You are on page 1of 8

Available online at www.sciencedirect.

com

ScienceDirect

Engineering immunogenic cell death with nanosized


drug delivery systems improving cancer immunotherapy
Wenlu Yan1,2,3, Tianqun Lang1,3, Xianrong Qi2 and Yaping Li1,3

Many anti-cancer therapies can induce or enhance the tumor microenvironment (ITM) [3]. One potential strat-
immunogenic cell death (ICD), a process that releases damage- egy to address these issues is promoting immunogenic
associated molecular patterns (DAMPs) to prime antigen cell death (ICD), a type of cell death releasing neoanti-
processing and presentation necessary for successful cancer gens as well as damage-associated molecular patterns
immunotherapy. However, the clinical potential of these (DAMPs) including calreticulin (CRT), high mobility
therapies, especially the chemotherapy, is limited by serious group box 1 (HMGB1), heat shock protein (HSP) and
systemic side effects, because of their non-specific adenosine-5’-triphosphate (ATP) [4]. CRT binding to
accumulation out of the tumors. Nanosized drug delivery CD91 receptor promotes phagocytosis together with
systems (NDDSs) can improve the specificity of anti-cancer HSP. Recruitment and activation of dendritic cells
therapies, which enhance ICD in the tumor while alleviating (DCs) are advanced after the recognition of ATP by
toxicities. In this review, we summarize recent progress of ICD- P2X7 receptor and HMGB1 by Toll-like receptor
inducing NDDSs with a focus on their enhanced safety and (TLR). Tumor-associated antigens (TAA) are then
efficacy for cancer immunotherapy. cross-presented to CD4+ and CD8+ T cells by matured
DCs, triggering adaptive immune responses against
Addresses tumors and turning tumor microenvironment (TME) to
1
State Key Laboratory of Drug Research & Center of Pharmaceutics, immunogenic state [5]. In summary, ICD-induction pro-
Shanghai Institute of Materia Medica, Chinese Academy of Sciences,
501 Haike Road, Shanghai 201203, China
vides a lymphocytes-enriched TME for enhancing immu-
notherapy (Figure 1a).
2
School of Pharmaceutical Sciences, Peking University, 38 Xueyuan
Road, Beijing 100191, China Meanwhile, non-specific deposition of ICD inducers
3
School of Pharmacy, University of Chinese Academy of Sciences,
or other immunotherapy agents is likely to cause irSEs
Beijing 100049, China
[6]. Encouragingly, nanosized drug delivery systems
Corresponding author: Li, Yaping (ypli@simm.ac.cn) (NDDSs) have shown great potential in addressing these
challenges with their abilities of improving the pharma-
cokinetic and bio-distribution profiles of drugs to accu-
Current Opinion in Biotechnology 2020, 66:36–43
mulate in tumors via the enhanced permeability and
This review comes from a themed issue on Tissue, cell and pathway retention (EPR) effect and decreasing the off-target
engineering
adverse effects [7,8]. NDDSs are also promising in the
Edited by Li Tang, Peng Xu and Haoran Zhang combined cancer therapy owning to their capacity of
For a complete overview see the Issue and the Editorial co-delivering drugs [9].
Available online 13th July 2020
https://doi.org/10.1016/j.copbio.2020.06.007
In this review, we will summarize the NDDSs-based
strategies that have been utilized to leverage ICD for
0958-1669/ã 2020 Elsevier Ltd. All rights reserved.
more efficient and safer cancer immunotherapy with a
focus on their design and mechanism of actions
(Figure 1b). Finally, the important contributions and
the possible directions of this field in the future will be
discussed.
Introduction
Immunotherapy revolutionizes the field of cancer therapy, Engineering ICD with NDDSs-based therapy
which normalizes or enhances immune surveillance to fight Treatments that can induce or enhance ICD include
against cancer [1]. Several immunotherapies including chemotherapy, phototherapy and radiotherapy [4].
immune checkpoint blockade (ICB) and T-cell transfer NDDSs transport ICD-inducing agents and immunother-
therapy have been used in the clinic. However, low response apy drugs to tumor sites together or separately, which
rate and severe immune-related side effects (irSEs) are two reduce adverse reactions and improve efficiency
major challenges for cancer immunotherapy [2]. (Figure 1c).

The low response rate of immunotherapy is likely associ- NDDSs-based chemotherapy


ated with reasons such as low immunogenicity of tumor, Some chemotherapeutics such as oxaliplatin (OXA) and
poor immune cell infiltration and immunosuppressive doxorubicin (DOX), as type Ⅰ ICD inducers, act on targets

Current Opinion in Biotechnology 2020, 66:36–43 www.sciencedirect.com


Engineering ICD with NDDSs for immunotherapy Yan et al. 37

Figure 1

(a) ICD improving different immunotherapies

(b) ICD-inducers deliveed by nanoplatforms improving immunotherapy (c) Different administration strategies

Current Opinion in Biotechnology

ICD-engineering treatments based on NDDSs improving immunotherapy.


Chemotherapy (CT), phototherapy (PT) and radiotherapy (RT) act on different targets (such as DNA, functional proteins or endoplasmic reticulum
(ER)) to induce ICD [10] ((a), left panel). Tumor cells undergoing ICD release DAMPs which activate DCs and T cells, improving the anti-tumor
effect of immunotherapy [4] ((a), middle panel). Three major immunotherapies are discussed in this review ((a), right panel). ICD inducers delivered
by functional nanoplatforms promote the efficiency and safety of immunotherapy [11] (b). ICD inducers and immunotherapy agents are targeted
delivered by nanoplatforms together or sequentially to tumor tissues and achieve tumor inhibition by triggering immune responses (c).

unrelated to endoplasmic reticulum (ER) such as DNA or therapies, anti-tumor effect will be greatly improved. Kuai
functional proteins to induce apoptosis and collateral ER et al. proposed a method to deliver DOX to tumors
stress effects [10]. Nanoparticles-encapsulated ICD indu- with a pH-sensitive high-density lipoprotein nanodisc
cers generated stronger anti-tumor effect than non-ICD (sHDL-DOX), increasing the intracellular uptake of
inducers or free ICD inducers [11], indicating that DOX by 2.8 times compared with free DOX injection
NDDSs-based chemotherapy was a feasible strategy to [13]. After sHDL-DOX and anti-PD-1 therapy, 88% mice’s
improve immunotherapy. tumors were eradicated, whereas anti-PD-1 monotherapy
could not control tumor growth of mice. The work of Zou
Combined with ICB et al. demonstrated similar results, in which catalase (CAT)
The low response rate due to limited T cell activation and and DOX were co-delivered via a biomimetic nanoplatform
infiltration in tumor is a main barrier of ICB. Chemotherapy [14]. DOX triggered ICD, while CAT catalyzed in situ
inducing ICD promotes the infiltration of CD8+ T cells, but H2O2 to relieve hypoxia of TME and downregulate the
upregulates the expression of PD-L1 through NF-kB expression of PD-L1 of tumor cells, thus creating an
signaling [12]. When combining these two complementary enabling environment for ICB.

www.sciencedirect.com Current Opinion in Biotechnology 2020, 66:36–43


38 Tissue, cell and pathway engineering

Since systemic ICB may cause irSEs, tumor-localized and homodimer of IDO inhibitor NLG919, with 30-fold
inhibition of PD-L1 is preferential. Su et al. utilized and 5.3-fold higher drug accumulation at the tumor site
copolymer of azide-terminated polyethylene glycol than free OXA and NLG919 [18]. Through dual regula-
(PEG) and poly-aspartic acid to assemble a pH-sensitive tion of ITM, the binary prodrug nanoparticle achieved
micelle with paclitaxel (PTX) in the core, which was then twofold stronger anti-cancer effect than free NLG919
decorated with anti-PD-1 antibodies (aPD-1) via matrix alone. The nanoparticle with simple preparation proce-
metalloproteinase-2 (MMP-2) -sensitive peptide linker dures is favorable for clinical transformation. Similar
and PEG chain [15]. The micelle became positively outcomes were shown that co-administration of chemo-
charged after PEG shedding and aPD-1 releasing in therapeutic agents and IDO inhibitor indoximod (IND)
weakly acidic (pH  6.5) and enzyme-rich TME, promot- regressed primary and metastatic tumors [19,20].
ing the internalization of tumor cells. PTX was finally
released in lysosomes (pH  5.0) and then induced ICD Combined with immunopotentiator
of tumor cells. This space-specific administration strategy Cancer vaccines triggering immune responses are likely
achieved synergistic anti-tumor profit, providing a feasi- to eliminate tumors. Besides biosafety, personalization of
ble idea for the co-delivery of ICB drugs and ICD vaccines and immune tolerance of TME are two barriers
inducers (Figure 2). The siRNA for PD-L1 knockdown that need to be overcome. In a recent work, curcumin-
or JQ1 for PD-L1 downregulation was co-delivered with loaded polymer nanoparticles with the particle size of
ICD inducers by nanocarriers, leading to successful anti- 98.73 nm and nanovaccines composed of antigen peptides
tumor chemo-immunotherapy as well [16,17]. and TLR agonists with the particle size of 91.23 nm self-
assembled into hydrogels [21]. Curcumin induced ICD
Combined with IDO inhibitor to turn ‘cold’ tumors into ‘hot’ tumors, and the vaccine
It has been confirmed that interferon-g (IFN-g) secretion further amplified the immune response. After combined
boosted by chemotherapeutics promotes overexpression of therapy, the infiltration of CD8+ T cells in tumors was
indoleamine 2,3-dioxygenase (IDO), a key factor leading to 4.89 and 2.11 times higher than that of nanomedicine and
ITM and immune escape. The IDO also promotes the nanovaccine, respectively.
proliferation of regulatory T cells (Tregs). Inhibitors of
IDO are thus able to reverse ITM and elicit the immune Adjuvants, as immune enhancers, are expected to be a
response with ICD. Inspired by this idea, Feng et al. con- new choice for cancer therapy. Cytosine-phosphate-
structed a prodrug nanoparticle with PEG-grafted OXA guanine (CpG) can induce the immune response and

Figure 2

Current Opinion in Biotechnology

Smart NDDS achieving space-specific administration of aPD-l and PTX.


MMP-2 and pH dual-sensitive NDDS co-delivered aPD-l and PTX, releasing aPD-l in weakly acidic and enzyme-rich TME. PTX induced ICD after
internalization by tumor cells, synergizing with ICB therapy to enhance the killing effect of CD8+ T cells on tumor cells [15].

Current Opinion in Biotechnology 2020, 66:36–43 www.sciencedirect.com


Engineering ICD with NDDSs for immunotherapy Yan et al. 39

promote the secretion of pro-inflammatory cytokines after photosensitizer) with aPD-L1 through a TME-sensitive
recognition by TLR-9. However, high dose and repeated nanocarrier, promoting DCs maturation and CTLs infil-
administration cause adverse reactions. To address the tration after laser irradiation [24]. Assisted by combining
problem, Wang et al. developed nanocomposites by con- therapy, over 80% 4T1 tumor-bearing mice survived until
necting DOX and CpG-silver nanocluster to MnO2 70 days, whereas mice treated aPD-L1 all died within
nanosheets, taking advantage of DOX as bi-functional 45 days.
modulator for ICD induction and TME transformation to
stimulate cancer immunotherapy of CpG [22]. The tumor Besides co-transportation, the lipid encapsulating the
inhibition rate of combined treatment was 95%, 3.9-fold Zn2+ ions coordinated with pyrophosphate as the core
higher than treatment without DOX. and the lipid layer loaded with photosensitizer assembled
the nanoparticles, which preferentially accumulated in
NDDSs-based phototherapy tumor owing to long circulation capacity with blood
Recent studies have shown that phototherapy including circulation half-life of 14.5 hours. Then the radiation
photodynamic therapy (PDT) and photothermal therapy boosted release of TAA, enabling complete tumor eradi-
(PTT) can also induce systemic activity by priming anti- cation with subsequent anti-PD therapy [25].
cancer immunity. As a type Ⅱ inducer, PDT selectively
targets to ER and generates reactive oxygen species PTT inducing ICD maintains the immunogenic micro-
(ROS) through photosensitizer, exposing CRT via PERK environment for ICB therapy. Under infrared light irra-
and BAX/BAK signal pathway [5,10]. PTT kills cancer diation, nanoplatforms loaded with photothermal agent
cells by generating local hyperthermia, leading to release and TLR receptor agonist [26], tubulin-stabilizer, or
of TAA without relying on the hypoxic microenvironment stimulator of interferon genes agonist [27] triggered spe-
of tumor tissue [23]. With the ability of immune stimula- cific immune response and achieved long-term tumor
tion, phototherapy is thus potential to synergize with suppression when combined with ICB therapy.
immunotherapy to achieve comprehensive suppression
of primary and metastatic tumors. Another innovative study reported a multi-functional
nanoparticle formed with light absorber, photosensitizer
Combined with ICB and DSPE-PEG-maleimide, with the ability of capturing
Promotion of intratumoral infiltration of cytotoxic T the TAA induced by thermal radiation and transporting
lymphocytes (CTLs) improves the efficacy of ICB ther- them to DCs [28] (Figure 3). From primary induction to
apy. Wang et al. combined indocyanine green (ICG, a accomplishing chain processing of the antigens, this

Figure 3

Current Opinion in Biotechnology

Nanoparticles capturing tumor antigens to improve immunotherapy.


Nanoparticles capture tumor antigens released from primary tumor cells undergoing ICD and transport them to DCs. Combining this strategy with
aCTLA-4 [28] or aPD-1 [29], suppression of metastatic tumor can be achieved.

www.sciencedirect.com Current Opinion in Biotechnology 2020, 66:36–43


40 Tissue, cell and pathway engineering

strategy provides a new insight for improving the immune of adhesion molecule and E-selectin to boost immune cell
response induced by ICD. recruitment and inflammatory cytokine signaling. The
chemokine gradient attracts effector T cells to irradiated
Combined with IDO inhibitor tumor with the promotion of vascular cell adhesion mol-
Physical therapy, including phototherapy, can also upreg- ecule (VCAM)-1, and the increased expression of major
ulate IDO. To address this issue, Gao et al. prepared a histocompatibility complex class Ⅰ (MHC-I) on the tumor
TME-sensitive prodrug vesicle for specific delivery of improves recognition and killing by immune system.
NLG919 and photosensitizer pheophorbide A (PPa) [30]. Effector T cells kill cancer cells through Fas-induced
ICD induction by PDT combined with IDO inhibition apoptosis or cytotoxic granules, releasing new TAA and
provoked 34.4  1.3% maturation of DCs and increased thus boosting the abscopal effect medicated by immune
the intratumoral infiltration of IFN-g+ CD8+ T cells by response [37,38].
30-fold higher than the control group. This work
addressed the negative feedback regulation of immuno- Low-dose hypo-fractionated radiation reprograms macro-
suppression caused by PDT. Moreover, a nanovesicle phages to M1 phenotype and induces more effective
assembled from block copolymer polyethylene glycol- ICD-based antigen release than single-dose radiotherapy.
b-cationic polypeptide not only carried photosensitizer High-atomic-number nanoparticles with high X-ray
and IND, but also induced ICD itself [31]. Through the absorption coefficients, as novel radiosensitizers, contrib-
cytotoxicity of ROS, immune activation, and regulation of ute to the local damage to tumor cells of radiotherapy.
ITM, PDT immunotherapy inhibited tumor growth with- Hafnium (Hf)-based nanoscale metal-organic frameworks
out causing obvious systemic toxicity, while free IND had which could promote ROS diffusion, were injected
minimal inhibitory effect on the tumor. In addition, a intratumorally followed by low-dose hypo-fractionated
redox-responsive disulfide nanoliposome activated radiation, extinguishing both the primary irradiated and
photoactivity by more than 100-fold without photosensi- secondary unirradiated tumor combined with aPD-L1 or
tizer and promoted anti-cancer treatment synergistically IDO inhibitor [39,40].
when encapsulating IND [32].
The effectiveness of radiotherapy depends on the con-
Combined with immunopotentiator centration of oxygen. Chen et al. designed a biocompati-
To solve the problems of limited light penetration depth ble nanoparticle with hydrophilic CAT in the core and
of PDT and low efficiency of photosensitizer loading, a hydrophobic immune adjuvant R837 in the poly(lactic-co-
large-pore mesoporous-silica-coated upconversion nano- glycolic) acid (PLGA) shell through double emulsification
particle was designed with the capability of loading method [41]. CAT relieved hypoxia of tumor tissue and
photosensitizers (12.28 wt%) and tumor cell fragments adjuvant promoted the recognition of tumor antigens by
as autoantigens (39.7 wt%) [33]. During PDT, endoge- immune cells under X-ray radiation. When combined
nous and exogenous antigens synergistically promoted radiotherapy with aCTLA-4, 60% of mice achieved
the proliferation and activation of T cells. Since intact 60-day survival, which indicated the nanoparticle was
cells were more immunogenic than cell fragments, Wang a feasible strategy for broadening the application of
et al. used the tumor-penetrating peptides to assemble radiotherapy.
nanofibers and transform into hydrogels. Then tumor
cells attached ICG and JQ1 were encapsulated [34]. Not only could nanoparticles mediate the production of
Local injection delayed 48.1% of the tumor recurrence tumor antigens, but also capture and deliver antigens
and eliminated 83.3% of metastatic tumor, showing the produced by radiotherapy to DCs to enhance cancer immu-
potential of this strategy in personalized immunotherapy. nity. Min et al. decorated nanoparticles formulated from
PLGA with amine polyethylene glycol, 1,2-dioleoyloxy-3-
High local hyperthermia predicts better outcome of PTT (trimethylammonium) propane or maleimide polyethylene
but also causes damages to healthy tissues and immune glycol for binding to TAA through different mechanisms, so
cells in the TME, leading to immune suppression. Chen as to promote antigen presentation and trigger a successful
et al. demonstrated that mild hyperthermia could induce adaptive immune response [29]. This approach is com-
durable immune memory and delay tumor growth using patible with clinical immunotherapy treatment options,
NDDSs loaded with TLR agonists, proving that balanc- and thus is of great translation potential.
ing tumor eradication and immune cell priming was
critical for an in situ vaccine [35]. Furthermore, to enhance the efficacy of radiation, opti-
mization of nanoparticles to maximize cellular uptake is
NDDSs-based radiotherapy highlighted [42]. Notably, attention should also be paid
Radiotherapy is the mainstream external treatment to timing and sequencing of radiotherapy and ICB,
for cancer by primarily disrupting the chemical bonds since radiation-induced acute increase of TAA produc-
between the bases in DNA [36]. The generation of ROS tion and tumor infiltrating lymphocytes only lasts for a
induced by ionizing radiation medicates the upregulation few days [43].

Current Opinion in Biotechnology 2020, 66:36–43 www.sciencedirect.com


Engineering ICD with NDDSs for immunotherapy Yan et al. 41

NDDSs-based combinatorial regimens [42]. Au et al. designed a DOX-encapsulated nanoparticle


Combination use of chemotherapy and physical therapy with the capacity of targeting to human leukocyte
(including phototherapy and radiotherapy) with different antigen-D related (HLA-DR) -overexpressed human
patterns of released DAMPs may complement with each lymphoma cells [48]. ICD induced by DOX and irradia-
other and exhibit stronger ICD. One requisition to tion resulted in the upregulation of HLA-DR, promoting
achieve synergistic effect is to deliver different drugs uptake of nanoparticles and exerting anti-tumor effi-
into the tumor and release them in a controlled manner. ciency with a positive feedback pathway. As a result,
NDDSs with multifunctionality, can fulfil this after about 70% of mice achieved long-term survival after the
optimization. concurrent chemo-immuno-radiotherapy.

The most commonly used strategy is the combination of In order to achieve ICD induction and endogenous IDO
phototherapy and chemotherapy. Feng et al. reported a inhibition, PEGylated OXA with ROS-liable thioketal
two-in-one nanoparticle loading ICG to medicate PDT spacer encapsulated GSH-liable PPa and NLG919 pro-
and PTX to reverse ITM efficiently without excipients drug, composing a light-inducible nanocargo (LINC)
(termed as ISPN) [44]. It could target to tumor tissue and [49]. LINC accumulated in tumor tissue and cleaved
secrete ATP after laser irradiation by twofold higher than PEG under the first near-infrared (NIR) laser. After
that of various control groups, indicating a more effective tumor cells internalization of LINC, NLG919 reversing
ICD induction. The combination treatment of ISPN with ITM and immune response triggered by the second
aPD-L1 caused a significant immune response against irradiation-medicated PDT and OXA eradicated 67%
tumor growth and metastasis (Figure 4). Other studies of the 4T1 tumors. The controllable drug delivery strat-
demonstrated that PDT and chemotherapy cumulatively egy enabled efficient and simple triple combination
induced ICD as well, delaying the growth of primary therapy.
tumors cooperated with aPD-1 or aCD47 [45,46].
Besides PDT, the combination of chemotherapy and Discussion and futures directions
PTT also facilitated the immune response, providing Recently, different categories of ICD-manipulating treat-
enough exposure of TAA for ICB therapy [47]. ments based on NDDSs have been developed and
showed great potential in cancer immunotherapy. The
Several studies have indicated that NDDS-based advantages of NDDSs-based ICD are as followed. Firstly,
chemotherapeutics can exacerbate DNA double-strand it targets to tumor site and induces the release of DAMPs
damage caused by radiotherapy with unobvious toxicity to act as an in situ vaccine, thereby achieving the effect of

Figure 4

Current Opinion in Biotechnology

Enhancing cancer immunotherapy by ISPN.


Phototherapy and chemotherapy synergistically induce ICD of tumor cells and eliminate immunosuppressive Tregs, inhibiting primary and
metastasis tumors when combined with ICB [44].

www.sciencedirect.com Current Opinion in Biotechnology 2020, 66:36–43


42 Tissue, cell and pathway engineering

personalized treatment. Secondly, it improves the 5. Rufo N, Garg AD, Agostinis P: The unfolded protein response in
immunogenic cell death and cancer immunotherapy. Trends
infiltration of immune cells to prime immune replete Cancer 2017, 3:643-658.
TME, accomplishing more effective immunotherapy 6. June CH, Warshauer JT, Bluestone JA: Is autoimmunity the
for tumor suppression while reducing side effects. achilles’ heel of cancer immunotherapy? Nat Med 2017,
Thirdly, the combined delivery of ICD inducers and 23:540-547.

immunotherapeutic agents with NDDSs improves their 7. Duan X, Chan C, Lin W: Nanoparticle-mediated immunogenic
cell death enables and potentiates cancer immunotherapy.
pharmacokinetic and bio-distribution profiles and Angew Chem Int Ed Engl 2019, 58:670-680.
achieves synergistic effects simultaneously.
8. Gao J, Wang WQ, Pei Q, Lord MS, Yu HJ: Engineering
nanomedicines through boosting immunogenic cell death for
More investigations are needed to transfer engineering improved cancer immunotherapy. Acta Pharmacol Sin 2020, 0:1-9.
ICD with NDDSs to clinical application such as devel- 9. Goldberg MS: Improving cancer immunotherapy through
oping highly biocompatible and biodegradable NDDSs.  nanotechnology. Nat Rev Cancer 2019, 19:587-602.
This review explores the application of nanotechnology in
In addition, how the interaction between nanoparticles immunotherapy.
and the immune system can be controlled by tuning the
10. Dudek AM, Garg AD, Krysko DV, De Ruysscher D, Agostinis P:
physicochemical properties of the NDDSs should be Inducers of immunogenic cancer cell death. Cytokine Growth
investigated. Furthermore, DAMPs released by ICD Factor Rev 2013, 24:319-333.
are a double-edged sword that may trigger chronic inflam- 11. Zhao X, Yang K, Zhao R, Ji T, Wang X, Yang X, Zhang Y, Cheng K,
mation and thus promote tumor development [50]. Thus,  Liu S, Hao J et al.: Inducing enhanced immunogenic cell death
with nanocarrier-based drug delivery systems for pancreatic
combination use of immune modulators is necessary, and cancer therapy. Biomaterials 2016, 102:187-197.
the type, dosage and release kinetic of these modulators The first report on the roles of ICD in treatment with nanomedicines.
should be carefully tuned. Therapy-resistant microevolu- 12. Peng J, Hamanishi J, Matsumura N, Abiko K, Murat K, Baba T,
tion under selection pressure in cancer is also a hurdle that Yamaguchi K, Horikawa N, Hosoe Y, Murphy SK et al.:
Chemotherapy induces programmed cell death-ligand
ICD-mediating therapies need to address. The properties 1 overexpression via the nuclear factor-kB to foster an
of ideal ICD inducers include non-susceptible to drug- immunosuppressive tumor microenvironment in ovarian
cancer. Cancer Res 2015, 75:5034-5045.
efflux channels and capable of inducing severe focused
ER stress, and so on [51]. For clinical translation, feasible 13. Kuai R, Yuan W, Son S, Nam J, Xu Y, Fan Y, Schwendeman A,
Moon JJ: Elimination of established tumors with nanodisc-
yet effective NDDSs design is required, which is neces- based combination chemoimmunotherapy. Sci Adv 2018, 4:
sary for large-scale production and quality control. We eaao1736.
believe that with the development of biotechnology and 14. Zou M, Liu W, Li C, Zheng D, Zeng J, Gao F, Ye J, Zhang X: A
biological engineering, the relevant mechanisms will be  multifunctional biomimetic nanoplatform for relieving hypoxia
to enhance chemotherapy and inhibit the PL-1/PD-L1 Axis.
further clarified. Engineering ICD based on NDDSs will Small 2018, 14:e1801120.
advance the innovation of cancer immunotherapy. Achieving dual inhibition to PD-1/PD-L1 Axis.
15. Su Z, Xiao Z, Wang Y, Huang J, An Y, Wang X, Shuai X: Codelivery
 of anti-PD-1 antibody and paclitaxel with matrix
Conflict of interest statement metalloproteinase and pH dual-sensitive micelles for
enhanced tumor chemoimmunotherapy. Small 2020, 16:
Nothing declared. e1906832.
Smart NDDS achieving controllable release of aPD-1 and PTX.
16. Tang X, Rao J, Yin S, Wei J, Xia C, Li M, Mei L, Zhang Z, He Q: PD-
Acknowledgements L1 knockdown via hybrid micelle promotes paclitaxel induced
National Natural Science Foundation of China (81871471, 81630052, cancer-immunity cycle for melanoma treatment. Eur J Pharm
81521005, 81690265), Natural Science Foundation of Shanghai Sci 2019, 127:161-174.
(19ZR1479900), and Key Scientific Research Program of CAS (QYZDJ- 17. Wang H, Tang Y, Fang Y, Zhang M, Wang H, He Z, Wang B, Xu Q,
SSW-SMC020) are gratefully acknowledged for financial support. Huang Y: Reprogramming tumor immune microenvironment
(TIME) and metabolism via biomimetic targeting codelivery of
shikonin/JQ1. Nano Lett 2019, 19:2935-2944.
References and recommended reading 18. Feng B, Zhou F, Hou B, Wang D, Wang T, Fu Y, Ma Y, Yu H, Li Y:
Papers of particular interest, published within the period of review,  Binary cooperative prodrug nanoparticles improve
have been highlighted as immunotherapy by synergistically modulating immune tumor
microenvironment. Adv Mater 2018, 30:e1803001.
 of special interest Activating ICD and reversing the ITM with a tumor acidity and reduction
 of outstanding interest dual activatable NDDS

1. Sanmamed MF, Chen L: A paradigm shift in cancer 19. Lu J, Liu X, Liao YP, Salazar F, Sun B, Jiang W, Chang CH, Jiang J,
immunotherapy: from enhancement to normalization. Cell Wang X, Wu AM et al.: Nano-enabled pancreas cancer
2018, 175:313-326. immunotherapy using immunogenic cell death and reversing
immunosuppression. Nat Commun 2017, 8:1811.
2. Riley RS, June CH, Langer R, Mitchell MJ: Delivery
technologies for cancer immunotherapy. Nat Rev Drug 20. Lu J, Liu X, Liao YP, Wang X, Ahmed A, Jiang W, Ji Y, Meng H,
Discov 2019, 18:175-196. Nel AE: Breast cancer chemo-immunotherapy through
liposomal delivery of an immunogenic cell death stimulus
3. Kalbasi A, Ribas A: Tumour-intrinsic resistance to immune plus interference in the IDO-1 pathway. ACS Nano 2018,
checkpoint blockade. Nat Rev Immunol 2020, 20:25-39. 12:11041-11061.
4. Li X: The inducers of immunogenic cell death for tumor 21. Liu X, Feng Z, Wang C, Su Q, Song H, Zhang C, Huang P, Liang XJ,
immunotherapy. Tumori 2018, 104:1-8.  Dong A, Kong D, Wang W: Co-localized delivery of

Current Opinion in Biotechnology 2020, 66:36–43 www.sciencedirect.com


Engineering ICD with NDDSs for immunotherapy Yan et al. 43

nanomedicine and nanovaccine augments the postoperative 35. Chen P, Pan W, Wu C, Yeh C, Korupalli C, Luo P, Chou C, Chia W,
cancer immunotherapy by amplifying T-cell responses. Sung H: Modulation of tumor microenvironment using a TLR-7/
Biomaterials 2020, 230:119649. 8 agonist-loaded nanoparticle system that exerts low-
Codelivery of nanomedicines and nanovaccines to overcome the limita- temperature hyperthermia and immunotherapy for in situ
tion of cancer immunotherapy. cancer vaccination. Biomaterials 2020, 230 119629.
22. Wang Z, Zhang Y, Liu Z, Dong K, Liu C, Ran X, Pu F, Ju E, Ren J, 36. Golden EB, Apetoh L: Radiotherapy and immunogenic cell
Qu X: A bifunctional nanomodulator for boosting CpG- death. Semin Radiat Oncol 2015, 25:11-17.
mediated cancer immunotherapy. Nanoscale 2017, 9:14236-
14247. 37. Formenti S, Demaria S: Systemic effects of local radiotherapy.
Lancet Oncol 2009, 10:718-726.
23. Ng CW, Li J, Pu K: Recent progresses in phototherapy-
synergized cancer immunotherapy. Adv Funct Mater 2018, 38. Ngwa W, Irabor OC, Schoenfeld JD, Hesser J, Demaria S,
28:1804688. Formenti SC: Using immunotherapy to boost the abscopal
effect. Nat Rev Cancer 2018, 18:313-322.
24. Wang D, Wang T, Yu H, Feng B, Zhou L, Zhou F, Hou B, Zhang H, 39. Ni K, Lan G, Chan C, Quigley B, Lu K, Aung T, Guo N, La Riviere P,
Luo M, Li Y: Engineering nanoparticles to locally activate T Weichselbaum RR, Lin W: Nanoscale metal-organic
cells in the tumor microenvironment. Sci Immunol 2019, 4: frameworks enhance radiotherapy to potentiate checkpoint
eaau6584. blockade immunotherapy. Nat Commun 2018, 9:2351.
25. Duan X, Chan C, Guo N, Han W, Weichselbaum RR, Lin W: 40. Lu K, He C, Guo N, Chan C, Ni K, Lan G, Tang H, Pelizzari C, Fu YX,
Photodynamic therapy mediated by nontoxic core-shell Spiotto MT, Weichselbaum RR et al.: Low-dose X-ray
nanoparticles synergizes with immune checkpoint blockade radiotherapy-radiodynamic therapy via nanoscale metal-
to elicit antitumor immunity and antimetastatic effect on organic frameworks enhances checkpoint blockade
breast cancer. J Am Chem Soc 2016, 138:16686-16695. immunotherapy. Nat Biomed Eng 2018, 2:600-610.
26. Ge R, Liu C, Zhang X, Wang W, Li B, Liu J, Liu Y, Sun H, Zhang D, 41. Chen Q, Chen J, Yang Z, Xu J, Xu L, Liang C, Han X, Liu Z:
Hou Y et al.: Photothermal-activatable Fe3O4 superparticle Nanoparticle-enhanced radiotherapy to trigger robust cancer
nanodrug carriers with PD-L1 immune checkpoint blockade immunotherapy. Adv Mater 2019, 31:e1802228.
for anti-metastatic cancer immunotherapy. ACS Appl Mater
Interfaces 2018, 10:20342-20355. 42. Her S, Jaffray DA, Allen C: Gold nanoparticles for applications in
cancer radiotherapy: mechanisms and recent advancements.
27. Wang J, Meng J, Ran W, Lee RJ, Teng L, Zhang P, Li Y: Adv Drug Deliv Rev 2017, 109:84-101.
Hepatocellular carcinoma growth retardation and PD-1
blockade therapy potentiation with synthetic high-density 43. Lamichhane P, Amin NP, Agarwal M, Lamichhane N: Checkpoint
lipoprotein. Nano Lett 2019, 19:5266-5276. inhibition: Will combination with radiotherapy and
nanoparticle-mediated delivery improve efficacy? Medicines
28. Wang M, Song J, Zhou F, Hoover AR, Murray C, Zhou B, Wang L, (Basel) 2018, 5:114.
Qu J, Chen WR: NIR-triggered phototherapy and
immunotherapy via an antigen-capturing nanoplatform for 44. Feng B, Niu Z, Hou B, Zhou L, Li Y, Yu H: Enhancing triple
metastatic cancer treatment. Adv Sci (Weinh) 2019, 6:1802157. negative breast cancer immunotherapy by ICG-templated
self-assembly of paclitaxel nanoparticles. Adv Funct Mater
29. Min Y, Roche KC, Tian S, Eblan MJ, McKinnon KP, Caster JM, 2020, 30:1906605.
 Chai S, Herring LE, Zhang L, Zhang T et al.: Antigen-capturing
nanoparticles improve the abscopal effect and cancer 45. Liu H, Hu Y, Sun Y, Wan C, Zhang Z, Dai X, Lin Z, He Q, Yang Z,
immunotherapy. Nat Nanotechnol 2017, 12:877-882. Huang P et al.: Co-delivery of bee venom melittin and a
One of first paper describing the development and use of antigen- photosensitizer with an organic-inorganic hybrid nanocarrier
capturing nanoparticles to improve cancer immunotherapy. for photodynamic therapy and immunotherapy. ACS Nano
2019, 13:12638-12652.
30. Gao A, Chen B, Gao J, Zhou F, Saeed M, Hou B, Li Y, Yu H:
Sheddable prodrug vesicles combating adaptive immune 46. Zhou F, Feng B, Yu H, Wang D, Wang T, Ma Y, Wang S, Li Y:
resistance for improved photodynamic immunotherapy of Tumor microenvironment-activatable prodrug vesicles for
cancer. Nano Lett 2020, 20:353-362. nanoenabled cancer chemoimmunotherapy combining
immunogenic cell death induction and CD47 blockade. Adv
31. Yang W, Zhang F, Deng H, Lin L, Wang S, Kang F, Yu G, Lau J, Mater 2019, 31:e1805888.
 Tian R, Zhang M et al.: Smart nanovesicle-mediated
immunogenic cell death through tumor microenvironment 47. Wen Y, Chen X, Zhu X, Gong Y, Yuan G, Qin X, Liu J:
modulation for effective photodynamic immunotherapy. ACS Photothermal-chemotherapy integrated nanoparticles with
Nano 2020, 14:620-631. tumor microenvironment response enhanced the induction of
Nanovesicle and PDT achieve dual ICB induction. immunogenic cell death for colorectal cancer efficient
treatment. ACS Appl Mater Interfaces 2019, 11:43393-43408.
32. Liu D, Chen B, Mo Y, Wang Z, Qi T, Zhang Q, Wang Y: Redox- 48. Au KM, Balhorn R, Balhorn MC, Park SI, Wang AZ: High-
activated porphyrin-based liposome remote-loaded with performance concurrent chemo-immuno-radiotherapy for the
indoleamine 2,3-dioxygenase (IDO) inhibitor for synergistic treatment of hematologic cancer through selective high-
photoimmunotherapy through induction of immunogenic cell affinity ligand antibody mimic-functionalized doxorubicin-
death and blockage of IDO pathway. Nano Lett 2019, 19:6964- encapsulated nanoparticles. ACS Cent Sci 2019, 5:122-144.
6976.
49. Feng B, Hou B, Xu Z, Saeed M, Yu H, Li Y: Self-amplified drug
33. Ding B, Shao S, Yu C, Teng B, Wang M, Cheng Z, Wong KL, Ma P,  delivery with light-inducible nanocargoes to enhance cancer
Lin J: Large-pore mesoporous-silica-coated upconversion immunotherapy. Adv Mater 2019, 31:e1902960.
nanoparticles as multifunctional immunoadjuvants with This study establishes NDDS that can co-load chemotherapy, photo-
ultrahigh photosensitizer and antigen loading efficiency for therapy and immunotherapy drugs.
improved cancer photodynamic immunotherapy. Adv Mater
2018, 30:e1802479. 50. Hernandez C, Huebener P, Schwabe RF: Damage-associated
molecular patterns in cancer: a double-edged sword.
34. Wang T, Wang D, Yu H, Feng B, Zhou F, Zhang H, Zhou L, Jiao S, Oncogene 2016, 35:5931-5941.
 Li Y: A cancer vaccine-mediated postoperative
immunotherapy for recurrent and metastatic tumors. Nat 51. Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P,
Commun 2018, 9:1532.  Vandenabeele P: Immunogenic cell death and DAMPs in cancer
This article reports that nanofibers assembled from cell-penetrating therapy. Nat Rev Cancer 2012, 12:860-875.
peptides are transformed into hydrogels to encapsulate cancer vaccines A review of the detailed mechanism of ICD and the properties of ideal ICD
modified with PD-1 inhibitors and photosensitizers. inducers.

www.sciencedirect.com Current Opinion in Biotechnology 2020, 66:36–43

You might also like