiotherapy can significantly promoted the CRT exposure and evoked ICD, and the
significantly enhance PDT-based antitumor immune response [111]. released antigens enabled the effective tumor infiltration of CD8+ T cells
Benefiting from the excellent tissue penetration of X-rays, researchers via radiotherapy-radiodynamic therapy in syngeneic mouse models of
have utilized scintillators and metals to transfer the energy of X-rays to breast and colorectal cancers [114]. Additionally, the distant tumor
PSs for boosting PDT efficacy [107,112–115]. For instance, Lu et al. growth was also remarkably suppressed, suggesting the excellent sys­
constructed 5, 15-di (p-benzoato) porphyrin-Hf (DBP-Hf) MOFs and 5, temic immune responses caused by IDOi@nMOFs [114].
10, 15, 20-tetra (p-benzoato) porphyrin-Hf (TBP-Hf) MOFs based on Hf Additionally, sonodynamic therapy (SDT) is also a local therapy
clusters and porphyrin-based PSs [114]. On the one hand, X-ray photos modality that uses ultrasound to activate sonosensitizers for ROS gen­
could be absorbed by Hf clusters and produced •OH radicals for eration [116]. The combination of PDT and SDT has been investigated to

9
S. Zhang et al. Biomaterials 282 (2022) 121433

enhance the anti-tumor effect and reduce the toxic side effects [117, [131,133]. For example, Im et al. designed a hypoxia-triggered meso­
118]. Notably, the fragments produced by SDT-mediated apoptotic or porous silica nanocarrier (CAGE) [131]. In the nanosystem, Ce6 was
necrotic cancer cells were able to act as antigens to activate host anti­ conjugated within the mesoporous silica nanoparticle decorated by Azo
tumor immunity [119–121]. More importantly, SDT-PDT synergistic linker, and then the adjuvant (CpG) was loaded into the CAGE complex
induction can efficiently evoke ICD, thereby activating a robust immune via electrostatic interaction [131]. Once CAGE was in a hypoxic condi­
response [122,123]. Particularly, two-dimensional (2D) carbon nitride tion within tumor tissues, CpG could be rapidly released by the cleavage
(g-C3N4) nanosheets with good biocompatibility and abundant surface of the Azo linkers [131]. Under local NIR light irradiation, ROS-induced
amino groups have been utilized for efficient delivery of photo/­ DAMPs from B16⋅Mo5 cells and the released CpG simultaneously
sonosensitizers [123,124]. For instance, Chen et al. designed a recruited DCs and promoted the maturation of DCs to present antigen
metal-free g-C3N4/Ce6 nanohybrid for combined immunotherapy [131]. As results, a robust antitumor response was evoked and a large
[123]. Ce6 was loaded in the nanosheets and acted as a dual-functional number of CTLs infiltration at tumor site was observed in the B16⋅Mo5
photo/sonosensitizer [123]. The nanohybrids not only displayed potent tumor bearing mice treated with CAGE + laser [131]. Moreover, Yang
cytotoxicity against 4T1 cells, but also effectively promoted the matu­ et al. also developed a multipurpose polymersomal nanoformulation
ration of DCs after NIR light and ultrasound treatment in vitro [123]. The encapsulating HPPH and Dox [132]. Interestingly, the chimeric
nanohybrids effectively stimulated immune responses in vivo, resulting cross-linked polymersome (CCPS) with primary and tertiary amines
in potent tumor immunotherapy in 4T1 tumor-bearing mice [123]. could act as an adjuvant, together with TAAs released from MC38 cells
In addition to combinations mentioned above, more interestingly, by PDT to enhance the level of DCs recruitment [132]. In vivo, high level
the researchers found that some versatile nanomaterials could also be of mature DCs in lymph node and CTLs at tumor site were observed due
used to enhance the ICD [91]. Yang et al. designed a unique to the “in situ vaccine” acted by CCPS/HPPH/Dox under NIR light irra­
nanovesicle-mediated and PDT-based nanoassemblies [91]. The diation [132]. It followed that the combination of ICD and adjuvant
pH-responsive nanovesicles (pRNVs) composed of block copolymer exerted potent PDT-synergized immunotherapy, leading to the highly
polyethylene glycol-b-cationic polypeptide, acted as not only a carrier effective suppression on primary and distant tumors in MC38
but also an ICD inducer [91]. Compared to HPPH-mediated PDT alone, tumor-bearing C57BL/6 mice [132].
such pRNVs loaded with HPPH could impressively elevate the ICD level In addition, the effective combination of ICD and vaccine could also
of tumor cells with high CRT exposure, TAAs secretion, DC recruitment, enhance immune stimulation and further potentiate immunotherapy.
maturation, migration, and CD8+ T cell activation, resulting in a potent Recently, Cheng et al. synthesized a chimeric peptide PpIX-PEG8-
suppression on primary and distant tumors in a B16F10 melanoma KVPRNQDWL, which could self-assembly into NPs (PPMA) with high
tumor model [91]. Overall, these findings suggest that the combination drug loading rate [134]. In this nanosystem, the melanoma specific
of PDT and other therapeutics could efficiently enhance the ICD levels of antigen (KVPRNQDWL) peptide could activate the infiltration of specific
tumor cells induced by PDT alone and stimulate an intense antitumor cytotoxic T cells into tumor tissues and synergistically amplify
immune response. PDT-based immunotherapy against malignant melanoma [134]. To
further elevate the level of antigen presentation, Xu et al. constructed a
4. Multimodal cancer immunotherapy in basis of PDT-induced biodegradable mesoporous silica nanoparticle (bMSN) nanoplatform
ICD with the co-loading of neoantigen-based vaccine, CpG oligodeox­
ynucleotide adjuvant, and photosensitizer Ce6 [135]. Compared with
Nowadays, diverse strategies have been explored for enhancing PDT single PDT, vaccine, or adjuvant, the multifunctional nanomaterial
and inducing ICD of tumor cells to activate the host immune response system recruited more tumor-infiltrating cytotoxic T-cells and exhibited
against cancer [125–127]. However, cancer immunotherapy is an a more potent antitumor efficacy on primary and contralateral MC-38
extremely complicated process, which can be broadly summarized in tumors in multiple murine models of bilateral tumor [135]. Therefore,
three stages: (i) antigen presenting stage; (ii) effector T cell proliferation the multiple-stimuli design offered a promising approach for eliciting
and differentiation stage; and (iii) tumor elimination stage [128]. The potent antitumor immune response [135].
ICD of tumor cells is regarded as the largest contributor to the first stage
of immunotherapy [128]. Whereas, the failure at any of these stages can 4.2. Combination with checkpoint blockade
easily diminish the effectiveness of cancer immunotherapy [128]. For
example, IDO can attenuate the activation of effector T cells [128]; Immune checkpoints are functional molecules that negatively regu­
cytotoxic T lymphocyte antigen 4 (CTLA-4) can suspend the activated T late the host immune response under physiological conditions, which
cells and mediate the inhibitory function of regulatory T (Treg) cells could maintain the host immune tolerance and reduce the damage to
[129]; and PD-1/PD-L1 can interfere with the recognition of tumor by tumor tissues in the process of immune response [136,137]. One of the
effector T cells, all of which can reduce the treatment efficiency of tumor mechanisms of tumors escaping immune attack is to down-regulate the
immunotherapy [130]. Therefore, single-modal cancer immunotherapy immune response through immune checkpoints, which modulate almost
based on PDT-driven ICD is not enough to attain a satisfactory therapy each step of the tumor immune cycle. Especially for tumor elimination
outcome. It is necessary to combine ICD-based PDT with other immu­ stage, once the PD-1 receptor expressed on the surface of immune T cells
notherapies for multimodal cancer immunotherapy. combines with the PD-L1 ligand expressed on the surface of tumor cells,
a negative control signals will be generated to induce T cells entering
4.1. Combination with adjuvants or vaccines into the resting state [128]. Following the conveyed signal, the prolif­
eration of CD8+ T cells in lymph nodes and the recognition of tumor cells
Antigen presentation, as the first stage of the immune response, is at tumor site will be reduced, and eventually result in the occurrence of
considered as the initiation of the immune response [128]. However, the immune escape. The combination of PDT and anti-PD-1/PD-L1 antibody
efficiency of antigen presentation alone from the stimulation of the ICD could effectively unshackle the immunosuppression of immune check­
is sometimes not enough to induce intense immunotherapy. Combining point PD-1/PD-L1 and enhance immune response against tumor [125,
ICD with vaccines or adjuvants might reinforce the generation of 138–140].
immunogenic fragments or other danger signals in tumor tissues [131, Recently, there have been many studies about the application of
132]. Cytosine-phosphate-guanine (CpG), as a Toll-like receptor 9 PDT-mediated ICD induction combined with anti-PD-1/PD-L1 antibody
(TLR9) agonist, can provoke the host immunity and stimulate cytokine [125,138–140]. Duan et al. designed nontoxic core-shell Zn-pyr­
secretion by priming immature DCs. Recently, CpG has been proved as a ophosphate (ZnP) NPs loaded with the photosensitizer pyrolipid
remarkable immune-adjuvant against solid tumors in clinical studies (ZnP@pyro) [139]. Under NIR light irradiation, the ZnP@pyro could

10
S. Zhang et al. Biomaterials 282 (2022) 121433

both kill tumor cells and trigger the ICD-based host immune response [129].
[139]. In vivo results, ZnP@pyro-mediated PDT not only successfully T cells will stop proliferating once exposed to low concentration of
inhibited the primary 4T1 tumor but also effectively prevented the lung tryptophan. IDO, as a neo-immune checkpoint protein, can discontinue
metastasis [139]. Furthermore, the combination of ZnP@pyro-mediated the activation of T cells by degrading tryptophan [143,144]. Both IDO
PDT and anti-PD-L1 antibody could sensitize tumor cells to effector T expressed in the tumor cells and some APCs can mediate the immune
cells, enhance the inhibition of primary 4T1 tumors and simultaneously escape of the tumor by inhibiting the proliferation of T cells [143,144].
suppress the growth of non-laser distant 4T1 tumors in both 4T1 and IDO inhibitors can regulate the immunosuppressive TME and promote
TUBO bilateral syngeneic mouse models [139]. Except for large­ the activation and proliferation of effector T cells [145,146]. The com­
–molecule PD-1/PD-L1 antibodies, small-molecule PD-1/PD-L1 in­ bination of PDT and IDO inhibitor can significantly enhance the
hibitors have also been widely investigated recently [130]. BMS-202, a immunotherapeutic effect [91,105,126,128,147,148]. For example, NPs
small-molecule PD-1/PD-L1 inhibitor developed by BMS, could prevent loaded with HPPH and IDO inhibitor indoximod (IND) could induce ICD
the PD-1/PD-L1 interaction and enhance immunotherapy [130]. Zhang of tumor under NIR light irradiation. The proliferation of CD8+ T cells
et al. constructed a co-assembly nanoplatform of Ce6 and BMS-202 to was promoted with the help of IND, resulting in an enhanced antitumor
accomplish the synergistic effect between PD-1/PD-L1 pathway and PDT immunotherapy in 4T1 tumor-bearing mice [91]. Song et al. synthesized
[130]. Ce6 and BMS-202 could assemble into NPs without other carrier a caspase-responsive chimeric peptide PpIX-1MT by integrating photo­
materials by one-step nanoprecipitation method. Due to the high drug sensitizer PpIX with IDO inhibitor 1-methyl-tryptophan (1 MT) [50].
loading, Ce6/BMS-202 NPs exhibited highly effective inhibition on The PpIX-1-mt could self-assemble into NPs and target to tumor tissue by
primary tumor, and concurrently caused ICD of 4T1 tumor cells with the enhanced penetration retention (EPR) effect [50]. Upon NIR light
CRT exposure under NIR light irradiation. Moreover, Ce6/BMS-202 NPs irradiation, the PpIX-1MT NPs generated ROS to induce the ICD of CT26
mediated combination therapy of ICB and PDT displayed significant tumor cells following with high CRT exposure and trigger the immune
suppression on distant tumor with efficient maturation of DCs and response [50]. Then, the subsequently released 1 MT via the cleavage of
infiltration of CTLs cells into the 4T1 tumors. More importantly, the the caspase could improve the activation efficiency of CD8+ cells [50].
combination therapy of PDT-based ICD and PD-L1 blockade was able to Consequently, a cascaded synergistic therapy inhibited both primary
successfully prevent lung metastasis and inhibit cancer relapse, making and lung metastasis tumor successfully in CT26 tumor-bearing mice,
a tumor prophylactic vaccination effect [130]. which might provide a promise for preventing tumor recurrence and
Another strategy to block PD-1/PD-L1 is to genetically regulate the metastasis clinically [50]. In addition to IND and IMT, some other small
PD-1/PD-L1 pathway via RNA interference (RNAi) [86,141]. Wang et al. molecule IDO inhibitors such as INCB24360 and NLG919 were also used
developed an acid-sensitive cationic micelleplex by integrating photo­ to strengthen immune response [105,126]. Recently, nanoassemblies
sensitizer (PPa) and siRNA [86]. When the micelleplex was caught in the with multiple therapy modalities have attracted more attention from
acid condition in tumor cells, both PPa and siRNA were rapidly released, researchers. Li et al. constructed a three-in-one immunotherapy nano­
and abundant ROS were generated under NIR irradiation [86]. The platform by a cascade enhancement on the three stages of immuno­
released siRNA specifically suppressed PD-L1 pathway and modulated therapy [128]. The multifunctional nanoplatform was composed of
immune tolerance via silencing the expression of PD-L1 on the surface of Ce6-conjugated HA (HC) with negative charge, dextro-1-methyl tryp­
tumor cells [86]. In a B16–F10 melanoma xenograft tumor model, the tophan (1-mt)-conjugated polylysine (PM) with positive charge and
combination of PDT and PD-1/PD-L1 block showed a more potent anti-PD-L1 monoclonal antibodies (aPD-L1) (Fig. 9) [128]. After accu­
immunotherapeutic effect on tumor growth, and the lung metastasis was mulation in tissues, the aPD-L1@HC/PM NPs firstly underwent charge
well suppressed [86]. In addition, Liu fabricated a CaCO3/MnO2-based reversal due to the overexpressed HAase, and aPD-L1 was released
nanoplatform with TME responsive biodegradable properties to enhance during this process. The positive charged nanoassemblies were easier to
PDT and strengthen PD-L1 blocking immunotherapy [142]. The NPs be endocytosed into cells, and could release the Ce6 and 1-mt under the
loaded with ICG and siRNA (Mn@CaCO3/ICG@siRNA) could relieve the action of cathepsin B. Under NIR light irradiation, Ce6 produced cyto­
hypoxic environment and accomplish the degradation of carrier by toxic ROS to induce robust ICD with higher CRT exposure on B16F10
consuming H+ and H2O2 in tumor cells [142]. Due to the oxygen sup­ cells [128]. Meanwhile, IDO inhibitor 1-mt could enhance the activation
plement, the enhanced PDT promoted the release and presentation of and proliferation of effector T cells, and aPD-L1 facilitated T cells
TAAs to induce intense ICD of tumor cells and activated the effector T eliminating tumor cells. As a result, the three-in-one immunotherapy
cells [142]. With the help of siRNA, the activated effector T cells nanoplatform not only inhibited the growth of primary tumor but also
effectively recognized and killed cancer cells, achieving synergistic ef­ prevented the distant tumor growth and lung metastasis, thereby per­
fect between PDT and PD-L1 blocking immunotherapy in the Lewis lung forming an excellent cascade-amplifying immunotherapy effect in
cancer-model [142]. B16F10 tumor-bearing mice [128].
In addition to PD-1/PD-L1 checkpoint, CTLA-4 overexpressed on In addition, the inhibitors or agonists of OX40 or TIM-3 have
activated CD4+ and CD8+ T cells, has a high homology with the co- received considerable attention in the field of immunotherapy. OX40
stimulating molecular receptor (CD28) on the surface of T cells. Anti- (CD134, TNFRSF4) was originally defined as a T cell activation marker,
CTLA-4 can block inactivation of effector T cells and CTLA-4- but it was later found to be a member of the co-activating NGFR/TNFR
dependent immune evasion [92,129]. Some researchers have demon­ superfamily [149]. OX40 is mainly expressed on the activated effector T
strated that combining PDT-inducted ICD with CTLA-4 checkpoint cells, regulatory T cells (Tregs), NK cells and neutrophils. Anti-OX40
blockade could improve the synergistic immunotherapy [92,129]. To strategies have been proved to improve the immune system responses
enhance the combination effect, Xu et al. designed Ce6-based image-­ via multiple routes, mainly including: (i) increasing the survival and
guided PDT in combination with PD-L1 and CTLA-4 dual checkpoint expansion of effector and memory T cells; (ii) promoting the secretion of
blockade [129]. They found that Ce6 + immunoglobulin G could cytokines, such as IL-2, IL-4, IL-5 and IFN-γ; and (iii) inhibiting the
assemble into nanostructure Chloringlobulin with the assist of poly­ immunosuppressive Tregs [149]. However, anti-OX40 agonists alone
vinylpyrrolidone (PVP) [129]. Subsequently, they utilized the αPD-L1 or could not yield satisfactory therapeutic outcomes, especially the meta­
anti-CTLA-4 antibody (αCTLA-4) to prepare αPD-L1 Chloringlobulin, static tumors. To address this challenge, combination of anti-OX40 ag­
αCTLA-4 Chloringlobulin and αPD-L1αCTLA-4 Chloringlobulin, and onists with other therapeutic modalities have been explored to enhance
further demonstrated the advantages of double-blocking immune the effectiveness of immunotherapy [150,151]. For instance, Alvim
checkpoint over single-blocking immune checkpoint, which could elicit et al. found that the combination of PDT with PD-1 inhibitor and OX40
robust host antitumor immunity and a persistent immune memory agonist significantly enhanced antitumor immune response [152]. In
against tumor rechallenge in GL261 orthotopic tumor-bearing mice mice allografted with MB-49 UTUC cells, the combination therapeutics

11
S. Zhang et al. Biomaterials 282 (2022) 121433

Fig. 9. The assembly and step-by-step release behavior f aPD-L1@HC/PM NPs (A) Assembly strategy for aPD-L1@HC/PM NPs. (B) Illustration of the step-by-step
detached release behavior of aPD-L1, Ce6, and 1-mt and the immunotherapy capability via the cascade-amplifying cancer-immunity cycle [128]. Reprinted with the
permission from (Li et al., 2019). Copyright (2019), American Chemical Society.

demonstrated distinct advantages over the single treatment in terms of [127].


tumor suppression, metastasis inhibition and immune response [152]. Sorafenib, a multi-kinase inhibitor, could suppress the immunosup­
Unlike other immune checkpoint molecules, TIM-3 was found to be pressive cells and potentiate tumor-specific cytotoxic T cell in TME,
specifically upregulated in CD4+ helper T cell 1 (Th1) and CD8+ cyto­ performing an augmented antitumor immune response [158]. Sun et al.
toxic T cells [153]. After being activated by its ligand galectin-9, TIM-3 designed ROS-responsive NPs (NPs-sorafenib/Ce6) co-loaded with sor­
inhibits the activity of effector T cell, resulting in T cell depletion in afenib and Ce6 to enhance antitumor immunotherapy [158]. Under NIR
tumors [154]. Furthermore, it has been found that animals with high light irradiation, large quantities of cytotoxic ROS were produced,
TIM3 expressions usually show resistance to anti-PD-1 therapy [155]. which induced the apoptosis of tumor cells and the release of TAAs, from
Therefore, a combination of anti-TIM3 and anti-PD-1 strategies repre­ dying tumor cells, and accelerated the release of intratumoral sorafenib
sents a promising tumor immunotherapy [156,157]. For instance, [158]. The released sorafenib subverted the immunosuppressive
Huang et al. developed a liposome encapsulated with ICG that could microenvironment and enhanced the antitumor immune response
eradicate the primary tumors upon NIR light irradiation [157]. When induced by PDT, improving the infiltration of cytotoxic CD8+ T cells in
combined with PD-1/TIM-3 blockade, the combination strategy effec­ tumor tissues and finally furnishing a potent suppression of 4T1 tumor
tively suppressed the distant tumors in two colon cancer animal models elimination and metastasis [158]. Moreover, the measure results of
(CT26 and MC38) [157]. tumor-infiltrating CD8+ demonstrated PDT-based combination with
sorafenib could induce strong and long-lasting antitumor immune
response against 4T1 tumor-bearing BALB/c mice [158].
4.3. Combination with other therapeutics
5. Conclusions
In addition to the combinations mentioned above, there are other
alternative combination approaches for enhancing PDT-synergized im­ Cancer immunotherapy is currently one of the most promising
mune response [59,127,158]. Imatinib (IMT), a novel inhibitor of Treg treatment modalities, but its application is limited by its low immune
cells activation, could readily cut down Treg cells-mediated immuno­ response and potential side effects. Non-invasive and spatiotemporal
suppressive function by inhibiting the expression of transcription factors PDT can induce ICD of tumor cells and further stimulate immune
STAT3, STAT5 and Foxp3 [127]. Ou et al. designed a pH-sensitive and response. Therefore, effective PDT enables to not only kill tumor cells
layer-by-layer hybrid nanoassembly loaded with IR-780 and IMT [127]. but also reverse the immunosuppressive TME, and further achieve a
Firstly, the GITR antibody with the function of targeting to Treg cells desired synergy with other immunotherapeutics. The application of
was connected to PLGA [127]. Subsequently, IMT and IR-780 were biomedical nanotechnology in PDT and PDT-based ICD induction has
incorporated into this nanosystem, which were further coated with shown many advantages, such as improving the physicochemical
pH-sensitive polymers [127]. Upon endocytosed into the acid tumor properties of PSs, extending the blood circulation time, facilitating
environment, the designed NPs would release IR-780 and IMT. Under tumor-targeting accumulation and enhancing PDT-induced ICD. More­
NIR light irradiation, IR-780-mediated PDT could induce the release of over, the integration of PSs with various nanomaterials can not only
TAAs and DAMPs, promote DCs maturation and antigen presentation to overcome challenges of PDT, such as hypoxia, ACQ effect, poor pene­
T cells [127]. Meanwhile, the introduction of IMT disrupted the tration and targeting ability, but also enhance the induction of ICD and
immunosuppressive function of Treg cells, and conclusively induced immunotherapy based on PDT through collaborative delivery strategy.
tremendous effector T cells towards tumor tissues [127]. As a result, the Among the diverse nano-vehicles, self-assembled nanoplatforms of small
combination treatment significantly extended survival of mice and molecules have exhibited unique advantages of high drug loading
inhibited the regrowth after therapy in B16BL/6 tumor-bearing mice

12
S. Zhang et al. Biomaterials 282 (2022) 121433

capacity, carrier-free self-delivering feature, tumor-specific drug acti­ [12] M.I. Carlo, M.H. Voss, R.J. Motzer, Checkpoint inhibitors and other novel
immunotherapies for advanced renal cell carcinoma, Nat. Rev. Urol. 13 (7)
vation or release and high security, which hold great promise for clinical
(2016) 420–431.
applications in the future. In this review, we outlined recent advanced [13] B.A. Inman, M.R. Harrison, D.J. George, Novel immunotherapeutic strategies in
photodynamic nanotherapeutics to induce ICD of tumor cells and development for renal cell carcinoma, Eur. Urol. 63 (5) (2013) 881–889.
potentiate cancer immunotherapy following three aspects: (i) the [14] J. Zhang, F. Dang, J. Ren, W. Wei, Biochemical aspects of PD-L1 regulation in
cancer immunotherapy, Trends Biochem. Sci. 43 (12) (2018) 1014–1032.
emerging photodynamic nanotherapeutics for ICD induction; (ii) PDT- [15] X. Zhang, C. Wang, J. Wang, Q. Hu, B. Langworthy, Y. Ye, W. Sun, J. Lin,
based combination strategies for potentiating ICD induction; (iii) T. Wang, J. Fine, H. Cheng, G. Dotti, P. Huang, Z. Gu, PD-1 blockade cellular
multimodal cancer immunotherapy based on photodynamic ICD vesicles for cancer immunotherapy, Adv Mater 30 (22) (2018), e1707112.
[16] H. Ledford, Therapeutic cancer vaccine survives biotech bust, Nature 519 (7541)
induction. (2015) 17–18.
Although the rapid development of PDT-synergized immunotherapy, [17] M. Hader, B. Frey, R. Fietkau, M. Hecht, U.S. Gaipl, Immune biological rationales
some limitations and challenges associated with PDT and immuno­ for the design of combined radio- and immunotherapies, Cancer Immunol.
Immunother. 69 (2) (2020) 293–306.
therapy remain to be solved. Firstly, the clinical application scopes of [18] A. Popovic, E.M. Jaffee, N. Zaidi, Emerging strategies for combination checkpoint
PDT and immunotherapy have certain boundedness for cancer patients, modulators in cancer immunotherapy, J. Clin. Invest. 128 (8) (2018) 3209–3218.
since PDT is still restricted to the treatment of epidermal tumors or tu­ [19] L. Galluzzi, A. Buque, O. Kepp, L. Zitvogel, G. Kroemer, Immunogenic cell death
in cancer and infectious disease, Nat. Rev. Immunol. 17 (2) (2017) 97–111.
mors near the skin. Moreover, the specificity and immunosuppression of [20] O. Kepp, L. Senovilla, I. Vitale, E. Vacchelli, S. Adjemian, P. Agostinis, L. Apetoh,
tumors can also result in great discrepancy of therapeutic outcomes and F. Aranda, V. Barnaba, N. Bloy, L. Bracci, K. Breckpot, D. Brough, A. Buque, M.
further limit the application of immunotherapy. Secondly, the mass G. Castro, M. Cirone, M.I. Colombo, I. Cremer, S. Demaria, L. Dini, A.
G. Eliopoulos, A. Faggioni, S.C. Formenti, J. Fucikova, L. Gabriele, U.S. Gaipl,
production and safe application of diverse biomaterials are still great
J. Galon, A. Garg, F. Ghiringhelli, N.A. Giese, Z.S. Guo, A. Hemminki,
challenges. Finally, the immune response against tumors is an extremely M. Herrmann, J.W. Hodge, S. Holdenrieder, J. Honeychurch, H.M. Hu, X. Huang,
complicated process, in which the interruption of any step will weaken T.M. Illidge, K. Kono, M. Korbelik, D.V. Krysko, S. Loi, P.R. Lowenstein, E. Lugli,
the effect of anti-tumor immunotherapy. In the future, it is necessary or Y. Ma, F. Madeo, A.A. Manfredi, I. Martins, D. Mavilio, L. Menger, N. Merendino,
M. Michaud, G. Mignot, K.L. Mossman, G. Multhoff, R. Oehler, F. Palombo,
rationally designing more effective nanotherapeutics to acquire desir­ T. Panaretakis, J. Pol, E. Proietti, J.E. Ricci, C. Riganti, P. Rovere-Querini,
able antitumor immunotherapy. A. Rubartelli, A. Sistigu, M.J. Smyth, J. Sonnemann, R. Spisek, J. Stagg, A.
Q. Sukkurwala, E. Tartour, A. Thorburn, S.H. Thorne, P. Vandenabeele, F. Velotti,
S.T. Workenhe, H. Yang, W.X. Zong, L. Zitvogel, G. Kroemer, L. Galluzzi,
Declaration of competing interest Consensus guidelines for the detection of immunogenic cell death,
OncoImmunology 3 (9) (2014), e955691.
[21] N. Casares, M.O. Pequignot, A. Tesniere, F. Ghiringhelli, S. Roux, N. Chaput,
The authors declare that they have no known competing financial E. Schmitt, A. Hamai, S. Hervas-Stubbs, M. Obeid, F. Coutant, D. Metivier,
interests or personal relationships that could have appeared to influence E. Pichard, P. Aucouturier, G. Pierron, C. Garrido, L. Zitvogel, G. Kroemer,
Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death,
the work reported in this paper. J. Exp. Med. 202 (12) (2005) 1691–1701.
[22] J. Zhou, G. Wang, Y. Chen, H. Wang, Y. Hua, Z. Cai, Immunogenic cell death in
Acknowledgements cancer therapy: present and emerging inducers, J. Cell Mol. Med. 23 (8) (2019)
4854–4865.
[23] L. Bezu, L.C. Gomes-de-Silva, H. Dewitte, K. Breckpot, J. Fucikova, R. Spisek,
This work was financially supported by the National Nature Science L. Galluzzi, O. Kepp, G. Kroemer, Combinatorial strategies for the induction of
Foundation of China (No. 82161138029), the Liaoning Revitalization immunogenic cell death, Front. Immunol. 6 (2015) 187.
[24] A.D. Garg, A.M. Dudek-Peric, E. Romano, P. Agostinis, Immunogenic cell death,
Talents Program (No. XLYC1907129), the Excellent Youth Science Int. J. Dev. Biol. 59 (1–3) (2015) 131–140.
Foundation of Liaoning Province (No. 2020-YQ-06), and the China [25] T. Mishchenko, E. Mitroshina, I. Balalaeva, O. Krysko, M. Vedunova, D.V. Krysko,
Postdoctoral Science Foundation (No. 2020M670794 and No. An emerging role for nanomaterials in increasing immunogenicity of cancer cell
death, Biochim. Biophys. Acta Rev. Canc 1871 (1) (2019) 99–108.
2021MD703858).
[26] O. Kepp, A. Tesniere, F. Schlemmer, M. Michaud, L. Senovilla, L. Zitvogel,
G. Kroemer, Immunogenic cell death modalities and their impact on cancer
References treatment, Apoptosis 14 (4) (2009) 364–375.
[27] D.V. Krysko, A.D. Garg, A. Kaczmarek, O. Krysko, P. Agostinis, P. Vandenabeele,
Immunogenic cell death and DAMPs in cancer therapy, Nat. Rev. Cancer 12 (12)
[1] R.L. Siegel, K.D. Miller, A. Goding Sauer, S.A. Fedewa, L.F. Butterly, J.
(2012) 860–875.
C. Anderson, A. Cercek, R.A. Smith, A. Jemal, Colorectal cancer statistics, 2020,
[28] G. Kroemer, L. Galluzzi, O. Kepp, L. Zitvogel, Immunogenic cell death in cancer
CA A Cancer J. Clin. 70 (3) (2020) 145–164.
therapy, Annu. Rev. Immunol. 31 (2013) 51–72.
[2] K.E. Pauken, M. Dougan, N.R. Rose, A.H. Lichtman, A. Sharpe, Adverse events
[29] R.J. Binder, Functions of heat shock proteins in pathways of the innate and
following cancer immunotherapy: obstacles and opportunities, Trends Immunol.
adaptive immune system, J. Immunol. 193 (12) (2014) 5765–5771.
40 (6) (2019) 511–523.
[30] M. Obeid, A. Tesniere, F. Ghiringhelli, G.M. Fimia, L. Apetoh, J.L. Perfettini,
[3] Y. Cao, X. Xu, S. Liu, L. Huang, J. Gu, Ganoderma: a cancer immunotherapy
M. Castedo, G. Mignot, T. Panaretakis, N. Casares, D. Metivier, N. Larochette,
review, Front. Pharmacol. 9 (2018) 1217.
P. van Endert, F. Ciccosanti, M. Piacentini, L. Zitvogel, G. Kroemer, Calreticulin
[4] J.S. McCune, Immunotherapy to treat cancer, Clin. Pharmacol. Ther. 100 (3)
exposure dictates the immunogenicity of cancer cell death, Nat. Med. 13 (1)
(2016) 198–203.
(2007) 54–61.
[5] Q. Chen, T. Sun, C. Jiang, Recent advancements in nanomedicine for ’cold’ tumor
[31] T. Panaretakis, N. Joza, N. Modjtahedi, A. Tesniere, I. Vitale, M. Durchschlag, G.
immunotherapy, Nano-Micro Lett. 13 (1) (2021) 92.
M. Fimia, O. Kepp, M. Piacentini, K.U. Froehlich, P. van Endert, L. Zitvogel,
[6] Y. Liu, J. Yang, B. Liu, W. Cao, J. Zhang, Y. Yang, L. Ma, J.M. de la Fuente,
F. Madeo, G. Kroemer, The co-translocation of ERp57 and calreticulin determines
J. Song, J. Ni, C. Zhang, D. Cui, Human iPS cells loaded with MnO2-based
the immunogenicity of cell death, Cell Death Differ. 15 (9) (2008) 1499–1509.
nanoprobes for photodynamic and simultaneous enhanced immunotherapy
[32] A.D.L. Souza, E. Rodrigues-Filho, A.Q.L. Souza, J.O. Pereira, A.K. Calgarotto,
against cancer, Nano-Micro Lett. 12 (1) (2020) 127.
V. Maso, S. Marangoni, S.L. Da Silva, Koninginins, phospholipase A2 inhibitors
[7] W. Ou, K.S. Nam, D.H. Park, J. Hwang, S.K. Ku, C.S. Yong, J.O. Kim, J.H. Byeon,
from endophytic fungus Trichoderma koningii, Toxicon 51 (2) (2008) 240–250.
Artificial nanoscale erythrocytes from clinically relevant compounds for
[33] A.D. Garg, D.V. Krysko, T. Verfaillie, A. Kaczmarek, G.B. Ferreira, T. Marysael,
enhancing cancer immunotherapy, Nano-Micro Lett. 12 (1) (2020) 90.
N. Rubio, M. Firczuk, C. Mathieu, A.J.M. Roebroek, W. Annaert, J. Golab, P. de
[8] Q. Sun, X. Bai, A.M. Sofias, R. van der Meel, E. Ruiz-Hernandez, G. Storm, W.
Witte, P. Vandenabeele, P. Agostinis, A novel pathway combining calreticulin
E. Hennink, B. De Geest, F. Kiessling, H.J. Yu, T. Lammers, Y. Shi, Cancer
exposure and ATP secretion in immunogenic cancer cell death, EMBO J. 31 (5)
nanomedicine meets immunotherapy: opportunities and challenges, Acta
(2012) 1062–1079.
Pharmacol. Sin. 41 (7) (2020) 954–958.
[34] I. Martins, Y. Wang, M. Michaud, Y. Ma, A.Q. Sukkurwala, S. Shen, O. Kepp,
[9] J. Wang, L. Fang, P. Li, L. Ma, W. Na, C. Cheng, Y. Gu, D. Deng, Inorganic
D. Métivier, L. Galluzzi, J.L. Perfettini, L. Zitvogel, G. Kroemer, Molecular
nanozyme with combined self-oxygenation/degradable capabilities for sensitized
mechanisms of ATP secretion during immunogenic cell death, Cell Death Differ.
cancer immunochemotherapy, Nano-Micro Lett. 11 (1) (2019) 74.
21 (1) (2013) 79–91.
[10] D. Karan, J.M. Holzbeierlein, P. Van Veldhuizen, J.B. Thrasher, Cancer
[35] L. Apetoh, F. Ghiringhelli, A. Tesniere, A. Criollo, C. Ortiz, R. Lidereau,
immunotherapy: a paradigm shift for prostate cancer treatment, Nat. Rev. Urol. 9
C. Mariette, N. Chaput, J.P. Mira, S. Delaloge, F. Andre, T. Tursz, G. Kroemer,
(7) (2012) 376–385.
L. Zitvogel, The interaction between HMGB1 and TLR4 dictates the outcome of
[11] M. Rescigno, F. Avogadri, G. Curigliano, Challenges and prospects of
anticancer chemotherapy and radiotherapy, Immunol. Rev. 220 (2007) 47–59.
immunotherapy as cancer treatment, Biochim. Biophys. Acta 1776 (1) (2007)
108–123.

13
S. Zhang et al. Biomaterials 282 (2022) 121433

[36] P. Scaffidi, T. Misteli, M.E. Bianchi, Release of chromatin protein HMGB1 by [60] Y. Yang, Z. Gu, J. Tang, M. Zhang, Y. Yang, H. Song, C. Yu, MnO2 nanoflowers
necrotic cells triggers inflammation, Nature 418 (6894) (2002) 191–195. induce immunogenic cell death under nutrient deprivation: enabling an
[37] S. Ladoire, D. Hannani, M. Vetizou, C. Locher, L. Aymeric, L. Apetoh, O. Kepp, orchestrated cancer starvation-immunotherapy, Adv. Sci. 8 (4) (2021) 2002667.
G. Kroemer, F. Ghiringhelli, L. Zitvogel, Cell-death-associated molecular patterns [61] R. Alzeibak, T.A. Mishchenko, N.Y. Shilyagina, I.V. Balalaeva, M.V. Vedunova, D.
as determinants of cancer immunogenicity, Antioxidants Redox Signal. 20 (7) V. Krysko, Targeting immunogenic cancer cell death by photodynamic therapy:
(2014) 1098–1116. past, present and future, J. Immun. Canc. 9 (1) (2021).
[38] R. Tang, J. Xu, B. Zhang, J. Liu, C. Liang, J. Hua, Q. Meng, X. Yu, S. Shi, [62] J. Ni, Y. Sun, J. Song, Y. Zhao, Q. Gao, X. Li, Artificial cell-mediated
Ferroptosis, necroptosis, and pyroptosis in anticancer immunity, J. Hematol. photodynamic therapy enhanced anticancer efficacy through combination of
Oncol. 13 (1) (2020) 110. tumor disruption and immune response stimulation, ACS Omega 4 (7) (2019)
[39] T.L. Aaes, A. Kaczmarek, T. Delvaeye, B. De Craene, S. De Koker, L. Heyndrickx, 12727–12735.
I. Delrue, J. Taminau, B. Wiernicki, P. De Groote, A.D. Garg, L. Leybaert, [63] H. Wang, K. Wang, L. He, Y. Liu, H. Dong, Y. Li, Engineering antigen as
J. Grooten, M.J. Bertrand, P. Agostinis, G. Berx, W. Declercq, P. Vandenabeele, D. photosensitiser nanocarrier to facilitate ROS triggered immune cascade for
V. Krysko, Vaccination with necroptotic cancer cells induces efficient anti-tumor photodynamic immunotherapy, Biomaterials 244 (2020) 119964.
immunity, Cell Rep. 15 (2) (2016) 274–287. [64] H.Y. Kim, M. Kang, Y.W. Choo, S.H. Go, S.P. Kwon, S.Y. Song, H.S. Sohn, J. Hong,
[40] T. Kang, Y. Huang, Q. Zhu, H. Cheng, Y. Pei, J. Feng, M. Xu, G. Jiang, Q. Song, B.S. Kim, Immunomodulatory lipocomplex functionalized with photosensitizer-
T. Jiang, H. Chen, X. Gao, J. Chen, Necroptotic cancer cells-mimicry nanovaccine embedded cancer cell membrane inhibits tumor growth and metastasis, Nano
boosts anti-tumor immunity with tailored immune-stimulatory modality, Lett. 19 (8) (2019) 5185–5193.
Biomaterials 164 (2018) 80–97. [65] G. Deng, Z. Sun, S. Li, X. Peng, W. Li, L. Zhou, Y. Ma, P. Gong, L. Cai, Cell-
[41] S.J. Dixon, K.M. Lemberg, M.R. Lamprecht, R. Skouta, E.M. Zaitsev, C.E. Gleason, membrane immunotherapy based on natural killer cell membrane coated
D.N. Patel, A.J. Bauer, A.M. Cantley, W.S. Yang, B. Morrison 3rd, B.R. Stockwell, nanoparticles for the effective inhibition of primary and abscopal tumor growth,
Ferroptosis: an iron-dependent form of nonapoptotic cell death, Cell 149 (5) ACS Nano 12 (12) (2018) 12096–12108.
(2012) 1060–1072. [66] W.L. Liu, M.Z. Zou, T. Liu, J.Y. Zeng, X. Li, W.Y. Yu, C.X. Li, J.J. Ye, W. Song,
[42] B. Zhou, J. Liu, R. Kang, D.J. Klionsky, G. Kroemer, D. Tang, Ferroptosis is a type J. Feng, X.Z. Zhang, Expandable immunotherapeutic nanoplatforms engineered
of autophagy-dependent cell death, Semin. Cancer Biol. 66 (2020) 89–100. from cytomembranes of hybrid cells derived from cancer and dendritic cells, Adv
[43] W. Wang, M. Green, J.E. Choi, M. Gijon, P.D. Kennedy, J.K. Johnson, P. Liao, Mater 31 (18) (2019), e1900499.
X. Lang, I. Kryczek, A. Sell, H. Xia, J. Zhou, G. Li, J. Li, W. Li, S. Wei, L. Vatan, [67] X. Xu, G. Deng, Z. Sun, Y. Luo, J. Liu, X. Yu, Y. Zhao, P. Gong, G. Liu, P. Zhang,
H. Zhang, W. Szeliga, W. Gu, R. Liu, T.S. Lawrence, C. Lamb, Y. Tanno, M. Cieslik, F. Pan, L. Cai, B.Z. Tang, A biomimetic aggregation-induced emission
E. Stone, G. Georgiou, T.A. Chan, A. Chinnaiyan, W. Zou, CD8(+) T cells regulate photosensitizer with antigen-presenting and hitchhiking function for lipid droplet
tumour ferroptosis during cancer immunotherapy, Nature 569 (7755) (2019) targeted photodynamic immunotherapy, Adv Mater 33 (33) (2021), e2102322.
270–274. [68] H. Wang, X. Han, Z. Dong, J. Xu, J. Wang, Z. Liu, Hyaluronidase with pH-
[44] S. Zhang, Y. Wang, Z. Kong, X. Zhang, B. Sun, H. Yu, Q. Chen, C. Luo, J. Sun, responsive dextran modification as an adjuvant nanomedicine for enhanced
Z. He, Pure photosensitizer-driven nanoassembly with core-matched PEGylation photodynamic-immunotherapy of cancer, Adv. Funct. Mater. 29 (29) (2019).
for imaging-guided photodynamic therapy, Acta Pharm. Sin. B (2021). [69] H. Liu, Y. Hu, Y. Sun, C. Wan, Z. Zhang, X. Dai, Z. Lin, Q. He, Z. Yang, P. Huang,
[45] S. Zhang, Z. Wang, Z. Kong, Y. Wang, X. Zhang, B. Sun, H. Zhang, Q. Kan, Z. He, Y. Xiong, J. Cao, X. Chen, Q. Chen, J.F. Lovell, Z. Xu, H. Jin, K. Yang, Co-delivery
C. Luo, J. Sun, Photosensitizer-driven nanoassemblies of homodimeric prodrug of bee venom Melittin and a photosensitizer with an organic–inorganic hybrid
for self-enhancing activation and synergistic chemo-photodynamic therapy, nanocarrier for photodynamic therapy and immunotherapy, ACS Nano 13 (11)
Theranostics 11 (12) (2021) 6019–6032. (2019) 12638–12652.
[46] N. Yang, W. Xiao, X. Song, W. Wang, X. Dong, Recent advances in tumor [70] J. Xu, L. Xu, C. Wang, R. Yang, Q. Zhuang, X. Han, Z. Dong, W. Zhu, R. Peng,
microenvironment hydrogen peroxide-responsive materials for cancer Z. Liu, Near-infrared-triggered photodynamic therapy with multitasking
photodynamic therapy, Nano-Micro Lett. 12 (1) (2020) 15. upconversion nanoparticles in combination with checkpoint blockade for
[47] E. Panzarini, V. Inguscio, G.M. Fimia, L. Dini, Rose Bengal acetate photodynamic immunotherapy of colorectal cancer, ACS Nano 11 (5) (2017) 4463–4474.
therapy (RBAc-PDT) induces exposure and release of Damage-Associated [71] Y. Shao, B. Liu, Z. Di, G. Zhang, L.-D. Sun, L. Li, C.-H. Yan, Engineering of
Molecular Patterns (DAMPs) in human HeLa cells, PLoS One 9 (8) (2014), upconverted metal–organic frameworks for near-infrared light-triggered
e105778. combinational photodynamic/chemo-/immunotherapy against hypoxic tumors,
[48] W. Li, J. Yang, L. Luo, M. Jiang, B. Qin, H. Yin, C. Zhu, X. Yuan, J. Zhang, Z. Luo, J. Am. Chem. Soc. 142 (8) (2020) 3939–3946.
Y. Du, Q. Li, Y. Lou, Y. Qiu, J. You, Targeting photodynamic and photothermal [72] S. Yan, X. Zeng, Y.a. Tang, B.F. Liu, Y. Wang, X. Liu, Activating antitumor
therapy to the endoplasmic reticulum enhances immunogenic cancer cell death, immunity and antimetastatic effect through polydopamine-encapsulated core-
Nat. Commun. 10 (1) (2019) 3349. shell upconversion nanoparticles, Adv. Mater. 31 (46) (2019).
[49] H. Deng, Z. Zhou, W. Yang, L.S. Lin, S. Wang, G. Niu, J. Song, X. Chen, [73] R. Liang, L. Liu, H. He, Z. Chen, Z. Han, Z. Luo, Z. Wu, M. Zheng, Y. Ma, L. Cai,
Endoplasmic reticulum targeting to amplify immunogenic cell death for cancer Oxygen-boosted immunogenic photodynamic therapy with gold nanocages@
immunotherapy, Nano Lett. 20 (3) (2020) 1928–1933. manganese dioxide to inhibit tumor growth and metastases, Biomaterials 177
[50] F. Yang, Z. Zhao, B. Sun, Q. Chen, J. Sun, Z. He, C. Luo, Nanotherapeutics for (2018) 149–160.
antimetastatic treatment, Trend. Canc. 6 (8) (2020) 645–659. [74] S. Bai, L.L. Yang, Y. Wang, T. Zhang, L. Fu, S. Yang, S. Wan, S. Wang, D. Jia, B. Li,
[51] S. Zhang, J. Guan, M. Sun, D. Zhang, H. Zhang, B. Sun, W. Guo, B. Lin, Y. Wang, P. Xue, Y. Kang, Z.J. Sun, Z. Xu, Prodrug-based versatile nanomedicine for
Z. He, C. Luo, J. Sun, Self-delivering prodrug-nanoassemblies fabricated by enhancing cancer immunotherapy by increasing immunogenic cell death, Small
disulfide bond bridged oleate prodrug of docetaxel for breast cancer therapy, 16 (19) (2020), e2000214.
Drug Deliv. 24 (1) (2017) 1460–1469. [75] D. Liu, B. Chen, Y. Mo, Z. Wang, T. Qi, Q. Zhang, Y. Wang, Redox-activated
[52] S. Zhao, X. Yu, Y. Qian, W. Chen, J. Shen, Multifunctional magnetic iron oxide porphyrin-based liposome remote-loaded with indoleamine 2,3-dioxygenase
nanoparticles: an advanced platform for cancer theranostics, Theranostics 10 (14) (Ido) inhibitor for synergistic photoimmunotherapy through induction of
(2020) 6278–6309. immunogenic cell death and blockage of Ido pathway, Nano Lett. 19 (10) (2019)
[53] Y. Li, Y. Miao, M. Chen, X. Chen, F. Li, X. Zhang, Y. Gan, Stepwise targeting and 6964–6976.
responsive lipid-coated nanoparticles for enhanced tumor cell sensitivity and [76] Z. Chen, L. Liu, R. Liang, Z. Luo, H. He, Z. Wu, H. Tian, M. Zheng, Y. Ma, L. Cai,
hepatocellular carcinoma therapy, Theranostics 10 (8) (2020) 3722–3736. Bioinspired hybrid protein oxygen nanocarrier amplified photodynamic therapy
[54] M. Saeed, J. Gao, Y. Shi, T. Lammers, H. Yu, Engineering nanoparticles to for eliciting anti-tumor immunity and abscopal effect, ACS Nano 12 (8) (2018)
reprogram the tumor immune microenvironment for improved cancer 8633–8645.
immunotherapy, Theranostics 9 (26) (2019) 7981–8000. [77] D. Li, J. Ren, J. Li, Y. Zhang, Y. Lou, J. Zhu, P. Liu, Y. Chen, Z. Yu, L. Zhao,
[55] G. Jin, R. He, Q. Liu, M. Lin, Y. Dong, K. Li, B.Z. Tang, B. Liu, F. Xu, Near-infrared L. Zhang, X. Chen, J. Zhu, J. Tao, Ferroptosis-apoptosis combined anti-melanoma
light-regulated cancer theranostic nanoplatform based on aggregation-induced immunotherapy with a NIR-responsive upconverting mSiO 2 photodynamic
emission luminogen encapsulated upconversion nanoparticles, Theranostics 9 (1) platform, Chem. Eng. J. 419 (2021).
(2019) 246–264. [78] H. He, L. Liu, R. Liang, H. Zhou, H. Pan, S. Zhang, L. Cai, Tumor-targeted
[56] Z. Yang, J. Wang, S. Ai, J. Sun, X. Mai, W. Guan, Self-generating oxygen enhanced nanoplatform for in situ oxygenation-boosted immunogenic phototherapy of
mitochondrion-targeted photodynamic therapy for tumor treatment with hypoxia colorectal cancer, Acta Biomater. 104 (2020) 188–197.
scavenging, Theranostics 9 (23) (2019) 6809–6823. [79] G. Lan, K. Ni, Z. Xu, S.S. Veroneau, Y. Song, W. Lin, Nanoscale metal-organic
[57] K. Siwawannapong, R. Zhang, H. Lei, Q. Jin, W. Tang, Z. Dong, R.Y. Lai, Z. Liu, framework overcomes hypoxia for photodynamic therapy primed cancer
A. Kamkaew, L. Cheng, Ultra-small pyropheophorbide-a nanodots for near- immunotherapy, J. Am. Chem. Soc. 140 (17) (2018) 5670–5673.
infrared fluorescence/photoacoustic imaging-guided photodynamic therapy, [80] T. Wang, H. Zhang, Y. Han, H. Liu, F. Ren, J. Zeng, Q. Sun, Z. Li, M. Gao, Light-
Theranostics 10 (1) (2020) 62–73. enhanced O2-evolving nanoparticles boost photodynamic therapy to elicit
[58] Y. Ma, Y. Zhang, X. Li, Y. Zhao, M. Li, W. Jiang, X. Tang, J. Dou, L. Lu, F. Wang, antitumor immunity, ACS Appl. Mater. Interfaces 11 (18) (2019) 16367–16379.
Y. Wang, Near-infrared II phototherapy induces deep tissue immunogenic cell [81] C.H. Chung, K.Y. Lu, W.C. Lee, W.J. Hsu, W.F. Lee, J.Z. Dai, P.W. Shueng, C.
death and potentiates cancer immunotherapy, ACS Nano 13 (10) (2019) W. Lin, F.L. Mi, Fucoidan-based, tumor-activated nanoplatform for overcoming
11967–11980. hypoxia and enhancing photodynamic therapy and antitumor immunity,
[59] X. Mai, Y. Zhang, H. Fan, W. Song, Y. Chang, B. Chen, J. Shi, X. Xin, Z. Teng, Biomaterials 257 (2020) 120227.
J. Sun, G. Teng, Integration of immunogenic activation and immunosuppressive [82] Z. Meng, X. Zhou, J. Xu, X. Han, Z. Dong, H. Wang, Y. Zhang, J. She, L. Xu,
reversion using mitochondrial-respiration-inhibited platelet-mimicking C. Wang, Z. Liu, Light-triggered in situ gelation to enable robust photodynamic-
nanoparticles, Biomaterials 232 (2020) 119699. immunotherapy by repeated stimulations, Adv Mater 31 (24) (2019), e1900927.

14
S. Zhang et al. Biomaterials 282 (2022) 121433

[83] M. Zhang, W. Wang, F. Wu, T. Zheng, J. Ashley, M. Mohammadniaei, Q. Zhang, [105] B. Feng, B. Hou, Z. Xu, M. Saeed, H. Yu, Y. Li, Self-amplified drug delivery with
M. Wang, L. Li, J. Shen, Y. Sun, Biodegradable Poly(gamma-glutamic acid) light-inducible nanocargoes to enhance cancer immunotherapy, Adv Mater 31
@glucose oxidase@carbon dot nanoparticles for simultaneous multimodal (40) (2019), e1902960.
imaging and synergetic cancer therapy, Biomaterials 252 (2020) 120106. [106] J. Ni, J. Song, B. Wang, H. Hua, H. Zhu, X. Guo, S. Xiong, Y. Zhao, Dendritic cell
[84] M. Zhao, X. Yang, H. Fu, C. Chen, Y. Zhang, Z. Wu, Y. Duan, Y. Sun, Immune/ vaccine for the effective immunotherapy of breast cancer, Biomed. Pharmacother.
hypoxic tumor microenvironment regulation-enhanced photodynamic treatment 126 (2020) 110046.
realized by pH-responsive phase transition-targeting nanobubbles, ACS Appl. [107] I. Villa, C. Villa, R. Crapanzano, V. Secchi, M. Tawfilas, E. Trombetta, L. Porretti,
Mater. Interfaces 13 (28) (2021) 32763–32779. A. Brambilla, M. Campione, Y. Torrente, A. Vedda, A. Monguzzi, Functionalized
[85] Z.Y. Yang, J.F. Wang, S. Liu, X.H. Li, L.Y. Miao, B. Yang, C.L. Zhang, J. He, S.C. Ai, scintillating nanotubes for simultaneous radio- and photodynamic therapy of
W.X. Guan, Defeating relapsed and refractory malignancies through a nano- cancer, ACS Appl. Mater. Interfaces 13 (11) (2021) 12997–13008.
enabled mitochondria-mediated respiratory inhibition and damage pathway, [108] L. Fu, X. Ma, Y. Liu, Z. Xu, Z. Sun, Applying nanotechnology to boost cancer
Biomaterials 229 (2020). immunotherapy by promoting immunogenic cell death, Chin. Chem. Lett. (2021).
[86] D. Wang, T. Wang, J. Liu, H. Yu, S. Jiao, B. Feng, F. Zhou, Y. Fu, Q. Yin, P. Zhang, [109] Y. Chao, L. Xu, C. Liang, L. Feng, J. Xu, Z. Dong, L. Tian, X. Yi, K. Yang, Z. Liu,
Z. Zhang, Z. Zhou, Y. Li, Acid-activatable versatile micelleplexes for PD-L1 Combined local immunostimulatory radioisotope therapy and systemic immune
blockade-enhanced cancer photodynamic immunotherapy, Nano Lett. 16 (9) checkpoint blockade imparts potent antitumour responses, Nat. Biomed. Eng. 2
(2016) 5503–5513. (8) (2018) 611–621.
[87] Z. Wang, F. Zhang, D. Shao, Z. Chang, L. Wang, H. Hu, X. Zheng, X. Li, F. Chen, [110] E.B. Golden, I. Pellicciotta, S. Demaria, M.H. Barcellos-Hoff, S.C. Formenti, The
Z. Tu, M. Li, W. Sun, L. Chen, W.F. Dong, Janus nanobullets combine convergence of radiation and immunogenic cell death signaling pathways, Front.
photodynamic therapy and magnetic hyperthermia to potentiate synergetic anti- Oncol. 2 (2012) 88.
metastatic immunotherapy, Adv. Sci. 6 (22) (2019) 1901690. [111] B. Doix, N. Trempolec, O. Riant, O. Feron, Low photosensitizer dose and early
[88] C. He, X. Duan, N. Guo, C. Chan, C. Poon, R.R. Weichselbaum, W. Lin, Core-shell radiotherapy enhance antitumor immune response of photodynamic therapy-
nanoscale coordination polymers combine chemotherapy and photodynamic based dendritic cell vaccination, Front. Oncol. 9 (2019) 811.
therapy to potentiate checkpoint blockade cancer immunotherapy, Nat. Commun. [112] H. Wang, B. Lv, Z. Tang, M. Zhang, W. Ge, Y. Liu, X. He, K. Zhao, X. Zheng, M. He,
7 (2016) 12499. W. Bu, Scintillator-based nanohybrids with sacrificial electron prodrug for
[89] D. Liu, H. Huang, B. Zhao, W. Guo, Natural melanin-based nanoparticles with enhanced X-ray-induced photodynamic therapy, Nano Lett. 18 (9) (2018)
combined chemo/photothermal/photodynamic effect induce immunogenic cell 5768–5774.
death (ICD) on tumor, Front. Bioeng. Biotechnol. 9 (2021) 635858. [113] W. Sun, L. Luo, Y. Feng, Y. Cai, Y. Zhuang, R.J. Xie, X. Chen, H. Chen,
[90] Y. Huang, Z. Guan, X. Dai, Y. Shen, Q. Wei, L. Ren, J. Jiang, Z. Xiao, Y. Jiang, Aggregation-induced emission gold clustoluminogens for enhanced low-dose X-
D. Liu, Z. Huang, X. Xu, Y. Luo, C. Zhao, Engineered macrophages as near- ray-induced photodynamic therapy, Angew. Chem. 132 (25) (2019)
infrared light activated drug vectors for chemo-photodynamic therapy of primary 10000–10007.
and bone metastatic breast cancer, Nat. Commun. 12 (1) (2021) 4310. [114] K. Lu, C. He, N. Guo, C. Chan, K. Ni, G. Lan, H. Tang, C. Pelizzari, Y.X. Fu, M.
[91] W. Yang, F. Zhang, H. Deng, L. Lin, S. Wang, F. Kang, G. Yu, J. Lau, R. Tian, T. Spiotto, R.R. Weichselbaum, W. Lin, Low-dose X-ray radiotherapy-
M. Zhang, Z. Wang, L. He, Y. Ma, G. Niu, S. Hu, X. Chen, Smart nanovesicle- radiodynamic therapy via nanoscale metal-organic frameworks enhances
mediated immunogenic cell death through tumor microenvironment modulation checkpoint blockade immunotherapy, Nat. Biomed. Eng. 2 (8) (2018) 600–610.
for effective photodynamic immunotherapy, ACS Nano 14 (1) (2020) 620–631. [115] K. Ni, T. Luo, A. Culbert, M. Kaufmann, X. Jiang, W. Lin, Nanoscale metal-organic
[92] M. Wang, J. Song, F. Zhou, A.R. Hoover, C. Murray, B. Zhou, L. Wang, J. Qu, W. framework Co-delivers TLR-7 agonists and anti-CD47 antibodies to modulate
R. Chen, NIR-triggered phototherapy and immunotherapy via an antigen- macrophages and orchestrate cancer immunotherapy, J. Am. Chem. Soc. 142 (29)
capturing nanoplatform for metastatic cancer treatment, Adv. Sci. 6 (10) (2019) (2020) 12579–12584.
1802157. [116] M. Xu, L. Zhou, L. Zheng, Q. Zhou, K. Liu, Y. Mao, S. Song, Sonodynamic therapy-
[93] X. Meng, B. Zhang, Y. Yi, H. Cheng, B. Wang, Y. Liu, T. Gong, W. Yang, Y. Yao, derived multimodal synergistic cancer therapy, Cancer Lett. 497 (2021) 229–242.
H. Wang, W. Bu, Accurate and real-time temperature monitoring during MR [117] N. Nomikou, K. Curtis, C. McEwan, B.M.G. O’Hagan, B. Callan, J.F. Callan, A.
imaging guided PTT, Nano Lett. 20 (4) (2020) 2522–2529. P. McHale, A versatile, stimulus-responsive nanoparticle-based platform for use in
[94] W. Zhang, K. Cai, X. Li, J. Zhang, Z. Ma, M.F. Foda, Y. Mu, X. Dai, H. Han, Au both sonodynamic and photodynamic cancer therapy, Acta Biomater. 49 (2017)
hollow nanorods-chimeric peptide nanocarrier for NIR-II photothermal therapy 414–421.
and real-time apoptosis imaging for tumor theranostics, Theranostics 9 (17) [118] L. Zheng, Y. Zhang, H. Lin, S. Kang, Y. Li, D. Sun, M. Chen, Z. Wang, Z. Jiao,
(2019) 4971–4981. Y. Wang, B. Dai, S. Zhuang, D. Zhang, Ultrasound and near-infrared light dual-
[95] X. Zhang, J. Tang, C. Li, Y. Lu, L. Cheng, J. Liu, A targeting black phosphorus triggered upconversion zeolite-based nanocomposite for hyperthermia-enhanced
nanoparticle based immune cells nano-regulator for photodynamic/photothermal multimodal melanoma therapy via a precise apoptotic mechanism, ACS Appl.
and photo-immunotherapy, Bioact. Mater. 6 (2) (2021) 472–489. Mater. Interfaces 12 (29) (2020) 32420–32431.
[96] P. Liu, W. Yang, L. Shi, H. Zhang, Y. Xu, P. Wang, G. Zhang, W.R. Chen, B. Zhang, [119] Q. Zhang, C. Bao, X. Cai, L. Jin, L. Sun, Y. Lang, L. Li, Sonodynamic therapy-
X. Wang, Concurrent photothermal therapy and photodynamic therapy for assisted immunotherapy: a novel modality for cancer treatment, Cancer Sci. 109
cutaneous squamous cell carcinoma by gold nanoclusters under a single NIR laser (5) (2018) 1330–1345.
irradiation, J. Mater. Chem. B 7 (44) (2019) 6924–6933. [120] H. Chen, L. Liu, A. Ma, T. Yin, Z. Chen, R. Liang, Y. Qiu, M. Zheng, L. Cai,
[97] L. Chen, L. Zhou, C. Wang, Y. Han, Y. Lu, J. Liu, X. Hu, T. Yao, Y. Lin, S. Liang, Noninvasively immunogenic sonodynamic therapy with manganese
S. Shi, C. Dong, Tumor-targeted drug and CpG delivery system for phototherapy protoporphyrin liposomes against triple-negative breast cancer, Biomaterials 269
and docetaxel-enhanced immunotherapy with polarization toward M1-type (2021) 120639.
macrophages on triple negative breast cancers, Adv Mater 31 (52) (2019), [121] Y. Yin, X. Jiang, L. Sun, H. Li, C. Su, Y. Zhang, G. Xu, X. Li, C. Zhao, Y. Chen,
e1904997. H. Xu, K. Zhang, Continuous inertial cavitation evokes massive ROS for
[98] Z. Liu, X. Yang, R. Liu, J. Bao, N. An, S. Jiang, S. Miao, C. Guo, G. Qu, H. Meng, reinforcing sonodynamic therapy and immunogenic cell death against breast
Phototherapy together with it triggered immunological response for Anti-HPV carcinoma, Nano Today 36 (2021).
treatment of oropharyngeal cancer: removing tumor and pathogenic virus [122] Z. Liu, J. Li, Y. Jiang, D. Wang, Multifunctional nanocapsules on a seesaw
simultaneously, Biomaterials 272 (2021) 120777. balancing sonodynamic and photodynamic therapies against superficial
[99] S. Yan, X. Zeng, Y. Tang, B.F. Liu, Y. Wang, X. Liu, Activating antitumor immunity malignant tumors by effective immune-enhancement, Biomaterials 218 (2019)
and antimetastatic effect through polydopamine-encapsulated core-shell 119251.
upconversion nanoparticles, Adv Mater 31 (46) (2019), e1905825. [123] M. Chen, Y. Zhang, L. Cui, Z. Cao, Y. Wang, W. Zhang, Y. Zheng, D. Sun, L. Zheng,
[100] Q. Yang, G. Shi, X. Chen, Y. Lin, L. Cheng, Q. Jiang, X. Yan, M. Jiang, Y. Li, S. Kang, D. Zhang, Protonated 2D carbon nitride sensitized with Ce6 as a smart
H. Zhang, H. Wang, Y. Wang, Q. Wang, Y. Zhang, Y. Liu, X. Su, L. Dai, M. Tang, metal-free nanoplatform for boosted acute multimodal photo-sono tumor
J. Li, L. Zhang, Z. Qian, D. Yu, H. Deng, Nanomicelle protects the immune inactivation and long-term cancer immunotherapy, Chem. Eng. J. 422 (2021).
activation effects of Paclitaxel and sensitizes tumors to anti-PD-1 Immunotherapy, [124] G. Liao, F. He, Q. Li, L. Zhong, R. Zhao, H. Che, H. Gao, B. Fang, Emerging
Theranostics 10 (18) (2020) 8382–8399. graphitic carbon nitride-based materials for biomedical applications, Prog. Mater.
[101] L. Hu, Z. Cao, L. Ma, Z. Liu, G. Liao, J. Wang, S. Shen, D. Li, X. Yang, The Sci. 112 (2020).
potentiated checkpoint blockade immunotherapy by ROS-responsive nanocarrier- [125] Z. Yuan, G. Fan, H. Wu, C. Liu, Y. Zhan, Y. Qiu, C. Shou, F. Gao, J. Zhang, P. Yin,
mediated cascade chemo-photodynamic therapy, Biomaterials 223 (2019) K. Xu, Photodynamic therapy synergize with PD-L1 checkpoint blockade for
119469. immunotherapy of colorectal cancer by multifunctional nanoparticle, Mol. Ther.
[102] B. Du, D.J. Waxman, Medium dose intermittent cyclophosphamide induces (2021).
immunogenic cell death and cancer cell autonomous type I interferon production [126] K. Lu, C. He, N. Guo, C. Chan, K. Ni, R.R. Weichselbaum, W. Lin, Chlorin-based
in glioma models, Cancer Lett. 470 (2020) 170–180. nanoscale metal-organic framework systemically rejects colorectal cancers via
[103] Y. Ge, C. Domschke, N. Stoiber, S. Schott, J. Heil, J. Rom, M. Blumenstein, synergistic photodynamic therapy and checkpoint blockade immunotherapy,
J. Thum, C. Sohn, A. Schneeweiss, P. Beckhove, F. Schuetz, Metronomic J. Am. Chem. Soc. 138 (38) (2016) 12502–12510.
cyclophosphamide treatment in metastasized breast cancer patients: [127] W. Ou, L. Jiang, R.K. Thapa, Z.C. Soe, K. Poudel, J.H. Chang, S.K. Ku, H.G. Choi,
immunological effects and clinical outcome, Cancer Immunol. Immunother. 61 C.S. Yong, J.O. Kim, Combination of NIR therapy and regulatory T cell
(3) (2012) 353–362. modulation using layer-by-layer hybrid nanoparticles for effective cancer
[104] A. Sharabi, N. Haran-Ghera, Immune recovery after cyclophosphamide treatment photoimmunotherapy, Theranostics 8 (17) (2018) 4574–4590.
in multiple myeloma: implication for maintenance immunotherapy, Bone Marrow [128] Q. Li, D. Zhang, J. Zhang, Y. Jiang, A. Song, Z. Li, Y. Luan, A three-in-one
Res. 2011 (2011) 269519. immunotherapy nanoweapon via cascade-amplifying cancer-immunity cycle

15
S. Zhang et al. Biomaterials 282 (2022) 121433

against tumor metastasis, relapse, and postsurgical regrowth, Nano Lett. 19 (9) stimulates mTOR: a novel Ido effector pathway targeted by D-1-methyl-
(2019) 6647–6657. tryptophan, OncoImmunology 1 (9) (2012) 1460–1468.
[129] J. Xu, S. Yu, X. Wang, Y. Qian, W. Wu, S. Zhang, B. Zheng, G. Wei, S. Gao, Z. Cao, [145] E. Vacchelli, F. Aranda, A. Eggermont, C. Sautes-Fridman, E. Tartour, E.
W. Fu, Z. Xiao, W. Lu, High Affinity of Chlorin e6 to Immunoglobulin G for P. Kennedy, M. Platten, L. Zitvogel, G. Kroemer, L. Galluzzi, Trial watch: Ido
Intraoperative Fluorescence Image-Guided Cancer Photodynamic and Checkpoint inhibitors in cancer therapy, OncoImmunology 3 (10) (2014), e957994.
Blockade Therapy, ACS Nano 13 (9) (2019) 10242–10260. [146] S. Qian, M. Zhang, Q. Chen, Y. He, W. Wang, Z. Wang, Ido as a drug target for
[130] R. Zhang, Z. Zhu, H. Lv, F. Li, S. Sun, J. Li, C.S. Lee, Immune checkpoint blockade cancer immunotherapy: recent developments in Ido inhibitors discovery, RSC
mediated by a small-molecule nanoinhibitor targeting the PD-1/PD-L1 pathway Adv. 6 (9) (2016) 7575–7581.
synergizes with photodynamic therapy to elicit antitumor immunity and [147] R. Liu, Y. An, W. Jia, Y. Wang, Y. Wu, Y. Zhen, J. Cao, H. Gao, Macrophage-mimic
antimetastatic effects on breast cancer, Small 15 (49) (2019), e1903881. shape changeable nanomedicine retained in tumor for multimodal therapy of
[131] S. Im, J. Lee, D. Park, A. Park, Y.M. Kim, W.J. Kim, Hypoxia-triggered breast cancer, J. Contr. Release 321 (2020) 589–601.
transforming immunomodulator for cancer immunotherapy via [148] W. Song, J. Kuang, C.X. Li, M. Zhang, D. Zheng, X. Zeng, C. Liu, X.Z. Zhang,
photodynamically enhanced antigen presentation of dendritic cell, ACS Nano 13 Enhanced immunotherapy based on photodynamic therapy for both primary and
(1) (2019) 476–488. lung metastasis tumor eradication, ACS Nano 12 (2) (2018) 1978–1989.
[132] W. Yang, G. Zhu, S. Wang, G. Yu, Z. Yang, L. Lin, Z. Zhou, Y. Liu, Y. Dai, F. Zhang, [149] S.M. Jensen, L.D. Maston, M.J. Gough, C.E. Ruby, W.L. Redmond, M. Crittenden,
Z. Shen, Y. Liu, Z. He, J. Lau, G. Niu, D.O. Kiesewetter, S. Hu, X. Chen, In situ Y. Li, S. Puri, C.H. Poehlein, N. Morris, M. Kovacsovics-Bankowski, T. Moudgil,
dendritic cell vaccine for effective cancer immunotherapy, ACS Nano 13 (3) C. Twitty, E.B. Walker, H.M. Hu, W.J. Urba, A.D. Weinberg, B. Curti, B.A. Fox,
(2019) 3083–3094. Signaling through OX40 enhances antitumor immunity, Semin. Oncol. 37 (5)
[133] Z. Cai, F. Xin, Z. Wei, M. Wu, X. Lin, X. Du, G. Chen, D. Zhang, Z. Zhang, X. Liu, (2010) 524–532.
C. Yao, Photodynamic therapy combined with antihypoxic signaling and CpG [150] H. Yokouchi, K. Yamazaki, K. Chamoto, E. Kikuchi, N. Shinagawa, S. Oizumi,
adjuvant as an in situ tumor vaccine based on metal-organic framework F. Hommura, T. Nishimura, M. Nishimura, Anti-OX40 monoclonal antibody
nanoparticles to boost cancer immunotherapy, Adv. Healthc. Mater. 9 (1) (2020), therapy in combination with radiotherapy results in therapeutic antitumor
e1900996. immunity to murine lung cancer, Cancer Sci. 99 (2) (2008) 361–367.
[134] H. Cheng, G.L. Fan, J.H. Fan, R.R. Zheng, L.P. Zhao, P. Yuan, X.Y. Zhao, X.Y. Yu, [151] D. Hirschhorn-Cymerman, G.A. Rizzuto, T. Merghoub, A.D. Cohen, F. Avogadri,
S.Y. Li, A self-delivery chimeric peptide for photodynamic therapy amplified A.M. Lesokhin, A.D. Weinberg, J.D. Wolchok, A.N. Houghton, OX40 engagement
immunotherapy, Macromol. Biosci. 19 (4) (2019), e1800410. and chemotherapy combination provides potent antitumor immunity with
[135] C. Xu, J. Nam, H. Hong, Y. Xu, J.J. Moon, Positron emission tomography-guided concomitant regulatory T cell apoptosis, J. Exp. Med. 206 (5) (2009) 1103–1116.
photodynamic therapy with biodegradable mesoporous silica nanoparticles for [152] R.G. Alvim, P. Georgala, L. Nogueira, A.J. Somma, K. Nagar, J. Thomas, L. Alvim,
personalized cancer immunotherapy, ACS Nano 13 (10) (2019) 12148–12161. A. Riegel, C. Hughes, J. Chen, A.B. Reis, S. Lebdai, A. Scherz, S. Zanganeh,
[136] C.M. Fares, E.M. Van Allen, C.G. Drake, J.P. Allison, S. Hu-Lieskovan, R. Gardner, K. Kim, J.A. Coleman, Combined OX40 agonist and PD-1 inhibitor
Mechanisms of resistance to immune checkpoint blockade: why does checkpoint immunotherapy improves the efficacy of vascular targeted photodynamic therapy
inhibitor immunotherapy not work for all patients? Am. Soc. Clin. Oncol. Edu. in a urothelial tumor model, Molecules 26 (12) (2021).
Book 39 (2019) 147–164. [153] O. Boenisch, F. D’Addio, T. Watanabe, W. Elyaman, C.N. Magee, M.Y. Yeung, R.
[137] J.J. Havel, D. Chowell, T.A. Chan, The evolving landscape of biomarkers for F. Padera, S.J. Rodig, T. Murayama, K. Tanaka, X. Yuan, T. Ueno, A. Jurisch,
checkpoint inhibitor immunotherapy, Nat. Rev. Cancer 19 (3) (2019) 133–150. B. Mfarrej, H. Akiba, H. Yagita, N. Najafian, TIM-3: a novel regulatory molecule of
[138] L. Gao, C. Zhang, D. Gao, H. Liu, X. Yu, J. Lai, F. Wang, J. Lin, Z. Liu, Enhanced alloimmune activation, J. Immunol. 185 (10) (2010) 5806–5819.
anti-tumor efficacy through a combination of integrin alphavbeta6-targeted [154] C. Zhu, A.C. Anderson, A. Schubart, H. Xiong, J. Imitola, S.J. Khoury, X.X. Zheng,
photodynamic therapy and immune checkpoint inhibition, Theranostics 6 (5) T.B. Strom, V.K. Kuchroo, The Tim-3 ligand galectin-9 negatively regulates T
(2016) 627–637. helper type 1 immunity, Nat. Immunol. 6 (12) (2005) 1245–1252.
[139] X. Duan, C. Chan, N. Guo, W. Han, R.R. Weichselbaum, W. Lin, Photodynamic [155] S. Koyama, E.A. Akbay, Y.Y. Li, G.S. Herter-Sprie, K.A. Buczkowski, W.
therapy mediated by nontoxic core-shell nanoparticles synergizes with immune G. Richards, L. Gandhi, A.J. Redig, S.J. Rodig, H. Asahina, R.E. Jones, M.
checkpoint blockade to elicit antitumor immunity and antimetastatic effect on M. Kulkarni, M. Kuraguchi, S. Palakurthi, P.E. Fecci, B.E. Johnson, P.A. Janne, J.
breast cancer, J. Am. Chem. Soc. 138 (51) (2016) 16686–16695. A. Engelman, S.P. Gangadharan, D.B. Costa, G.J. Freeman, R. Bueno, F.S. Hodi,
[140] G. Yang, L. Xu, J. Xu, R. Zhang, G. Song, Y. Chao, L. Feng, F. Han, Z. Dong, B. Li, G. Dranoff, K.K. Wong, P.S. Hammerman, Adaptive resistance to therapeutic PD-1
Z. Liu, Smart nanoreactors for pH-responsive tumor homing, mitochondria- blockade is associated with upregulation of alternative immune checkpoints, Nat.
targeting, and enhanced photodynamic-immunotherapy of cancer, Nano Lett. 18 Commun. 7 (2016) 10501.
(4) (2018) 2475–2484. [156] J.E. Kim, M.A. Patel, A. Mangraviti, E.S. Kim, D. Theodros, E. Velarde, A. Liu, E.
[141] F. Sun, Q. Zhu, T. Li, M. Saeed, Z. Xu, F. Zhong, R. Song, M. Huai, M. Zheng, W. Sankey, A. Tam, H. Xu, D. Mathios, C.M. Jackson, S. Harris-Bookman,
C. Xie, L. Xu, H. Yu, Regulating glucose metabolism with prodrug nanoparticles T. Garzon-Muvdi, M. Sheu, A.M. Martin, B.M. Tyler, P.T. Tran, X. Ye, A. Olivi, J.
for promoting photoimmunotherapy of pancreatic cancer, Adv. Sci. 8 (4) (2021) M. Taube, P.C. Burger, C.G. Drake, H. Brem, D.M. Pardoll, M. Lim, Combination
2002746. therapy with anti-PD-1, anti-TIM-3, and focal radiation results in regression of
[142] Y. Liu, Y. Pan, W. Cao, F. Xia, B. Liu, J. Niu, G. Alfranca, X. Sun, L. Ma, J.M. de la murine gliomas, Clin. Cancer Res. 23 (1) (2017) 124–136.
Fuente, J. Song, J. Ni, D. Cui, A tumor microenvironment responsive [157] T.Y. Huang, G.L. Huang, C.Y. Zhang, B.W. Zhuang, B.X. Liu, L.Y. Su, J.Y. Ye,
biodegradable CaCO3/MnO2- based nanoplatform for the enhanced M. Xu, M. Kuang, X.Y. Xie, Supramolecular photothermal nanomedicine mediated
photodynamic therapy and improved PD-L1 immunotherapy, Theranostics 9 (23) distant tumor inhibition via PD-1 and TIM-3 blockage, Front. Chem. 8 (2020) 1.
(2019) 6867–6884. [158] X. Sun, Z. Cao, K. Mao, C. Wu, H. Chen, J. Wang, X. Wang, X. Cong, Y. Li, X. Meng,
[143] M. Platten, W. Wick, B.J. Van den Eynde, Tryptophan catabolism in cancer: X. Yang, Y.G. Yang, T. Sun, Photodynamic therapy produces enhanced efficacy of
beyond Ido and tryptophan depletion, Cancer Res. 72 (21) (2012) 5435–5440. antitumor immunotherapy by simultaneously inducing intratumoral release of
[144] R. Metz, S. Rust, J.B. Duhadaway, M.R. Mautino, D.H. Munn, N.N. Vahanian, C. sorafenib, Biomaterials 240 (2020) 119845.
J. Link, G.C. Prendergast, Ido inhibits a tryptophan sufficiency signal that

16

You might also like