Chem Soc Rev: Review Article

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 50

Chem Soc Rev

View Article Online


REVIEW ARTICLE View Journal

Nanomedicine embraces cancer radio-


Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

immunotherapy: mechanism, design, recent


Cite this: DOI: 10.1039/d2cs00437b
advances, and clinical translation
Haonan Li, †a Qiang Luo,†a Hu Zhang, c
Xuelei Ma,a Zhongwei Gu, a

Qiyong Gong*ab and Kui Luo *ab

Cancer radio-immunotherapy, integrating external/internal radiation therapy with immuno-oncology treatments,


emerges in the current management of cancer. A growing number of pre-clinical studies and clinical trials have
recently validated the synergistic antitumor effect of radio-immunotherapy, far beyond the ‘‘abscopal effect’’, but
it suffers from a low response rate and toxicity issues. To this end, nanomedicines with an optimized design
have been introduced to improve cancer radio-immunotherapy. Specifically, these nanomedicines are elegantly
prepared by incorporating tumor antigens, immuno- or radio-regulators, or biomarker-specific imaging agents
into the corresponding optimized nanoformulations. Moreover, they contribute to inducing various biological
effects, such as generating in situ vaccination, promoting immunogenic cell death, overcoming radiation
resistance, reversing immunosuppression, as well as pre-stratifying patients and assessing therapeutic response
Received 21st July 2022 or therapy-induced toxicity. Overall, this review aims to provide a comprehensive landscape of nanomedicine-
DOI: 10.1039/d2cs00437b assisted radio-immunotherapy. The underlying working principles and the corresponding design strategies for
these nanomedicines are elaborated by following the concept of ‘‘from bench to clinic’’. Their state-of-the-art
rsc.li/chem-soc-rev applications, concerns over their clinical translation, along with perspectives are covered.

a
Department of Radiology, Department of Biotherapy, Huaxi MR Research Center 1. Introduction
(HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular
Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan In recent years, cancer immunotherapy, harnessing the innate
University, No. 37 Guoxue Alley, Chengdu 610041, China. or adaptive host immune systems to combat cancer, has
E-mail: qiyonggong@hmrrc.org.cn, luokui@scu.edu.cn enjoyed a flourishing growth rate. Generally, it is divided into
b
Functional and Molecular Imaging Key Laboratory of Sichuan Province and two major antineoplastic approaches: active cancer vaccines for
Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu
prophylactic or therapeutic use and passive immuno-oncology
610041, China
c
Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA 91711, USA treatments.1 Passive treatments include administration of
† These authors contributed equally. immune checkpoint inhibitors (ICIs) targeting programmed

Haonan Li received his M. M. Qiang Luo received his PhD degree


degree from Sichuan University in from Sichuan University in 2021.
2020 under the supervision of Prof. He then joined the postdoctoral
Kui Luo and Prof. Qiyong Gong. position at West China Hospital,
Currently, he is pursuing a PhD Sichuan University, under the
degree under the guidance of Prof. guidance of Prof. Kui Luo and
Kui Luo at the Huaxi MR Research Prof. Qiyong Gong. His research
Center at West China Hospital, focuses on nanomedicine-assisted
Sichuan University. His research tumor therapy, radionuclide-based
interests focus on the smart cardiac interventional radio-
nanomedicine-aided cancer radio- therapy, and the treatment of
immunotherapy, theranostics, and immunotherapy-induced cardio-
Haonan Li molecular imaging. Qiang Luo toxicity using intelligent nano-
medicine.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

death-1 (PD-1),2 programmed death ligand-1 (PD-L1) or cyto- to the above concerns.12–14 Briefly, RT and immunotherapy
toxicity T-lymphocyte-associated protein 4 (CTLA-4),3,4 injection share a solid biological basis for their synergistic antitumor
of chimeric antigen receptor T-cell therapy (CAR T),5 bispecific effect. RT, consisting of external beam irradiation (X-rays, g-
antibodies (bsAbs, e.g., blinatumomab),6 costimulatory recep- rays, protons, or carbon ions) and internal radioisotope-
tors (e.g., CD137, CD134, and toll-like receptor) agonists,7,8 and mediated brachytherapy,15 is a prevalent cancer treatment
interferons/immunocytokines (e.g., interleukin-12).9 However, method as nearly 50% of cancer patients have received RT.16
as indicated by increasing evidence, this cancer treatment For antitumor therapy, RT exhibits a local tumoricidal effect
suffers from a low response rate on several types of solid and induces stromal, immunological, and vascular changes,
tumors, limited patient benefits, immune-related adverse but it often fails to treat metastases. In this context, systemic
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

events, and pseudoprogression.10,11 immunotherapy is an excellent addition to RT to achieve a


Inspired by enormous benefits achieved by combined ther- whole-body anti-primary-tumor and anti-metastasis therapy. In
apy, the combination of immunotherapy with radiation therapy addition, RT can evoke various immunogenic responses, parti-
(RT), termed as radio-immunotherapy, has recently thrived as a cularly immunogenic cell death (ICD) and the ‘‘abscopal
potent weapon fighting against cancer and a potential solution effect’’.17 Thus, RT may act as a response enhancer for

Prof. Hu Zhang received his PhD Prof. Zhongwei Gu is a Professor


degree in Biochemical Engineering at the West China Hospital of
from the Department of Bio- Sichuan University. Prof. Gu has
chemical Engineering at University been appointed as a Chief
College London (UK) in 2004. He is Scientist of the National Basic
currently an Adjunct Professor in Research Program of China (the
Bioprocessing at Keck Graduate 973 Program) for three five-year
Institute (USA). Prof. Zhang has periods since 1999. He has
published over 130 peer-viewed awarded numerous awards,
papers. His research focuses on including the Fellow of the
applying emerging techniques to International Union of Societies
biological processes and systems to for Biomaterials Science and
Hu Zhang produce valuable biological or Zhongwei Gu Engineering (FBSE) and the
chemical products, as well as Distinguished Visiting Fellow-
smart nanomedicine for drug and ship Award of the Royal Academy of Engineering. He has
gene delivery. published over 300 peer-reviewed scientific manuscripts and held
more than 30 issued patents. His research activities focus on
nanobiomaterials, biomimetic delivery systems, and tissue
engineering.

Prof. Qiyong Gong is currently a Prof. Kui Luo received his PhD
Full Professor of Clinical degree in Biomedical Engineering
Radiology at the West China (2009) from Sichuan University
Hospital of Sichuan University, under the supervision of Prof.
Director of the Key Laboratory of Zhongwei Gu. From 2009 to 2011,
Functional and Molecular he carried out postdoctoral work on
Imaging of Sichuan Province, polymeric nanomedicines at the
and the President of West China University of Utah, USA. Dr Luo
Xiamen Hospital of Sichuan was promoted to an Associate
University. Prof. Gong has Professor in 2012 and a Full
published more than 600 peer- Professor in 2013 in Sichuan
viewed papers, with an h-index University. From 2016, he is a Full
Qiyong Gong of 91, and he is the 2018–19, Kui Luo Professor and PI in West China
2022 Highly Cited Researcher by Hospital, Sichuan University,
Web of Science Group of the Clarivate Analytics. The main research China. He has authored over 140 peer-reviewed publications with an
in his lab focuses on magnetic resonance imaging of psychiatric h-index of 43. His research interests include nanomedicines, polymers-
disorders and brain tumors, as well as functional nanomedicines based imaging agents and drug/gene delivery systems.
for molecular imaging and cancer therapy.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

immuno-oncology treatments in a systemic immune-associated have been found to be associated with an enhancement in
manner or a local therapy-focused manner. Specifically, multi- the immune response. Furthermore, RT at a low dose has been
ple effects induced by RT, such as inducing in situ cancer demonstrated to help in normalizing the tumor vasculature,
vaccinations for tumor eradication and immune memory evoking the systemic immune response, and reprogramming
maintenance,18 overcoming the barrier of the dense tumor the tumor stroma.21 However, high-dose RT is capable of
matrix and normalizing tumor vascular for easy access of ICIs causing severe blood vessel damage, overcoming a large tumor
and CAR T cells to the tumor tissue,19 or activating the cGMP- burden, and converting ‘‘cold’’ tumors to ‘‘hot’’ ones that are
AMP synthase-stimulator of interferon gene (cGAS-STING) path- much more sensitive to immunotherapy.22,23 For example,
way for the recruitment of pro-inflammatory chemokines,20 brachytherapy at a high dose of 10 Gy was found to convert
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Fig. 1 Scheme for nanomedicine-assisted cancer radio-immunotherapy. Optimized strategies in designing nanomedicines include surface modification
for eliciting immune response, structure modification for RT-responsive drug release, as well as incorporating chemical moieties for prolonging
circulation, enhancing cell uptake, and promoting excretion after the treatment. Nanomedicines have played a critical role in cancer radio-
immunotherapy: (i) pre-stratification at a stimulation dose for selecting patients for radio-immunotherapy or therapeutic response assessment for
adjusting treatment plans. (ii) Improving cancer vaccination through (a) nanomedicine-aided capture and presentation of RT-induced released antigens;
(b) RT-treated tumor cell membranes as a component of personalized cancer nanovaccines; (c) RT-induced priming of the tumor microenvironment
(TME) for exogenous cancer nanovaccines. (iii) Prompting immunogenic cell death through imposing endoplasmic reticulum stress and releasing
immunological regulating agents. (iv) Mitigating therapy resistance through addressing biological or pathological barriers via active targeting and
extracellular matrix/vascular normalization, as well as reversing immunosuppression and radioresistance. (v) Reducing radio-immunotherapy-induced
toxicities through accelerating blood clearance of antibodies, pre-protecting immunocytokines, and addressing radiation proctopathy.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

80% of ‘‘cold’’ prostate cancer cells into ‘‘intermediate’’ or therapies, particularly the integration of renascent immu-
‘‘hot’’ immune subtypes in 24 prostate cancer patients.24 Mean- notherapy with radiotherapy, have great potential to replace
while, several studies have implied that immunotherapy can mono-immunotherapy and become the mainstream workforce
aid in prompting the ‘‘abscopal effect’’ induced by RT.25,26 in immuno-oncology-based treatments.43,44
Recent years have witnessed encouraging results from clinical Non-irradiated remote tumor recession on patients who are
trials of radio-immunotherapy; however, cancer radio- subjected to RT is the clinical basis to develop cancer radio-
immunotherapy is hindered by three critical issues. First, the immunotherapy. This phenomenon, termed as the ‘‘abscopal
aggregate antitumor potency is still inadequate to treat a sum of effect’’, is well-known, but it is a low incidence event in clinical
indications, for example, dormant lesions in cancer patients and practice, mainly due to insufficient antitumor immune
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

immunosuppressive tumor types.27 Second, innate or adaptive response elicited by RT and an immunosuppressive TME.45 In
resistance to radio- and/or immuno-therapy, as well as these a recent clinical trial, about 29% of 168 patients with advanced
therapies-related adverse effects, has been reported.28,29 Third, tumors receiving anti-PD-1 and RT showed the ‘‘abscopal
there is a lack of valid criteria to help in guiding and assessing effect’’.46 The underlying mechanism of this effect is complex
this radio-immunotherapy. For example, imaging manifestations, and remains to be understood. Immunomodulatory signals or
like hyperprogression and pseudoprogression, could display a false agents including DNA damage-induced inflammatory signals,
signal, leading to a significant delay in the treatment plan.30 released tumor neoantigens, and dendritic cell maturation
To address the above issues, nanomedicines have garnered stimulators have been found to contribute to the effect,47,48
increasing interest for cancer radio-immunotherapy due to their but the effect was not seen in immunodeficient athymic mice
improved pharmacokinetics and incredible auxiliary functions that received RT.49 Thus, this effect confirms the immunomo-
(Fig. 1). Unique physicochemical properties and functional mod- dulatory properties of RT and multiple biological processes are
ifications endow these nanomedicines with the ability to overcome involved in the effect.
the obstacles encountered by conventional chemo- or immuno-
therapeutic drugs, such as a low therapeutic drug concentration in
the region of interest, non-specific distribution in healthy organs or 2.1 Biological synergistic basis of cancer radio-
tissues, and a rapid excretion rate.31–34 Effectiveness and a low-toxic immunotherapy
profile of liposome- or albumin-based nanomedicines have been The primary biological synergistic basis of cancer radio-
demonstrated in routine cancer treatment.35–37 A myriad of nano- immunotherapy is shown in Fig. 2, which includes the immu-
medicines have been explored for radio-immunotherapy. Basically, nomodulating effect of RT and the radio-sensitization effect of
they are designed to promote the interaction between RT and immunotherapy.
antitumor immunity. For example, in one aspect, nano- (i) RT induces ICD. RT directly triggers the release of
radiosensitizers augment local radiation deposition and improve tumor antigens and damage-associated molecular patterns
radioresistance, resulting in increased ICD and in situ (DAMPs) including calreticulin (CRT), adenosine 5 0 -triphos-
vaccination.38,39 In other aspects, nanocarriers with/without intrin- phate (ATP), and high mobility group protein B1 (HMGB1).50
sic bioactive properties have been applied to deliver therapeutic/ It indirectly increases the level of reactive oxygen species (ROS)
diagnostic radionuclides, tumor antigens, ICIs, and immunomo- and endoplasmic reticulum (ER) stress, leading to ICD.17
dulators to provoke strong, durable antitumor immunity, address (ii) RT induces vaccination. As a means of deeply-
immunosuppression, and realize imaging-aided therapy.40,41 penetrated, damage-generating stimulation, RT significantly
Besides, incorporation of nanomedicines into adoptive cells or enriches the variety and amount of tumor antigens including
CAR T cells to deliver ICIs represents a clinical translational path tumor-associated antigens (TAAs) and tumor-specific antigens
for radio-immunotherapy.42 Encouragingly, nanomedicines have (TSAs) and subsequently induces their release, thereby improv-
been applied in routine clinical practices and more in clinical trials ing tumoral antigenicity and promoting in situ vaccination.
or pre-clinical studies for radio-, immuno-, or combinational radio- Upregulated expression and enhanced presentation of TAAs
immuno-therapy. after RT result in potent cancer vaccination, and TSAs origi-
Overall, this review provides a comprehensive coverage of nated from RT-induced or somatic nonsynonymous mutations
nanomedicine-assisted cancer radio-immunotherapy, includ- may not easily develop immune tolerance and can be further
ing design strategies and synergetic antitumor mechanisms utilized to develop neoantigen-based peptide cancer vaccines.51
of nanomedicines for radio-immunotherapy. Their recent Specifically, four gene mutations (Adgrf5, Cand1, Dhx58, and
applications in solid tumors and considerations over their Raet1e) upregulated by RT (8 Gy  3 fractions) and their
clinical translation have also been elaborated. encoded immunogenic neoepitopes in 4T1 cells were recently
identified.52 In addition, it was reported that immunogenic
mutation in a gene, KPNA2, upon radiation-induced exposure
2. Cancer immunotherapy embraces occurred in a lung cancer patient after surgical removal of two
radiotherapy brain metastases.53 RT has also been found to increase the
amount of major histocompatibility complex I in the tumor cell
After years of rapid expansion of cancer immunotherapy, membrane, facilitating recognition of effector T cells towards
accumulative evidence indicates that the combinational tumor neoantigens.54 Moreover, RT-induced tumor debris

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article


Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Fig. 2 Illustration of the biological basis of cancer radio-immunotherapy. (a) RT elicits immunogenic cell death (ICD). RT increases the reactive oxygen
species (ROS) level and endoplasmic reticulum (ER) stress, as well as releases damage-associated molecular patterns to activate dendritic cells (DCs). The
release of various chemokines from DCs helps induction and recruitment of immune cells. (b) RT induces cancer vaccination. RT helps releasing tumor
antigens and upregulating major histocompatibility complex I (MHC-I), contributing to enhancing the antitumor effect of the antigen-specific
cytotoxicity T cells. (c) RT activates the cGAS-STING pathway to generate type I interferons, which can enhance the antitumor immunity and increase
the PD-L1 expression that are favorable for anti-PD-L1-based immunotherapy. (d) RT primes the tumor microenvironment (TME), allowing easy access of
immunotherapeutic agents (ICIs, CAR T, or bsAbs) to tumor sites and establishing favorable microenvironmental conditions for immunotherapy. ECM,
extracellular matrix; CAF, cancer-associated fibroblast; TAM, tumor-associated macrophage.

releases DAMPs and proinflammatory cytokines, resulting in tumor burden, loosening the dense tumor matrix, normalizing
the activation of antigen-presenting cells (APCs).55 tumor vascular structures, and inducing an inflammatory TME.
(iii) RT activates the cGAS-STING signaling axis. RT can This TME priming effect allows effective infiltration of high-
induce breakage of double-stranded DNA in tumor cells. The molecule-weight ICIs, large-size artificially modified cells, and
broken DNA is sensed by cyclic GMP-AMP synthase (cGAS), effector T cells, contributing to an immunotherapy-friendly
resulting in the activation of the stimulator of interferon gene immune phenotype in the TME.59,60 Specifically, RT helps
(STING), an innate immune sensor that can induce the genera- increasing pattern recognition receptors and immune check-
tion of type I interferons and pro-inflammatory factors to points on tumor cells and thereby improves their sensitivity to
activate DCs.56 These DCs acquire the expression of the C–C immunotherapy.61 Furthermore, markedly increased infiltra-
chemokine receptor 7 and they migrate to the tumor-draining tion of eosinophils after RT treatment has been recently iden-
lymph nodes for tumor neoantigen presentation.26,57 Conver- tified to improve the recruitment and anti-tumor response of
sely, another report revealed that STING after RT exerted an CD8+ T cells.62 To note, it has been unveiled that RT induces
immunosuppressive effect by inducing an influx of CCR2+M- pyroptosis, leading to an enhanced antitumor immunity in a
MDSCs.58 Caspase 9/Caspase 3/GSAME-mediated manner.63 Another
(iv) RT mediates priming of the TME as a neoadjuvant to recent study has indicated that pre-existing inflammatory T
immunotherapy. RT has been found to help in reducing the cells that survived after RT treatment could mediate the

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

Table 1 Selected clinical trials for the combinational cancer radio-immunotherapy

Combinational Trial registration Enroll-


strategies no. Phase Status Indications ment Arms description Treatment outcome
RT + immune checkpoint inhibitor (ICI)
SBRT + durvalumab NCT02311361 I/II Completed Unresectable 65a Four cohorts: durvalumab +Acceptable safety profile;
(aPD-L1) or tremeli- (REF72) pancreatic 8 Gy  1 f (A1) or 5 Gy  modest treatment benefits:
mumab (aCTLA-4) cancer 5 f (A2), durvalumab + tre-
median PFS (1.7 vs. 2.5 vs.
melimumab + 8 Gy  1 f (B1)
0.9 vs. 2.3 months) and OS
or 5 Gy  5 f (B2) (3.3 vs. 9 vs. 2.1 vs. 4.2
months) in the cohorts A1,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

A2, B1, and B2, respectively


SBRT + pem- NCT02492568 II Completed Non-small 92a Two arms: pembrolizumab Similar toxicity profile;
brolizumab (aPD-1) (REF73) cell lung can- (A); SBRT (8 Gy  3 f) + clinical benefits: ORR (18%
cer (NSCLC) pembrolizumab (B) vs. 36%, p = 0.07), median
PFS (1.9 vs. 6.6 months, p =
0.19), and median OS (7.6
vs. 15.9 months, p = 0.16)
in the arm A and B,
respectively
EBRT + vaginal bra- NCT04214067 III Recruiting Stage I–II 168b Two arms: brachytherapy + Not available
chytherapy + endometrial EBRT; EBRT + brachyther-
pembrolizumab cancer apy + pembrolizumab
RT + chimeric antigen receptor T (CAR T) cells
Yttrium-90 micro- NCT02416466 I Completed Liver 8a One arm: anti-CEA CAR T No grade 4/5 toxicity
spheres + anti-CEA (REF74) metastases (three hepatic artery infu- events; clinical benefits:
CAR T sions) + Yttrium-90 median OS (6.9 months)
microspheres
RT + CAR T NCT04790747 I/II Recruiting Hematologi- 50b One arm: sequential RT + Not available
cal intravenous infusion of
malignancies CAR T
RT + bispecific antibody (bsAb)
131
I + omburtamab NCT01099644 I Active, not Peritoneal 54a One arm: intraperitoneal No dose-dependent toxi-
(REF75) recruiting cancer injection of 131I-8H9 cities; transient adverse
(omburtamab) effect; phase II activity was
established at 2.96 GBq
m2
Anti-CEA  anti- NCT01221675 I/II Completed CEA- 18a Two arms: optimization Absorbed doses predicted
HSG TF2 bsMAb + (REF76) expressing study and escalating activ- from pre-therapeutic ima-
IMP-288-Luteium + SCLC or ity phase I/II study ging session for therapy
IMP-288-Indium NSCLC session; a shorter pre-
targeting delay (24 h) and
the highest TF2 molar dose
were the best parameters
RT + cancer vaccine
EBRT + sipuleucel-T NCT01807065 II Completed Castrate- 51a Two arms: sipuleucel-T (A); Both arms were well-
(REF77) resistant EBRT + sequential tolerated; median PFS (2.46
prostate sipuleucel-T (B). vs. 3.65 months, p = 0.06)
cancer in the Arm A and B
Radium-233 + NCT02463799 II Completed Prostate 36a Two arms: radium-233 + No synergistic toxicity;
sipuleucel-T (REF78) cancer sipuleucel T (A); sipuleucel- median PFS (10.7 vs. 3.1
T (B). months, p = 0.02), PSA
response (33% vs. 0%, p =
0.04), and AlkPhos
response (60% vs. 7%, p =
0.01) in the arm A and B,
respectively
153
Sm-EDTMP + NCT00450619 II Completed Prostate 44a Two arms: 153Sm-EDTMP Both arms have similar
vaccine (REF79) cancer radiation (A); 153Sm- toxicity; median PFS (1.7
EDTMP + recombinant vs. 3.7 months, p = 0.034)
fowlpox- and vaccina- and a 430% PSA decline (0
TRICOM vaccine + sar- vs. 19%, p = 0.073) in the
gramostim (B). arm A and B, respectively
RT + autologous dendritic cells (DCs)
EBRT + autologous NCT01347034 II Completed Soft tissue 20a Two arms: EBRT alone; One in fourteen had ser-
DCs sarcoma EBRT + autologous DCs ious adverse events in the
(i.t.). treatment group with EBRT
+ DC injection; therapeutic
outcome is not given.
RT + DC immuniza- NCT03548571 II/III Recruiting Glioblastoma 60b Two arms: DC immuniza- Not available
tion + tion (i.d.) + subsequent RT
temozolomide (2 Gy  30 f) and temozo-
lomide; RT (2 Gy  30 f)
and temozolomide

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

Table 1 (continued)

Combinational Trial registration Enroll-


strategies no. Phase Status Indications ment Arms description Treatment outcome
RT + costimulatory receptor agonist
RT + GLA-SE (TLR4 NCT02180698 I Completed Metastatic 16a One arm: glucopyranosyl Grade 1 or 2 toxicity; local
agonist) (REF80) sarcoma lipid A-stable-emulsion tumor control (14/14), and
(GLA-SE) + RT 1 CR, 1 PR, and 11 SD on
156 days post-trial
RT + imiquimod NCT01421017 I/II Completed Breast cancer 31a Three arms: RT + imiqui- All but one adverse event
(TLR7 agonist) (REF81) with chest mod; RT + cyclopho- was grade 1 or 2; abscopal
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

wall recur- sphamide; RT + response was generated in


rence or skin cyclophosphamide + 3/9 cases
metastases imiquimod
RT + interferon/immunocytokine
SBRT + L19-IL2 NCT02086721 I Completed Oligometa- 6a One arm: SBRT + sub- Recommended dose in
(REF82) static solid sequent L19-IL2 (i.v.) phase II is 15 million
tumors international Units
SABR + Darleukin NCT03705403 II Recruiting Stage IV 126b Two arms: standard of care Not available
(L19-IL2) (REF83,84) NSCLC SABR and/or RT; SABR and/
or RT + L19-IL2
Abbreviations: SBRT, stereotactic body radiation therapy; PFS, progression free survival; OS, overall survival; ORR, overall response rate; EBRT,
external beam radiation therapy; CEA, carcinoembryonic antigen; SCLC, small cell lung cancer; HSG, histamine–succinyl–glycine; 153Sm-EDTMP,
samarium 153 lexidronam pentasodium; TLR, toll-like receptor; CR, complete response; PR, partial response; SD, stable disease; SABR, stereotactic
ablative body radiotherapy. a Represents the actual participants. b Indicates the estimated number (All data but therapeutic outcome were
accessed from ClinicalTrials.gov).

activation of local antitumor immune response and exert a 2.2 Current landscape of cancer radio-immunotherapy
highly potent therapeutic effect in comparison with freshly Recent years have witnessed the successful development of
infiltrating T cells.64 Moreover, it was reported that in an cancer radio-immunotherapy. Hundreds of clinical trials on
orthotopic glioblastoma mice model, long-term intravital this topic have been registered, and their recent progress has
microscopy fluorescence images confirmed that RT aided in been summarized elsewhere.70,71 Various RT methods, such as
rapid extravasation of CAR T cells from vasculature and their
external beam radiation therapy or brachytherapy with hypo- or
expansion in the TME.65
hyper-fractionation at a high- or low-radiation dose using
(v) Immunotherapy curbs non-irradiated distant tumors.
different radiation sources, have been combined with different
One significant unique advantage of radio-immunotherapy is
immuno-oncology treatments including ICIs, CAR T, bsAbs, or
that two individual therapies can complement in controlling
cytokines via different modes of administration (i.v., s.c., i.p.,
primary tumors and distant metastases. Immunotherapy can
and i.d.) sequentially or concurrently. The safety and efficacy of
be an excellent addition to RT in advanced cancers since multi-
these combination treatments have been evaluated, and their
site radiation cannot completely cover distant tumors in
indications include but are not limited to non-small cell lung
advanced cancers.66
cancer, glioblastoma, pancreatic cancer, soft tissue sarcoma,
(vi) Immunotherapy induces TME priming as a neoadju-
and prostate cancer. Herein, a few representative clinical trials
vant to RT. Sufficient intratumoral infiltration of immune
are listed in Table 1.
effector cells or reduction of immunosuppressive cells induced
Beyond these selected clinical trials, analysis results of two
by immunotherapy contributes to a supportive TME for sub-
large-datasets provide a current summary of the therapeutic
sequent RT treatment. For example, in a study conducted on
outcomes in the combinational treatment of immunotherapy
two orthotopic head and neck cancer murine models, a murine
MOC2-bearing C57BL/6 model and a murine LY2-bearing and RT. An analysis of the National Cancer Database reveals
BALB/c model, targeting regulatory T cells by inhibiting STAT3 that the addition of immunotherapy to RT contributes to an
was found to improve the therapeutic efficacy of RT.67 improved overall survival time (OS) in melanoma brain metas-
Additionally, a recent study based on murine tumor models tases according to the data from 1104 patients. 192 patients
and the cancer patient samples after treatment with RT and received both RT and immunotherapy with a median OS of 11.1
anti-PD-L1 immunotherapy revealed that both treatments months, while the rest were treated with RT alone with a
shared a similar adaptive immune response by eliminating median OS of 6.2 months.85 However, a retrospective analysis
tumor-promoting erythroid progenitor cells (Ter cells), which of patients with metastatic NSCLC, among which 6383 received
secreted artemin to promote tumor growth.68 In summary, RT plus immunotherapy and 170 479 received hypofractionated
their synergistic effects on alleviating early-stage/late-stage RT alone, reveals no difference in the overall survival time
small/large tumor burdens, curbing local foci/distant metas- between two groups.86 Thus, this combined therapy displayed
tases, activating the antitumor immunity at dormant tumor encouraging therapeutic outcomes for a few cancer types, but it
sites, and preventing tumor recurrence can be harnessed for is not effective against other indications. Additionally, several
combating advanced cancers.69 issues, including ambiguity between pseudoprogression and

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

tumor progression, the off-target effect of immune-adjuvants four categories: immunomodulators, radiosensitizers, regula-
and radio-enhancers, and therapeutic resistance, contribute tors for improving both therapies, and probes for diagnosis,
to under-expected therapeutic efficacies and severe systemic therapeutic response evaluation, or imaging-assisted treat-
toxicities. ments. In this combinational treatment, nanomedicines may
play multiple roles through incorporation of different func-
tional groups (Fig. 3).
3. Nanomedicines aid in cancer radio- Nanomedicine-enabled immunomodulation. The nano-
medicine-mediated immunomodulation effect has been sum-
immunotherapy marized by Nam et al. and Irvine et al.87,88 The effect is
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

3.1 Basic mechanisms predominantly related to single or multiple phases of the


Overall, application of nanomedicines improves the safety and cancer immunity cycle, such as improving antigen presenta-
efficacy of cancer radio-immunotherapy. Potential roles of tion, restoring T cell function, and promoting cancer
nanomedicines in this combined therapy can be divided into vaccination.87,88

Fig. 3 Illustration of four major roles of nanomedicine in cancer radio-immunotherapy. (a) Nanomedicine-mediated immunomodulation. The role of
nanomedicines in improving immunotherapy includes: (i) acting as exogenous nanovaccines and antigen-capturing NPs; (ii) delivery of small-molecule
immunomodulatory agents; (iii) delivery of ICIs & cytokines; (iv) nano-modifiers for immune cells. (b) Nanomedicine-aided radiation sensitization through
increasing radiation-dose deposition and ROS generation, improving tumor oxygenation and exacerbating DNA damage, and decreasing GSH generation
and preventing DNA repair. (c) Nanomedicine-contributed assistance in improving both radiotherapy and immunotherapy through interfering with their
shared signaling pathways (e.g., cGAS-STING) and reducing therapy resistance. (d) Nanomedicine-participated imaging visualizes the tumoral
accumulation of nanomedicines and semi-quantifies specific biomarkers for radio-immunotherapy in pre-selecting patients for this combinational
treatment or assessing therapeutic response.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

In cancer radio-immunotherapy, there are four main speci- mitochondria- or endoplasmic reticulum-targeting nanomedi-
fic immunomodulatory roles of nanomedicines. 1. They act as cines could contribute to highly-selective damage to tumor
cancer nanovaccines or antigen-capturing NPs. Briefly, exo- cells;112–114 ii. Delivering tumor hypoxic radiosensitizing
somes or irradiated cell membranes of RT-treated tumor cells reagents. Hypoxia is a major contributor to radio-resistance.
are used as personalized cancer nanovaccines due to the Thus, tumor re-oxygenation strategies, including the use of
abundance of tumor neoantigens and DAMPs in them. The oxygen-saturated nanomaterials (perfluorocarbon),115 oxygen-
use of antigen-capturing NPs can help in adsorbing and pre- generators (MnO2),116 NO-releasing prodrugs,117 HIF-1a
senting released antigens after RT, favoring DC activation inhibitor-containing carriers,118 catalase, catalase-like metals
and enhancing antigen-specific T cell-mediated antitumor or H2O2,119 have been employed to address tumor hypoxia-
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

immune response.89,90 2. They deliver immunomodulatory induced radioresistance. iii. Carrying inhibitors for DNA repair
agents in the manner of physical encapsulation or chemical and radioresistance signaling. Inhibitors for vital proteins
conjugation.91–93 Agonists of STING and TLR, as well as inhi- (mTOR) and signaling pathways (hedgehog signaling
bitors of indoleamine-2,3-dioxygenase (IDO), have been incor- or CD73/adenosine) have been incorporated into nanomedi-
porated into the nanoscale delivery system, contributing to cines to indirectly increase ROS deposition and augment the
evading immune escape or potentiating ICI-based antitumor microenvironmental stress.120,121 Inhibitors for DNA repair
immune response for RT.94,95 3. They deliver ICIs and cyto- (olaparib),122 enhancers for RT-induced apoptosis
kines. Inefficient delivery and unpleasant systemic toxicity of (perifosine),123 and disrupters for the cell cycle (proflavine
immuno-drugs hamper their clinical application, while nano- hemisulfate, gemcitabine, and pentoxifylline) and the NAD+
technology may help in addressing these issues.96–98 For metabolism are the family members of radiosensitizers,124,125
instance, the result of a phase II clinical trial (NCT00396019) as they directly or indirectly improve the therapeutic
with 86 participants indicates the administration of PEGylated outcome of RT.
interferon Alfa-2b (PEG-Intron) to patients with plexiform neu- Nanomedicine-contributed assistance in radio-immuno-
rofibromas could significantly delay the onset of tumor pro- therapy. Nanomedicines aid in improving the synergistic inter-
gression in comparison to the placebo group.99 Additionally, action of radiotherapy and immunotherapy. Two distinctive
advanced nanotechnology-aided immunomodulatory nanome- approaches have been explored: one is to interfere with their
dicines have been developed, such as PD-L1 antagonist peptide- synergistic or shared signaling pathways, and the other is to
decorated polymeric nanoparticles,100 a dendrimer–ICI address radioresistance or immunotherapy resistance.
conjugate,101,102 and a cytokine or an antibody modified with In the first approach, the STING pathway is one of the most
an inner stimuli-responsive mask agent.103,104 4. They can be studied pathways. Generally, nanomedicines can be used to
used to modify immune cells. Nanomedicines, mostly in the increase RT-induced ruptured micronuclei with cytosolic DNA,
form of nanogels, can be modified to attach to effector T cells as well as deliver exogenous STING agonists, which augment
or macrophages via a backpacking approach to simultaneously the STING activation for a robust antitumor immune response.
realize multiple functions, such as controlled release of IL15 to For instance, cGAMP/MOL, comprising a STING agonist cGAMP
selectively expand tumor-infiltrated T cells or IFN-g to maintain and an Hf12-Ir metal organic layer, elicited robust STING
proinflammatory M1 macrophages.105,106 activation with low-dose radiation. Significant response from
Nanomedicine-aided radiation sensitization. One signifi- interferon regulatory factors and excretion of STING-IFN axis-
cant advantage of nanomedicine-aided radiation sensitization related inflammatory cytokines (IFN-b and IL-6) were observed
is the ability to maximize the immunogenic or therapeutic after treatment with the nano-radiosensitizer compared to the
effect of RT, but reduce the radiation dose to a safe level.107 one with cGAMP alone.126 In another study, an exogenous
There are three major means of realizing nanomedicine- STING agonist, c-di-AMP, was integrated into a Mn2+ chelated
aided radiation sensitization: (i) amplifying local radiation tannic acid-based nanoplatform to treat large tumors. The level
deposition and ROS generation by using high-Z nanomedi- of the second messenger of STING, cGAMP, was significantly
cines. Metal elements and their nano-derivatives with a high increased by 2-fold in the 4T1 tumor tissue in this nano-
atomic number and efficient nano-catalytic properties, particu- combinational treatment group in comparison with that in
larly in the form of metal–organic frameworks, including the RT-treated one on day 12 post-treatment.127
hafnium oxide, gadolinium, gold, bismuth, platinum, polyox- Strategies for overcoming physiological barriers during
otungstate, titanium oxide, and tantalum, have been explored nano-medicinal delivery and addressing hypoxia, a main
for this radiation sensitization effect.38,108–110 For example, factor conducive to the resistance to radio-immunotherapy,
hafnium oxide-containing nanoparticles (NBTXR3) have been have been employed to improve the therapeutic efficacy of
evaluated in 15 clinical trials and they have shown a promising radio-immunotherapy. Briefly, modification with active target-
therapeutic effect. The working principle of this nanoparticle is ing moieties, vascular normalization, and ECM normalization
dose partitioning. An increased radiation dose is deposited are three common nanomedicine-assisted TME-modulating
close to this high-Z radiosensitizer, resulting in improved methods. Tumor re-oxygenation to address hypoxia is
photoelectric interaction.111 In addition, the high-Z radiosensi- achieved via exogenous delivery of oxygen-generating
tizer can contribute to non-oxygen-dependent ROS generation agents or endogenous generation promoted by delivered
by catalyzing a great amount of H2O2 in the TME. Furthermore, reagents.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

Nanomedicine-assisted imaging. Nanomedicine-assisted type, and irradiated filed), immunotherapy (interval before or
bioimaging could play a critical role in cancer radio- after RT, type of administration, and immuno-oncology drugs),
immunotherapy. Its real-time in situ visual observations could and nanomedicines (functional role, formulation type, admin-
help in formulating or adjusting treatment plans, longitudin- istration route, and delivery sequence). To achieve optimal
ally monitoring tumor progression, and acting as a predictor to nanomedicine-assisted radio-immunotherapies, we will pre-
assess therapeutic response or a participator in multiple sent the current landscape and discuss up-to-date nanoformu-
imaging-guided precision radiation platforms.128 lations and concepts in this emerging field.
In contrast to current clinical routine imaging modalities,
nanomedicine-assisted imaging is devoted to improving the
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Current landscape. Formulations of nanomedicines and their


sensitivity and specificity of imaging signal of tumors under-
potential role in the current practice of this combined therapy
going radio-immunotherapy. Internal/external radiation expo-
are selected and shown in Table 2. The roles of these nanome-
sure may alter the immune phenotype of tumors and specific
dicines are very different depending on their formulations and
biomarkers on the cell surface (such as PD-1, PD-L1, and
indications. They can be a nanocarrier for an exogenous anti-
CTLA-4) or the related cells (cancer-associated fibroblast and
gen (OVA) or a radiosensitizer to improve radiosensitivity and
tumor-associated macrophages) may have thereafter been
overcome radiotherapy resistance, a scaffold for immunomo-
changed.129,130 This dynamic process can be monitored with
dulator drugs, or a container for in situ generated neoantigens.
the help of engineered site-specific probes, which consist of
As shown in Table 2, these nanomedicines not only offer
corresponding targeting moieties and imaging moieties. Multi-
therapeutic benefits, but also act as an adjuvant medium to
functional probes labelled with different signal sources could
enhance radio-immunotherapy.
be constructed thanks to advances in nanotechnology.131 They
can be designed to reach multiple targets or deliver multiple
imaging agents to obtain a comprehensive coverage of the Advances in nanoformulations for nanomedicines. A library of
disease status. A dual positron emission tomography (PET)/ cancer nanomedicines for immunotherapy or radiotherapy has
near-infrared fluorescent probe, which was prepared from 89Zr, been extensively summarized elsewhere.147,148 This review con-
near-infrared fluorophore (CF-MPTMS)-attached silica nano- centrates on advances in nanoformulations for nanomedicines
particles with protamine on the surface and a heparin coating that promote radio-immunotherapy.
layer, was used to in vitro label CAR T cells and in vivo track The incorporation of a multi-functional nanocarrier into the
these cells in mice models bearing ovarian peritoneal nanomedicine for cancer radio-immunotherapy has led to the
carcinomatosis.132 Many ex vivo cell labelling methods, such maximum therapeutic effect with reduced toxicity to a large
as radionuclide-labelling via copper-free click chemistry,133 and extent. Four major types of nanocarriers have been explored for
superparamagnetic iron oxide labelling,134 have been estab- these nanoformulations: organic, inorganic, organic–inorganic
lished to visualize immune effector T or NK cells during the hybrid, and biomimetic nanocarriers (Fig. 4). The classification
treatment process.135 is established on the basis of the body structure of the nano-
Unfortunately, there is no well-defined, accurate biomarker formulation. To note, biomimetic nanoformulations refer to
yet for cancer radio-immunotherapy. A few critical indicators, those biological products originating from mammalian cells or
such as CD8,136 granzyme B,137 and lymphocyte activation bacteria, while natural or synthetic proteins are considered as
gene-3,138 show positive correlation between their expression organic nanoformulations.
and the therapeutic outcome. Additionally, the use of imaging Liposomes, polymers, and albumin are the most widely used
probes for multiple biomarkers could be a promising solution organic nanocarriers in clinical drug delivery practice due to
for combination therapy. Challenges remain for interpretation their non-immunogenic or low-immunogenic properties
of these images and correlation of these imaging signals with and great biocompatibility.149 In the preclinical studies, den-
the exact therapeutic response to this combination treatment. drimers and natural polysaccharides have recently gained
Support of evidence-based medicines and the use of artificial popularity compared to other polymers. Specifically, multiva-
intelligence in medical imaging analysis could help in addres- lent binding properties of dendrimers can be harnessed to
sing these challenges.139 increase the binding kinetics of ICIs to their corresponding cell
surface receptors.150 In a recent study, hyperbranched G7
3.2 Design strategies of nanomedicines for radio- poly(amidoamide) dendrimers were conjugated with aPD-L1,
immunotherapy with an estimate of 3.7  0.5 antibodies per one dendrimer
Elegant and rational design of nanomedicines is a critical step and a one-order-of-magnitude increase in the binding kinetics
for their application in radio-immunotherapy. Clinical require- with PD-L1 in comparison with free aPD-L1.101 Natural poly-
ments, preparation, and delivery routes for these nanomedi- saccharides, particularly hyaluronic acid, stand out for
cines should be considered. their immunomodulatory effects.97,98 Notably, several hydro-
3.2.1 Nanoformulations for the combined therapy of RT phobic immunological agents or radiosensitizers, such as cyclic
and immunotherapy. Many factors play a role in the application dinucleotides and 2-(2-nitroimidazol-1-yl) acetic acid, can act as
of nanomedicines in this combined therapy, including para- the hydrophobic domain in self-assembled polymeric
meters associated with RT (duration, dose, fraction, radiation conjugates.151,152

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

Table 2 Selected nanomedicines for combinational radio-immunotherapy

Ref. Nanoformulation RT IT Comments


Preclinical studies
Organic
Zhang et al.140 Smac-TLR7/8 peptide g-ray EBRT TLR7/8 agonist This peptide self-assembled into a nanofibrous and porous
hydrogel, overcoming the radioresistant TME and repolariz-
ing TAMs
Li et al.141 AmpFY9 peptide g-ray EBRT IDO-1 inhibitor This pH-responsive transformable peptide aided in reversing
immunosuppression via the GSH-responsive release of
NLG919
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Inorganic
Wang et al.95 4PI-Zn@CaCO3 X-ray EBRT IDO-1 inhibitor Neutralizing tumor acidity via biocompatible biomineral
CaCO3 and depleting Kyn via 4PI potentiated RT against CT26
and 4T1 tumors by inducing potent antitumor immune
response
Dong et al.142 WO2.9-WSe2-PEG NPs X-ray EBRT aPD-L1 Semiconductor-based nanoscale radiosensitizers combined
with aPD-L1 in a triple-therapy manner to treat local and
distal tumors
Organic–inorganic hybrid
Pei et al.143 177
Lu-APPs-PEG RIT RT-induced The radioactive nano-oxygen generator relieved tumor
immune hypoxia, promoted CTL infiltration, and inhibited tumor cells
response and Tregs
Sang et al.144 AHSC nanoparticles X-ray EBRT aCTLA-4; aCTLA- NP-reinforced RT and aCTLA-4 increased PD-L1 expression,
4 + aPD-L1 promoting anti-PD-L1 therapy
Biomimetic
Tian et al.145 PD-L1 siRNA loaded RGD- X-ray EBRT PD-L1 siRNA An in situ EV-mediated ICI therapy was applied to glio-
EV blastoma with short-burst radiation
Qin et al.146 Au@MC38 X-ray EBRT aPD-1 Live MC38 cell-derived biogenetic AuNPs were formed; these
Au@MC38 NPs displayed homologous targeting of tumor
cells, thus enhancing RT and immune response
Clinical trials
Liposome
NCT04580771 Liposomal HPV-16 E6/E7 EBRT Vaccine This phase II trial aimed to improve the therapeutic response
Multipeptide Vaccine against HPV-infected cervical tumors by using a liposomal
vaccine (six HPV-16 peptide-containing lipid R-DOTAP)
NCT00828009 BLP25 liposome vaccine EBRT Vaccine This phase II trial aimed to treat lung cancer with a liposomal
vaccine (lyophilization of BLP25 lipopeptide, monopho-
sphoryl lipid A, and three lipids: cholesterol, DMPG, and
DPPC) and bevacizumab after EBRT
PEG
NCT04936841 NKTR-214 (PEGylated IL-2) EBRT Immuno- This phase II trial aimed to deliver PEGylated IL-2 and aPD-1
cytokine and in combination with RT to treat head and neck cancer
aPD-1
Protein
NCT04756505 Bintrafusp Alfa EBRT Fused protein A fused protein composed of aPD-L1 and TGF-beta, an
and NHS-IL12 immunocytokine, and the protein was combined with RT to
treat breast cancer in the phase I trial
Inorganic nanoparticles
NCT05039632 Hafnium oxide-containing EBRT aCTLA-4/aPD-1 This phase I/II trial investigated the adverse effects and
nanoparticles (NBTXR3) therapeutic benefits of a combined therapy, consisting of
NBTXR3-augmented RT, aCTLA-4, and aPD-1 for metastatic
solid tumors
Abbreviations: IT, immunotherapy; IDO-1, indoleamine 2,3-dioxygenase-1; APPs, Au-Pt@PCN-224; RIT, internal radioisotope therapy; CTLs,
cytotoxic T lymphocytes; AHSC, atovaquone, hafnium, sabutoclax, and chlorin Ce6-containing metal-phenolic networks; RGD-EV, cyclic RGDyK
peptide-modified extracellular vesicles; HPV, human papillomavirus; R-DOTAP, R-enantiomer of 1,2-dioleoyl-3-trimethylammonium-propane
chloride; DMPG, dimyristoyl phosphatidylglycerol; DPPC, dipalmitoyl phosphatidylcholine.

Protein-derivatives are another essential source of organic monovalent 2Rb17c-R3B23) and mAb (trastuzumab), mono-
nanomedicines,153 which can be divided into protein-based meric nanobodies (2Rb17c) turned out to be the most effective
nanocarriers, protein-based therapeutics (e.g., antibody or anti- one in homogenous intratumoral distribution and rapid renal
genic peptide), TME-modulating enzymes, and multifunctional clearance.156 In the regime of protein-based therapeutics, frag-
hybrid proteins.154 For protein-based nanocarriers, mussel ments or entire antibodies/antigens can be engineered with
adhesive proteins have recently emerged as an efficient nano- nanomedicines. For example, tumor antigens and Fc fragments
carrier for localized stable retention of anti-PD-L1 because were fused to the C-terminal of the protein nanocarrier to form
of their substantial tissue-adhesion properties.155 Besides, a cancer nanovaccine, enabling DC activation and tumor-
their size plays a critical role when they are delivered in a specific targeting.157 In addition, bispecific T-cell engagers
systemic manner. In contrast to dimeric-nanobodies (anti- (BiTEs) were nano-engineered with two single-chain variable
HER2 2Rb17c-2Rb17c, control R3B23-R3B23, and dimeric fragments (scFvs) that targeted T cells and tumor cells,

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev


Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Fig. 4 Representation of four major types of nanoformulations (organic, inorganic, organic–inorganic hybrid, biomimetic nanoformulations) that are
employed in radio-immunotherapy. (a) Organic nanoformulations. (i) Dendrimers aid in carrying immune checkpoint inhibitors or tumor antigens
released from RT-treated tumor tissues; (ii) protein-derivatives, ranging from ICIs, targeting antibodies, albumin, cytokines to adhesive proteins, are
modified with radionuclides or immunomodulatory agents via a cleavable linker to simultaneously improve the therapeutic efficacy and address their
toxicity issues; (iii) liposomes act as a conventional tool to deliver various immune-stimulation agents; (iv) polymeric nanoparticles carrying small-
molecule radiosensitizers are surface modified with ligands or therapeutic proteins via a DNA scaffold and other linkers. (b) Inorganic nanoformulations.
(i) Two representative radiosensitizers in clinical trials: hafnium oxide NPs and gadolinium-chelated polysiloxane-based NPs, (ii) various inorganic
nanoagents acting as immuno-adjuvants or carriers. (c) Organic–inorganic hybrid nanoformulations. Nanoscale coordination polymers or metal–organic
frameworks are representative structures. In these nanoformulations, metal ions can amplify local radiation and transfer energy to organic ligands upon
radiation, exacerbating ROS production. Meanwhile, immunomodulatory agents can bind to these formulations via noncovalent encapsulation or
covalent binding. (d) Biomimetic nanoformulations. Membrane-vesicles extracted from various mammalian cells or bacteria, exosomes, and fused
membranes take advantage of the unique properties of these individual cells (e.g., stealth, tumor-homing, or immune-stimulation) to realize desired
tumor control in the context of radio-immunotherapy.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

respectively.158,159 Similarly, synthetic nanoparticle antibodies internal stimulation, metal oxide-based nanoparticles can neu-
were prepared from a Janus nanoplatform with a cell-targeting tralize tumor acidity or catalytically decompose over-expressing
ligand on one ‘‘face’’ and a Fc-mimicking ligand on the H2O2 to relieve the immunosuppression in the TME (e.g.,
opposite ‘‘face’’.160 Both BiTEs and synthetic nanoparticle CaCO3 or MnO2) for enhancing the RT efficacy,95,173,174 or
antibodies were engineered onto the surface of the nanomedi- generating adequate ROS to induce in situ vaccination (e.g.,
cine with multivalent contact. In addition, a short-chain syn- HfO2).175 More importantly, with the rapid progress of biode-
thetic DNA scaffold was demonstrated as a versatile tool to gradable strategies in designing inorganic nanomedicines,
optimize the surface of an organic nanocarrier or a cell to their toxicity issues related to long-term retention and tough
strengthen the effect of their immunomodulation on immune clearance can be elegantly addressed.176
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

cells.161 In a previous study, via complementary DNA reaction, Metal–organic frameworks (MOFs), consisting of high Z
different therapeutic protein molecules (anti-CD3 and anti- metal ions/clusters and coordinated organic molecules, are a
CD28, IL-2, or synthetic priming antigens) were ratiometrically typical organic–inorganic hybrid nanocarrier used in radio-
loaded onto the surface of poly(lactic-co-glycolic acid) (PLGA) immunotherapy. They have enjoyed a blooming growth in this
polymer–DNA nanoparticles. Encouragingly, this biotechnology field due to their homogeneous porous structure, the radio-
realized intact bioactivity of protein molecules, in vivo CAR T sensitization effect, and their therapeutic enhancements in
activation and tumor clearance via an ‘‘AND’’ logic-gate, and vaccination and ICI treatment.177,178 For example, cationic
ex vivo T cell activation and expansion.162 nMOFs were applied to deliver anionic CpGs via electrostatic
Inorganic nanoformulations have been widely used in clin- interactions to perform X-ray-activated vaccination.179 A wide
ical practice or trials, including aluminum salt (aluminum range of metal ions/clusters and coordinated organic ligands in
hydroxide and aluminum phosphate), graphene or silica. They preparing MOFs endow this hybrid nanocarrier with a diversity
act as cancer vaccine adjuvants for antigen reservoir and DC of functions. For instance, the high-Z element Hf or Bi has the
maturation,163 as well as radiosensitizers. NBTXR3 (phase III, radiation amplified effect, metals (Mn, Fe) are able to act as an
status: recruiting) and AGuIX (phase II, status: recruiting) are imaging and immune- or redox-modulating agent, and organic
two examples of inorganic nano-radiosensitizers.39 In preclini- ligands such as Ir(DBB)[dF(CF3)ppy]2+, Ir(bpy)[dF(CF3)ppy]2+,
cal studies, a great sum of inorganic nanomedicines, such as DBA, and DBP (corresponding chemical nomenclature: DBB,
aluminum NPs, CaCO3 NPs, metal oxide NPs (e.g., MnO2), and 4,4 0 -di(4-benzoato)-2,2 0 -bipyridine; dF(CF3)ppy, 2-(2,4-difluo-
mesoporous silica NPs, emerge as potent radiosensitizers, rophenyl)-5-(trifluoromethyl)pyridine; bpy, 2,2 0 -bipyridine;
immuno-adjuvants or carriers. These inorganic nanomedicines DBA, 2,5-di(p-benzoato)aniline; DBP, 5,15-di(p-benzoato)-
exhibit high Z metal- or hypoxia relief-mediated radiation porphyrin) are photosensitizing.180–182 A study has witnessed
sensitization, display inherent or acquired immunomodulatory more than 99% of tumor regression in a MC38 mice model
effects, and possess a high drug-loading capacity.164,165 In the treated with Hf-based nMOFs at a low radiation dosage of
term of radiation sensitization, several high-Z element-based 0.5 Gy  5 fractions. Mechanistically, the secondary building
nanomedicines with efficient nano-catalytic properties, such as units (SBUs) of electron-dense Hf12 and Hf6 absorbed X-rays to
hafnium oxide, gadolinium, gold, bismuth, platinum, and produce hydroxyl radicals. The generated energy was trans-
titanium oxide, have been reported to augment radiation ferred to the photosensitizing ligand Ir(DBB)[dF(CF3)ppy]2+ to
deposition and ROS generation.108–110,166 Besides, these inor- produce 1O2 and O2.183 Monte Carlo simulation results
ganic nanomedicines can be doped or labelled with radio- revealed that the radiosensitization effect of lattices in the
nuclides or self-assembled from radionuclides themselves MOF exceeded solid NPs due to enhanced scattering of photons
(such as b-particle emitter 198Au) as an internal radioisotope and electrons within the lattices regardless of the radiation
therapeutic agent.167 Several radiolabeling methods have there- source and particle size. Thus, tuning the lattice parameters,
after been prompted. A recent study proposed a universal such as the SBU size and/or the inter-SBU distance could
chelator-free radiolabeling method, i.e., the use of a SnCl2/ contribute to an optimal radiation dose.184 Meanwhile, other
HCl solution and Tween 80 for labelling therapeutic 188Re with inorganic–organic hybrid nanocarriers, such as organic
both inorganic (SiO2, Au, and Fe3O4) and organic (PLA) NPs ligands, polymers or biomacromolecule-modified inorganic
with a high labelling efficiency up to 98% and an in vitro nanoformulations could be potential candidates of radio-
radiochemical stability of 95%.168 Their adjuvant roles in immunotherapy.
immunotherapy are generally divided into two types. One is Biomimetic nanocarriers have the reputation for their bio-
adsorption or delivery of antigenic peptides or mRNA for a compatibility, the intrinsic tumor-homing effect, and inherent
sustained effect.169 The other one is to realize immune- immunogenic properties.185–187 A myriad of engineered biomi-
stimulation by their biological active structures. For instance, metic membrane-based vesicles (single membrane-based vesi-
PEGylated mesoporous silica nanoparticles with a tunable pore cles, fused cell membrane-based vesicles, and exosomes),
diameter and a large internal surface have been previously originating from blood cells, platelets, tumor cells, immune
reported to trigger the TLR4/NF-kB pathway in macrophages cells, and bacteria, have been utilized to deliver antigenic
and recruit T cells to inflame cold tumors.170,171 b-alanine- agents or other therapeutic agents to elicit an immune
modified Gd@C82 was also demonstrated to reprogram TAMs response in the practice of cancer vaccination or radio-
to the tumor-killing M1 type.172 Notably, upon external or immunotherapy.188–192 These biomimetic nanocarriers provide

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

a strong shelter to prevent premature leakage and enzymatic Optimization of nanomedicines. Optimization of nanomedi-
lysis of cargos such as nucleic acids or proteins. Among single cines can be obtained by modulating their size, shape, elasti-
membrane-based vesicles, one typical and prevalent applica- city, composition, surface charge, and surface chemistry.213,214
tion is to use tumor cell membranes or bacteria-derived outer In cancer radio-immunotherapy, size-changeability and shape-
membrane vesicles (OMVs) as a biomimetic coating layer to deformability were found to play an important role in deliver-
realize targeted delivery of ICIs or radiosensitizers, as well as ing antitumor vaccines to lymph nodes or immunoregulatory
immune-activation.193,194 Fused cell membrane-based vesicles agents to tumors.215 It was reported that a rod-like shape of
integrate characteristic biochemical receptors and surface func- mesoporous silica nanoparticles outperformed in targeting
tional groups of each individual cells, and thereafter can act as tumor cells, including MCF7 cells and pancreas cancer cells
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

a potent nanocarrier to achieve multiple functions in radio- (PANC-1 cells), rather than healthy cells (MCF10A and CAFs) in
immunotherapy. For example, fusion of the cytomembranes of comparison with the spherical one.216 For tuning the surface
mature DCs and tumor cells achieved co-expression of tumor charge, a charge-reversal modification strategy by harnessing a
antigens and immunological costimulatory molecules.195 Other tumoral acidity-responsive bond that links a positively-charged
hybrid vesicles have also been widely explored, including fusion inner ligand and a neutral or negative anti-fouling outer ligand
of membranes from erythrocytes and tumor cells,196 hybridiza- (e.g., zwitterion polymer or PEG) has been applied to construct
tion of the membranes of macrophages and cancer cells,197 or a nanomedicine with negligible surface protein adsorption and
blending of autologous tumor cell membranes and OMVs.198 high tumor cellular uptake.217 In the case of lymphatic trans-
And recently, liposomes/lipid reagents or polymeric nanostruc- port, a negatively-charged nanocarrier was found to be superior
tures have been introduced to cell membrane-based vesicles to to the positively-charged one.218 Moreover, active surface mod-
improve their stability and address their issues, such as size- or ifications with sufficiently accessible ligands enhance selective
shape-controllability and reproducibility.199,200 Lipid-bilayer targetability of nanomedicines towards tumor cells or tumor-
extracellular vesicles are also employed to enhance cell–cell associated macrophages.219,220 In one study, surface modifica-
communication, strengthen their interaction with non-cellular tion of glucose moieties in generation 4 hydroxyl PAMAM
TME constituents, and deliver bioactive cargos.201,202 HER2+ dendrimers endowed them with the targetability towards TAMs
extracellular vesicles from BT-474 cells were successfully via interaction between glucose and glucose transporters.
attached to the surface of MDA-MB-231 cells from triple- Meanwhile, replacement of glucose with galactose facilitated
negative breast cancer tissues. This tissue has no therapeutic targeting galactins on the surface of glioblastoma cells.221 In
receptors, and the introduction of extracellular vesicles into addition, layer-by-layer (LDL) surface modification is an emer-
these cells helped in achieving targeted therapy of HER2 ging method to simultaneously allow specific targeting and
positive cancer.203 Immunomodulatory drugs including BDC- toxicity mitigation of high-dose immunocytokine therapy.
1001 (TLR7/8 agonists) and pembrolizumab (PD-1 antibody), Single-chain interleukin-12 was coated onto a liposome, and
as well as HER2-targeting radionuclide drugs (e.g., 131I-GMIB- the prepared product was wrapped with a second layer of poly-L-
Anti-HER2-VHH1, 177Lu-DOTA-ADAPT6-ABD035), have been arginine and a third layer of poly-L-glutamic acid or hyaluronic
employed in this case.204–207 acid. Even at an increasing dose, a significant toxicity reduction
Overall, the above four major types of nanocarriers, particu- and an enhanced therapeutic efficiency were observed in the
larly liposomes, protein-derivatives, inorganic radiosensitizers, treatment of this LDL-modified nanoparticle toward murine
and biomimetic vesicles, have great potential in clinical trans- colorectal and ovarian tumors.222
lation in consideration of their drug-loading capacity, biologi- To conclude, the optimization of size, shape, structure,
cal safety, encouraging preliminary clinical trial results, and surface charge or surface chemistry of nanomedicines can
acceptance by patients and doctors. Meanwhile, the interaction endow them with the ability of loading onco-immunological
between nanocarriers and biological cells, immunotoxicity, and agents (tumor antigens, STING agonists, or ICD inducers) and
immunogenicity remain to be understood.208 Thus, intensive radio-enhancers to tumors or lymph nodes in a more safe and
studies on these aspects of nanocarriers should be investigated. effective manner.223–225
For instance, graphdiyne oxide as a nanocarrier helped polariz- Additionally, nanomedicines can be optimized according to
ing M2 macrophages to pro-inflammatory ones.209,210 their systemic or local administration routes. A high level of
3.2.2 Optimization of nanomedicine-assisted radio-immu- drug accumulation in lesions, stability during blood circula-
notherapy. Achieving an effective localized drug concentration tion, and a low level of distribution in normal tissues are
in tumors and reducing non-specific distribution outside the essential for nanomedicines in a systemic administration man-
tumors could be hindered by several obstacles: the blood–brain ner. Thus, nanomedicines are often tuned or modified to have a
barrier, the blood-tumor barrier, a dense extracellular high drug loading, specific tumor-targetability, and resistance
tumor matrix, a high interstitial fluid pressure, non-specific to protein corona formulation. Meanwhile, for local adminis-
adhesion, and uptake by the liver reticuloendothelial tration of nanomedicines via intratumoral injection, transder-
system.211,212 Nanomedicines can be thereafter optimized with mal administration, transarterial embolization, or portal vein
a few engineering strategies to overcome these above obstacles embolization, they should have a prolonged residence time
to enhance their therapeutic efficacy in radio-immunotherapy with controlled release of antitumor drugs including immu-
(Fig. 5). notherapeutic agents or radiosensitizers in the region of

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

interest.226 Strategies including size-tunability for local reten- immunotherapeutic agents or radiosensitizers at the target
tion and chemical conjugation for prolonged retention are tumor site and reducing their toxicity to normal tissues.228–232
preferable in the design of these nanomedicines. For instance, External therapeutic radiation can be an excellent external
in a recent study, thermal-responsive phase transition of stimulus for controlled release of tumor antigens and immu-
elastin-like polypeptides and electrostatic interaction between notherapeutic agents in radio-immunotherapy.50 For instance,
their oligolysine tail and CpG were utilized to promote the PhotoCORM MnBr(CO)5, a photo-sensitive moiety to release CO
formation of a local complexation depot, achieving long-term in a lanthanide scintillator NP (ScNPs:NaLuF4:Gd,Tb@NaLuF4),
local retention of GpG for more than 3 weeks and therapeutic was indirectly activated by external X-rays to subsequently
131
I for 2 weeks.227 realize the release of the CO gas in a deep-tissue (about 5 cm)
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

and simultaneously achieve CO-mediated ROS generation and


Optimization of stimuli-responsive drug-release from nanome- ICD.233 The common radiation-responsive chemical bonds or
dicines. Stimuli-responsive drug release from nanomedicines structures are summarized in Fig. 5b. The underlying working
triggered by external stimuli (ultrasound, magnetic, light, or principle of radiation-responsive drug release is not well under-
X-ray radiation) or internal stimuli (low pH, high ROS, and stood and different mechanisms have been proposed, for
over-expressed proteases in the TME) can help in releasing example, radiation triggered photolytic degradation of a PLA

Fig. 5 Illustration of nanomedicine optimizations based on administration routes and reported radiation-responsive bonds or structures in RT-based
combined therapies. (a) Optimization of nanomedicines via their administration routes comprise two major steps: the first step is to meet basic
requirements of nanoformulations for systemic routes or local routes; the second one is to optimize the structure and surface chemistry of
nanomedicines to realize protein resistance, size/shape/charge transformation, active targeting, and controlled release. (b) Optimization of radiation-
responsive drug release: the mechanism scheme and a group of reported radiation-responsive bonds or structures in RT-based therapies.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

polymer.234,235 In another case, g-rays triggered the cleavage of solid tumors among all breast cancers,243 has the highest
hydrogen bonds between the antitumor drug, pemetrexed, and number of tumor-infiltrating lymphocytes during the anti-
cytosine-containing diselenide in diselenide-pemetrexed PD1 treatment of 40 breast cancer patients.244 Metastatic
assemblies, and the released drug activated NK cells and melanoma and lung cancer with a high mutation rate may be
exerted its antitumor activity.236 ROS-sensitive bonds can also a better candidate for this combined radio-immunotherapy
respond to g-ray radiation since they induce ROS generation. since it facilitates neoantigen discovery and selection.245,246
For example, hydrophobic poly-(propylene sulfide) was Meanwhile, glioblastoma, characterized with a low nonsynon-
reported to be oxidized to hydrophilic sulfoxides/sulfone in ymous mutation rate and insufficient infiltration of immune
the presence of OH that was generated upon g-ray radiation, cells in tumors,247 is not sensitive to conventional RT or
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

and a robust release of encapsulated drugs was achieved during immunotherapy, but it may be effectively treated with
the oxidation process.237 nanomedicine-mediated radio-immunotherapy.
Intensive studies have been devoted to establishing and
Optimization of administration routes for nanomedicines. The selecting in vitro/in vivo models for evaluating the therapeutic
administration routes of nanomedicines into the body can be potency of nanomedicines for radio-immunotherapy. Ideal
categorized into systemic delivery (intravenous or oral admin- models should be able to map the actual tumor microenviron-
istration) and localized supply (intra-tumoral, intra-nodular, ment in the human body, including the immune system.
transdermal, intra-peritoneal, intranasal, or intraocular admin- Discoveries from these models could accelerate clinical transla-
istration). As reported, the ability of inducing vaccination via tion. While, in reality, a cancer tissue consisting of tumor cells
intradermal/subcutaneous injection, intramuscular injection, and at least one immune cell population from a systemic-
and i.v. injection/oral administration of nanomedicines is in stimulated mouse model (genetically-engineered or therapy-
the descending order.238 induced) are often selected to determine the therapeutic effi-
The optimal administration route for a nanomedicine is cacies of radio-immunotherapy.
heavily dependent on its therapeutic mechanism and the Generally, animal tumor models consist of subcutaneous- or
characteristics of indications.239 It is suggested that cancer orthotopic-grafted cancer, metastasis-induced cancer (intrave-
nanovaccines should reach lymphatic sites to augment the nous or intracardiac engraftment), and artificially induced
presentation of antigens to APCs, the maturation of APCs, spontaneous cancer.248 The orthotopic animal model offers a
and the activation of antigen-specific cytotoxicity T cells. There biomimicking tumor microenvironment similar to that of the
are two major routes for delivery of nanomedicines into lym- original cancer development. A transgene-driven model may
phatic sites: direct access through parenteral injection (sub- provide a great insight on early oncogenesis and tumor
cutaneous and intradermal), or mucosal administration mutation-induced neoantigens, while a carcinogen-driven
(enteral, pulmonary, and intravaginal).240 The sequence of model can display constitutional heterogeneity in tumor
administration is essential to the final vaccination effect. In a tissues.249 Notably, the selection of the mouse species is a
recent study, simultaneous or sequential intravenous/subcuta- crucial parameter in evaluating the effectiveness of cancer
neous (IV/SC) vaccination was carried out using antigen/CpG- radio-immunotherapy. It has been found that C57BL/6 mice
loaded layered double hydroxide nanoparticles in a tumoral prefer to developing Th1 immune response, which is vital to
murine model bearing E.G7-OVA-lymphoma or B16F10- CD8+ T cell-participated antitumor immune response.250
melanoma. The antitumor effect of IV-priming + SC-boosting Hence, C57BL/6 mice species is widely used for evaluating the
was much stronger than that of the untreated group, and more efficacy of radiotherapy and immunotherapy. The ratio of M1/
than 75–90% of the tumor volume shrank after sequential M2 phenotype of TAMs varies among mice models and it is
IV/SC vaccination. Simultaneous IV/SC injection of this cancer higher in the model bearing 4T1 murine breast cancer than the
nanovaccine contributed to a one-week delay of tumor progres- model with CT26 tumors.118 In addition, these cancer models
sion to the end point when the tumor volume reached should be reproducible and practical to be built. Tracking of
1000 mm3 in both tumor models at an early-stage with an the cell lineage could be essential to realize reproducibility.251
initial volume of 50 mm3 and a late-stage with an initial volume In the preclinical trial of treating cancer with radio-
of 500 mm3, respectively.241 immunotherapy, bilateral tumor models and re-challenged
3.2.3 Determination of indications and preclinical models tumor models are the most frequently used. To develop a re-
for radio-immunotherapy. To date, oncology-related indica- challenged model for a prophylactic nanovaccine against can-
tions have been trialed with radio-immunotherapy and their cer or pre-immunization, the nanovaccine or an immune-
corresponding murine cellular lines are listed in the brackets, stimulation agent is primarily injected into the left and right
including non-small cell lung cancer (LLC), melanoma (B16- footpads of mice models. After seven-day immunization, cancer
F10, Cloudman S91), prostate cancer (TRAMP-C2), colon carci- cells are subsequently injected.252 It has been reported that the
noma (MC38 and CT26), liver cancer (rabbit VX2, H22, and efficacy of the vaccine depends on the survival rate of the
Hepa 1–6), triple-negative breast cancer (4T1, EMT6, and injected cancer cells. Additionally, thanks to rapid progress of
TUBO), and bladder cancer (MB49).241,242 Very impressively, a microfluidic chips, in vitro silico-based models, such as patient-
map of intratumoral changes at the single-cell level indicated specific glioblastoma models, have been established as a robust
that triple-negative breast cancer, the most aggressive form of predictive tool for radio-immunotherapy.253

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

However, there are very few reliable, reproducible and potency and biosafety of nanomedicine-assisted caner radio-
cheap models covering metastasis, cancer prevention, tumor immunotherapy can be properly and widely assessed.
dormancy or quiescence, and immune- or radiation-resistance.
More importantly, due to inherent limitations, the animal
models used in most pre-clinical studies on radio- 4. State-of-the-art nanomedicine-
immunotherapy are murine cell line-derived models, not assisted cancer radio-immunotherapy
human tumor-originated ones. Three emerging humanized
murine models for immune-oncology-based therapy, including Nanomedicines have assisted in cancer radio-immunotherapy
the Hu-PBL model, Hu-CD34 model, and BLT model, have been in two major aspects. One is that nanomedicines have aided in
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

built on immunodeficient mice via injection of human periph- in vitro diagnosis, in vivo pre-selection, real-time monitoring,
eral blood mononuclear cells (PBMCs), human CD34+ hemato- and evaluation of the therapeutic response of cancer patients.
poietic stem cells, and human fetal liver and thymus along with They have also contributed to increasing the therapeutic effi-
stem cells, respectively.254 However, these humanized models cacy and reducing toxicity. Their state-of-the-art advances are
suffer from a few issues: induction of a severe graft-verse-host summarized in Table 3.
disease, inability to induce MHC-restricting tumor antigen-
specific immune response, and a complex modeling 4.1 Nanomedicine-assisted imaging for radio-
process.255 Efforts should be devoted to building models to immunotherapy
capture accurate and dynamic biological information of human Imaging cancer tissues is an integral part of cancer treatment and
cancer tissues while maintaining accessibility so that the it has been realized through a wide range of nanomedicines. For

Table 3 Representative examples of nanomedicine-assisted radio-immunotherapy

Main function Interventions Remarks Ref.


64
Pre-stratification and Cu-labeled polyglucose NPs Tumor associated macrophage imaging; 256
response assessment Monitoring TAM response to adjuvant therapy;
Correlating imaging intensity with TAM densities.
68
Ga-NOTA-Nb109 Nonblocking imaging of PD-L1. 257
ROS-responsive IO-Gd Early stratification of RT response;
nanovesicle aiMRI approach is developed;
Acute oxidative stress bridge antitumor immunity. 258
Nanovaccine and in situ Bi2O3@OVA@DC + RT OVA as a synthesis template for Bi2O3 NPs; 259
cancer vaccination Improved efficiency compared with OVA@DC;
Augmenting the STING signaling by Bi2O3 NPs.
PLGA/CpG@PDA-Au + RT Rapid SPG membrane emulsification; 89
Radio-sensitization;
In situ capture of RT-induced antigens.
RP@RMs Irradiated tumor cell membranes as a vaccine; stronger 54
immunogenicity of RMs.
Prompting ICD Phy@PLGdH nanosheets + RT + Shape affecting radiation deposition; 260
aPD-L1 Nanosheets outperforming spherical one;
Inhibiting the pentose phosphate pathway.
H@Gd-NCPs + RT + aPD-L1/ Radiation deposition & GSH depletion; 261
aCTLA-4 Decomposition of H2O2 by Hemin;
Sensitized RT potentiating ICI therapy.
Hf-CpG MXF + RT CpG as DNA components of MXF; 262
Maintaining long-term immune-memory.
Overcoming therapy GDYO nanosheets Inherent immunomodulatory properties; 210
resistance Polarizing M2- to M1-type TAMs;
Stimulating NF-kB & MAPK pathways.
NIA-D1@R848 + RT NIA reducing radioresistance of hypoxia cells;
D
PPA-1 relieving suppression of T cells. 263
AuDAP + RT Dual targeting with AS1411 aptamer & M2pep;
Repolarizing M2 to M1 via NF-kB signaling axis. 264
Reducing therapy-induced Masked IL-12 Protein domain as a mask agent; 96
toxicity Tumor protease-cleavable linker;
Encouraging efficacy without systemic irAEs.
Three click-antidote: Ab-antidotes derived from NPs; 265
phospholipid-PEG micelles, BSA, Short-circulating CLIO NPs perform better than other two
CLIO NPs + click-antibody clicked NPs and CLIO nanoworms.
Polydopamine NPs Oral administration; 266
Scavenging ROS and suppressing inflammation; curing RT-
induced intestinal injury.
Abbreviations: IO, iron oxide; aiMRI, activatable inflammation magnetic resonance imaging; OVA, ovalbumin; SPG, Shirasu porous glass;
RP@RMs, irradiated cancer cell membrane coated on R837-loaded PLGA; Phy@PLGdH, physcion@layered gadolinium hydroxide; GDYO,
graphdiyne oxide; NIA, 2-(2-nitroimidazol-1-yl) acetic acid; AuDAP, dual-functional Au nanoparticle; CLIO, cross-linked dextran iron oxide.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

cancer radio-immunotherapy, nanomedicines have effectively and an accumulation amount of 4.43  0.33% ID g1 in the OS-
helped pre-selecting patients for this treatment, assessing ther- 732 tumor tissue at 24 h post-injection in vivo, confirming its
apeutic response of patients during the treatment, and identifying biological effect on its application in bioimaging.276 In another
immune-related adverse events (irAEs). Furthermore, in vivo ima- study, 64Cu-labelled polyglucose nanoparticles, termed as
ging biomarkers of cancer tissues or tumor-infiltrating immune Macrin, were constructed for quantitative analysis of TAMs
cells could provide guidelines for preparing nano-formulations to via imaging; this probe with a size of B20 nm exhibited a high
achieve effective radio-immunotherapy in pre-selected patients. selectivity (490%) towards macrophages. Since the amount of
4.1.1 Pre-stratification of patients for radio-immu- a radionuclide-labelled model drug accumulated in TAM-rich
notherapy. Pre-stratification of patients can help in avoiding tumors was more than 7-fold that of TAM-deficient tumors, this
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

ineffective treatment, preventing delays in therapeutic inter- TAM-monitoring approach showed a potent ability of pre-
ventions, and reducing undesirable adverse effects.267 In this selecting patients (Fig. 6a).256 In whole, imaging PD-L1 expres-
context, imaging helps semi-quantitative analysis of character- sion on tumor cell and the infiltration density of TAMs is
istic components in the TME, such as tumor-associated macro- currently the most feasible pre-stratification strategy for
phages and tumor-infiltrating lymphocytes.268 Imaging also immuno-based therapy.
reveals tumor kinetics/dynamics for evaluating the extracellular Tumor biomarkers in different cancer types, such as tissue
matrix (ECM) density and the enhanced permeability and factors and L1-cell adhesion molecules, have also been explored
retention (EPR) effect. The vasculature density can be observed as promising pre-selection targets in assessing the accumulation
by serial imaging, and these EPR-indicated radiographical level of nanomedicines at the tumor sites. The tissue factor (TF)
features are crucial for patient stratifications in nanomedicine- was chosen as a potential target for anaplastic thyroid cancer.
assisted treatment. ALT-836 as a TF-specific mAb was thereafter developed. According
Nanomedicine-assisted imaging strategies for pre- to quantitative flow cytometry analysis, ALT-836 displayed a much
stratification of patients include: (a) imaging the expression higher level of cellular uptake in TF-abundant THJ-16T cells than
of biomarkers that are associated with positive therapeutic TF-deficient TPC-1 cells. Its radionuclide-labelled derivative, 64Cu-
outcomes, (b) monitoring dynamic changes in biomarkers NOTA-ALT-836, was reported to achieve high accumulation in
upon therapeutic interventions at a stimulating dose for rapid subcutaneous and orthotopic ATCs; the peak of tumor uptake
pre-evaluation of a treatment plan, and (c) assessing the reached 19.93  2.17% and 37.20  1.71% ID g1 in subcuta-
accumulation level of therapeutic agents in the tumor neous and orthotopic models, respectively (Fig. 6b).277 The L1-cell
tissue.269 These obtained images can help in initially discrimi- adhesion molecule (L1CAM) in cholangiocarcinoma was investi-
nating responders from non-responders towards radio- gated in another study. A diagnostic radioisotope, 64Cu, was
immunotherapy. Among all imaging modalities, nuclear med- conjugated to NH2-terminated chimeric anti-L1CAM (cA10-A3)
ical imaging techniques, such as PET and single photon emis- through a bifunctional chelator, 2-S-(4-isothiocyanatoenzyl)-1,4,7-
sion computed tomography (SPECT), have ultra-high imaging triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA). In vivo
sensitivity and are a preferred choice for imaging in radio- biodistribution of 64Cu-NOTA-cA10-A3 (37 MBq/100 mg) peaked
immunotherapy.270 In detail, radionuclides emit b+ or g rays for with 18.9  2.6% ID g1 at 48 h post-injection at the SCK-L1
PET or SPECT imaging signal, respectively, as well as b or a tumor site, while its tumor accumulation was much lower in other
rays for radiation therapy. These radionuclides can be chelated mice tumor models (Choi-CK, SCK, and JCRB1033). The in vivo
to a nanomedicine to diagnose and pre-stratify cancer patients imaging result was in line with the in vitro cell-bound assay and
simultaneously.271 For instance, PET imaging of intercellular the L1CAM expression level in these cell lines, which was con-
adhesion molecule-1, an up-regulated inducible glycoprotein in firmed from western blotting and flow cytometry analysis, indicat-
non-irradiated tumors in mice receiving RT, can be a valuable ing the feasibility of using this 64Cu-NOTA-cA10-A3 probe for
tool to pre-select patients with the ‘‘abscopal effect’’ at an early detecting L1CAM-positive patients.278
stage.272 In summary, nanomedicine-assisted imaging strategies
A sum of factors, such as a high tumor mutational burden, T broaden the way to pre-select patients for nanomedicine-
cell-inflamed gene expression, PD-1/PD-L1 expression, muta- based radio-immunotherapy. Moreover, classification methods
tions in the repair or correction pathways for DNA damage or to distinguish tumor immune states have emerged, such as
mismatch, and microsatellite instability, have been taken into transcriptomic-based analytical platforms for TME subtypes
consideration as reliable clinical biomarkers, as well as inde- and tumor immunity in the microenvironment.279,280 These
pendent indicators to jointly stratify human cancer to some methods may dramatically accelerate the discovery of specific
extent.273–275 In a recent report, an anti-hPD-L1 heavy chain- and sensitive biomarkers for pre-stratification of cancer
only antibody, Nb6, was labelled with a PET signal-emitting patients and facilitate the development of their corresponding
radionuclide, 124I, to select positive-responsive responders in nanomedicine-assisted imaging probes.
an osteosarcoma OS-732 tumor for the following anti-PD-L1- 4.1.2 Response assessment. Decision on the continuation,
based immunotherapy. This Nb6 antibody with a high affinity suspension, or modification of radio-immunotherapy depends
for hPD-L1 was first selected from 95 monoclones with the help on patients’ response, including biomarker levels from multi-
of phage display technology. 124I-anti-hPD-L1 displayed a high ple sources (blood, lymphatic system, and tumor tissues),
binding affinity value (2.19 nM) towards OS-732 cells in vitro specific radiographic manifestations, and clinical symptoms.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article


Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Fig. 6 Pre-stratification of murine tumor models through nanomedicine-enabled PET imaging of tumor-associated biomarkers. (a) 64Cu-labelled
polyglucose nanoparticles were constructed for quantitative analysis of tumor-associated macrophages: (i) the chemical structure of this probe and its
working principle; (ii) PET/CT imaging of KP lung adenocarcinoma-bearing C57BL/6 mice at 24 h post-injection of this probe. Reproduced with
permission. Ref. 256 Copyright 2018, American Chemical Society. (b) A 64Cu-labelled tissue factor-specific mAb was developed to image tissue factors:
(i) flow cytometry confirmed a high level of the tissue factor in THJ-16 anaplastic thyroid cancer (ATC) cells; (ii) representative maximum intensity
projection (MIP) images of orthotopic ATC murine models using 64Cu-NOTA-ALT-836. Reproduced with permission. Ref. 277 Copyright 2020 the
Authors, Published by Wiley-VCH.

In general, the response patterns to cancer radio- value from in vitro detection is restricted due to a lack of unified
immunotherapy can be defined as follows: hyperprogression, criteria for sampling time, sampling location, multi-biopsy, and
pseudoprogression, stable disease, partial response, and com- tumor heterogeneity,286 while in vivo nanomedicine-mediated
plete response.281 imaging is free from these limitations. It can provide accurate
A clear, early understanding of tumor response to therapies immune profiles of patients and its non-invasive imaging
in holistic cancer treatment is essential to improve clinical features cause less harm to the body.
outcomes. The nanomedicine-assisted in vitro diagnosis PD-L1 (B33 kDa) is recognized as a well-known biomarker
approach and in vivo functional imaging emerge as powerful for radio-immunotherapy. After RT intervention, it is highly
and timely tools for evaluating and predicting body response to expressed in tumor cells and antigen-presenting cells to realize
radio-immunotherapy. Because of a relatively slow therapeutic immune escape, which may facilitate aPD-L1 therapy.20,287 A
response towards radiotherapy and immunotherapy due to diagnostic radionuclide 89Zr-conjugated anti-PD-L1 (atezolizu-
changes in tumor size, invasion, and metastasis, this mab), was utilized in a recent clinical study to image 22
nanomedicine-mediated approach can unveil timely, accurate patients. Each patient had one unique tumor and there were
and specific information about tumor tissues after RT treat- three different tumor types among 22 patients including meta-
ment, such as dynamic changes in immune cell infiltration and static bladder cancer, non-small cell lung cancer, and triple-
early indicators (e.g., elevated ROS or caspase-3).258,282 negative breast cancer. Compared to immunohistochemistry
Circulating tumor DNA (ctDNA) acts an early evaluation (IHC)- or RNA-Seq-based in vitro predictive biomarkers, the
indicator for in vitro diagnosis and provides valuable prognostic generated PET signal from this 89Zr-labelled mAb was better
information about patients treated with antitumor immu- correlated with the therapeutic responses.288 In a previous pre-
notherapy and/or radiotherapy.283,284 A feasible approach for clinical study, 89Zr-desferrioxamine(Df)-atezolizumab was
the in vitro detection of ctDNA could be the use of employed for non-invasive quantitation of PD-L1 expression
nanomedicine-aided detection probes, such as gold nano- after RT treatment. Two lung cancer cell lines, PD-L1+ H460
particles and DNA nanomedicines.285 However, the prognostic cells and PD-L1 A549 cells, were selected in this study. After

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

delivering external RT (5 fractions of 2 Gy) to tumor sites, tumor domain antibody with a different binding epitope from that
uptake of 89Zr-Df-atezolizumab in PD-L1+ H460 bearing mice of anti-PD-L1 and a chelated PET tracer (68Ga-NOTA), was
was significantly increased, while its uptake was slightly applied to quantitatively assess PD-L1 expression in tumors.
improved in mice models with PD-L1 A549 cells. Of note, this Successfully, this probe had a relatively high equilibrium dis-
RT treatment significantly improved the PD-L1/b-actin ratio by sociation constant of 2.9  109 M and a maximum uptake
nearly six fold in H460 cells. These results suggested that the ratio of 5.0  0.35% ID g1 could be achieved in A375-hPD-L1
PD-L1 expression was upregulated after RT, and this 89Zr- tumors at 1 h post-injection (Fig. 7a).257 Therefore, PD-L1
labelled antibody could be used to monitor the change in PD-L1 peptide antagonists that do not affect the binding of PD-L1
expression.289 Moreover, some engineering strategies haven antibodies could be explored to modify imaging probe-labeled
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

been developed for PD-L1 targeting. In one report, a or non-labeled immunomodulatory nanomedicines after RT
radionuclide-labelled lipid-PD-L1 aptamer was applied for the treatment.
immunoimaging of PD-L1. A 99mTc-labelled and PD-L1 Other emerging technologies, including multiple
aptamer-modified C18 chain (C18-apPDL1) was constructed biomarker-targeting multimodal probes and radiomic analysis,
and exhibited two-fold tumoral accumulation at 24 h post- allow biomarker-driven predictions of tumor response to radio-
injection (about 0.88% ID g1) compared to the PD-L1 aptamer immunotherapy. A dual PET/SERS imaging nanoprobe was
and a random sequence-modified C18.290 Notably, nonblocking used to simultaneously detect PD-L1+ tumor cells and CD8+ T
imaging of PD-L1 on tumor cells, a novel and advanced imaging cells (Fig. 7b). A Raman reporter, 5,5-dithiobis(2-nitrobenzoic
method, emerges and allows real-time monitoring of PD-L1 acid) with a peak wavenumber at 1325 cm1 or para-
expression without interfering with the PD-L1 blockade-based mercaptobenzoic acid with a feature peak at 1580 cm1, was
therapy. 68Ga-NOTA-Nb109, composed of a specific single- incorporated on the surface of gold nanostars through a thiol–

Fig. 7 PET imaging via nano-probes aids in accurate and specific monitoring of therapeutic responses towards immune-related therapies. (a)
Nonblocking PET imaging of PD-L1 using 68Ga-NOTA-NB109: (i) illustration of the nano-probe and its working principle; (ii) dynamic PET imaging of
three murine tumor models using this nano-probe. Reproduced with permission. Ref. 257 Copyright 2020, Society of Nuclear Medicine and Molecular
Imaging. (b) Dual-model surface-enhanced Raman spectroscopy (SERS)/PET imaging of PD-L1 and CD8+ in immuno-oncology-treated YUMM2.1
tumors via an engineered immunoactive probe prepared from gold nanostars: (i) scheme of the preparation process of this nano-probe; (ii) SERS spectra
of murine tumor models receiving anti-CD137 + anti-PD-L1 treatment and an IgG control, as well as their corresponding SERS quantification; (iii) PET-CT
imaging of the treated groups at 24 h post-injection of this nano-probe and their corresponding PET quantification. Reproduced with permission. Ref.
291 Copyright 2019, American Chemical Society.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

Au reaction. Correspondingly, PEGylated anti-CD8 or anti-PD- advanced imaging tools, can provide a precise and clear land-
L1 was then conjugated to gold nanostars, followed by DOTA scape of tumor response to radio-immunotherapy. Neverthe-
chelator conjugation and 64Cu chelation. After seven days from less, one should note that not all these indices have clinical
the initial treatment of both PD-L1 and CD137 agonists, two significance and some of them can only be applied to disease-
immunoreactive gold nanostars (IGNs) were injected to moni- specific evaluation. Meanwhile, a few principal endpoints such as
tor the response towards immunotherapy. This treatment was overall survival, progression-free survival, and time to form new
repeated three times and the administration of IgG was set as a metastases may be overestimated for radio-immunotherapy, and
control. PET/CT imaging revealed a relatively high level of other endpoints for radio-immunotherapy should be explored for
tumor accumulation of IGNs in the combinational treatment better evaluation of the therapeutic outcome.
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

group (0.58% ID g1) compared to that in the IgG-treated


control group (0.31% ID g1). Furthermore, the averaged SERS
spectral intensity in the treated and control groups supported 4.2 Cancer nanovaccine-participated radio-immunotherapy
that much more CD8+ T cells were seen in the treatment group, Cancer nanovaccine-mediated vaccination, including exogen-
which was consistent with the immunohistochemistry staining ous vaccination, in situ cancer vaccination (ISV), or both, has
result via 3,3 0 diaminobenzidine, an agent for CD8+ T cell- emerged as a hot topic in cancer research to eradicate residual
specific immunostaining. In all, this imaging platform provides cancer tissues after surgical treatment, induce tumor regres-
a versatile scaffold for multiple biomarker-specific imaging.291 sion and durable antitumor immune response, and prevent
In another study, novel radiomic analysis based on quantitative tumor progression.299,300 Currently, three major categories of
imaging after incorporation of iodixanol-encapsulated lipo- clinically-available personalized cancer vaccines are whole cell
somes was employed to assess myeloid-derived suppressor cell lysates of DCs, synthetic long peptides, and mRNA.301 So far,
(MDSC)-directed immunotherapy. A prolonged blood circula- there are two clinically-approved therapeutic cancer vaccines:
tion time and a high level of nanoparticle accumulation Bacillus Calmette-Guerin (BCG) for early-stage bladder cancer
in perivascular regions facilitated spatiotemporal visualiza- and Sipuleucel-T (Provenge) for prostate cancer, and a few
tion of MDSC-influenced vascular structures. Furthermore, preventive cancer vaccines against HPV-induced cancer (Cer-
nanoparticle-mediated CT images were extracted for radiomic varix, Gardasil, and Gardasil-g) or hepatitis B virus-induced
analysis, and the results indicated that the texture-based fea- cancer (HEPLISAV-B).302 The nanomedicine-based biological
ture was helpful in differentiating treatment groups.292 negative or biological active vaccination adjuvants, which are
Inspried by these findings, it could be inferred that dual beyond conventionally-used liposomes or lipidoid nano-
targeting of MDSCs or M2-like TAMs could be valuable to particles, along with RT, a potent inducer of ISV,303 have been
determine the therapeutic resistance at the tumor site. Dual demonstrated to boost innate or adaptive antitumor immune
targeting strategies to inhibit these cells have been demon- response and increase the therapeutic efficacy by improving
strated to improve the antitumor therapeutic efficay,293,294 and antigenicity and adjuvanticity.304–307 Their working principle is
depletion of both of them was reported to poteniate the ICI concisely demonstrated in Fig. 8.
therapy,295 indicating the benefit of imaging both MDSCs Exogenous cancer vaccination is realized by cancer nano-
and TAMs. vaccines that carry model antigens or identified antigens. These
Pseudoprogression and hyperprogression are two typical nanovaccines carrying identified antigens are ready for large-
outcomes in radio-immunotherapy, which are multiple factor- scale production and clinical translation since these biological
driven and not fully understood yet. It is reported that the active agents are well understood. In contrast, individualized
overall incident rate of pseudoprogression is less than 10% in in situ vaccination relies mainly on the antigens from immuno-
cancer patients receiving ICIs, for instance, 6.4% in patients genic dying tumor cells induced by radiation therapy or other
with melanoma, 5% in patients with NSCLC, and 7% in therapies.308 Treatments, such as radiotherapy, Bacillus
patients with genitourinary cancer.296 Five major hypotheses Calmette-Guerin, TLR agonists, oncolytic viruses, immune
are proposed for pseudoprogression, including (a) Treg expan- cytokines, and Doxil, can elicit in situ cancer vaccination.309
sion; (b) T-cell exhaustion; (c) modulation of pro-tumorigenic Compared with exogenous cancer nanovaccines, this in situ
immune subsets; (d) oncogenic-pathway activation; and (e) vaccination approach can avoid a complex preparation proce-
aberrant inflammation.297 Meanwhile, hyperprogression refers dure, possible contaminations, and immune tolerance induced
to two distinct pathophysiologic phenomena: (1) rapid tumor by the use of universal tumor-associated model antigens.
progression may not be correlated with immune checkpoint Notably, antigen-capturing nanoparticles, such as maleimide-
inhibition; and (2) accelerated tumor growth may be induced by modified dendrimers or bacteria outer membrane vesicles and
ICI-related premature death (e.g., apoptosis).298 With regard dopamine-coated PLGA polymeric nanoparticles, aid in form-
to its multiplex imaging and specific-targeting properties, ing in situ cancer vaccination by capturing antigens released
nanoprobes may be an effective strategy to aid in the non- from the above therapy-treated tumors.89,90
invasive discrimination until more relevant biomarkers have Several typical nanomedicines derived from polymers,
been found. MOFs, tumor-derived membrane-based vesicles or exosomes,
Overall, these advanced technologies, including in vitro or radionuclide-labelled ones are presented below and their
diagnosis of circulating biomarkers or in vivo non-invasive potent vaccination effects are discussed.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev


Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Fig. 8 Scheme for underlying mechanisms of exogenous and in situ cancer vaccination induced by RT-aided cancer nanovaccines. (a) Engineered
nanomedicines are i.v. or i.t. administrated to the body for capturing and presenting RT-induced released tumor antigens. (b) Tumor membrane vesicles
isolated and extruded from RT-treated in vitro tumor cells or in vivo tumor tissues are utilized as personalized cancer nanovaccines. (c) RT primes the
tumor microenvironment, contributing to a preferable condition for exogenous cancer nanovaccine-initiated immune response.

Polymers, including micelles, polymer–epitope conjugates, vaccination, which is primarily attributed to its porous and
and lipid–polymer hybrids, are the leading nanocarriers for hollow structure for holding antigens or immune-stimulating
cancer vaccination apart from liposomes. A pH-responsive agents, as well as its radio-sensitization effect to aid in RT-
PC7A nanovaccine, consisting of E7 or OVA antigenic peptide- induced in situ vaccination.313 Two nanoscale MOFs, DBP-Hf
loaded polyethylene glycol-b-poly(2-hexamethyleneimino ethyl and DBA-Hf, were prepared from an Hf cluster and a porphyrin-
methacrylate), was developed by Gao et al. for treating a size- based photosensitizer ligand, 5,15-di(p-benzoato)porphyrin
able solid tumor by incorporating local RT (20 Gy). In the tumor (DBP) and 5,10,15,20-tetra(p-benzoato)porphyrin (DBA), respec-
mice model of TC-1 (murine lung cancer) and B16-OVA (murine tively. These prepared MOFs were applied to deliver an
melanoma cancer), this combined treatment exhibited a sig- INCB024360 analogue, an IDO inhibitor, to realize synergistic
nificant volume reduction in both primary and distal tumors radio-immunotherapy via an in situ vaccination approach. Both
via systemic and local STING-activation mechanisms.310 Their nanomedicines, IDOi@nMOF, exhibited distinctive radiation
following study on a library of melanoma antigens (Trp1, enhancement factors: 2.13–3.36 for DBP-Hf and 1.47–1.66 for
Tnpo3, Obsl1, Pbk, and Gp100) revealed that phase transition DBA-Hf, in comparison with 1.00–1.22 for HfO2 at the same Hf
of these antigen structures from random coils to a-helix con- concentration. Intratumoral injection of the IDOi-loaded DBP-
tributed to efficient antigenic peptide loading in PC7A micelles, Hf MOF in combination with low-dose X-rays (0.5 Gy  6
which may provide a new means of selecting peptide antigens fractions) to treat bilateral murine breast cancer (TUBO) and
for nanovaccines.311 In another study, this PC7A polymer was colorectal cancer (CT26) demonstrated a superior antitumor
applied to combine with CpG, while maleimide-decorated effect.177 In a similar study, anionic CpG was loaded on the
bacterial membrane was coated on them to form an in situ surface of a cationic nMOF with a photosensitizing ligand via
vaccine. This antigen-capturing nanoparticle aided in absorb- electrostatic interaction to obtain Hf-DBBF-Ir@CpG. Intratu-
ing released neoantigens from tumor tissues after RT interven- moral injection of this nanomedicine and local X-ray irradia-
tion and then cross-presenting to lymph nodes for subsequent tion were applied to a primary tumor during the treatment
DC maturation and CTL infiltration.312 In summary, the tun- process. In situ vaccination was initiated by CpG incorporated
able physiochemical properties of polymer-based nanovaccines in Hf-DBBF-Ir@CpG as a pathogen-associated molecular pat-
allow improving antigen transportation to immune follicles or tern agent and released agents (DAMPs and tumor neoanti-
tumors and protecting antigens from pre-degradation or rapid gens) from tumor cells induced by this treatment.
clearance. Consequently, this approach efficiently activated antigen-
A high-Z element-containing MOF is a potent nanostructure presenting cells and facilitated the infiltration of cytotoxic T
candidate for boosting exogenous and in situ cancer cells in tumor regions in immunosuppressive murine colorectal

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

MC38 and pancreatic Panc02 models. When this treatment was DC, and activating cytotoxic T lymphocytes. It is challenging
combined with aPD-L1, the cure rate reached 83.3% in a T cell- for an exogeneous nanovaccine to be effective in these multiple
absent MC38 model.179 One major advantage of MOFs is easy procedures. Thus, cancer nanovaccines usually have insuffi-
and broad modulation of their compositions (metal ions, cient vaccine potency for tumor eradication, and they are
organic ligands, and loaded agents) to realize different pur- often accompanied by other therapies, including ICIs or
poses. In a recent study, MRI-visible Mn2+ was chosen as chemotherapeutic drugs.
metal ions, meso-2,6-diaminopimelic acid (DAP), a nucleotide
oligomerization binding domain 1 agonist, was selected as
organic ligands, and ovalbumin (OVA) acted as a model anti- 4.3 Prompting immunogenic cell death (ICD)
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

gen. The combination of these compositions achieved an ICD is referred to cell death that elicits an immune response by
imaging-guided nanovaccine delivery to B16-OVA melanoma recruiting and activating DCs to antigens released from dead or
tumors.314 dying tumor cells.319,320 ICD is often induced after physical/
Tumor cell membranes or tumor-derived exosomes isolated chemical stresses on the endoplasmic reticulum and it is
from X-ray irradiated tumor cells could be employed as perso- featured with a high production level of ROS and an apoptotic
nalized nanovaccines due to their upregulated expression of cell morphology that maintains membrane integrity.321,322 ICD
MHC-I, IFN-g, and tumor neoantigens.54,315 For instance, sub- inducers either prompt tumor cell death or induce the release
cutaneous injection of microvesicles at an average size of of DAMPs. They are normally divided into two main types: type
339.9  139.0 nm produced from irradiated C6 glioma cells I acts as an ROS generator; and type II exerts escalated collateral
at a single dose of 50 Gy led to 450% tumoral volume stresses on the endoplasmic reticulum.323 Besides, the hall-
reduction in a subcutaneous glioma C6-bearing rat model marks of ICD are exposure of calreticulin (CRT) and heat shock
compared to the group treated with microvesicles at a size of proteins on the cell surface, active release of high mobility
395.0  202.6 nm from cells without irradiation. Apoptotic tumor group protein B1 (HMGB1) and ATP, and late-stage presenta-
cells in the irradiated microvesicle-treated group were three times tion of uric acid.324,325 ATP provides a ‘‘find me’’ signal to
higher than in the control group, thus the predominant antitumor recruit DCs. The DC phagocytoses the generated ERp57/CRT
mechanism of this irradiated microvesicle-based vaccine was complexes to offer an ‘‘eat me’’ signal through their interaction
attributed to apoptosis of tumor cells.316 with CD91, a DC receptor, and the NF-kB pathway to produce
In addition to the above external radiation-participated IL-6 and TNF-a. Notably, ICD is also strongly correlated with
vaccination methods, injection of therapeutic radioisotope- cancer vaccination.326,327
incorporated nanomedicines is another practical and feasible Among ICD inducers from a physical energy source, such as
approach to induce in situ vaccination. Clinically available 131I a magnetic field,328 ultrasound,329 and near-infrared light,330
was selected as an internal radiation source, and alginate RT is the most accessible, acceptable, and well-studied ther-
(ALG), a natural polysaccharide as an endogenous Ca2+- apeutic modality in clinics. When immunotherapy is intro-
responsive gelation and fixed agent, was chosen to locally hold duced simultaneously, RT outperforms other inducers.
131
I-labelled catalase (CAT) and an immune-adjuvant CpG Specifically, RT holds the following advantages: (a) deep tumor
oligonucleotide at the tumor site. This ‘‘vaccine-alike’’ 131I- penetration; (b) high power energy;331 (c) localized therapeutic
CAT/CpG/ALG displayed excellent immune memory pro- radioisotopes for internal radioisotope therapy that can mimic
tection with no apparent secondary tumor growth. No animal an irradiation field to reduce damage to normal tissues;332 (d)
death was observed until the end observation time point imaging-guided precise therapy via one integral instrument
at day 90. The percentage of effector memory T cells (IGRT or MR-Linac);333,334 (f) tunable biological effects induced
(CD3+CD8+CD62LCD44+ T cells) in the spleen and the level at different doses;21,22 and (g) routine use without sensitizers.
of cytokines (TNF-a and TNF-g) in the serum were significantly In the context of radio-immunotherapy, RT induces upregu-
higher in CT26 tumor models treated with 131I-CAT/CpG/ALG lated expression of tumor antigens and inducible or constitu-
than those treated with surgery and 131I-CAT/ALG.317 In another tive DAMPs,335 but it generates too weak an effect to elicit
study, 131I-MnO2-BSA was systemically administered to CT26 sufficient ICD due to several endogenous resistance mechan-
murine colorectal tumor models. Compared with other thera- isms and poor radiation absorption in some of tumor tissues
pies (control, MnO2-BSA, and free 131I), this radionuclide-based (e.g., sarcoma).336 Insufficient ICD or low immunogenicity is
nanoformulation contributed to a 1–3-fold increase in matured also often seen in conventional ICI-based immunotherapy.337
DCs and a 1–2-fold increase in CD3+CD8+ cytotoxic T cells at the The use of nanomedicines can boost the effect of ICD in radio-
tumor sites.318 immunotherapy by either delivering adequate immune adju-
Overall, there remain a few challenges in this vaccination- vants to elicit strong immune response or escalating the anti-
mediated treatment. There may be a potential risk for in situ tumor effect of radiation, thus enhancing the therapeutic
vaccination: burst release of tumor-associated antigens that are efficacies of immunotherapy. Cisplatin-loaded poly(L-glutamic
also expressed in non-tumoral tissues may result in nonspecific acid)-graft-methoxy poly(ethylene glycol) complex nanoparticles
enhanced immune response to healthy tissues. Exogenous (CDDP-NPs) were reported to significantly enhance RT-induced
nanovaccination involves multiple procedures including load- ICD. The percentage of CRT-positive Lewis lung cancer cells
ing tumor neoantigens, transporting to lymph nodes, maturing increased from 16.47% by treatment with RT to 27.03% after

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

treatment with RT + CDDP-NPs, in comparison with 20.53% in all the treatment groups on murine Tramp C1 prostate cancer
the group treated with RT + cisplatin.338 cells, RT plus S-AuNC induced sufficient ICD with a
A few nanosized ICD inducers, including nanoscale coordi- HMGB1+CRT+ cell population of 51.4%, in comparison with
nation polymers, high Z radiosensitizers, and natural polysac- 37.4% in the group treated with RT. Additionally, the PD-L1
charides, have been developed in combination with strategies expression in the RT + S-AuNC-treated group was significantly
of amplifying oxidative stress to induce a boosted ICD effect. increased by nearly 4 fold compared to that in the RT group,
Nanoscale coordination polymers (NCPs) in which ROS- facilitating the following therapy with released anti-PD-L1
generating metal ions (e.g., Hf, Gd, Cu, or Bi) are incorporated from the nanomedicine.343 Similarly, a heterojunction
into functional ligands or biomacromolecules (CpG oligodeoxy- WO2.9–WSe2–PEG nanostructure (WSP NP) with high photo-
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

nucleotides) have been reported to induce potent ICD via an catalytic activity was developed to boost the intracellular ROS
elevated ER stress.262,339 For instance, in a recent study, gado- level, which could induce ICD. It was noticed that the high
linium and zoledronic acid self-assembled to form a nanorod- photocatalytic properties of this heterojunction structure
like NCP, ZGd-NRs, with a long and short diameter of 200 and were ascribed to the inherent properties of the high-Z ele-
20 nm, respectively. Evaluation of intracellular ROS and g-H2Aw ment and the heterojunction structure because this structure
in CT26 cells indicated that ZGd-NRs could efficiently amplify was able to prevent the recombination of electrons and holes.
ROS generation and enhance X-ray deposition. The incorpo- The fluorescence intensity of ROS generated in the group
rated Gd3+ ions accounted for most of the radio-sensitization treated with WSP NPs and X-rays was strengthened by 1.5
effect. The CT26 cells treated with ZGd-NRs and RT displayed a times compared to that in the X-ray-treated group. Confocal
1–4-fold increase in the ICD induction indicators including the imaging and flow cytometry also confirmed sufficient induc-
level of CRT, HMGB1, and ATP in comparison with other three tion of an ICD biomarker, CRT, in 4T1 cells treated with WSP
groups treated with saline, ZGd-NRs, and saline + RT. When the NPs and X-rays.142 To conclude, high-Z nano-radiosensitizers
treatment with ZGd-NRs and RT was combined with aPD-L1, can be used to prepare potent nanoformulations for promot-
the infiltrated CD4+ and CD8+ T cells in both primary and ing induction of ICD through increasing the ROS-mediated
distant tumors were nearly doubled. Furthermore, the func- endoplasmic reticulum stress under low-dose external irra-
tional ligand, zoledronic acid, helped in depleting TAMs to diation. Moreover, these nano-radiosensitizers in combi-
reprogram an immunosuppressive TME by causing apoptosis nation with ICIs could significantly improve antitumor
of macrophages (Fig. 9a).340 Another strategy to boost potent immune response.
ICD was recently proposed. Mixed-valence copper (Cu+/Cu2+) Natural polysaccharides including chitosan, hyaluronic
was chosen as a metal ion candidate. 5 0 -Guanosine monopho- acid, dextran, alginate, and Ganoderma lucidum polysaccharides
sphate (5 0 -GMP), widely presented in the body, acted as a have been reported to be immunoactive and promote DC
coordination ligand for this Cu-NCP. Amplified oxidase stress maturation, a critical immunogenic indicator of ICD.98,344 For
was attributed to a synergistic effect via Cu+-mediated ROS example, sulfhydryl-modified Ganoderma lucidum polysacchar-
generation and Cu2+-triggered GSH elimination. This nanome- ides (GLP) were covalently conjugated to bismuth sulfide
dicine in combination with RT increased CD8+ cell infiltration nanoparticles (BiNPs) to prepare GLP-BiNPs. BiNPs were
and IFN-g expression in a 4T1 murine breast model. Further- synthesized via a BSA-mediated template approach. DC matura-
more, in a 4T1 primary and metastatic tumor model, the tion was then investigated in the spleen/tumor of 4T1 breast
addition of aPD-L1 showed a better antitumor effect and a tumor-bearing mice treated with a negative control, BiNP, and
greater level of biosafety than other control groups, as evi- GLP-BiNP. In contrast to groups treated with BiNP and the
denced by the primary tumor volume, the mice body weight, control, the percentages of mature DCs in the tumor/spleen at a
counts of lung metastatic sites, and the survival percentage post-injection time point of 24 h or 72 h were higher in the mice
(Fig. 9b).341 Similarly, gadolinium, 5 0 -GMP, and hemin (a group receiving GLP-BiNP. In the group treated with 4 Gy X-ray
peroxidase mimic), self-assembled to form H@Gd-NCPs. radiation at 4 h after intravenous injection of GLP-BiNP, the
Hemin has a similar function to Cu2+, utilizing overproduced tumor volume was effectively reduced compared to other
H2O2 to deplete GSH via peroxidase-mimic catalytic activity.261 groups treated with a negative control, GLP-BiNP, X-rays, and
To conclude, NCPs offer a versatile scaffold or model carrier for BiNP + X-rays. The percentage of CD4+/CD8+ T cells in spleno-
a vast library of ICD-inducing ligands, metal ions and immune- cytes and the concentration of IFN-g and IL-4 in the group
activation agents. treated with GLP-BiNP + X-rays were significantly higher than
High-Z element radiosensitizers contribute to an enhanced those in other treatment groups.345 In another study, the
performance of RT-induced ICD. They generate ROS in an Astragalus membranaceus polysaccharide was reported to induce
intracellular oxygen-independent manner, contributing to the DC activation through the TLR4 signaling pathway.195 Since the
amplification of the endoplasmic reticulum stress, a crucial potential bio-effect induced by these immunoactive agents is
inducer for ICD.342 A snowflake-like Au nanocarrier with RT- very promising, their biological mechanism and biosafety
responsive degradability, S-AuNC, was constructed to load anti- should be examined in great detail, such as the triggered
PD-L1. Under X-ray irradiation, the high-Z gold-based nanos- signaling pathways for inducing immune activation and the
tructure produced a high ROS level, which irreversibly disin- intensity, durability, and universality of induced immune
tegrated the structure to release the loaded anti-PD-L1. Among activation.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article


Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Fig. 9 Nanoscale coordination polymer-based ICD inducers for improving antitumor therapeutic outcomes. (a) Zoledronic acid–gadolinium coordina-
tion polymer nanorods (ZGd-NRs) aided in improving radio-immunotherapy: (i) scheme of the preparation procedure and TEM images of ZGd-NRs;
three typical ICD biomarkers in CT26 tumor cells, (ii) CRT, (iii) HMGB1, and (iv) ATP, upon four different treatments (saline, ZGd-NRs, saline + RT,
ZGd-NRs + RT). Reproduced with permission. Ref. 340 Copyright 2021, American Chemical Society. (b) Mixed-valence copper-based nanoscale
coordination polymers (Cu-NCPs) for augmenting radio-immunotherapy: (i) preparation process and TEM images of Cu-NCPs; (ii) synergistic
mechanism of Cu-NCPs with RT to promote ICD; (iii) growth curves of distant tumors and (iv) counts of lung metastases sites in tumor-bearing murine
models after various treatments including saline, Cu-NCPs + RT + aCD8a, aPD-L1 + RT, Cu-NCPs + RT, and Cu-NCPs + RT + aPD-L1. Reproduced with
permission. Ref. 341 Copyright 2021, Wiley-VCH.

Overall, the approaches to using NCPs, radiosensitizers, potential role of mono-immunomodulator agents as the
or natural polysaccharides to escalate the tumor ROS ICD inducer.
level and induce ICD-related immune response have been
demonstrated to be able to induce potent ICD. At present, 4.4 Overcoming resistance to radio-immunotherapy
RT-induced ICD is vastly studied in cancer radio- Biological or pathological barriers, insensitivity to radiation
immunotherapy, while a few studies focus on the therapy, and immunosuppression are three major resistances

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

to cancer radio-immunotherapy. These three resistances are peptide, Cys–Arg–Glu–Lys–Ala peptide, and twin arginine trans-
tightly interwoven due to their common drivers, such as location peptide), small molecules (e.g., folic acid, biotin,
hypoxia and myeloid-derived suppressor cells. As a result, mannose, galactose, and sialic acid), and other active targeting
biological or pathological barriers are addressed to mitigate ligands (e.g., epithelial cell adhesion molecule aptamer, diben-
the resistance to radio-immunotherapy in most cases, and zocyclooctyne, and hyaluronic acid) have been used for improv-
mitigation in the radiation resistance may be accompanied by ing accumulation and retention of nanomedicines in the region
the effect of reversing immunosuppression, and vice versa. of interest. This strategy is well summarized elsewhere.350,351
4.4.1 Addressing biological or pathological barriers to To cross the brain–blood barriers, recently, an adenosine 2A
radio-immunotherapy. Before active drugs arrive at the sites receptor agonist has been reported to affect both F-actin and
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

of interest, there exist a number of different barriers along their tight junctions of endothelial cells, and it could be a potential
delivery paths. These barriers can be divided into two types:346 immunotherapeutic candidate for active targeting of brain
one is physiological barriers, such as the blood–brain barriers malignancies.352 Similarly, an ApoE-peptide-modified polymer-
in the treatment of malignant brain lesions and nonspecific some was shown to successfully penetrate the blood–brain
accumulation in the liver reticuloendothelial system,347,348 and barrier in a mimicking in vitro model, a bEnd.3 murine
the other is disease-specific, for example, the presence of endothelial cell monolayer. Furthermore, its immunoregulatory
few blood vessels in large tumor burdens, a dense, highly cargos (granzyme B and CpG) contributed to a prolonged
desmoplastic extracellular tumoral matrix, and blood vessel survival duration in orthotopic glioma-bearing mice.353 In
compression.349 addition, an in vivo click chemistry-mediated approach was
To address the physiological barriers, active targeting moi- reported by harnessing the interaction between pre-azide
eties are often incorporated or conjugated onto nanomedicines group-modified lymphatic endothelial cells and a dibenzo-
for precision therapy (Fig. 10a). Antibodies/nanobodies (e.g., cyclooctyhne-functionalized cancer nanovaccine to increase
anti-epidermal growth factor receptor and anti-human the accumulation of the nanovaccine in the lymph node.354
epidermal growth factor receptor-2), peptides (e.g., intercellular Furthermore, pre-occupying the sites in the liver reticuloen-
adhesion molecule-1 peptide, arginine–glycine–aspartic acid dothelial system is another approach to improving active

Fig. 10 Scheme for three major strategies of addressing barriers in nanomedicine-assisted radio-immunotherapy. (a) Active targeting of cell-based
vesicle or nanocarrier is realized by surface modification with peptides, aptamers, tumor cell membranes, click agents, antibodies, small molecules, and/
or carbohydrates. (b) Vascular normalization is achieved through nitric oxide-releasing agents, low-dose radiation, inhibition of the IGFBP7-CD93 axis or
anti-VEGF, use of folic acid (FA)-modified gold NPs, and copper depletion and aggregation. (c) ECM normalization is achieved by depleting cancer-
associated fibroblasts, inhibiting hyaluronan and collagen via various strategies. Abbreviations: ICAM-1, intercellular adhesion molecule-1; CREKA, Cys–
Arg–Glu–Lys–Ala; EPCAM, epithelial cell adhesion molecule; TCO, trans-cyclooctene; DBCO, dibenzoazacyclooctyne; VEGF, vascular endothelial
growth factor; ECM, extracellular matrix; FAP, fibroblast activation protein; PDAC, pancreatic ductal adenocarcinoma; HBO, hyperbaric oxygen.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

targeting and bioavailability of drugs.355 In summary, these (FAPI-46).368,369 Moreover, SLC7A11, a cystine transporter in
active targeting strategies, such as cancer nanovaccines pancreatic ductal adenocarcinoma (PDAC) tumor stroma-
targeting lymph nodes and radiosensitizers or nano- derived CAF, was identified to be independently prognostic of
immunomodulators targeting the TME,356–358 could enhance poor overall survival. A gene-silencing nanomedicine against
the accumulation of the nanomedicines to exert their biological SLC7A11 was thereafter developed and proved to be able to
effects. reduce tumor growth, CAF activation, and fibrosis in orthotopic
Besides RT-aided blood flow improvement, strategies of murine PDAC models.370 For stroma-rich collagen and hyalur-
using nanomedicines for vascular normalization and ECM onan, a significant decrease in ECM levels (e.g., collagen) in 4T1
normalization are conducive to addressing the disease-specific metastases nodules was recently observed after the treatment of
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

barriers.359,360 Vascular normalization could be achieved by tranilast and liposomal Doxil. Specifically, the fraction areas of
utilizing antiangiogenic agents (e.g., anti-VEGF and nitric collagen I, hyaluronan, and hypoxia in this treatment group
oxide-based prodrugs) or other therapeutic interventions (e.g., were significantly reduced, while the density of the perfused
low-dose radiation) to address issues originating from abnor- vessels increased in contrast to other treatment groups. As a
mal tumor vasculature, allowing adequate perfusion of medic- result, this nanomedicine-induced vessel decompression effect
inal drugs and tumor oxygenation (Fig. 10b).361 These resulted in an elevated antitumor effect of ICI in two
therapeutic benefits are often dose- and time-dependent.362 It immunotherapy-insensitive metastatic breast cancer models
was reported that blocking the insulin-like growth factor- (4T1 and E0771).371 In addition, hyperbaric oxygen (HBO)
binding protein 7 (IGFBP7)/CD93 axis using mAbs in two mice therapy could leverage high-pressure oxygen (2.5 atm) to over-
models, an orthotopic KPC tumor model and a B16 tumor come stroma-rich solid tumors, including PDAC, hepatocellular
model, contributed to tumor vascular normalization, which in carcinoma, and triple-negative breast cancer, through depleting
turn sensitized tumor cells to ICI therapy.363 Besides, folic acid- main components of the extracellular matrix (collagen) via
incorporated gold nanoparticles (AuNPP-FA), composed of multiple mechanisms, such as downregulating the gene of
folate, a PEG block, a PDEAEA block with tertiary amines, TGF-b as well as the genes related to the CXCL12/CXCR4
and stabilized gold nanoparticles, were applied to normalize signaling axis and disrupting hypoxia-mediated immunosup-
the tumor vascular structure through strengthening tight junc- pression. Thus, this physical therapy may be a potential addition
tions by upregulating the VE-cadherin level and increasing the to current nanomedicine-assisted radio-immunotherapy.372
pericyte coverage. Notably, a correlation between tumor vascu- 4.4.2 Overcoming resistance to radiation therapy. Radia-
lar normalization realized by AuNPP-FA and tumor metastasis tion resistance, featured with tumor relapse, progression, or
inhibition was established, indirectly supporting the potential spread after RT, remains one major obstacle in cancer radio-
effect of vascular normalization on reducing metastasis.364 In immunotherapy. From a theoretical point of view, tumor
another report, imidazole-incorporated organosilica nanoche- hypoxia, altered mitochondrial metabolism and energy genera-
lators with a copper/phosphate responsive aggregation beha- tion, immune evasion, inflammation, and the presence of
vior were developed to realize antiangiogenesis and vascular cancer stem cells may be the major constitutional factors for
obstruction after depleting Cu2+. An elevated level of Cu2+ and the radioresistant phenotype of tumors.373
phosphate in the tumor environment was significantly reduced Hypoxia is the primary factor that leads to radiotherapy
because the coordination interaction between imidazole or insensitivity or radiation resistance because the indirect DNA-
phosphate and Cu2+ led to aggregation of these dispersed damaging effect of RT predominantly relies on oxygen in the
sub-6 nm nanochelators. A significant antitumor effect was TME.374 Hence, improving tumor oxygenation or enhancing
achieved in both 4T1 breast cancer and CT26 murine colon oxygen-independent free radical generation are two promising
cancer models after treatment with nanochelators compared to approaches for relieving hypoxia-induced radioresistance in the
other groups treated with saline or a copper chelator TM in the TME. Improving tumor oxygenation can be realized by increas-
clinical trial.365 ing intratumoral blood flow, delivering or generating oxygen,
ECM normalization regulates the dense, aberrant tumor and reducing the expression of hypoxia-inducible factors (e.g.,
ECM and vessel compression, enabling homogenous distribu- HIF-1a) in the TME.375 The use of a high-Z radiosensitizer for
tion and enhanced penetration of nanomedicines (Fig. 10c). selective RT irradiation on hypoxia tumor regions or a nano-
Abundant cancer-associated fibroblasts (CAFs), collagen I, and catalyzer (e.g., catalase and holo-lactoferrin) contributes to
hyaluronan may be the main therapeutic targets. CAF, a driver oxygen-independent free radical generation.376,377
for fibrosis generation, matrix remodeling and immune cross- Exogenous oxygen carriers, for instance, hemoglobin (Hb)
talk, may exert negative effects on radio-immunotherapy and and perfluorocarbon (PFC), as well as in situ oxygen generators,
hence become a leading therapeutic target.366 Specific targets like the combined use of H2O2 and catalase, have been
and inhibitors for CAFs have been discovered. Cyclopamine, a employed for tumor re-oxygenation to relieve hypoxia for
sonic hedgehog inhibitor, was reported to deplete stroma- radio-immunotherapy.115 PFC@lipo, Hb@lipo, and PX-478, a
producing CAFs in a polymeric micelle nanoformulation.367 HIF-1a inhibitor, were investigated on their hypoxia relief
Fibroblast activation protein (FAP) that is overexpressed in performance. Hb@lipo treatment was found to be the best
CAFs was reported to be successfully targeted by nanomedi- among these three candidates because of its more moderate
cines modified with a quinoline-based FAP-inhibitor variant oxygen release, a higher level of tumor accumulation through

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

systemic administration, and a safer profile compared to the successfully used to prepare NO-generating nanomedicines to
other two treatments.118 In another study, Hb, a high-Z radio- alleviate tumor hypoxia. A NO-generating micelle was formed
sensitizer (hafnium, Hf), gallic acid, a natural metal chelator, via self-assembly through thin-film hydration from D-a-
and catechol/Ce6-modified 8-arm PEG, self-assembled into Tocopheryl polyethylene glycol 1000 succinate (TPGS)-NO. Under
Hb@Hf-Ce6 nanoparticles. The fluorescence intensity of the a high GSH tumor environment,388,389 this micelle could release
generated 1O2 by Hb@Hf-Ce6 nanoparticles under X-ray irra- NO in a reductive-responsive manner. In vivo PET-CT imaging
diation (8 Gy) was 7.29 times higher than that of free Ce6, with a hypoxia-specific probe (18F-MISO) confirmed that TPGS-NO
indicating this combined strategy achieved a synergistic ROS- and TPGS-NO + RT could effectively reduce tumor hypoxia regions
generating effect. Furthermore, treatment with Hb@Hf-Ce6 + in LLC tumor-bearing mice. Consistently, immunofluorescence
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

RT + anti-PD-1 in a bilateral 4T1 murine breast tumor model imaging and western blotting analysis demonstrated a significant
and its lung metastatic model exhibited remarkable control of decline in HIF-1a and Glut1 expression in tumor tissues after i.v.
tumor progression and an apparent reduction in lung injection of TPGS-NO (50 mg kg1) every two days in the first week
metastasis.378 Moreover, a polydopamine-NP-stabilized oxygen and RT (8 Gy) on day 5. Consequently, this TPGS-NO + RT
microcapsule was also investigated as a new oxygen carrier to approach contributed to a pronounced antitumor effect over other
alleviate tumor hypoxia. Oxygen was sheared into microbub- groups treated with PBS, TPGS-NO, and RT.390 D-Arginine, another
bles, and dopamine was then oxidized in an alkaline environ- source of NO, was loaded onto the surface of hyaluronic acid-
ment. Amino-rich polylysine and chitosan were used to aid in modified MIL-100 (Fe) to form HA@MOF/D-Arg, which was used
accelerating the interfacial polymerization of dopamine nano- to improve the radiosensitivity of K7M2 murine osteosarcoma. It
particles. Finally, glutaraldehyde was used for the permanent was shown that by down-regulating HIF-1a expression, tumor
cross-linking of these NPs. Immunohistochemistry results hypoxia was alleviated in the mice group treated with HA@MOF/D-
showed that the expression of HIF-1a, an indirect indicator of Arg + X-rays. More importantly, the lung metastasis sites of
the hypoxia degree, was much lower in the hep1–6 murine osteosarcoma were found to be significantly reduced compared
tumor mice group treated with local oxygen microcapsules and to other treatment groups including PBS + X-rays, D-Arg + X-rays,
local oxygen microcapsules + RT (8 Gy  2 fractions) in and HA@MOF + X-rays. In addition, this combined therapy
comparison with other groups, indicating that oxygen micro- demonstrated an encouraging tumoricidal effect over other thera-
capsules could effectively relieve tumor hypoxia.379 pies. The control of tumor growth was found to be in a descending
Catalase (CAT), a nano-catalyzer and an in situ oxygen order of HA@MOF + X-rays, D-Arg + X-rays, and PBS + X-rays
generator, can catalytically accelerate the decomposition of (Fig. 11a).117 In another report, a 32P-labelled single-layer 2D
H2O2 into oxygen at the tumor site. External delivery of CAT nanosheet, ZnNO(32P), was fabricated using Zn ions, sodium
and H2O2 into tumors can contribute to robust tumor oxygena- nitroprusside (Na2Fe(CN)5NO) as a NO donor, and 32P radio-
tion. For instance, CAT@liposome and H2O2@liposome were isotopes. In their design, the therapeutic radioisotope 32P with
intravenously injected into 4T1 murine breast tumor-bearing b-ray emission efficiently activated water to generate strong
mice in sequence at a 4 h interval. The hypoxia positive areas in Cerenkov luminescence (CL) and subsequently triggered NO
ex vivo immunofluorescence staining images of tumors after release. Radioisotope tracing and CL intensity monitoring in the
different treatments were semi-quantitatively assessed. The tumor suggested that the tumoral retention time of
percentage of hypoxia was 61% for the group treated with intratumorally-injected ZnNO(32P) was slightly longer than that
PBS and 63% for the group treated with H2O2@liposome, while of ZnCN(32P) but much longer than that of free 32P. Meanwhile,
it reduced to B27% and B9% for the group treated with this sustainable NO-releasing strategy significantly improved the
CAT@liposome and CAT@liposome + subsequent H2O2@lipo- hypoxia-mediated immunosuppressive tumor environment. The
some, respectively. As a result, the tumor oxygenation method immunofluorescence images of tumor slices from different mice
via CAT@liposome + subsequent H2O2@liposome significantly groups confirmed that the tumors treated with ZnNO(32P) exhib-
improved the antitumor effect after treatment with radio- ited the smallest HIF-1a and Treg positive areas and the highest
immunotherapy (X-rays + anti-CTLA-4).380 A similar improve- CTL positive area or the highest CTL/Treg ratio. Furthermore, the
ment in tumor re-oxygenation was observed in a PLGA- combinational treatment with ZnNO(32P) and anti-PD-1 elicited a
R837@CAT nanoparticle. In this nanoparticle, water-soluble potent and durable immune response. The CTL positive percen-
CAT was encapsulated into the core of PLGA, and a TLR-7 tages in local and distant tumors in a CT26 colon tumor model
agonist, imiquimod (R837), was loaded onto the shell of PLGA. were the highest after this combinational treatment. Meanwhile,
Cancer radio-immunotherapy of a subcutaneous CT26 model the concentrations of immunomodulators, including IFN-g and
and an orthotopic 4T1 metastasis model via X-rays + aCTLA4 TNF-a, continuously increased and they surpassed those in other
resulted in a distinctive antitumor effect after application of groups including PBS, anti-PD-1, and ZnNO(32P) on day 0, day 7,
PLGA-R837@CAT nanoparticles.381 and day 12 (Fig. 11b).382
Additionally, nitric oxide (NO), an exogenous regulating Overall, nanomedicines have been developed for tackling
agent, aids in relieving tumor hypoxia by modulating angiogen- tumor hypoxia and other radio-resistance factors to precisely
esis. A few NO donors, including sodium nitroprusside,383 S- deliver/generate oxygen, nitric oxide, HIF-1a inhibitors, radio-
nitrosothiols,384 D-arginine,117 L-arginine,385 DETA NONOate,386 sensitizers, or other similar functional agents in the tumor
and 3-morpholinosydnonimine hydrochloride,387 have been hypoxia region. Besides, strategies to overcome radioresistance

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article


Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Fig. 11 NO-generating MOFs for enhancing radio-immunotherapy through sensitizing the TME. (a) D-Arginine-loaded MOFs increased the radio-
sensitivity of osteosarcoma: (i) illustration of the synthetic route for HA@MOF/D-Arg; (ii) scheme of the radio-sensitization mechanism via HA@MOF/D-
Arg; (iii) free radicals and (iv) NO gas generated from HA@MOF/D-Arg, which were measured from the intensity of their specific in vitro probes;
(v) antitumor effects of the combination therapy of HA@MOF/D-Arg with X-ray RT (indicated with arrows), PBS + X-ray, D-Arg + X-ray, and HA@MOF + X-
ray were used as controls. Reproduced with permission. Ref. 117 Copyright 2021, Elsevier. (b) 32P-Labelled ZnFe(CN)5NO nanosheets aided in improving
radioisotope-immunotherapy: (i) illustration of the lattice structure, cerenkov luminescence-induced NO release, and TME priming effect of 32P-labelled
ZnFe(CN)5NO nanosheets; (ii) immunofluorescence images of tumor slices after various treatments. Blue DAPI for cell nucleus, and green HIF-1a for
hypoxia; (iii) released NO concentrations from ZnNO(32P), ZnNO, and free 32P at different incubation time points; quantitative analysis of (iv) HIF-1a
positive area and (v) CTL/Treg ratio in tumor slices resected from murine tumor models at 12 h post-treatment with various agents, including PBS, ZnNO,
free 32P, ZnCn(32P), and ZnNO(32P). Reproduced with permission. Ref. 382 Copyright 2019, Elsevier.

of tumors have been formulated. For example, dual blockage of immunotherapy.392–395 Moreover, tumor indications, including
RT-induced upregulated CD47 and HER2 eliminates radiore- glioblastoma and liver metastasis, have been frequently
sistant HER2+ breast cancer.391 However, challenges remain to described as immune resistant.396,397
be addressed, including rapid diffusion and clearance of gas
(oxygen and nitric oxide), a low amount of gas generated from Inhibiting TAMs, MDSCs, or Tregs. TAMs, MDSCs, and Tregs
these nanomedicines, and delivery of these nanomedicines to are critical drivers for immunosuppression in tumors.398 TAMs
the tumor hypoxia regions that are often located far from blood include pro-tumoral M2-like macrophages in a large population
vessels. and anti-tumoral M1-like macrophages in a minor
4.4.3 Reversing immunosuppression. Absence of T cells or population.399 Depleting or reducing the recruitment of M2-
insufficiency in T cell infiltration, recruitment and accumula- like TAMs, MDSCs, or Tregs has been demonstrated to success-
tion of myeloid-derived suppressor cells (MDSCs), as well as the fully reverse immunosuppression.400–403
presence of hypoxia, hypermetabolism, regulatory T cells TAM-depleting bisphosphonates, a clinically-used drug,404
(Tregs) and prostaglandin E2, the loss of MHC-I molecules on were reacted with calcium ions via a reverse microemulsion
tumor cells and IFN-g sensing, and vascular occupation or method. The product was modified with PEG for labelling with
compression result in immune evasion and resistance to diagnostic or therapeutic radioisotopes. Cationic 99mTc4+ was

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

labelled onto this nanomedicine with a radiolabelling yield of BP, respectively. Furthermore, the addition of therapeutic
70% by coordination between technetium and phosphonate for radioisotope 32P to CaBP-PEG NPs showed an excellent syner-
single photon emission computed tomography imaging, while gistic antitumor effect with a combination therapeutic index of
32
P, in the form of anionic 32PO43 as a pharmaceutic active 0.48. These results confirmed that CaBP-PEG NPs could effec-
ingredient, was labelled via anion exchange. This radiolabelled tively deplete TAMs and offer a favorable tumor microenviron-
CaBP(99mTc4+)-PEG nanomedicine displayed a potent tumor- ment for the following radiotherapy (Fig. 12a).405
homing ability with a tumor uptake amount of 5.4% ID g1 at In an attempt to deplete MDSCs, chitosan/poly(g-glutamic
24 h post-injection. The F4/80-positive area for TAMs was acid) nanoparticles, Ch/g-PGA NPs, were developed as a func-
significantly reduced to 0.23% after CaBP-PEG treatment in tional adjuvant to RT. RT was used to reduce the primary tumor
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

comparison with 10.74%, 11.3%, and 3.67% in the groups burden, while Ch/g-PGA NPs were applied to decrease spleno-
treated with PBS, CaP-PEG, and free BP, respectively. Similarly, megaly and lung metastases and relieve systemic immunosup-
the TAM-stimulated EGFR-positive area shrank to 35% in the pression. Consequently, the percentage of immunosuppressive
CaBP-PEG-treated group in comparison with 67%, 64%, and myeloid cells (CD45+CD11b+) in the spleen and the weight of
52% in the control groups treated with PBS, CaP-PEG, and free the spleen in the 4T1 tumor-bearing mice group were

Fig. 12 Nanomedicine-assisted approaches to eliminating or polarizing TAMs. (a) Chelator-free, radionuclides-labelled calcium bisphosphonate
enhanced cancer radiotherapy through depleting TAMs: (i) illustration of the preparation procedure of radio-labelled CaBP-PEG nanoparticles; (ii)
release of bisphosphonate (BP), a macrophage-depleting agent, from this NP under different pH conditions; quantitative analysis of (iii) F4/80-positive
and (iv) EGFR-positive areas in tumor slices from a murine tumor model after different treatments; (v) tumor growth curves of different treatment groups
upon administration two times indicated with arrows below the horizontal axis. Reproduced with permission. Ref. 405 Copyright 2018, American
Chemical Society. (b). Gadofullerene (GF-Ala) nanoparticles repolarized TAMs into M1-type ones: (i) scheme of the synthetic route for GF-Ala; (ii) the flow
chart of a co-culture cell system and quantitative analysis of M1 or M2 markers-associated protein expression after different incubation conditions.
Reproduced with permission. Ref. 172 Copyright 2020, American Chemical Society. (c) Triangle-shaped tellurium nanostars enabled TAM repolarization
to potentiate radio-immunotherapy: (i) illustration of the preparation procedure of GTe at different reaction durations, as well as its biological function; (ii)
the percentage of M2-type TAMs in the tumor and spleen tissue under different treatments. Reproduced with permission. Ref. 406 Copyright 2020,
Elsevier.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

reduced.407 In a murine model of GL261 glioma, the PD-L1 level surface-decoration with a targeting ligand, RGD (Arg–Gly–Asp),
in tumor-associated myeloid cells (TAMCs), including M- and biocompatible chitosan constituted a radiation-driven
MDSCs, PMN-MDSCs, and TAMs, was found to be the highest immunotherapeutic enhancer, GTe-RGD. In vitro experiments
compared to other glioma-infiltrating immune cells (microglia, demonstrated that GTe-RGD could improve the radiation sen-
DC, CD8+ CTL, CD4+ Teff, and Tregs). Based on this finding, a sitization of tumor cells including A375, HeLa, and 4T1 cells.
TAMC-targeting lipid-based nanoscale system with surface- GTe-RGD exhibited GSH-responsive degradability, promoting
modification by aPD-L1 and encapsulation of dinaciclib (a its in vivo clearance after its therapeutic function. Furthermore,
cyclin-dependent kinase 5 inhibitor) was developed to treat the percentage of the M2 phenotype was much lower in the
TAMCs. Specifically, flow cytometric analysis confirmed that via tumor tissue and slightly lower in the spleen tissue in the 4T1
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

the Rhod-PE-labelled approach, aPD-L1-LNP exhibited a high breast murine mice group treated with GTe-RGD + X-rays/anti-
efficiency in targeting in vitro-generated TAMCs, as well as PD-1 than that in the groups receiving PBS, GTe-RGD, X-rays,
human-derived TAMCs, which were isolated from the periph- and GTe-RGD + X-rays (Fig. 12c).406 Additionally, the joint use
eral blood and tumor tissue of glioblastoma patients. In addi- of hydrophobic imiquimod and hydrophilic aCD47 co-
tion, the viabilities and immunosuppressive activities of delivered by an Hf-DBP nMOF exerted a TAM repolarization
TAMCs were effectively impaired by this aPD-L1-LNP. Further- effect in bilateral CT26 tumor models. In this study, imiqui-
more, thanks to the effective scavenging of TAMCs to prevent mod, a TLR-7 agonist, was loaded in the pore of the MOF and
an increase in their PD-L1 level after RT, the combination of RT aCD47 was attached onto the surface of the Hf-DBP nMOF with
with aPD-L1-LNP effectively extended the survival time of two acetate capping groups. In contrast to the PBS-treated group,
syngeneic gliomas models of GL261 and CT2A compared to that this nanomedicine-mediated treatment increased the M1 popu-
treated with RT.408 lation by 7.6% and reduced the M2 population by 15.7%. When
the nanomedicine was further combined with RT, a nearly one-
Polarizing tumor-associated macrophages. Polarizing pro- fold increase and drop was observed in the percentage of M1
tumor M2-like TAMs to the pro-inflammatory ones is another and M2 cells, respectively.415
promising approach to overcoming immunosuppression.
Immunomodulatory agents, such as NO donor S-nitrosothiol, Disturbing immunosuppression-related pathways and immuno-
anti-CD40, toll-like receptor agonists (resiquimod or imiqui- suppressive agents. In cancer sites, such as liver metastasis sites,
mod), bexarotene, chlorogenic acid, and TMP195, have been TAMs, MDSCs, Kupffer cells, and hepatic dendritic cells collec-
reported to aid in this transformation process.409–412 For tively form and support a network of active immunosuppressive
instance, it has been reported that lipidic polyplex-aided deliv- pathways, suppressing the activation of CD8+ effector and CD4+ T
ery of the plasmid of IL12, a pro-inflammatory chemokine, cells and inducing immune tolerance to immunotherapy.396,416,417
could increase the ratio of M1/M2 by more than four times.413 In addition, an increase in the level of TNFa and TGF-b induced
Moreover, Ginseng-derived nanoparticles (GDNPs), isolated by RT, and the rapid formation of adenosine from ATP released by
from Panax ginseng C.A. Mey, were found to polarize TAMs to RT could be the culprits of immunosuppressive agent-mediated
the M1-like one via the TLR-4/myeloid differentiation antigen immune tolerance.418–420 In detail, RT-induced TGF-b exerts
88 signaling pathway.414 multiple immune-suppressive activities, such as suppressing
Specifically, b-alanine-modified Gd@C82 (GF-Ala) nano- effector T cells, promoting tumoral infiltration of Tregs, and
particles were reported to re-educate TAMs to a tumoricidal driving DCs to the immune-tolerogenic phenotypes.421 To con-
M1-like phenotype through the activation of NF-kB and IRF5 clude, these immunosuppression-related signaling pathways and
pathways. This fabricated GF-Ala nanomedicine had an average immunosuppressive agents could be harnessed to overcome
diameter of 68.1  2.3 nm and a negative surface potential of resistance to immuno-oncology-associated treatments.
37.7  0.3 mV. After treatment of RAW264.7 cells with the Specifically, it has been reported that the inhibition of the
nanomedicine, the expression of TNF-a, a classical M1 marker, BTK-PI3Kg signaling pathway and blocking the CD47-
was sharply increased over 3.6 times than that in the untreated SIRPa axis contribute to reverse immunosuppression.422
RAW264.7 cells. Meanwhile, the level of IL-10, a classic M2- Furthermore, the blockade of the CCL2/CCR2, CXCR4-
related maker, was 32.6% lower in the GF-Ala-treated group CXCL12, CSF-1/CSF-1R signaling pathways effectively prevented
than that in control cells without any treatment. The nanome- macrophage infiltration in the tumor, and the inhibition of
dicine in combination with anti-PD-L1 to treat a 4T1 CSF-1R in macrophages promoted therapeutic antibody-
tumor model exhibited a pronounced synergistic antitumor dependent phagocytosis of cancer cells.423–426 For example, a
effect. Given the radio-sensitization properties of high-Z synthetic protein nanoparticle, comprising human serum albu-
gadolinium, its antitumor effect could be further improved min, fluorescently-labelled bovine serum albumin, iRGD,
with the intervention of RT (Fig. 12b).172 In another report, a AMD3100 (a CXCR4 antagonist), and bifunctional macromer,
metallic radiosensitizer-assisted radio-immunotherapy (exter- has been shown to block the CXCR4-CXCL12 pathway in the
nal RT + anti-PD-1) was developed and effectively reduced the orthotopic genetically engineered glioblastoma murine models.
percentage of M2-like macrophages. L-Glutathione-modified Furthermore, its combination with RT (2 Gy  10 fractions)
triangle-shaped tellurium nanostars were prepared via a promoted an extended survival time. Three mice survived over
shape-control one-pot hydrothermal method. Their following 60 days, while all the mice in other treated groups were dead.426

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

Overall, despite a small number of studies on the use of or B1 fold in RT + PDA-NPs on day 1 or day 3.5 post-radiation,
nanomedicines to intervene immunosuppression-related sig- respectively, compared to that with radiation only. Moreover,
naling pathways and immunosuppressive agents,420,426 inhibi- from immunohistochemical assays and fluorescence imaging,
tors for these pathways and agents can be incorporated into the PDA-NPs + RT-treated mice group exhibited superior
nanocarriers for overcoming immunosuppression, improving protection of intestinal-associated cells (Lgr5+ intestinal
their solubility and bioavailability while reducing their toxicity stem cells, lysozyme+ Paneth cells, ki67+ crypt base columnar
induced from their accumulation in the normal tissue. cells, or villus+ intestinal cells length) than the radiation-only
group.266
4.5 Remission of the therapy-induced toxicity issue The application of customized masking agents to develop
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Nonspecific deposition of high radiation energy and distribu- immunotherapeutic prodrugs for switchable immune modula-
tion of immuno-oncology agents during cancer radio- tion is another example of using nanomedicines to realize
immunotherapy are the primary toxicity-inducing factors. RT- tumor-specific activation of antitumor immune response and
induced acute and late-onset adverse events include brain prevention of undesirable normal tissue accumulation.438 Engi-
damage (edema, radionecrosis, neurocognitive impairment, neering interleukin-based prodrugs is a preferable method to
and brain inflammation), lung injury (pneumonitis and fibro- reduce immune-toxicity without compromising their therapeu-
genesis), and lymphotoxicity.427 These adverse events have tic efficiency. For instance, an IL-2 mutein/Fc fusion protein, a
been reported to be associated with the cGAS-STING signaling biological binding site for CD8 T cells, was masked with an IL-2
pathway,428 while the toxicities from the immuno-oncology receptor via a matrix metalloproteinase (MMP)-cleavable linker.
treatment depend on immunotherapeutic drugs. The use of Similarly, a recent study employed the masking strategy to
ICIs is often accompanied by multi-organ inflammation reduce the immunotoxicity of IL-12. They fused a domain of
(derma, gastrointestinal, and endocrine),429,430 while CAR T the IL-12 receptor to IL-2 via a MMP-responsive linker.96 There-
cell therapy and cytokine therapy may lead to the cytokine fore, the masking strategy via nano-engineering protease-
release syndrome and/or the immune effector cell-associated sensitive linkers significantly reduces the toxicity in cytokine-
neurotoxicity syndrome.431 based immunotherapy, meanwhile, the specific-site drug dis-
From the above discussions, it can be seen that the adverse tribution and the antitumor effect have been greatly improved.
events induced by RT and immunotherapy are strongly related In addition, a significant shortcoming of current antibody-
to inflammation. High-dose steroid therapy can effectively based immunotherapy is their excessive blood circulation time,
relieve the severity of inflammation. However, steroid- which may lead to the formation of thrombus and other
refractory irAEs, such as nonendocrine irAEs, are insensitive adverse effects. These adverse effects and their immunogenic
to this therapy.432 Therefore, protective drugs, detoxicant properties may cause damage to healthy tissues after the
agents, or other strategies to reduce radio- or immuno-related administration of these antibodies. Nanosized click antidotes,
toxicities have been explored. A couple of agents, including avidin chases, and other nano-agents have been employed for
memantine, selenium, and anti-CD47 antibody, confer a radio- blood clearance of antibodies or nanomedicine-mediated stra-
protective effect to the brain tissue undergoing whole-brain tegies have emerged to accelerate blood clearance of these
radiotherapy and the adjacent normal tissues predominantly antibodies (Fig. 13).219,439–442
via a ROS-scavenging manner.433–435 Meanwhile, masking Overall, a few toxicity-inducing mechanisms of cancer radio-
agents or antibody-clearance agents are dedicated to relieving immunotherapy are well-studied, while the majority of them
immune-associated toxicity. are still unknown. Improved biodistribution and pleiotropic
Radiation proctopathy (RP) is the result of inflammation of immunomodulatory effects of nanomedicines, as well as
the colorectal tissue after RT, in which the platelet-derived employment of specific radio- or immune-protective drugs,
growth factor C (PDGF-C) and excessive ROS can act as ther- can help in addressing issues of already identified toxicities.
apeutic targets.436 In one study, upregulation of PDGF-C was Non-specific body retention of nanomedicines should be
found in the tissues with RP either from patients or animal reduced for their long-term safety. Renal- or hepato-clearance
models and genetic deletion of PDGF-C in mice could relieve strategies have been applied to the design of these nanomedi-
RP-associated adverse effects. In their following experiments, cines. Quantitative structure–activity relationships of the nano-
the use of crenolanib, a selective agonist for PDGF receptors, in medicines could be examined to reduce their toxicities. More
a PEG-400-based solubilizer, successfully prevented and/or importantly, toxicity from the common composition of nano-
ameliorated RP in mice, supported with a 20% thickness medicines should be seriously considered. Take PEG for exam-
reduction of the submucosal layer and a decreased level of ple, the PEGylated approach can greatly alleviate the systemic
expression of fibrosis markers (a-SMA and fibronectin) in a toxicity of IL-2 (extensive inflammation and a fatal vascular leak
mice model 8 weeks after RT.437 Another study employed syndrome), a cytokine used in adoptive cell transfer for T-cell
spherical polydopamine nanoparticles (PDA-NPs) at 50 mg expansion in clinical practice, which originates from non-
kg1 via an oral administration route to scavenge ROS and specific distribution of IL-2 receptors.443 According to a recent
suppress inflammation in RT-induced intestinal injury murine report, PEGylated liposomes containing TLR agonists induced
models. Successfully, the content of MDA, an indicator of hypersensitivity reaction upon multiple dosing, which may be
the lipid peroxidation of intestines, was reduced by B3 fold attributed to the generation of anti-PEG IgGs.444

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article


Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Fig. 13 Scheme for emerging nanomedicine-mediated strategies to accelerate blood clearance of antibodies. Various scavenging nano-agents have
been designed to combine excessive engineered antibodies in the blood stream for rapid clearance via five main strategies, including tco-tetrazine,
azides-cyclooctynes, complementary DNA, biotin–streptavidin, and bsAbs-haptens. Ref. 219 and 439–442.

5. Considerations for clinical (i) Current nanoscale delivery systems are complex. Excess-
translation and future directions engineering of the chemical structure and equipping with many
functional moieties are often sought in the preparation of nano-
Clinical translation of nanomedicine-mediated radio- medicines for realizing radio-immunotherapy. The primary con-
immunotherapy will benefit more cancer patients. However, cern of these nanomedicines is the reproducibility during their
numerous factors need to be carefully considered before these preparation process. Furthermore, the interaction between nano-
nanomedicines enter clinical trials. We will summarize the formulations (e.g., PEGylated nanomedicines) and biological
challenges in this field, discuss lessons learned from ongoing organs/tissues/cells still largely remains unknown.449 In particular,
or completed clinical trials on radio-immunotherapy, and great efforts are needed to discover the interaction between these
provide insights into challenges and future research direction nanomedicines with a complex structure and the systemic immu-
of this combined therapeutic methods.445–448 nity in the patient. More importantly, incorporation of multiple
biological active agents in one single nanoplatform may increase
5.1 Summary of the challenging issues for radio- the complexity of unveiling their synergistic mechanisms of
immunotherapy action. Thorough investigations of a single promising active agent
In the past five years, a myriad of nanoformulations have been in the nanomedicine should be conducted to reveal its working
explored for radio-immunotherapy. This nanotechnology-aided mechanisms and additional active agents could be expanded to
therapy, mostly in the preclinical stage, helps in addressing a realize synergistic therapeutic benefits. In this context, MnO2 NPs
low response rate and systemic toxicity, as well as realizing may be a great candidate for radio-immunotherapy of cancer.
therapeutic benefits, including effective systemic tumor control MnO2 nanomedicine-mediated immunomodulation and radio-
and long-term immune memory protection. However, chal- sensitization have been discovered to some extent: Mn2+ aug-
lenges remain in the current trials of nanomedicines for ments the STING signaling and hypoxia-mediated radioresistance
radio-immunotherapy. Three prominent challenges are is relieved by oxygen generation in a MnO2-mimic catalyst decom-
highlighted below. position of over-expressing H2O2 in TME.116

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

(ii) There are too many combinational treatment options. of this nanomedicine-assisted combined therapy. Particularly,
Many factors could play a role in the application of the lessons from the the terminated, withdrawn, or suspended
nanomedicine as a mono-therapeutic agent for radio- trials can help in designing and preparing nanomedicines,
immunotherapy or an adjuvant to RT and immunotherapy. building patient cancer-mimicking animal models, and formu-
These factors include administration routes, sequence, admin- lating treatment procedures for this radio immunotherapy. We
istration intervals, radiation doses, fractionation, the radiation have listed a few lessons from these trials.
source, and doses of the therapeutic agent(s) and the nanocar- (i) Recruitment of inadequate patients for trials. A high risk
rier. Consequently, superfluous treatment plans could be for- of a new treatment may deter patients from participating these
mulated from the combination of the above factors, but these trials. Some of the enrollment criteria are set too high and very
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

plans for a nanoformulation could not be fully executed con- few patients can meet the enrollment criteria. A clinical trial
sidering the experimental cost and resources. It is advised to screening tool to analyze 16 095 potential participants revealed
narrow down these factors based on previous and ongoing that 13% of them did not enroll into the trial because they did
clinical trials and practice. In view of the complex radiation- not meet the trial eligibility.458 Solid preclinical data should be
immunity net, we suggest establishing a radiation dose and obtained to provide consistent and efficacious treatment in
fractions for one indication. Robust immunity-eliciting radia- animal models. The clinical benefits of this treatment should
tion schemes (e.g., 8 Gy  3 fractions) have been applied to be easily understood by these patients.
compare the synergistic effects of various immunomodulatory (ii) Poor or slow accrual. Many clinical trials are terminated
nanoagents.52 Meanwhile, a hypofractionated low-dose radia- or suspended due to poor or slow accrual. An unoptimized trial
tion plan (e.g., 2 Gy  5 fractions) is combined with one protocol, chronic and acute comorbidities (e.g., hypertension or
immune-drug, such as an ICI, to compare radiosensitivity and diabetes) of participants, a low recruitment momentum, a slow
the synergistic abscopal effect of different metal or non-metal screening process for the first trial participants, and a better
radiosensitizers.450,451 Additionally, the initial tumor volume, alternative therapy may result in poor or slow accrual.459
administration frequency, dosing, administration route, and (iii) Insufficient supply of nanomedicines. For instance,
therapeutic sequence could be standardized to establish the CCX872-B, a CCR2 inhibitor, was in shortage in a phase I/II
evaluation criteria for these nanomedicines. trial (NCT03778879). Challenging issues with chemistry, man-
(iii) Complete profiles of the immunogenicity and immuno- ufacturing and control (CMC) of nanomedicines, a high man-
toxicity of nanocarriers need to be established. The immuno- ufacture cost, or an insufficient manufacturing capacity could
toxicity of PEGylated liposomes used as a vaccine was recently contribute to inadequate supply of nanomedicines for clinical
revealed,452,453 including the anti-PEG IgG-induced hypersensi- trials. Facile preparation of nanomedicines with simple chem-
tivity reaction.444 However, there are very few reports on thor- istry or structures could be readily transferred to large-scale
ough characterization of immunogenicity and immunotoxicity production. Great reproducibility, a high yield in a large scale, a
of polymer-, protein-, or inorganic-based nanocarriers. The high purity of pharmaceutic active ingredients, and fewer
standardized characterization methods should be established preparation steps are essential for the CMC of these nanome-
to accelerate the translation of nanomedicines for clinical dicines in good manufacturing practice (GMP). Importantly,
trials. Recently, polysaccharides,454 cell membranes of treated CQAs of nanomedicines in terms of potency and safety should
tumor or immune cells,455,456 and bacterial outer membranes be thoroughly studied.
are pursued as personalized nanocarriers because they can (iv) Ineffective treatment and high toxicity. A low biological
elicit robust adaptive and innate immune response.457 The efficacy and CD19 CAR CTL persistence result in terminating a
immunogenicity and immunotoxicity of these personalized phase I/II trial (NCT01195480). A tumor lysate vaccine, Imiqui-
nanocarriers are two of the most critical quality attributes mod, and RT were used in another phase I trial (NCT01400672)
(CQAs). The impurities in the preparation of these nanocar- to treat diffuse intrinsic pontine glioma and glioblastoma
riers, such as endotoxin and chemical residuals, may also multiforme and this trial terminated due to ineffective treat-
induce innate immune response, which could provide an ment and high toxicity.
inaccurate profile of immunogenicity and immunotoxicity of Overall, many factors contribute to the successful clinical
personalized nanocarriers. translation of a nanomedicine to a specific indication. Treat-
ment effectiveness and tolerant toxicity are two critical factors.
5.2 Lessons learned from ongoing clinical trials Currently, there are few clinical trials in nanomedicine-
Over 500 clinical trials have been registered in the NCT data- mediated radio-immunotherapy. An approach to combining
base (https://clinicaltrial.gov/ct2/home) on cancer radio- clinically used radiation therapeutic methods and approved
immunotherapy during the past five years. Although there are immunotherapeutic nanoformulation agents should be consid-
a very few trials on nanomedicine-assisted radio-immuno- ered for nanomedicine-mediated radio-immunotherapy first,
therapy, the number of trials has been steadily increasing due and this approach can accelerate the clinical translation of this
to unique advantages and attributes of these nanomedicines as new treatment and the ensuing other nanomedicine-assisted
well as an increase in the approval of nanomedicines for other radio-immunotherapy. In addition, feasible and precise cri-
therapies. Strengths and weaknesses of these clinical trials teria, along with reliable nanomedicine-aided imaging meth-
could provide great insights into effective clinical translation ods to differentiate the disparity in the treatment response and

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

predict the degree of toxic, adverse effects have not been peptides obtained from patients’ biological samples, involves
established due to the slow response of immunotherapy and superfluous less controllable steps and has reproducible issues,
radiotherapy. but increasing evidence from these preclinical studies or clin-
ical trials contribute to developing potent, simple, personalized
5.3 Challenges for clinical translation nanomedicines.467,468
Despite the inspiring results from preclinical studies and a few
clinical trials, there are many challenges for clinical translation
of the nanomedicines in radio-immunotherapy. 5.4 Future directions
Generally, good manufacturing practice (GMP), gram or Based on the challenging issues, lessons drawn from ongoing,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

kilogram-scale production, and facile, reproducible prepara- completed, or withdrawn trials, along with recent advances in
tion procedures are the three major hurdles on the way of this field, we suggest three promising research topics on
nanomedicines’ ‘‘from bench to clinic’’.460,461 In the context of nanomedicine-assisted cancer radio-immunotherapy: radiola-
nanomedicine-mediated cancer radio-immunotherapy, there belled immunogenic antibodies for theranostics, harnessing inflam-
are other specific issues. mation, and metastases-directed combined therapy.
(i) Challenges in building in vitro cell assays. Since the Radiolabeled, immunogenic antibodies for theranostics.
combinational biological basis of radiotherapy and immu- Introducing theranostic radionuclide pairs into the antibodies
notherapy is in a systemic manner, conventional in vitro cell with immunological regulation effects, such as ICIs, non-
assays fail to evaluate their therapeutic efficacy. Establishment Hodgkin lymphoma-treating monoclonal antibody (mAb) (Bex-
of specifically designed cellular assays is always time- and xar and Zevalin), bispecific mAbs, and mAb conjugates,469
resource-consuming. For instance, the main procedure of the could achieve precise SPECT/PET-assisted radio-immuno-
in vitro cross-presentation assay usually takes 1–2 weeks to therapy. These radiolabeled antibodies may amplify antitumor
complete, and this does not include the duration for isolating immune response by simultaneously revitalizing effector T
and stimulating bone marrow-derived dendritic cells and CD8+ cells, reprogramming the immunosuppressive tumor stroma,
T cells from the murine bone marrow or spleen.462 and monitoring and killing tumor cells. One concern over this
(ii) Lack of investigation of large-scale human tumor-derived novel treatment lies in that the energy emitted from the loaded
samples in animal models. Due to the inherent limitation, radionuclides may affect the inherent function of antibodies. It
murine cell lines are often employed for most pre-clinical has been demonstrated in previous studies that the function of
studies in radio-immunotherapy. A few humanized murine mAbs was not compromised after the addition of diagnostic
models still experience issues, such as rejection, poor or low radionuclides (32P or 131I) to anti-PD-1 or anti-PD-L1
MHC-restricting antigen-specific immune response, and a mAbs.382,470 However, since therapeutic radionuclides generate
complex modeling process, thus hampering their wide relatively high radiation energy than diagnostic ones, low-dose
application.463,464 So far, there is a lack of large-scale studies therapeutic radionuclides could be selected to regulate the
on human tumor-derived cell lines or tissues or organoids in tumor immunological microenvironment. This issue deserves
animal models. Effective humanized murine models could help more efforts for preparing clinically-translatable radiolabeled,
in shortening the gap between preclinical studies and clinical immunogenic antibodies.
trials or practices. Additionally, expanding the repertoire of theranostic radio-
(iii) Simple design and facile preparation of nanomedicines. nuclides can be critical for this treatment method. Visible
Several adjuvant agents or pharmaceutical preparations for isotopes of currently used therapeutic radionuclides may be
cancer vaccines and inorganic radiosensitizers have undergone such a candidate. For instance, thyroid tumor ablation was
clinical trials.175,465,466 Their chemical structure is simple and realized with the help of 131I, and liver cancer with 90Y micro-
their prepare process is readily implemented in a GMP environ- spheres. Their corresponding isotopes, such as 124I and 86Y,
ment, thus their pharmacokinetics, therapeutic efficacy, safety, could be utilized to achieve imaging function.471 Other typical
and tolerability can be monitored or evaluated via currently theranostic pairs include 64Cu/67Cu, 68Ga/67Ga, 83Sr/89Sr,
203
established methods. Pb/212Pb, and 72As/77As.472 Their radioactive potencies and
To conclude, this research area is still in its fetal period and biological half-lives should be considered during the selection
its great potential remains to be explored. Great efforts need to of these theranostic pairs: the half-lives should be slightly long
be devoted to this research area. Currently, over-engineered enough to cover the duration including customized labelling,
nanomedicines in a controlled manner could provide a average cargo delivery, and peak tumor accumulation; and after
potential means of accelerating their clinical translation for adequate tumoral accumulation they can exert their tumor-
simultaneously exploring the biological function and synergis- killing effect in a rational time range without noticeable
tic biological effect of multiple agents. After the mechanistic adverse effects. Chelator binding prevails in the current radi-
action of these over-engineered nanomedicines is revealed, olabeling methods.473 In addition to clinically used chelators,
their structure and preparation could be simplified. More such as 1,4,7,10-tetraazacyclododecane-tetraacetic acid (DOTA)
importantly, personalized treatment in this emerging research and 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA), other
field, consisting of nanotechnology-aided modification and tightly-bound and selective chelating ligands with high specific
optimized CAR T cells, DCs, tumor cells, or antigenic tumoral activities could be explored. Moreover, tumor-specific targeting

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

moieties of these ligands should be modified to avoid potential regression, or recurrence of both primary and distant
damage to immune cells. tumors.487,488 In a recent report, phosphatidylserine-coated
Harnessing inflammation. Prolonged inflammation, a hall- liposomes loaded with cyclic guanosine monophosphate-
mark of cancer, is closely correlated with excessive ROS- adenosine monophosphate, NP-cGAMP, were aerosolized for
induced oxidative stress and it may account for tumor progres- inhalation administration to treat multifocal lung metastases
sion since an elevated ROS level in the TME may weaken ICD in combination with fractionated RT (24 Gy/3 fractions). Phy-
and tumoral infiltration of CTL cells.474,475 Cancer treatment siochemically, this NP-cGAMP had a size of around 119 nm and
methods, such as radiation therapy and immunotherapy, can a zeta potential of 41 mV. Two distinctive in vivo lung
also induce inflammation, and a key step of inducing inflam- metastasis models were built through intravenously injected
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

mation is the vascular reaction. B16-OVA cells and orthotopically injected 4T1-Luc cells into
The inflammation process originating from RT may have mammary fat pads. Lung metastasis sites were significantly
‘‘double-sword’’ impacts on cancer immunity. On the one hand, reduced in both tumor models after the combined treatment of
RT induces rapid inflammatory response via recruiting TAMs, NP-cGAMP and RT compared to other therapies. Monitoring of
leading to pseudo-progression and hindrance of timely and 4T1 lung metastases with 39 day IVIS and MRI revealed that the
effective treatment.476 Furthermore, immune analysis reveals that metastasis sites after this treatment were under well control
incomplete radiofrequency ablation induces sustained local and the ratio of CD8+ T/Treg in 4T1 lung metastases was
inflammation with predominant myeloid suppressor cells, which improved.489
inhibit T cell function in tumors.477 On the other hand, RT- Currently, a CREKA (Cys–Arg–Glu–Lys–Ala) peptide, target-
induced inflammation contributes to shaping the antigenicity of ing fibrin–fibronectin complexes in the tumor stroma and
tumor cells, as well as the phenotype, immune-polarity, and vessel walls, has been widely used as a metastasis-specific
density of inflammatory immune cells.476 ligand.490 After more therapeutic targets for metastases, includ-
Notably, inflammation can be a therapeutic target.478,479 ing melanocortin 1 receptor for metastatic melanoma and
Therapeutic intervention of chronic inflammation in tumors leucine-rich alpha-2-glycoprotein 1,491,492 have been discovered
could enhance the clinical efficacy of therapeutic vaccines and and confirmed, other reliable and feasible targeting ligands can
adoptive T cells,480 because inflammation in the tumor can be thereafter designed and applied to modify nanomedicines
lead to caspase-1-dependent pyroptosis, a highly inflammatory for treating multiple metastases, especially the tiny one. In
form of programmed cell death.481,482 On basis of this finding, addition, tumor metabolism can be exploited as a potential
a few nanomedicines have been designed to augment inflam- target to halt tumor cell growth and prevent tumor seeding
mation. For example, the phospholipid-coated Na2S2O8 NP, an since a few metabolic pathways experience dynamic changes
H2O2-independent ROS-generating agent, was in situ degraded during tumor migration.493
to toxic  SO4 and  OH. The osmotic pressure induced by
excess released Na+ from this peroxydisulfate NP led to
caspase-1 dependent pyroptosis, evidenced by a significant 6. Conclusions
increase in the caspase-1 activity and secretion of IL-1b in
4T1 and CT26 cells treated with this NP.483 A body of evidence from preclinical studies and clinical trials has
Overall, inflammation could be harnessed during radio- indicated that cancer radio-immunotherapy has distinctive advan-
immunotherapy of cancer. The inflammatory TME could be tages over monotherapy in improving therapeutic outcomes and
remodeled via RT, and anti-inflammation drugs are then used mitigating immune- or radio-induced toxicity. Furthermore,
to avoid adverse events. However, the antitumor inflammatory unique properties of nanomedicines, such as tumor/lymphatic
effect is in a transient therapeutic window and anti- system-specific targeting, radio-sensitization, accommodation of
inflammation drugs should be administered to avoid long- multiple immunomodulators, and size/shape/surface tunability,
term inflammation and inflammation-induced toxicity. A endow them with multiple indispensable and supplementary
highly efficient indicator for the therapeutic window is to be roles in this synergistic radio-immunotherapy. Among these roles,
established for maximizing the positive effect of therapy- nanomedicine-mediated imaging could help in the differentiation
inducing inflammation but minimizing its adverse effects. of the tumor immune state, patient stratification, diagnosis,
Metastasis-directed combined therapy. Metastases are the monitoring and timely assessment of therapeutic response and
primary culprit of high mortality in cancer patients. After prediction of long-term clinical benefits. In addition, amplifying
migrating to the secondary or more distant sites, plastic metas- the tumoricidal effect and reducing therapy-induced toxicities to a
tasizing tumor cells can colonize and modulate the local tolerant level have been achieved through rational design of
environment. A dormancy site that is insensitive to therapeutic nanomedicines with flexible modification in overcoming therapy
interventions can be thereafter formed.484 It has been reported resistance, eliciting solid innate or adaptive immune response,
that oligo-metastasis with 3–5 metastasis sites has been shown reinvigorating cytotoxic effectors, and increasing immune-
to gain distinctive clinical benefits by combined radio- metabolic interplays.
immunotherapy.485,486 Although the working principles of synergistic radio-
Nanomedicine-mediated radio-immunotherapy could be immunotherapy and mechanisms of reversing immune- and/
potent to this deadly indication, and it could detain growth, or radio-suppression have been unveiled to some extent for the

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

nanomedicines in animal models, there are many challenges in M. Huse, R. C. Hendrickson, X. Wang, I. Rivière and
the transitional process of ‘‘from bench to clinic’’ for nanome- M. Sadelain, Nature, 2019, 568, 112–116.
dicines in radio-immunotherapy. A consortium convened from 6 B. Blanco, C. Domı́nguez-Alonso and L. Alvarez-Vallina,
multi-disciplinary teams could be formed to establish the Clin. Cancer Res., 2021, 27, 5457–5464.
criteria for therapeutic response, clinical benefits, acceptable 7 Y. Otani, J. Y. Yoo, C. T. Lewis, S. Chao, J. Swanner, T.
toxicity level, patient selection and the library of suitable Shimizu, J. M. Kang, S. A. Murphy, K. Rivera-Caraballo,
nanomedicine candidates for cancer radio-immunotherapy. B. Hong, J. C. Glorioso, H. Nakashima, S. E. Lawler,
Y. Banasavadi-Siddegowda, J. D. Heiss, Y. Yan, G. Pei,
M. A. Caligiuri, Z. Zhao, E. A. Chiocca, J. Yu and B. Kaur,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

Author contributions Clin. Cancer Res., 2022, 28, 1460–1473.


H.-N. Li, Q. Luo, and K. Luo conceived the outline of this 8 W. Li, X. Zhang, C. Zhang, J. Yan, X. Hou, S. Du, C. Zeng,
review. H.-N. Li and Q. Luo collected papers for writing. H.-N. Li W. Zhao, B. Deng, D. W. McComb, Y. Zhang, D. D. Kang,
drafted the manuscript and performed the creation of figures. J. Li, W. E. Carson III and Y. Dong, Nat. Commun., 2021,
H. Zhang, X.-L. Ma, and Z.-W. Gu helped revise the manuscript 12, 7264.
and discuss the content. Q.-Y. Gong and K. Luo supervised this 9 D. Xue, B. Moon, J. Liao, J. Guo, Z. Zou, Y. Han, S. Cao,
project and finalized this manuscript. Y. Wang, Y. X. Fu and H. Peng, Sci. Immunol., 2022,
7, eabi6899.
10 B. J. Schneider, J. Naidoo, B. D. Santomasso, C. Lacchetti,
Conflicts of interest S. Adkins, M. Anadkat, M. B. Atkins, K. J. Brassil, J. M.
Caterino, I. Chau, M. J. Davies, M. S. Ernstoff, L. Fecher,
There are no conflicts to declare.
M. Ghosh, I. Jaiyesimi, J. S. Mammen, A. Naing,
L. J. Nastoupil, T. Phillips, L. D. Porter, C. A. Reichner,
Acknowledgements C. Seigel, J. M. Song, A. Spira, M. Suarez-Almazor,
U. Swami, J. A. Thompson, P. Vikas, Y. Wang,
Fig. 1–5, 7b(i), 8, 10 and 13 and table of contents entry were J. S. Weber, P. Funchain and K. Bollin, J. Clin. Oncol.,
created with BioRender.com. This work was financially sup- 2021, 39, 4073–4126.
ported by the National Natural Science Foundation of China 11 C. Valero, M. Lee, D. Hoen, A. Zehir, M. F. Berger,
(52073193, 51873120, 81621003), 135 project for disciplines V. E. Seshan, T. A. Chan and L. G. T. Morris, JAMA Oncol.,
of excellence, West China Hospital, Sichuan University 2021, 7, 739–743.
(ZYJC21013). 12 K. B. Pointer, S. P. Pitroda and R. R. Weichselbaum, Trends
Cancer, 2022, 8, 9–20.
13 J. D. Schoenfeld, A. Giobbie-Hurder, S. Ranasinghe, K. Z.
Notes and references
Kao, A. Lako, J. Tsuji, Y. Liu, R. C. Brennick, R. D. Gentzler,
1 U. Wiedermann, E. Garner-Spitzer, Y. Chao, M. Maglakelidze, C. Lee, J. Hubbard, S. M. Arnold, J. L. Abbruzzese, S. K.
I. Bulat, A. Dechaphunkul, W. Arpornwirat, C. Charoentum, Jabbour, N. V. Uboha, K. L. Stephans, J. M. Johnson, H. Park,
C. J. Yen, T. C. Yau, S. Tanasanvimon, J. Maneechavakajorn, L. C. Villaruz, E. Sharon, H. Streicher, M. M. Ahmed, H. Lyon,
A. Sookprasert, L. Y. Bai, W. C. Chou, T. Ungtrakul, M. Drinic, C. Cibuskis, N. Lennon, A. Jhaveri, L. Yang, J. Altreuter,
J. Tobias, C. C. Zielinski, L. Chong, N. J. Ede, M. T. Marino L. Gunasti, J. L. Weirather, R. H. Mak, M. M. Awad,
and A. J. Good, Clin. Cancer Res., 2021, 27, 3649–3660. S. J. Rodig, H. X. Chen, C. J. Wu, A. M. Monjazeb and
2 G. Terrero, J. Datta, J. Dennison, D. A. Sussman, I. Lohse, F. S. Hodi, Lancet Oncol., 2022, 23, 279–291.
N. B. Merchant and P. J. Hosein, JAMA Oncol., 2022, 8, 938–940. 14 S. George, B. I. Rini and H. J. Hammers, JAMA Oncol., 2019,
3 J. Gao, N. Navai, O. Alhalabi, A. Siefker-Radtke, M. T. 5, 411–421.
Campbell, R. S. Tidwell, C. C. Guo, A. M. Kamat, S. F. Matin, 15 A. Gomez-Iturriaga, M. Keyes, J. Martin and D. E. Spratt,
J. C. Araujo, A. Y. Shah, P. Msaouel, P. Corn, J. Wang, J. N. Lancet Oncol., 2022, 23, 23–25.
Papadopoulos, S. S. Yadav, J. M. Blando, F. Duan, S. Basu, 16 N. G. Zaorsky, J. B. Yu, S. M. McBride, R. T. Dess,
W. Liu, Y. Shen, Y. Zhang, M. D. Macaluso, Y. Wang, J. Chen, W. C. Jackson, B. A. Mahal, R. Chen, A. Choudhury,
J. Zhang, A. Futreal, C. Dinney, J. P. Allison, S. Goswami and A. Henry, I. Syndikus, T. Mitin, A. Tree, A. U. Kishan and
P. Sharma, Nat. Med., 2020, 26, 1845–1851. D. E. Spratt, Adv. Radiat. Oncol., 2020, 5, 26–32.
4 N. K. Altorki, T. E. McGraw, A. C. Borczuk, A. Saxena, 17 M. E. Rodrı́guez-Ruiz, C. Vanpouille-Box, I. Melero,
J. L. Port, B. M. Stiles, B. E. Lee, N. J. Sanfilippo, R. J. Scheff, S. C. Formenti and S. Demaria, Trends Immunol., 2018,
B. B. Pua, J. F. Gruden, P. J. Christos, C. Spinelli, J. Gakuria, 39, 644–655.
M. Uppal, B. Binder, O. Elemento, K. V. Ballman and 18 S. Rottenberg, C. Disler and P. Perego, Nat. Rev. Cancer,
S. C. Formenti, Lancet Oncol., 2021, 22, 824–835. 2021, 21, 37–50.
5 M. Hamieh, A. Dobrin, A. Cabriolu, S. J. C. van der Stegen, 19 H. B. Barsoumian, R. Ramapriyan, A. I. Younes, M. S.
T. Giavridis, J. Mansilla-Soto, J. Eyquem, Z. Zhao, Caetano, H. Menon, N. I. Comeaux, T. R. Cushman,
B. M. Whitlock, M. M. Miele, Z. Li, K. M. Cunanan, J. E. Schoenhals, A. P. Cadena, T. P. Reilly, D. Chen,

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

F. Masrorpour, A. Li, D. S. Hong, A. Diab, Q. N. Nguyen, 31 Q. Luo, Z. Duan, X. Li, L. Gu, L. Ren, H. Zhu, X. Tian,
I. Glitza, R. Ferrarotto, S. G. Chun, M. A. Cortez and R. Chen, H. Zhang, Q. Gong, Z. Gu and K. Luo, Adv. Funct.
J. Welsh, J. Immunother. Cancer, 2020, 8, e000537. Mater., 2022, 32, 2110408.
20 S. S. Du, G. W. Chen, P. Yang, Y. X. Chen, Y. Hu, 32 X. Zheng, D. Pan, G. Zhu, L. Zhang, A. Bhamra, R. Chen,
Q. Q. Zhao, Y. Zhang, R. Liu, D. X. Zheng, J. Zhou, J. Fan H. Zhang, Q. Gong, Z. Gu and K. Luo, Adv. Mater., 2022,
and Z. C. Zeng, Int. J. Radiat. Oncol., Biol., Phys., 2022, 112, 34, 2201200.
1243–1255. 33 M. B. Atkins, M. J. Robertson, M. Gordon, M. T. Lotze,
21 F. G. Herrera, C. Ronet, M. Ochoa de Olza, D. Barras, M. DeCoste, J. S. DuBois, J. Ritz, A. B. Sandler, H. D.
I. Crespo, M. Andreatta, J. Corria-Osorio, A. Spill, Edington, P. D. Garzone, J. W. Mier, C. M. Canning,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

F. Benedetti, R. Genolet, A. Orcurto, M. Imbimbo, L. Battiato, H. Tahara and M. L. Sherman, Clin. Cancer
E. Ghisoni, B. Navarro Rodrigo, D. R. Berthold, Res., 1997, 3, 409–417.
A. Sarivalasis, K. Zaman, R. Duran, C. Dromain, J. Prior, 34 H. Cai, Y. Xiang, Y. Zeng, Z. Li, X. Zheng, Q. Luo, H. Zhu,
N. Schaefer, J. Bourhis, G. Dimopoulou, Z. Tsourti, Q. Gong, Z. Gu, Y. Liu, H. Zhang and K. Luo, Acta Pharm.
M. Messemaker, T. Smith, S. E. Warren, P. Foukas, Sin. B, 2021, 11, 544–559.
S. Rusakiewicz, M. J. Pittet, S. Zimmermann, C. 35 S. S. Sridhar, N. Blais, B. Tran, M. N. Reaume, S. A. North,
Sempoux, U. Dafni, A. Harari, L. E. Kandalaft, S. J. M. R. Stockler, K. N. Chi, N. E. Fleshner, G. Liu,
Carmona, D. Dangaj Laniti, M. Irving and G. Coukos, J. W. Robinson, S. D. Mukherjee, Y. Rahim, E. Winquist,
Cancer Discovery, 2022, 12, 108–133. C. M. Booth, N. T. Nguyen, E. K. Beardsley, N. S.
22 R. J. Griffin, M. M. Ahmed, B. Amendola, O. Belyakov, Alimohamed, G. T. McDonald, K. Ding and W. R. Parulekar,
S. M. Bentzen, K. T. Butterworth, S. Chang, C. N. Coleman, JAMA Oncol., 2020, 6, 1751–1758.
V. Djonov, S. C. Formenti, E. Glatstein, C. Guha, 36 H. Park, A. Otte and K. Park, J. Controlled Release, 2022,
S. Kalnicki, Q. T. Le, B. W. Loo, Jr., A. Mahadevan, M. 342, 53–65.
Massaccesi, P. G. Maxim, M. Mohiuddin, M. Mohiuddin, 37 E. Pujade-Lauraine, K. Fujiwara, J. A. Ledermann, A. M. Oza,
N. A. Mayr, C. Obcemea, K. Petersson, W. Regine, R. Kristeleit, I.-L. Ray-Coquard, G. E. Richardson, C. Sessa,
M. Roach, P. Romanelli, C. B. Simone 2nd, J. W. Snider, K. Yonemori and S. Banerjee, Lancet Oncol., 2021, 22,
D. R. Spitz, B. Vikram, M. C. Vozenin, M. Abdel-Wahab, 1034–1046.
J. Welsh, X. Wu and C. L. Limoli, Int. J. Radiat. Oncol. Biol. 38 C. Ji, M. Zhao, C. Wang, R. Liu, S. Zhu, X. Dong, C. Su and
Phys., 2020, 107, 766–778. Z. Gu, ACS Nano, 2022, 16, 9428–9441.
23 S. Demaria, C. Guha, J. Schoenfeld, Z. Morris, A. Monjazeb, 39 H. Song, H. Sun, N. He, C. Xu, Y. Wang, L. Du, Y. Liu,
A. Sikora, M. Crittenden, S. Shiao, S. Khleif, S. Gupta, Q. Wang, K. Ji, J. Wang, M. Zhang, Y. Gu, Y. Zhang, L. Feng,
S. C. Formenti, B. Vikram, C. N. Coleman and M. M. O. Tillement, W. Wang and Q. Liu, Nanoscale, 2022, 14,
Ahmed, J. Immunother. Cancer, 2021, 9, e002038. 11429–11442.
24 S. P. Keam, H. Halse, T. Nguyen, M. Wang, N. Van Kooten 40 T. L. Nguyen, B. G. Cha, Y. Choi, J. Im and J. Kim,
Losio, C. Mitchell, F. Caramia, D. J. Byrne, S. Haupt, G. Ryland, Biomaterials, 2020, 239, 119859.
P. K. Darcy, S. Sandhu, P. Blombery, Y. Haupt, S. G. Williams 41 C. A. Ferreira, S. Goel, E. B. Ehlerding, Z. T. Rosenkrans,
and P. J. Neeson, J. Immunother. Cancer, 2020, 8, e000792. D. Jiang, T. Sun, E. Aluicio-Sarduy, J. W. Engle, D. Ni and
25 M. A. Postow, M. K. Callahan, C. A. Barker, Y. Yamada, W. Cai, Nano Lett., 2021, 21, 4692–4699.
J. Yuan, S. Kitano, Z. Mu, T. Rasalan, M. Adamow and 42 Q. Hu, H. Li, E. Archibong, Q. Chen, H. Ruan, S. Ahn,
E. Ritter, New Engl. J. Med., 2012, 366, 925–931. E. Dukhovlinova, Y. Kang, D. Wen, G. Dotti and Z. Gu, Nat.
26 W. Ngwa, O. C. Irabor, J. D. Schoenfeld, J. Hesser, Biomed. Eng., 2021, 5, 1038–1047.
S. Demaria and S. C. Formenti, Nat. Rev. Cancer, 2018, 43 F. G. Herrera, J. Bourhis and G. Coukos, Ca-Cancer J. Clin.,
18, 313–322. 2017, 67, 65–85.
27 J. S. O’Donnell, M. W. L. Teng and M. J. Smyth, Nat. Rev. 44 J. C. Jagodinsky, P. M. Harari and Z. S. Morris, Int. J. Radiat.
Clin. Oncol., 2019, 16, 151–167. Oncol. Biol. Phys., 2020, 108, 6–16.
28 A. X. Lozano, A. A. Chaudhuri, A. Nene, A. Bacchiocchi, 45 H. Zhuang, Cancer Commun., 2020, 40, 649–654.
N. Earland, M. D. Vesely, A. Usmani, B. E. Turner, C. B. 46 M. Trommer, S. Y. Yeo, T. Persigehl, A. Bunck, H. Grüll,
Steen, B. A. Luca, T. Badri, G. S. Gulati, M. R. Vahid, M. Schlaak, S. Theurich, M. von Bergwelt-Baildon,
F. Khameneh, P. K. Harris, D. Y. Chen, K. Dhodapkar, J. Morgenthaler and J. M. Herter, Front. Pharmacol., 2019,
M. Sznol, R. Halaban and A. M. Newman, Nat. Med., 2022, 10, 511.
28, 353–362. 47 S. Demaria, B. Ng, M. L. Devitt, J. S. Babb, N. Kawashima,
29 N. Gabriel, K. Balaji, K. Jayachandran, M. Inkman, L. Liebes and S. C. Formenti, Int. J. Radiat. Oncol. Biol.
J. Zhang, S. Dahiya and M. Goldstein, Cancer Res., 2022, Phys., 2004, 58, 862–870.
82, 2019–2030. 48 T. P. Lippert and R. A. Greenberg, J. Clin. Invest., 2021,
30 S. B. Elsherif, M. Anderson, A. A. Chaudhry, S. P. Kumar, 131, e148274.
D. R. Gopireddy, C. Lall and P. R. Bhosale, Eur. J. Radiol., 49 L. Zhu, S. Hu, Q. Chen, H. Zhang, J. Fu, Y. Zhou, Y. Bai,
2022, 146, 110062. Y. Pan and C. Shao, Int. J. Biol. Sci., 2021, 17, 926–941.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

50 W. Wang, H. Xu, Q. Ye, F. Tao, I. Wheeldon, A. Yuan, Y. Hu 70 I. Minn, S. P. Rowe and M. G. Pomper, Lancet Oncol., 2019,
and J. Wu, Nat. Biomed. Eng., 2022, 6, 44–53. 20, 443–451.
51 E. Mizukoshi, H. Nakagawa, T. Tamai, M. Kitahara, 71 R. S. Goedegebuure, C. Vonk, L. P. Kooij, S. Derks and
K. Fushimi, K. Nio, T. Terashima, N. Iida, K. Arai, V. L. Thijssen, Int. J. Radiat. Oncol. Biol. Phys., 2020, 108,
T. Yamashita, T. Yamashita, Y. Sakai, M. Honda and 56–69.
S. Kaneko, Nat. Commun., 2022, 13, 3123. 72 C. Xie, A. G. Duffy, G. Brar, S. Fioravanti, D. Mabry-Hrones,
52 C. Lhuillier, N. P. Rudqvist, T. Yamazaki, T. Zhang, M. Walker, C. M. Bonilla, B. J. Wood, D. E. Citrin, E. M. Gil
M. Charpentier, L. Galluzzi, N. Dephoure, C. C. Clement, Ramirez, F. E. Escorcia, B. Redd, J. M. Hernandez,
L. Santambrogio, X. K. Zhou, S. C. Formenti and J. L. Davis, B. Gasmi, D. Kleiner, S. M. Steinberg,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

S. Demaria, J. Clin. Invest., 2021, 131, e138740. J. C. Jones and T. F. Greten, Clin. Cancer Res., 2020, 26,
53 S. C. Formenti, N.-P. Rudqvist, E. Golden, B. Cooper, 2318–2326.
E. Wennerberg, C. Lhuillier, C. Vanpouille-Box, 73 W. Theelen, H. M. U. Peulen, F. Lalezari, V. van der Noort,
K. Friedman, L. F. de Andrade and K. W. Wucherpfennig, J. F. de Vries, J. Aerts, D. W. Dumoulin, I. Bahce,
Nat. Med., 2018, 24, 1845–1851. A. N. Niemeijer, A. J. de Langen, K. Monkhorst and
54 Z. Tuo, Q. He, Z. Zhang, Y. Wang, J. Sun, Q. Wei, Y. Hu, P. Baas, JAMA Oncol., 2019, 5, 1276–1282.
J. F. Lovell, H. Jin and K. Yang, Chem. Eng. J., 2022, 74 S. C. Katz, E. Prince, M. Cunetta, P. Guha, A. Moody,
433, 134437. V. Armenio, L. J. Wang, N. J. Espat and R. P. Junghans,
55 Z. Bian, L. Shi, K. Kidder, K. Zen, C. Garnett-Benson and Cancer Res., 2017, 77, CT109.
Y. Liu, Nat. Commun., 2021, 12, 3229. 75 S. Modak, P. Zanzonico, M. Grkovski, E. K. Slotkin,
56 K. M. Garland, T. L. Sheehy and J. T. Wilson, Chem. Rev., J. A. Carrasquillo, S. K. Lyashchenko, J. S. Lewis,
2022, 122, 5977–6039. I. Y. Cheung, T. Heaton, M. P. LaQuaglia, N. V. Cheung
57 H. du Bois, T. A. Heim and A. W. Lund, Sci. Immunol., 2021, and N. Pandit-Taskar, J. Clin. Oncol., 2020, 38, 4283–4291.
6, eabg3551. 76 C. Bodet-Milin, L. Ferrer, A. Rauscher, D. Masson, L. Rbah-
58 H. Liang, L. Deng, Y. Hou, X. Meng, X. Huang, E. Rao, Vidal, A. Faivre-Chauvet, E. Cerato, C. Rousseau,
W. Zheng, H. Mauceri, M. Mack, M. Xu, Y. Fu and J. Hureaux, O. Couturier, P. Y. Salaün, D. M. Goldenberg,
R. R. Weichselbaum, Nat. Commun., 2017, 8, 1736. R. M. Sharkey, F. Kraeber-Bodéré and J. Barbet, Front.
59 N.-P. Rudqvist, K. A. Pilones, C. Lhuillier, E. Wennerberg, Med.-Lausanne, 2015, 2, 84.
J.-W. Sidhom, R. O. Emerson, H. S. Robins, J. Schneck, 77 P. Twardowski, J. Y. C. Wong, S. K. Pal, P. H. Frankel,
S. C. Formenti and S. Demaria, Cancer Immunol. Res., 2018, K. Franklin, M. Junqueira, M. R. Prajapati, D. Harwood and
6, 139–150. N. Agarwal, J. Clin. Oncol., 2017, 35, 222.
60 M. O. de Olza, B. N. Rodrigo, S. Zimmermann and 78 C. H. Marshall, J. C. Park, T. L. DeWeese, S. King, M. Afful,
G. Coukos, Lancet Oncol., 2020, 21, 419–430. J. Hurrelbrink, C. Manogue, P. Cotogno, N. P. Moldawer,
61 A. Arina, S. I. Gutiontov and R. R. Weichselbaum, Clin. P. C. Barata, C. G. Drake, E. M. Posadas, A. J. Armstrong,
Cancer Res., 2020, 26, 2777–2782. A. O. Sartor and E. S. Antonarakis, J. Clin. Oncol., 2020,
62 J. N. Cheng, W. Luo, C. Sun, Z. Jin, X. Zeng, P. B. Alexander, 38, 130.
Z. Gong, X. Xia, X. Ding, S. Xu, P. Zou, Y. Y. Wan, Q. Jia, 79 C. R. Heery, R. A. Madan, M. Bilusic, J. W. Kim, N. K. Singh,
Q. J. Li and B. Zhu, Sci. Adv., 2021, 7, eabc7609. M. Rauckhorst, S. M. Steinberg, W. L. Dahut, C. Chen,
63 W. Cao, G. Chen, L. Wu, K. N. Yu, M. Sun, M. Yang, Y. Jiang, R. S. DiPaola, M. N. Stein, D. Panicali, J. W. Hodge,
Y. Jiang, Y. Xu, S. Peng and W. Han, Int. J. Radiat. Oncol. Biol. J. Schlom and J. L. Gulley, J. Clin. Oncol., 2013, 31, 102.
Phys., 2022, DOI: 10.1016/j.ijrobp.2022.07.1841. 80 Y. D. Seo, J. Zhou, K. Morse, J. Patino, S. Mackay, E. Y. Kim,
64 A. Arina, M. Beckett, C. Fernandez, W. Zheng, S. Pitroda, E. U. Conrad, R. B. O’Malley, L. D. Cranmer, H. Lu,
S. J. Chmura, J. J. Luke, M. Forde, Y. Hou and B. Burnette, F. J. Hsu, Y. Xu, E. T. Loggers, T. Hain, V. G. Pillarisetty,
Nat. Commun., 2019, 10, 3959. G. Kane, S. Riddell, J. H. ter Meulen, R. L. Jones and
65 S. Murty, S. T. Haile, C. Beinat, A. Aalipour, I. S. Alam, S. Pollack, J. Clin. Oncol., 2018, 36, 71.
T. Murty, T. M. Shaffer, C. B. Patel, E. E. Graves and 81 M. D. Janosky, S. Demaria, Y. Novik, R. Oratz, A. Tiersten,
C. L. Mackall, OncoImmunology, 2020, 9, 1757360. J. D. Goldberg, E. Wang, F. Marincola, S. Formenti and
66 E. D. Brooks and J. Y. Chang, Nat. Rev. Clin. Oncol., 2019, S. Adams, J. Clin. Oncol., 2013, 31, TPS3122.
16, 123–135. 82 E. J. Van Limbergen, A. Hoeben, R. I. Y. Lieverse,
67 A. J. Oweida, L. Darragh, A. Phan, D. Binder, S. Bhatia, R. Houben, C. Overhof, A. Postma, J. Zindler, F. Verhelst,
A. Mueller, B. V. Court, D. Milner, D. Raben and L. J. Dubois, D. De Ruysscher, E. G. C. Troost and
R. Woessner, J. Natl. Cancer Inst., 2019, 111, 1339–1349. P. Lambin, Int. J. Radiat. Oncol. Biol. Phys., 2021, 109,
68 Y. Hou, H. L. Liang, X. Yu, Z. Liu, X. Cao, E. Rao, X. Huang, 1421–1430.
L. Wang, L. Li and J. Bugno, Sci. Transl. Med., 2021, 83 R. I. Y. Lieverse, E. J. Van Limbergen, C. J. G. Oberije,
13, eabb0130. E. G. C. Troost, S. R. Hadrup, A. C. Dingemans, L. E. L.
69 D. F. Boreel, P. N. Span, S. Heskamp, G. J. Adema and Hendriks, F. Eckert, C. Hiley, C. Dooms, Y. Lievens,
J. Bussink, Clin. Cancer Res., 2021, 27, 2970–2978. M. C. de Jong, J. Bussink, X. Geets, V. Valentini, G. Elia,

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

D. Neri, C. Billiet, A. Abdollahi, D. Pasquier, P. Boisselier, 101 J. Bu, A. Nair, M. Iida, W. J. Jeong, M. J. Poellmann,
A. Yaromina, D. De Ruysscher, L. J. Dubois and P. Lambin, K. Mudd, L. J. Kubiatowicz, E. W. Liu, D. L. Wheeler and
BMC Cancer, 2020, 20, 557. S. Hong, Nano Lett., 2020, 20, 4901–4909.
84 V. Olivo Pimentel, D. Marcus, A. M. van der Wiel, N. G. 102 W. J. Jeong, J. Bu, Y. Han, A. J. Drelich, A. Nair, P. Král and
Lieuwes, R. Biemans, R. I. Lieverse, D. Neri, J. Theys, S. Hong, J. Am. Chem. Soc., 2020, 142, 1832–1837.
A. Yaromina, L. J. Dubois and P. Lambin, J. Immunother. 103 A. Mansurov, P. Hosseinchi, K. Chang, A. L. Lauterbach,
Cancer, 2021, 9, e001764. L. T. Gray, A. T. Alpar, E. Budina, A. J. Slezak, S. Kang and
85 P. Gabani, B. W. Fischer-Valuck, T. M. Johanns, S. Cao, Nat. Biomed. Eng., 2022, 6, 819–829.
L. F. Hernandez-Aya, J. W. Keller, K. M. Rich, A. H. Kim, 104 I. Etxeberria, E. Bolaños, A. Teijeira, S. Garasa, A. Yanguas,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

G. P. Dunn, C. G. Robinson and M. R. Chicoine, Radiother. A. Azpilikueta, W. M. Kavanaugh, O. Vasiljeva, M. Belvin,


Oncol., 2018, 128, 266–273. B. Howng, B. Irving, K. Tipton, J. West, L. Mei,
86 J. E. Bates, C. G. Morris, M. T. Milano, A. R. Yeung and A. J. Korman, E. Sega, I. Olivera, A. Cirella, M. C. Ochoa,
B. S. Hoppe, Radiother. Oncol., 2019, 138, 75–79. M. E. Rodriguez, A. Melero, M. F. Sanmamed,
87 J. Nam, S. Son, K. S. Park, W. Zou, L. D. Shea and J. J. Engelhardt and I. Melero, Proc. Natl. Acad. Sci. U. S. A.,
J. J. Moon, Nat. Rev. Mater., 2019, 4, 398–414. 2021, 118, e2025930118.
88 D. J. Irvine and E. L. Dane, Nat. Rev. Immunol., 2020, 20, 105 L. Tang, Y. Zheng, M. B. Melo, L. Mabardi, A. P. Castaño,
321–334. Y. Q. Xie, N. Li, S. B. Kudchodkar, H. C. Wong, E. K. Jeng,
89 J. Lu, Z. Guo, R. Zheng, W. Xie, X. Gao, J. Gao, Y. Zhang, M. V. Maus and D. J. Irvine, Nat. Biotechnol., 2018, 36,
W. Xu, J. Ye, X. Guo, J. Tang, J. Yu, L. Wang, B. Xu, 707–716.
G. Zhang and L. Zhao, ACS Appl. Mater. Interfaces, 2022, 106 C. W. Shields IV, M. A. Evans, L. L. Wang, N. Baugh, S. Iyer,
14, 4995–5008. D. Wu, Z. Zhao, A. Pusuluri, A. Ukidve, D. C. Pan and
90 Y. Li, K. Zhang, Y. Wu, Y. Yue, K. Cheng, Q. Feng, X. Ma, S. Mitragotri, Sci. Adv., 2020, 6, eaaz6579.
J. Liang, N. Ma, G. Liu, G. Nie, L. Ren and X. Zhao, Small, 107 D. Manoharan, L. C. Chang, L. C. Wang, Y. S. Shan,
2022, 18, 2107461. F. C. Lin, L. C. Wu, H. S. Sheu, W. P. Su and C. S. Yeh,
91 C. Czysch, C. Medina-Montano, N. K. Dal, T. Dinh, ACS Nano, 2021, 15, 9084–9100.
Y. Fröder, P. Winterwerber, K. Maxeiner, H. J. Räder, 108 P. Zhang, A. Darmon, J. Marill, N. M. Anesary and S. Paris,
D. Schuppan, H. Schild, M. Bros, B. Biersack, F. Feranoli, Int. J. Nanomed., 2020, 15, 3843–3850.
S. Grabbe and L. Nuhn, Macromol. Rapid Commun., 2022, 109 L. Zheng, R. Zhu, L. Chen, Q. Fu, J. Li, C. Chen, J. Song and
43, 2200095. H. Yang, Nano Res., 2021, 14, 3744–3755.
92 H. Liu, Z. Wen, H. Chen, Z. Yang, Z. Le, Z. Liu, Y. Chen and 110 P. Pan, X. Dong, Y. Chen, X. Zeng and X. Z. Zhang, ACS
L. Liu, J. Controlled Release, 2022, 352, 497–506. Nano, 2022, 16, 801–812.
93 M. Wehbe, L. Wang-Bishop, K. W. Becker, D. Shae, 111 L. Maggiorella, G. Barouch, C. Devaux, A. Pottier,
J. J. Baljon, X. He, P. Christov, K. L. Boyd, J. M. Balko and E. Deutsch, J. Bourhis, E. Borghi and L. Levy, Future Oncol.,
J. T. Wilson, J. Controlled Release, 2021, 330, 1118–1129. 2012, 8, 1167–1181.
94 H. Zheng, B. Guo, X. Qiu, Y. Xia, Y. Qu, L. Cheng, F. Meng 112 Y. Chen, P. Gao, T. Wu, W. Pan, N. Li and B. Tang, Chem.
and Z. Zhong, Bioact. Mater., 2022, 16, 1–11. Commun., 2020, 56, 10621–10630.
95 C. Wang, Z. Dong, Y. Hao, Y. Zhu, J. Ni, Q. Li, B. Liu, 113 J. Xue, D. Duosiken, S. Zhong, J. J. Cao, L. Y. Hu, K. Sun,
Y. Han, Z. Yang, J. Wan, K. Yang, Z. Liu and L. Feng, Adv. K. Tao and S. J. Pan, Acta Biomater., 2021, 131, 508–518.
Mater., 2022, 34, 2106520. 114 H. Deng, Z. Zhou, W. Yang, L. S. Lin, S. Wang, G. Niu,
96 A. Mansurov, P. Hosseinchi, K. Chang, A. L. Lauterbach, J. Song and X. Chen, Nano Lett., 2020, 20, 1928–1933.
L. T. Gray, A. T. Alpar, E. Budina, A. J. Slezak, S. Kang, 115 Z. Duan, Q. Luo, L. Gu, X. Li, H. Zhu, Z. Gu, Q. Gong,
S. Cao, A. Solanki, S. Gomes, J. M. Williford, M. A. Swartz, H. Zhang and K. Luo, Nanoscale, 2021, 13, 13681–13692.
J. L. Mendoza, J. Ishihara and J. A. Hubbell, Nat. Biomed. 116 Z. Lu, S. Bai, Y. Jiang, S. Wu, D. Xu, Y. Chen, Y. Lan, Y. An,
Eng., 2022, 6, 819–829. J. Mao, X. Liu and G. Liu, Adv. Funct. Mater., 2022,
97 F. Zamboni, S. Vieira, R. L. Reis, J. Miguel Oliveira and 32, 2207749.
M. N. Collins, Prog. Mater. Sci., 2018, 97, 97–122. 117 C. Du, M. Zhou, F. Jia, L. Ruan, H. Lu, J. Zhang, B. Zhu,
98 Y. Zeng, Y. Xiang, R. Sheng, H. Tomás, J. Rodrigues, Z. Gu, X. Liu, J. Chen, Z. Chai and Y. Hu, Biomaterials, 2021,
H. Zhang, Q. Gong and K. Luo, Bioact. Mater., 2021, 6, 269, 120642.
3358–3382. 118 M. Jiang, B. Qin, L. Luo, X. Li, Y. Shi, J. Zhang, Z. Luo,
99 R. I. Jakacki, E. Dombi, S. M. Steinberg, S. Goldman, C. Zhu, G. Guan, Y. Du and J. You, J. Controlled Release,
M. W. Kieran, N. J. Ullrich, I. F. Pollack, A. Goodwin, 2021, 335, 408–419.
P. E. Manley, J. Fangusaro, R. Allen and B. C. Widemann, 119 X. Zhou, M. You, F. Wang, Z. Wang, X. Gao, C. Jing, J. Liu,
Neuro. Oncol., 2017, 19, 289–297. M. Guo, J. Li, A. Luo, H. Liu, Z. Liu and C. Chen, Adv.
100 Y. Yu, J. Li, B. Song, Z. Ma, Y. Zhang, H. Sun, X. Wei, Y. Bai, Mater., 2021, 33, 2100556.
X. Lu, P. Zhang and X. Zhang, Biomaterials, 2022, 120 S. de Leve, F. Wirsdörfer and V. Jendrossek, Front. Immu-
280, 121312. nol., 2019, 10, 698.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

121 W. Li, J. Yan, H. Tian, B. Li, G. Wang, W. Sang, Z. Zhang, 141 M. Li, Z. Wang, X. Liu, N. Song, Y. Song, X. Shi, J. Liu, J. Liu
X. Zhang and Y. Dai, Bioact. Mater., 2023, 22, 34–46. and Z. Yu, J. Controlled Release, 2021, 340, 35–47.
122 Y. Jiang, J. Martin, M. Alkadhimi, K. Shigemori, 142 X. Dong, R. Cheng, S. Zhu, H. Liu, R. Zhou, C. Zhang,
P. Kinchesh, S. Gilchrist, V. Kersemans, S. Smart, K. Chen, L. Mei, C. Wang, C. Su, X. Liu, Z. Gu and Y. Zhao,
J. M. Thompson, M. A. Hill, M. J. O’Connor, B. R. Davies ACS Nano, 2020, 14, 5400–5416.
and A. J. Ryan, Br. J. Cancer, 2021, 124, 1809–1819. 143 P. Pei, W. Shen, Y. Zhang, Y. Zhang, Z. Qi, H. Zhou, T. Liu,
123 S. R. Vink, S. Lagerwerf, E. Mesman, J. H. Schellens, L. Sun and K. Yang, Biomaterials, 2022, 280, 121326.
A. C. Begg, W. J. van Blitterswijk and M. Verheij, Clin. 144 W. Sang, Z. Zhang, G. Wang, L. Xie, J. Li, W. Li, H. Tian and
Cancer Res., 2006, 12, 1615–1622. Y. Dai, Adv. Funct. Mater., 2022, 32, 2113168.
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

124 M. Landry, D. Nelson, E. Choi, A. DuRoss and C. Sun, 145 T. Tian, R. Liang, G. Erel-Akbaba, L. Saad, P. J. Obeid,
Transl. Oncol., 2022, 16, 101336. J. Gao, E. A. Chiocca, R. Weissleder and B. A. Tannous, ACS
125 S. Clement, J. M. Campbell, W. Deng, A. Guller, S. Nisar, Nano, 2022, 16, 1940–1953.
G. Liu, B. C. Wilson and E. M. Goldys, Adv. Sci., 2020, 146 X. Qin, C. Yang, H. Xu, R. Zhang, D. Zhang, J. Tu, Y. Guo,
7, 2003584. B. Niu, L. Kong and Z. Zhang, Small, 2021, 17, 2103984.
126 T. Luo, G. T. Nash, X. Jiang, X. Feng, J. Mao, J. Liu, 147 Y. Shi and T. Lammers, Acc. Chem. Res., 2019, 52,
A. Juloori, A. T. Pearson and W. Lin, Adv. Mater., 2022, 1543–1554.
34, 2110588. 148 A. N. DuRoss, M. J. Neufeld, S. Rana, C. R. Thomas, Jr. and
127 D. Wang, T. Nie, C. Huang, Z. Chen, X. Ma, W. Fang, C. Sun, Adv. Drug Delivery Rev., 2019, 144, 35–56.
Y. Huang, L. Luo and Z. Xiao, Small, 2022, 18, 2203227. 149 J. Wolfram and M. Ferrari, Nano Today, 2019, 25, 85–98.
128 J. Shi, K. Xu, A. Keyvanloo, T. S. Udayakumar, A. Ahmad, 150 J. H. Myung, M. J. Eblan, J. M. Caster, S. J. Park,
F. Yang and Y. Yang, Int. J. Radiat. Oncol. Biol. Phys., 2020, M. J. Poellmann, K. Wang, K. A. Tam, S. M. Miller,
108, 1063–1072. C. Shen, R. C. Chen, T. Zhang, J. E. Tepper, B. S. Chera,
129 K. R. Patel, A. Martinez, J. M. Stahl, S. J. Logan, A. J. A. Z. Wang and S. Hong, Clin. Cancer Res., 2018, 24,
Perricone, M. J. Ferris, Z. S. Buchwald, M. Chowdhary, 2539–2547.
K. A. Delman, D. K. Monson, S. V. Oskouei, N. B. Reimer, 151 X. Zhu, X. Wang, B. Li, Y. Zhang, Y. Chen, W. Zhang,
K. Cardona, M. A. Edgar and K. D. Godette, OncoImmunol- Y. Wang, W. Zhai, Z. Liu, S. Liu, J. Sun, Z. Chen and Y. Gao,
ogy, 2018, 7, 1442168. Small, 2022, 18, 2107001.
130 C. Grassberger, S. G. Ellsworth, M. Q. Wilks, F. K. Keane 152 E. L. Dane, A. Belessiotis-Richards, C. Backlund, J. Wang,
and J. S. Loeffler, Nat. Rev. Clin. Oncol., 2019, 16, 729–745. K. Hidaka, L. E. Milling, S. Bhagchandani, M. B. Melo,
131 X. Xiao, H. Cai, Q. Huang, B. Wang, X. Wang, Q. Luo, Y. Li, S. Wu, N. Li, N. Donahue, K. Ni, L. Ma, M. Okaniwa,
H. Zhang, Q. Gong, X. Ma, Z. Gu and K. Luo, Bioact. Mater., M. M. Stevens, A. Alexander-Katz and D. J. Irvine, Nat.
2023, 19, 538–549. Mater., 2022, 21, 710–720.
132 S. Harmsen, E. I. Medine, M. Moroz, F. Nurili, J. Lobo, 153 Q. Zhang, J. Zhang, J. Song, Y. Liu, X. Ren and Y. Zhao, ACS
Y. Dong, M. Turkekul, N. V. K. Pillarsetty, R. Ting, Nano, 2021, 15, 8001–8038.
V. Ponomarev, O. Akin and O. Aras, Biomaterials, 2021, 154 Z. Wang, K. Zhi, Z. Ding, Y. Sun, S. Li, M. Li, K. Pu and
269, 120630. J. Zou, Semin. Cancer Biol., 2021, 69, 77–90.
133 E. Kim and H. Koo, Chem. Sci., 2019, 10, 7835–7851. 155 K. I. Joo, Y. Jeong, S. M. Hwang, M. Shin, J. Lee, G. Sharma,
134 M. Geppert and M. Himly, Front. Immunol., 2021, H. Lee, S. H. Im and H. J. Cha, Biomaterials, 2020,
12, 688927. 263, 120380.
135 O. Martinez, J. Sosabowski, J. Maher and S. Papa, J. Nucl. 156 P. Debie, C. Lafont, M. Defrise, I. Hansen, D. M. van
Med., 2019, 60, 730–735. Willigen, F. W. van Leeuwen, R. Gijsbers, M. D’huyvetter,
136 J. W. Seo, R. Tavaré, L. M. Mahakian, M. T. Silvestrini, N. Devoogdt and T. Lahoutte, J. Controlled Release, 2020,
S. Tam, E. S. Ingham, F. B. Salazar, A. D. Borowsky, 317, 34–42.
A. M. Wu and K. W. Ferrara, Clin. Cancer Res., 2018, 24, 157 W. Chen, Y. Yuan and X. Jiang, J. Controlled Release, 2020,
4976–4987. 328, 395–406.
137 A. Nguyen, A. Ramesh, S. Kumar, D. Nandi, A. Brouillard, 158 Z. Tian, M. Liu, Y. Zhang and X. Wang, J. Hematol. Oncol.,
A. Wells, L. Pobezinsky, B. Osborne and A. A. Kulkarni, Sci. 2021, 14, 75.
Adv., 2020, 6, eabc2777. 159 W. J. Cheng, K. H. Chuang, Y. J. Lo, M. Chen, Y. J. Chen,
138 Q. Lecocq, R. M. Awad, Y. De Vlaeminck, W. De Mey, S. R. Roffler, H. O. Ho, S. Y. Lin and M. T. Sheu,
T. Ertveldt, C. Goyvaerts, G. Raes, K. Thielemans, J. Controlled Release, 2022, 344, 235–248.
M. Keyaerts, N. Devoogdt and K. Breckpot, J. Nucl. Med., 160 J. Liu, R. Toy, C. Vantucci, P. Pradhan, Z. Zhang, K. M. Kuo,
2021, 62, 1638–1644. K. P. Kubelick, D. Huo, J. Wen and J. Kim, Nano Lett., 2021,
139 C. Shi, Z. Zhou, H. Lin and J. Gao, Small Methods, 2021, 21, 875–886.
5, 2001025. 161 L. Sun, F. Shen, Z. Xiong, H. Yang, Z. Dong, J. Xiang, Q. Gu,
140 Y. Zhang, Z. Feng, J. Liu, H. Li, Q. Su, J. Zhang, P. Huang, Q. Ji, C. Fan and Z. Liu, J. Am. Chem. Soc., 2022, 144,
W. Wang and J. Liu, Bioact. Mater., 2022, 16, 359–371. 7634–7645.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

162 X. Huang, J. Z. Williams, R. Chang, Z. Li, C. E. Burnett, 181 K. Ni, G. Lan, Y. Song, Z. Hao and W. Lin, Chem. Sci., 2020,
R. Hernandez-Lopez, I. Setiady, E. Gai, D. M. Patterson and 11, 7641–7653.
W. Yu, Nat. Nanotechnol., 2021, 16, 214–223. 182 K. Ni, Z. Xu, A. Culbert, T. Luo, N. Guo, K. Yang, E.
163 J. Li, H. Ren and Y. Zhang, Coordin. Chem. Rev., 2022, Pearson, B. Preusser, T. Wu, P. La Riviere, R. R.
455, 214345. Weichselbaum, M. T. Spiotto and W. Lin, Nat. Biomed.
164 Z. Liang, Y. Yang, G. Yu, H. Zhu, X. Xia, C. Chen, D. Fu, Eng., 2022, 6, 144–156.
M. Li, G. Cheng, C. Xue, L. Shi, H. Zeng and B. Sun, 183 G. Lan, K. Ni, S. S. Veroneau, Y. Song and W. Lin, J. Am.
Biomaterials, 2021, 275, 120960. Chem. Soc., 2018, 140, 16971–16975.
165 J. Lu, Y. Jiao, G. Cao and Z. Liu, Chem. Eng. J., 2021, 184 Z. Xu, K. Ni, J. Mao, T. Luo and W. Lin, Adv. Mater., 2021,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

420, 129746. 33, 2104249.


166 R. Chai, L. Yu, C. Dong, Y. Yin, S. Wang, Y. Chen and 185 R. Li, Y. He, S. Zhang, J. Qin and J. Wang, Acta Pharm. Sin.
Q. Zhang, Bioact. Mater., 2022, 17, 276–288. B, 2018, 8, 14–22.
167 P. Pei, T. Liu, W. Shen, Z. Liu and K. Yang, Mater. Horiz., 186 L. Chen, W. Hong, W. Ren, T. Xu, Z. Qian and Z. He, Signal
2021, 8, 1348–1366. Transduction Targeted Ther., 2021, 6, 225.
168 T. Karpov, A. Postovalova, D. Akhmetova, A. R. Muslimov, 187 Z. Li, H. Cai, Z. Li, L. Ren, X. Ma, H. Zhu, Q. Gong,
E. Eletskaya, M. V. Zyuzin and A. S. Timin, Chem. Mater., H. Zhang, Z. Gu and K. Luo, Bioact. Mater., 2023, 21,
2022, 34, 6593–6605. 299–312.
169 X. Hong, X. Zhong, G. Du, Y. Hou, Y. Zhang, Z. Zhang, 188 F. Combes, E. Meyer and N. N. Sanders, J. Controlled
T. Gong, L. Zhang and X. Sun, Sci. Adv., 2020, 6, eaaz4462. Release, 2020, 327, 70–87.
170 P. A. Bielecki, M. E. Lorkowski, W. M. Becicka, 189 D. Xia, D. Hang, Y. Li, W. Jiang, J. Zhu, Y. Ding, H. Gu and
P. U. Atukorale, T. J. Moon, Y. Zhang, M. Wiese, Y. Hu, ACS Nano, 2020, 14, 15654–15668.
G. Covarrubias, S. Ravichandran and E. Karathanasis, 190 M. Mehanny, C.-M. Lehr and G. Fuhrmann, Adv. Drug
Nanoscale Horiz., 2021, 6, 156–167. Delivery Rev., 2021, 173, 164–180.
171 M. Sun, P. Gu, Y. Yang, L. Yu, Z. Jiang, J. Li, Y. Le, Y. Chen, 191 S. R. Kang, D. H. Nguyen, S. W. Yoo and J. J. Min, Adv. Drug
Q. Ba and H. Wang, J. Immunother. Cancer, 2021, Delivery Rev., 2021, 181, 114085.
9, e002508. 192 Y. Zeng, S. Li, S. Zhang, L. Wang, H. Yuan and F. Hu, Acta
172 L. Li, M. Zhen, H. Wang, Z. Sun, W. Jia, Z. Zhao, C. Zhou, Pharm. Sin. B, 2022, 12, 3233–3254.
S. Liu, C. Wang and C. Bai, Nano Lett., 2020, 20, 4487–4496. 193 S. K. Alsaiari, S. S. Qutub, S. Sun, W. Baslyman,
173 X. Zhang, C. He, Y. Sun, X. Liu, Y. Chen, C. Chen, R. Yan, M. Aldehaiman, M. Alyami, A. Almalik, R. Halwani,
T. Fan, T. Yang, Y. Lu, J. Luo, X. Ma and G. Xiang, Acta J. Merzaban and Z. Mao, Sci. Adv., 2021, 7, eabe7174.
Pharm. Sin. B, 2021, 11, 3608–3621. 194 L. Gong, Y. Zhang, J. Zhao, Y. Zhang, K. Tu, L. Jiao, Q. Xu,
174 X. Guan, L. Sun, Y. Shen, F. Jin, X. Bo, C. Zhu, X. Han, X. Li, M. Zhang and S. Han, Small, 2022, 18, 2107656.
Y. Chen, H. Xu and W. Yue, Nat. Commun., 2022, 13, 2834. 195 G. Pang, C. Chen, Y. Liu, T. Jiang, H. Yu, Y. Wu, Y. Wang,
175 S. Bonvalot, P. L. Rutkowski, J. Thariat, S. Carrère, F. J. Wang, Z. Liu and L. W. Zhang, ACS Appl. Mater.
A. Ducassou, M. P. Sunyach, P. Agoston, A. Hong, Interfaces, 2019, 11, 42661–42670.
A. Mervoyer, M. Rastrelli, V. Moreno, R. K. Li, B. Tiangco, 196 X. Han, S. Shen, Q. Fan, G. Chen, E. Archibong, G. Dotti,
A. C. Herraez, A. Gronchi, L. Mangel, T. Sy-Ortin, P. Z. Liu, Z. Gu and C. Wang, Sci. Adv., 2019, 5, eaaw6870.
Hohenberger, T. de Baère, A. Le Cesne, S. Helfre, 197 C. Gong, X. Yu, W. Zhang, L. Han, R. Wang, Y. Wang,
E. Saada-Bouzid, A. Borkowska, R. Anghel, A. Co, S. Gao and Y. Yuan, J. Nanobiotechnol., 2021, 19, 58.
M. Gebhart, G. Kantor, A. Montero, H. H. Loong, 198 M. Z. Zou, Z. H. Li, X. F. Bai, C. J. Liu and X. Z. Zhang, Nano
R. Vergés, L. Lapeire, S. Dema, G. Kacso, L. Austen, Lett., 2021, 21, 8609–8618.
L. Moureau-Zabotto, V. Servois, E. Wardelmann, 199 M. Hu, J. Zhang, L. Kong, Y. Yu, Q. Hu, T. Yang, Y. Wang,
P. Terrier, A. J. Lazar, J. Bovée, C. Le Péchoux and K. Tu, Q. Qiao, X. Qin and Z. Zhang, ACS Nano, 2021, 15,
Z. Papai, Lancet Oncol., 2019, 20, 1148–1159. 3123–3138.
176 X. Wang, X. Zhong, J. Li, Z. Liu and L. Cheng, Chem. Soc. 200 J. H. Park, Y. Jiang, J. Zhou, H. Gong, A. Mohapatra, J. Heo,
Rev., 2021, 50, 8669–8742. W. Gao, R. H. Fang and L. Zhang, Sci. Adv., 2021,
177 K. Lu, C. He, N. Guo, C. Chan, K. Ni, G. Lan, H. Tang, 7, eabf7820.
C. Pelizzari, Y. X. Fu, M. T. Spiotto, R. R. Weichselbaum 201 D. X. Zhang, L. T. Vu, N. N. Ismail, M. T. N. Le and
and W. Lin, Nat. Biomed. Eng., 2018, 2, 600–610. A. Grimson, Semin. Cancer Biol., 2021, 74, 24–44.
178 K. Ni, T. Luo, G. T. Nash and W. Lin, Acc. Chem. Res., 2020, 202 Y. Guo, G. Hu, Y. Xia, H. Li, J. Yuan, J. Zhang, Y. Chen,
53, 1739–1748. H. Guo, Y. Yang, Y. Wang and Z. Deng, Bioact. Mater., 2022,
179 K. Ni, G. Lan, N. Guo, A. Culbert, T. Luo, T. Wu, R. R. 16, 204–217.
Weichselbaum and W. Lin, Sci. Adv., 2020, 6, eabb5223. 203 Z. Quinn, W. Mao, Y. Xia, R. John and Y. Wan, Bioact.
180 K. Ni, G. Lan, C. Chan, B. Quigley, K. Lu, T. Aung, N. Guo, Mater., 2021, 6, 749–756.
P. La Riviere, R. R. Weichselbaum and W. Lin, Nat. Com- 204 M. D’Huyvetter, J. Vos, V. Caveliers, I. Vaneycken,
mun., 2018, 9, 2351. J. Heemskerk, F. P. Duhoux, C. Fontaine, M. Vanhoeij,

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

A. D. Windhorst, F. V. Aa, N. H. Hendrikse, J. L. E. Eersels, 225 B. Q. Chen, Y. Zhao, Y. Zhang, Y. J. Pan, H. Y. Xia,
H. Everaert, P. Gykiere, N. Devoogdt, G. Raes, T. Lahoutte R. K. Kankala, S. B. Wang, G. Liu and A. Z. Chen, Bioact.
and M. Keyaerts, J. Nucl. Med., 2021, 62, 1097–1105. Mater., 2023, 21, 1–19.
205 J. Garousi, E. von Witting, J. Borin, A. Vorobyeva, M. Altai, 226 Q. Chen, M. Chen and Z. Liu, Chem. Soc. Rev., 2019, 48,
O. Vorontsova, M. W. Konijnenberg, M. Oroujeni, 5506–5526.
A. Orlova, V. Tolmachev and S. Hober, Biomaterials, 2021, 227 G. Kelly, J. J. Milligan, E. M. Mastria, S. Kim, S. R. Zelenetz,
266, 120381. J. Dobbins, L. Y. Cai, X. Li, S. K. Nair and A. Chilkoti,
206 M. E. Kieffer, A. M. Patel, S. A. Hollingsworth and J. Controlled Release, 2022, 343, 267–276.
W. M. Seganish, Expert Opin. Ther. Pat., 2020, 30, 825–845. 228 J. Xu, R. Saklatvala, S. Mittal, S. Deshmukh and
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

207 Y. Y. Janjigian, A. Kawazoe, P. Yañez, N. Li, S. Lonardi, A. Procopio, Adv. Sci., 2020, 7, 1903394.
O. Kolesnik, O. Barajas, Y. Bai, L. Shen, Y. Tang, 229 A. B. Cook and P. Decuzzi, ACS Nano, 2021, 15, 2068–2098.
L. S. Wyrwicz, J. Xu, K. Shitara, S. Qin, E. Van Cutsem, 230 C. Zhang and K. Pu, Chem. Soc. Rev., 2020, 49, 4234–4253.
J. Tabernero, L. Li, S. Shah, P. Bhagia and H. C. Chung, 231 H. Ding, P. Tan, S. Fu, X. Tian, H. Zhang, X. Ma, Z. Gu and
Nature, 2021, 600, 727–730. K. Luo, J. Controlled Release, 2022, 348, 206–238.
208 Z. Tang, Y. Xiao, N. Kong, C. Liu, W. Chen, X. Huang, 232 M. Liu, L. Huang, W. Zhang, X. Wang, Y. Geng, Y. Zhang,
D. Xu, J. Ouyang, C. Feng, C. Wang, J. Wang, H. Zhang and L. Wang, W. Zhang, Y. J. Zhang, S. Xiao, Y. Bao, M. Xiong
W. Tao, Acta Pharm. Sin. B, 2021, 11, 3447–3464. and J. Wang, Nat. Nanotechnol., 2022, 17, 541–551.
209 G. Peng, T. Duan, M. Guo, Y. Xue, C. Chen, Y. Li, K. Leifer 233 Y. Li, M. Jiang, Z. Deng, S. Zeng and J. Hao, Adv. Sci., 2021,
and B. Fadeel, Nanoscale, 2021, 13, 13072–13084. 8, 2004391.
210 M. Guo, J. Liu, X. Chen, Z. You, F. Gao, T. Liu, J. Ren, J. Liu, 234 R. Misra, K. Sarkar, J. Lee, V. J. Pizzuti, D. S. Lee,
Z. Xiong and Y. Liu, Nano Today, 2022, 45, 101543. M. P. Currie, S. E. Torregrosa-Allen, D. E. Long,
211 D. Huo, X. Jiang and Y. Hu, Adv. Mater., 2020, 32, 1904337. G. A. Durm and M. P. Langer, J. Controlled Release, 2019,
212 L. Jia, X. Li, H. Liu, J. Xia, X. Shi and M. Shen, Nano Today, 303, 237–252.
2021, 36, 101022. 235 W. Fan, W. Tang, J. Lau, Z. Shen, J. Xie, J. Shi and X. Chen,
213 J. Li, X. Jiang, H. Li, M. Gelinsky and Z. Gu, Adv. Mater., Adv. Mater., 2019, 31, 1806381.
2021, 33, 2004172. 236 T. Li, S. Pan, S. Gao, W. Xiang, C. Sun, W. Cao and H. Xu,
214 Y. Wang, J. Wang, D. Zhu, Y. Wang, G. Qing, Y. Zhang, Angew. Chem., Int. Ed., 2020, 59, 2700–2704.
X. Liu and X. J. Liang, Acta Pharm. Sin. B, 2021, 11, 237 Y. Yu, Z. Feng, J. Liu, X. Hou, X. Zhou, J. Gao, W. Wang,
886–902. Y. Zhang, G. Li and J. Liu, ACS Omega, 2021, 6,
215 T. Song, Y. Xia, Y. Du, M. W. Chen, H. Qing and G. Ma, Adv. 19445–19457.
Mater., 2021, 33, 2100106. 238 L. Zhang, W. Wang and S. Wang, Expert Rev. Vaccines,
216 V. T. Cong, W. Wang, R. D. Tilley, G. Sharbeen, 2015, 14, 1509–1523.
P. A. Phillips, K. Gaus and J. J. Gooding, Adv. Funct. Mater., 239 A. Marabelle, R. Andtbacka, K. Harrington, I. Melero,
2021, 31, 2007880. R. Leidner, T. de Baere, C. Robert, P. A. Ascierto,
217 M. Zhang, X. Chen, C. Li and X. Shen, J. Controlled Release, J.-F. Baurain and M. Imperiale, Ann. Oncol., 2018, 29,
2020, 319, 46–62. 2163–2174.
218 J. W. Breslin, Y. Yang, J. P. Scallan, R. S. Sweat, 240 O. M. Feeney, D. H. Brundel, N. L. Trevaskis, E. Cao,
S. P. Adderley and W. L. Murfee, Compr. Physiol., 2018, 9, L. M. Kaminskas and C. J. Porter, Adv. Drug Delivery Rev.,
207–299. 2020, 160, 115–135.
219 Y. Yang, J. Xu, Y. Sun, L. Mo, B. Liu, X. Pan, Z. Liu and 241 L. Zhang, X. Sun, Y. Jia, X. Liu, M. Dong, Z. P. Xu and
W. Tan, J. Am. Chem. Soc., 2021, 143, 8391–8401. R. Liu, Nano Today, 2020, 35, 100923.
220 Y. Kim, H. J. Jung, Y. Lee, S. Koo, R. Thangam, W. Y. Jang, 242 Y. Wang, W. Deng, N. Li, S. Neri, A. Sharma, W. Jiang and
S. Y. Kim, S. Park, S. Lee, G. Bae, K. D. Patel, Q. Wei, S. H. Lin, Front. Pharmacol., 2018, 9, 185.
K. B. Lee, R. Paulmurugan, W. K. Jeong, T. Hyeon, D. Kim 243 S. Ghosh, A. Javia, S. Shetty, D. Bardoliwala, K. Maiti,
and H. Kang, J. Am. Chem. Soc., 2022, 144, 5769–5783. S. Banerjee, A. Khopade, A. Misra, K. Sawant and
221 R. Sharma, K. Liaw, A. Sharma, A. Jimenez, M. Chang, S. Bhowmick, J. Controlled Release, 2021, 337, 27–58.
S. Salazar, I. Amlani, S. Kannan and R. M. Kannan, 244 A. Bassez, H. Vos, L. Van Dyck, G. Floris, I. Arijs,
J. Controlled Release, 2021, 337, 179–192. C. Desmedt, B. Boeckx, M. V. Bempt, I. Nevelsteen and
222 A. E. Barberio, S. G. Smith, S. Correa, C. Nguyen, B. Nhan, K. Lambein, Nat. Med., 2021, 27, 820–832.
M. Melo, T. Tokatlian, H. Suh, D. J. Irvine and 245 H. Jung, H. S. Kim, J. Y. Kim, J. M. Sun, J. S. Ahn, M. J. Ahn,
P. T. Hammond, ACS Nano, 2020, 14, 11238–11253. K. Park, M. Esteller, S. H. Lee and J. K. Choi, Nat. Commun.,
223 W. Yang, H. Deng, S. Zhu, J. Lau, R. Tian, S. Wang, Z. Zhou, 2019, 10, 4278.
G. Yu, L. Rao, L. He, Y. Ma and X. Chen, Sci. Adv., 2020, 246 H. Lam, L. K. McNeil, H. Starobinets, V. L. DeVault,
6, eabd1631. R. B. Cohen, P. Twardowski, M. L. Johnson, M. L.
224 Y. Wang, S. Li, X. Wang, Q. Chen, Z. He, C. Luo and J. Sun, Gillison, M. N. Stein, U. N. Vaishampayan, A. P. DeCillis,
Biomaterials, 2021, 271, 120737. J. J. Foti, V. Vemulapalli, E. Tjon, K. Ferber,

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

D. B. DeOliveira, W. Broom, P. Agnihotri, E. M. Jaffee, 265 W. J. Smith, G. Wang, H. Gaikwad, V. P. Vu, E. Groman,
K. K. Wong, C. G. Drake, P. M. Carroll, T. A. Davis and D. W. Bourne and D. Simberg, ACS Nano, 2018, 12,
J. B. Flechtner, Cancer Discovery, 2021, 11, 696–713. 12523–12532.
247 M. Gromeier, M. C. Brown, G. Zhang, X. Lin, Y. Chen, 266 S. Jia, S. Dong, H. Liu, H. Yu, Z. Chen, S. Wang, W. Li,
Z. Wei, N. Beaubier, H. Yan, Y. He, A. Desjardins, R. Peng, F. Li, Q. Jiang and J. Liu, Biomater. Sci., 2022, 10,
J. E. Herndon, 2nd, F. S. Varn, R. G. Verhaak, J. Zhao, 3309–3322.
D. P. Bolognesi, A. H. Friedman, H. S. Friedman, 267 H. K. Angell, D. Bruni, J. C. Barrett, R. Herbst and J. Galon,
F. McSherry, A. M. Muscat, E. S. Lipp, S. K. Nair, Clin. Cancer Res., 2020, 26, 332–339.
M. Khasraw, K. B. Peters, D. Randazzo, J. H. Sampson, 268 T. S. C. Ng, V. Gunda, R. Li, M. Prytyskach, Y. Iwamoto,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

R. E. McLendon, D. D. Bigner and D. M. Ashley, Nat. R. H. Kohler, S. Parangi, R. Weissleder and M. A. Miller,
Commun., 2021, 12, 352. Radiology, 2021, 298, 123–132.
248 M. V. Guerin, V. Finisguerra, B. J. Van den Eynde, 269 R. Tian, C. Ke, L. Rao, J. Lau and X. Chen, Adv. Drug
N. Bercovici and A. Trautmann, Elife, 2020, 9, e50740. Delivery Rev., 2020, 161–162, 145–160.
249 E. Schültke, S. Bayat, S. Bartzsch, E. Bräuer-Krisch, 270 W. Wei, Z. T. Rosenkrans, J. Liu, G. Huang, Q. Y. Luo and
V. Djonov, S. Fiedler, C. Fernandez-Palomo, F. Jaekel, W. Cai, Chem. Rev., 2020, 120, 3787–3851.
P. Pellicioli, V. Trappetti and G. Hildebrandt, Int. 271 K. Herrmann, M. Schwaiger, J. S. Lewis, S. B. Solomon,
J. Radiat. Oncol. Biol. Phys., 2021, 110, 521–525. B. J. McNeil, M. Baumann, S. S. Gambhir, H. Hricak and
250 H. Watanabe, K. Numata, T. Ito, K. Takagi and R. Weissleder, Lancet Oncol., 2020, 21, 146–156.
A. Matsukawa, Shock, 2004, 22, 460–466. 272 Y. Zhao, T. Zhang, Y. Wang, D. Lu, J. Du, X. Feng, H. Zhou,
251 S. Zaaijer, S. C. Groen and N. E. Sanjana, Nat. Biotechnol., N. Liu, H. Zhu and S. Qin, Proc. Natl. Acad. Sci. U. S. A.,
2021, 39, 666–670. 2021, 118, e2010333118.
252 N. Tateshita, N. Miura, H. Tanaka, T. Masuda, S. Ohtsuki, 273 R. Cristescu, R. Mogg, M. Ayers, A. Albright, E. Murphy,
K. Tange, Y. Nakai, H. Yoshioka and H. Akita, J. Controlled J. Yearley, X. Sher, X. Q. Liu, H. Lu and M. Nebozhyn,
Release, 2019, 310, 36–46. Science, 2018, 362, eaar3593.
253 Z. Zhang, L. Liu, C. Ma, X. Cui, R. H. W. Lam and W. Chen, 274 T. A. Chan, M. Yarchoan, E. Jaffee, C. Swanton,
Small Methods, 2021, 5, 2100197. S. A. Quezada, A. Stenzinger and S. Peters, Ann. Oncol.,
254 P. De La Rochere, S. Guil-Luna, D. Decaudin, G. Azar, 2019, 30, 44–56.
S. S. Sidhu and E. Piaggio, Trends Immunol., 2018, 39, 275 C. Valero, M. Lee, D. Hoen, J. Wang, Z. Nadeem, N. Patel,
748–763. M. A. Postow, A. N. Shoushtari, G. Plitas and V. P. Bala-
255 C. Peres, A. I. Matos, L. I. F. Moura, R. C. Acúrcio, chandran, Nat. Genet., 2021, 53, 11–15.
B. Carreira, S. Pozzi, D. Vaskovich-Koubi, R. Kleiner, 276 H. F. Huang, H. Zhu, G. H. Li, Q. Xie, X. T. Yang, X. X. Xu,
R. Satchi-Fainaro and H. F. Florindo, Adv. Drug Delivery X. B. Tian, Y. K. Wan and Z. Yang, Bioconjugate Chem.,
Rev., 2021, 172, 148–182. 2019, 30, 2614–2623.
256 H.-Y. Kim, R. Li, T. S. Ng, G. Courties, C. B. Rodell, 277 W. Wei, Q. Liu, D. Jiang, H. Zhao, C. J. Kutyreff, J. W. Engle,
M. Prytyskach, R. H. Kohler, M. J. Pittet, M. Nahrendorf J. Liu and W. Cai, Adv. Sci., 2020, 7, 1903595.
and R. Weissleder, ACS Nano, 2018, 12, 12015–12029. 278 I. H. Song, M. S. Jeong, H. J. Hong, J. I. Shin, Y. S. Park,
257 G. Lv, X. Sun, L. Qiu, Y. Sun, K. Li, Q. Liu, Q. Zhao, S. Qin S.-K. Woo, B. S. Moon, K. I. Kim, Y. J. Lee and J. H. Kang,
and J. Lin, J. Nucl. Med., 2020, 61, 117–122. Clin. Cancer Res., 2019, 25, 6148–6159.
258 Z. Zhou, H. Deng, W. Yang, Z. Wang, L. Lin, 279 A. Bagaev, N. Kotlov, K. Nomie, V. Svekolkin, A. Gafurov,
J. Munasinghe, O. Jacobson, Y. Liu, L. Tang, Q. Ni, O. Isaeva, N. Osokin, I. Kozlov, F. Frenkel and
F. Kang, Y. Liu, G. Niu, R. Bai, C. Qian, J. Song and O. Gancharova, Cancer Cell, 2021, 39, 845–865.
X. Chen, Nat. Commun., 2020, 11, 3032. 280 Y. Zhang and L. Chen, JAMA Oncol., 2016, 2, 1403–1404.
259 H. Yu, H. Guo, H. Zu, H. Ding, S. Lin, Y. Wang, L. W. Zhang 281 E. Borcoman, Y. Kanjanapan, S. Champiat, S. Kato,
and Y. Wang, Aggregate, 2022, 3, 194. V. Servois, R. Kurzrock, S. Goel, P. Bedard and C. Le
260 Y. Wang, J. Chen, R. Duan, R. Gu, W. Wang, J. Wu, H. Lian, Tourneau, Ann. Oncol., 2019, 30, 385–396.
Y. Hu and A. Yuan, Adv. Mater., 2022, 34, 2109726. 282 Q. Fu, H. Feng, L. Su, X. Zhang, L. Liu, F. Fu, H. Yang and
261 Z. Huang, Y. Wang, D. Yao, J. Wu, Y. Hu and A. Yuan, Nat. J. Song, Angew. Chem., Int. Ed., 2022, 61, 202112237.
Commun., 2021, 12, 145. 283 S. B. Goldberg, A. Narayan, A. J. Kole, R. H. Decker, J. Teysir,
262 Y. Yang, B. Liu, Y. Liu, J. Chen, Y. Sun, X. Pan, J. Xu, S. Xu, N. J. Carriero, A. Lee, R. Nemati, S. K. Nath, S. M. Mane,
Z. Liu and W. Tan, Nano Lett., 2022, 22, 2826–2834. Y. Deng, N. Sukumar, D. Zelterman, D. J. Boffa, K. Politi,
263 X. Zhu, X. Wang, B. Li, Y. Zhang, Y. Chen, W. Zhang, S. N. Gettinger, L. D. Wilson, R. S. Herbst and A. A. Patel,
Y. Wang, W. Zhai, Z. Liu, S. Liu, J. Sun, Z. Chen and Y. Gao, Clin. Cancer Res., 2018, 24, 1872–1880.
Small, 2022, 18, 2107001. 284 E. Sanz-Garcia, E. Zhao, S. V. Bratman and L. L. Siu, Sci.
264 Y. Chen, S. Liu, P. Gao, W. Pan, M. Shi, J. Wang, N. Li and Adv., 2022, 8, eabi8618.
B. Tang, Sci. China: Mater., 2022, DOI: 10.1007/s40843-022- 285 Z. Dong, X. Xue, H. Liang, J. Guan and L. Chang, Anal.
2140-7. Chem., 2021, 93, 1855–1865.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

286 G. De Rubis, S. Rajeev Krishnan and M. Bebawy, Trends 305 M. Fusciello, F. Fontana, S. Tähtinen, C. Capasso, S. Feola,
Pharmacol. Sci., 2019, 40, 172–186. B. Martins, J. Chiaro, K. Peltonen, L. Ylösmäki,
287 Y. Liu, J. Zugazagoitia, F. S. Ahmed, B. S. Henick, E. Ylösmäki, F. Hamdan, O. K. Kari, J. Ndika, H. Alenius,
S. N. Gettinger, R. S. Herbst, K. A. Schalper and A. Urtti, J. T. Hirvonen, H. A. Santos and V. Cerullo, Nat.
D. L. Rimm, Clin. Cancer Res., 2020, 26, 970–977. Commun., 2019, 10, 5747.
288 F. Bensch, E. L. van der Veen, M. N. Lub-de Hooge, 306 N. Gong, Y. Zhang, X. Teng, Y. Wang, S. Huo, G. Qing,
A. Jorritsma-Smit, R. Boellaard, I. C. Kok, S. F. Oosting, Q. Ni, X. Li, J. Wang, X. Ye, T. Zhang, S. Chen, Y. Wang,
C. P. Schröder, T. J. N. Hiltermann, A. J. van der Wekken, J. Yu, P. C. Wang, Y. Gan, J. Zhang, M. J. Mitchell, J. Li and
H. J. M. Groen, T. C. Kwee, S. G. Elias, J. A. Gietema, X. J. Liang, Nat. Nanotechnol., 2020, 15, 1053–1064.
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

S. S. Bohorquez, A. de Crespigny, S. P. Williams, 307 J. Xu, J. Lv, Q. Zhuang, Z. Yang, Z. Cao, L. Xu, P. Pei,
C. Mancao, A. H. Brouwers, B. M. Fine and E. G. E. de C. Wang, H. Wu, Z. Dong, Y. Chao, C. Wang, K. Yang,
Vries, Nat. Med., 2018, 24, 1852–1858. R. Peng, Y. Cheng and Z. Liu, Nat. Nanotechnol., 2020, 15,
289 E. B. Ehlerding, H. J. Lee, T. E. Barnhart, D. Jiang, L. Kang, 1043–1052.
D. G. McNeel, J. W. Engle and W. Cai, Bioconjugate Chem., 308 X. Xiong, J. Zhao, J. Pan, C. Liu, X. Guo and S. Zhou, Nano
2019, 30, 1434–1441. Lett., 2021, 21, 8418–8425.
290 L. Tian, M. Shao, Y. Gong, Y. Chao, T. Wei, K. Yang, 309 M. Saxena, S. H. van der Burg, C. J. M. Melief and
Q. Chen and Z. Liu, Sci. China: Chem., 2022, 65, 574–583. N. Bhardwaj, Nat. Rev. Cancer, 2021, 21, 360–378.
291 Y. C. Ou, X. Wen, C. A. Johnson, D. Shae, O. D. Ayala, 310 M. Luo, Z. Liu, X. Zhang, C. Han, L. Z. Samandi, C. Dong,
J. A. Webb, E. C. Lin, R. C. DeLapp, K. L. Boyd, A. B. D. Sumer, J. Lea, Y. X. Fu and J. Gao, J. Controlled
Richmond, A. Mahadevan-Jansen, M. Rafat, J. T. Wilson, Release, 2019, 300, 154–160.
J. M. Balko, M. N. Tantawy, A. E. Vilgelm and R. Bardhan, 311 J. Wilhelm, M. Quiñones-Pérez, J. Wang, X. Wang,
ACS Nano, 2020, 14, 651–663. V. S. Basava and J. Gao, J. Controlled Release, 2021, 329,
292 L. Devkota, Z. Starosolski, C. H. Rivas, I. Stupin, 353–360.
A. Annapragada, K. B. Ghaghada and R. Parihar, Sci. 312 R. B. Patel, M. Ye, P. M. Carlson, A. Jaquish, L. Zangl,
Adv., 2020, 6, eaba6156. B. Ma, Y. Wang, I. Arthur, R. Xie, R. J. Brown, X. Wang,
293 T. Li, D. Chen, H. Liu, Y. Tao, X. He, S. Zang, J. Li, L. Zhang, R. Sriramaneni, K. Kim, S. Gong and Z. S. Morris, Adv.
M. Li, J. Liu and Q. He, Nanoscale, 2022, 14, 13098–13112. Mater., 2019, 31, 1902626.
294 G. Delfanti, F. Cortesi, A. Perini, G. Antonini, L. Azzimonti, 313 X. Li, X. Wang, A. Ito and N. M. Tsuji, Nat. Commun., 2020,
C. de Lalla, C. Garavaglia, M. L. Squadrito, M. Fedeli, 11, 3858.
M. Consonni, S. Sesana, F. Re, H. Shen, P. Dellabona 314 H. Zhao, J. Xu, Y. Li, X. Guan, X. Han, Y. Xu, H. Zhou,
and G. Casorati, Sci. Immunol., 2022, 7, eabn6563. R. Peng, J. Wang and Z. Liu, ACS Nano, 2019, 13,
295 E. Loeuillard, J. Yang, E. Buckarma, J. Wang, Y. Liu, 13127–13135.
C. Conboy, K. D. Pavelko, Y. Li, D. O’Brien, C. Wang, 315 C. Wan, Y. Sun, Y. Tian, L. Lu, X. Dai, J. Meng, J. Huang,
R. P. Graham, R. L. Smoot, H. Dong and S. Ilyas, J. Clin. Q. He, B. Wu, Z. Zhang, K. Jiang, D. Hu, G. Wu, J. F. Lovell,
Invest., 2020, 130, 5380–5396. H. Jin and K. Yang, Sci. Adv., 2020, 6, eaay9789.
296 H. J. Park, K. W. Kim, J. Pyo, C. H. Suh, S. Yoon, H. Hatabu 316 B. Pineda, F. J. Sánchez Garcı́a, N. K. Olascoaga, V. Pérez de
and M. Nishino, Radiology, 2020, 297, 87–96. la Cruz, A. Salazar, S. Moreno-Jiménez, N. Hernández
297 L. B. Costa, M. A. Queiroz, F. G. Barbosa, R. F. Nunes, E. C. Pedro, A. Márquez-Navarro, A. Ortiz Plata and J. Sotelo,
Zaniboni, M. M. Ruiz, D. Jardim, J. F. Gomes Marin, G. G. Mol. Ther., 2019, 27, 1612–1620.
Cerri and C. A. Buchpiguel, Radiographics, 2021, 41, 120–143. 317 Y. Chao, L. Xu, C. Liang, L. Feng, J. Xu, Z. Dong, L. Tian,
298 B. Kas, H. Talbot, R. Ferrara, C. Richard, J. P. Lamarque, X. Yi, K. Yang and Z. Liu, Nat. Biomed. Eng., 2018, 2,
S. Pitre-Champagnat, D. Planchard, C. Balleyguier, 611–621.
B. Besse, L. Mezquita, N. Lassau and C. Caramella, JAMA 318 J. Zhang, M. Yang, X. Fan, M. Zhu, Y. Yin, H. Li, J. Chen,
Oncol., 2020, 6, 1039–1046. S. Qin, H. Zhang, K. Zhang and F. Yu, J. Nanobiotechnol.,
299 M. R. Sheen and S. Fiering, Wiley Interdiscip. Rev.: 2022, 20, 103.
Nanomed. Nanobiotechnol., 2019, 11, e1524. 319 G. Kroemer, L. Galluzzi, O. Kepp and L. Zitvogel, Annu. Rev.
300 L. Qin, H. Zhang, Y. Zhou, C. S. Umeshappa and H. Gao, Immunol., 2013, 31, 51–72.
Small, 2021, 17, 2006000. 320 Z. Li, X. Lai, S. Fu, L. Ren, H. Cai, H. Zhang, Z. Gu, X. Ma
301 W. H. Li and Y. M. Li, Chem. Rev., 2020, 120, 11420–11478. and K. Luo, Adv. Sci., 2022, 9, 2201734.
302 S. Cuzzubbo, S. Mangsbo, D. Nagarajan, K. Habra, A. G. 321 Q. Li, C. Chen, J. Kong, L. Li, J. Li and Y. Huang, Acta
Pockley and S. E. B. McArdle, Front. Immunol., 2020, 11, 615240. Pharm. Sin. B, 2022, 12, 2533–2549.
303 E. B. Golden, A. E. Marciscano and S. C. Formenti, Int. 322 R. Mezzapelle, M. E. Bianchi and M. P. Crippa, Cell Death
J. Radiat. Oncol. Biol. Phys., 2020, 108, 891–898. Dis., 2021, 12, 869.
304 P. H. Lizotte, A. M. Wen, M. R. Sheen, J. Fields, 323 A. Banstola, K. Poudel, J. O. Kim, J. H. Jeong and S. Yook,
P. Rojanasopondist, N. F. Steinmetz and S. Fiering, Nat. J. Controlled Release, 2021, 337, 505–520.
Nanotechnol., 2016, 11, 295–303. 324 D. R. Green, Cell, 2019, 177, 1094–1107.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

325 S. Sen, M. Won, M. S. Levine, Y. Noh, A. C. Sedgwick, 346 X. Liu, D. Wang, P. Zhang and Y. Li, Nano Today, 2019,
J. S. Kim, J. L. Sessler and J. F. Arambula, Chem. Soc. Rev., 29, 100801.
2022, 51, 1212–1233. 347 S. Nizzero, H. Shen, M. Ferrari and B. Corradetti, Trends
326 A. D. Garg, L. Vandenberk, C. Koks, T. Verschuere, L. Boon, Cancer, 2020, 6, 40–48.
S. W. Van Gool and P. Agostinis, Sci. Transl. Med., 2016, 348 H. Wang, Y. Chao, H. Zhao, X. Zhou, F. Zhang, Z. Zhang,
8, 328ra27. Z. Li, J. Pan, J. Wang, Q. Chen and Z. Liu, ACS Nano, 2022,
327 M. Z. Jin and X. P. Wang, Front. Immunol., 2021, 16, 664–674.
12, 697964. 349 T. R. Cox, Nat. Rev. Cancer, 2021, 21, 217–238.
328 J. Li, Y. Luo and K. Pu, Angew. Chem., Int. Ed., 2021, 60, 350 A. K. Pearce and R. K. O’Reilly, Bioconjugate Chem., 2019,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

12682–12705. 30, 2300–2311.


329 J. Li, Y. Luo, Z. Zeng, D. Cui, J. Huang, C. Xu, L. Li, K. Pu 351 P. Mi, H. Cabral and K. Kataoka, Adv. Mater., 2020,
and R. Zhang, Nat. Commun., 2022, 13, 4032. 32, 1902604.
330 C. Zhang, Z. Zeng, D. Cui, S. He, Y. Jiang, J. Li, J. Huang 352 L. Meng, C. Wang, Y. Lu, G. Sheng, L. Yang, Z. Wu, H. Xu,
and K. Pu, Nat. Commun., 2021, 12, 2934. C. Han, Y. Lu and F. Han, ACS Appl. Mater. Interfaces, 2021,
331 S. Son, J. Kim, J. Kim, B. Kim, J. Lee, Y. Kim, M. Li, H. Kang 13, 11657–11671.
and J. S. Kim, Chem. Soc. Rev., 2022, 51, 8201–8215. 353 J. Wei, D. Wu, Y. Shao, B. Guo, J. Jiang, J. Chen, J. Zhang,
332 P. Pei, T. Liu, W. Shen, Z. Liu and K. Yang, Mater. Horiz., F. Meng and Z. Zhong, J. Controlled Release, 2022, 347,
2021, 8, 1348–1366. 68–77.
333 B. A. McDonald, S. Vedam, J. Yang, J. Wang, P. Castillo, 354 H. Qin, R. Zhao, Y. Qin, J. Zhu, L. Chen, C. Di, X. Han,
B. Lee, A. Sobremonte, S. Ahmed, Y. Ding, A. S. R. K. Cheng, Y. Zhang, Y. Zhao, J. Shi, G. J. Anderson, Y. Zhao
Mohamed, P. Balter, N. Hughes, D. Thorwarth, and G. Nie, Adv. Mater., 2021, 33, 2006007.
M. Nachbar, M. E. P. Philippens, C. H. J. Terhaard, D. Zips, 355 Y. Tang, X. Wang, J. Li, Y. Nie, G. Liao, Y. Yu and C. Li, ACS
S. Böke, M. J. Awan, J. Christodouleas and C. D. Fuller, Int. Nano, 2019, 13, 13015–13026.
J. Radiat. Oncol. Biol. Phys., 2021, 109, 1606–1618. 356 Y. Chu, L. Qian, Y. Ke, X. Feng, X. Chen, F. Liu, L. Yu,
334 P. Keall, D. T. Nguyen, R. O’Brien, E. Hewson, H. Ball, L. Zhang, Y. Tao and R. Xu, J. Nanobiotechnol., 2022,
P. Poulsen, J. Booth, P. Greer, P. Hunter, L. Wilton, 20, 190.
R. Bromley, J. Kipritidis, T. Eade, A. Kneebone, G. Hruby, 357 D. Zhu, Z. Liu, Y. Li, Q. Huang, L. Xia and K. Li, Biomater-
T. Moodie, A. Hayden, S. Turner, S. Arumugam, ials, 2021, 274, 120894.
M. Sidhom, N. Hardcastle, S. Siva, K. H. Tai, V. Gebski 358 K. Luo, Y. Lian, M. Zhang, H. Yu, G. Wang and J. Li, Chem.
and J. Martin, Int. J. Radiat. Oncol. Biol. Phys., 2020, 107, Eng. J., 2021, 412, 128659.
530–538. 359 V. Sheth, L. Wang, R. Bhattacharya, P. Mukherjee and
335 M. Zhu, M. Yang, J. Zhang, Y. Yin, X. Fan, Y. Zhang, S. Qin, S. Wilhelm, Adv. Funct. Mater., 2021, 31, 2007363.
H. Zhang and F. Yu, Front. Immunol., 2021, 12, 705361. 360 M. Izci, C. Maksoudian, B. B. Manshian and S. J. Soenen,
336 H. K. Birdi, A. Jirovec, S. Cortés-Kaplan, J. Werier, Chem. Rev., 2021, 121, 1746–1803.
C. Nessim, J. S. Diallo and M. Ardolino, J. Immunother. 361 F. Mpekris, C. Voutouri, J. W. Baish, D. G. Duda,
Cancer, 2021, 9, e001580. L. L. Munn, T. Stylianopoulos and R. K. Jain, Proc. Natl.
337 L. Xie, G. Wang, W. Sang, J. Li, Z. Zhang, W. Li, J. Yan, Acad. Sci. U. S. A., 2020, 117, 3728–3737.
Q. Zhao and Y. Dai, Biomaterials, 2021, 269, 120638. 362 Y. Huang, T. Stylianopoulos, D. G. Duda, D. Fukumura and
338 Y. Wang, N. Shen, Y. Wang, M. Li, W. Zhang, L. Fan, L. Liu, R. K. Jain, Cancer Res., 2013, 73, 7144–7146.
Z. Tang and X. Chen, Biomater. Sci., 2021, 9, 3019–3027. 363 Y. Sun, W. Chen, R. J. Torphy, S. Yao, G. Zhu, R. Lin,
339 G. Hou, J. Qian, M. Guo, W. Xu, J. Wang, Y. Wang and R. Lugano, E. N. Miller, Y. Fujiwara, L. Bian, L. Zheng,
A. Suo, Chem. Eng. J., 2022, 435, 134778. S. Anand, F. Gao, W. Zhang, S. E. Ferrara, A. E. Goodspeed,
340 Z. Huang, D. Yao, Q. Ye, H. Jiang, R. Gu, C. Ji, J. Wu, Y. Hu A. Dimberg, X. J. Wang, B. H. Edil, C. C. Barnett,
and A. Yuan, ACS Nano, 2021, 15, 8450–8465. R. D. Schulick, L. Chen and Y. Zhu, Sci. Transl. Med.,
341 Y. Wang, Y. Ding, D. Yao, H. Dong, C. Ji, J. Wu, Y. Hu and 2021, 13, eabc8922.
A. Yuan, Small, 2021, 17, 2006231. 364 N. Huang, Y. Liu, Y. Fang, S. Zheng, J. Wu, M. Wang,
342 J. R. Cubillos-Ruiz, S. E. Bettigole and L. H. Glimcher, Cell, W. Zhong, M. Shi, M. Xing and W. Liao, ACS Nano, 2020,
2017, 168, 692–706. 14, 7940–7958.
343 B. Choi, H. Choi, B. Yu and D. H. Kim, ACS Nano, 2020, 14, 365 Y. Yang, J. Tang, M. Zhang, Z. Gu, H. Song, Y. Yang and
13115–13126. C. Yu, Nano Lett., 2019, 19, 7750–7759.
344 B. V. Lima, M. J. Oliveira, M. A. Barbosa, R. M. Gonçalves 366 R. Chen, L. Huang and K. Hu, Acta Pharm. Sin. B, 2020, 10,
and F. Castro, Mater. Today Adv., 2022, 15, 100252. 2140–2155.
345 H. Yu, Y. Yang, T. Jiang, X. Zhang, Y. Zhao, G. Pang, 367 J. Zhao, H. Wang, C. H. Hsiao, D. S. Chow, E. J. Koay,
Y. Feng, S. Zhang, F. Wang, Y. Wang, Y. Wang and Y. Kang, X. Wen, Q. Huang, Y. Ma, J. A. Bankson,
L. W. Zhang, ACS Appl. Mater. Interfaces, 2019, 11, S. E. Ullrich, W. Overwijk, A. Maitra, D. Piwnica-Worms,
27536–27547. J. B. Fleming and C. Li, Biomaterials, 2018, 159, 215–228.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

368 L. Zhao, B. Niu, J. Fang, Y. Pang, S. Li, C. Xie, L. Sun, 388 L. Luo, Y. Qi, H. Zhong, S. Jiang, H. Zhang, H. Cai, Y. Wu,
X. Zhang, Z. Guo, Q. Lin and H. Chen, J. Nucl. Med., 2022, Z. Gu, Q. Gong and K. Luo, Acta Pharm. Sin. B, 2022, 12,
63, 862–868. 424–436.
369 K. Hu, J. Li, L. Wang, Y. Huang, L. Li, S. Ye, Y. Han, 389 J. Huang, C. Zhuang, J. Chen, X. Chen, X. Li, T. Zhang,
S. Huang, H. Wu, J. Su and G. Tang, Acta Pharm. Sin. B, B. Wang, Q. Feng, X. Zheng, M. Gong, Q. Gong, K. Xiao,
2022, 12, 867–875. K. Luo and W. Li, Adv. Mater., 2022, 34, 2201516.
370 G. Sharbeen, J. A. McCarroll, A. Akerman, C. Kopecky, 390 J. Tu, K. Tu, H. Xu, L. Wang, X. Yuan, X. Qin, L. Kong,
J. Youkhana, J. Kokkinos, J. Holst, C. Boyer, M. Erkan, Q. Chu and Z. Zhang, Eur. J. Pharm. Biopharm., 2020, 150,
D. Goldstein, P. Timpson, T. R. Cox, B. A. Pereira, 96–107.
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

J. L. Chitty, S. K. Fey, A. K. Najumudeen, A. D. Campbell, 391 D. Candas-Green, B. Xie, J. Huang, M. Fan, A. Wang,
O. J. Sansom, R. M. C. Ignacio, S. Naim, J. Liu, N. Russia, C. Menaa, Y. Zhang, L. Zhang, D. Jing, S. Azghadi,
J. Lee, A. Chou, A. Johns, A. J. Gill, E. Gonzales-Aloy, W. Zhou, L. Liu, N. Jiang, T. Li, T. Gao, C. Sweeney,
V. Gebski, Y. F. Guan, M. Pajic, N. Turner, M. V. Apte, R. Shen, T. Y. Lin, C. X. Pan, O. M. Ozpiskin,
T. P. Davis, J. P. Morton, K. S. Haghighi, J. Kasparian, G. Woloschak, D. J. Grdina, A. T. Vaughan, J. M. Wang,
B. J. McLean, Y. F. Setargew and P. A. Phillips, Cancer Res., S. Xia, A. M. Monjazeb, W. J. Murphy, L. Q. Sun,
2021, 81, 3461–3479. H. W. Chen, K. S. Lam, R. R. Weichselbaum and J. J. Li,
371 F. Mpekris, M. Panagi, C. Voutouri, J. D. Martin, R. Samuel, Nat. Commun., 2020, 11, 4591.
S. Takahashi, N. Gotohda, T. Suzuki, P. Papageorgis, 392 C. M. Jackson, J. Choi and M. Lim, Nat. Immunol., 2019, 20,
P. Demetriou, C. Pierides, L. Koumas, P. Costeas, 1100–1109.
M. Kojima, G. Ishii, A. Constantinidou, K. Kataoka, 393 L. Feng, L. Yang, L. Li, J. Xiao, N. Bie, C. Xu, J. Zhou, H. Liu,
H. Cabral and T. Stylianopoulos, Adv. Sci., 2021, 8, 2001917. L. Gan and Y. Wu, Nano Res., 2022, 15, 593–602.
372 X. Liu, N. Ye, S. Liu, J. Guan, Q. Deng, Z. Zhang, C. Xiao, 394 L. You, W. Wu, X. Wang, L. Fang, V. Adam, E. Nepovimova,
Z. Y. Ding, B. X. Zhang, X. P. Chen, Z. Li and X. Yang, Adv. Q. Wu and K. Kuca, Med. Res. Rev., 2021, 41, 1622–1643.
Sci., 2021, 8, 2100233. 395 K. Roehle, L. Qiang, K. S. Ventre, D. Heid, L. R. Ali,
373 A. M. Buckley, N. Lynam-Lennon, H. O’Neill and P. Lenehan, M. Heckler, S. J. Crowley, C. T. Stump,
J. O’Sullivan, Nat. Rev. Gastroenterol. Hepatol., 2020, 17, G. Ro, A. Godicelj, A. M. Bhuiyan, A. Yang, M. Quiles Del
298–313. Rey, T. Biary, A. M. Luoma, P. T. Bruck, J. F. Tegethoff,
374 M. P. Krafft, Curr. Opin. Pharmacol., 2020, 53, 117–125. S. L. Nopper, J. Li, K. T. Byrne, M. Pelletier,
375 Z. Wang, X. Gong, J. Li, H. Wang, X. Xu, Y. Li, X. Sha and K. W. Wucherpfennig, B. Z. Stanger, J. J. Akin,
Z. Zhang, ACS Nano, 2021, 15, 5405–5419. J. D. Mancias, J. Agudo, M. Dougan and S. K. Dougan,
376 C. Zhang, L. Yan, Z. Gu and Y. Zhao, Chem. Sci., 2019, 10, Sci. Transl. Med., 2021, 13, eabf5058.
6932–6943. 396 J. Yu, M. D. Green, S. Li, Y. Sun, S. N. Journey, J. E. Choi,
377 Z. Zhang, J. Yang, Q. Min, C. Ling, D. Maiti, J. Xu, L. Qin S. M. Rizvi, A. Qin, J. J. Waninger, X. Lang, Z. Chopra, I. El
and K. Yang, Small, 2019, 15, 1803703. Naqa, J. Zhou, Y. Bian, L. Jiang, A. Tezel, J. Skvarce,
378 W. Sang, L. Xie, G. Wang, J. Li, Z. Zhang, B. Li, S. Guo, R. K. Achar, M. Sitto, B. S. Rosen, F. Su, S. P. Narayanan,
C. X. Deng and Y. Dai, Adv. Sci., 2021, 8, 2003338. X. Cao, S. Wei, W. Szeliga, L. Vatan, C. Mayo, M. A. Morgan,
379 X. Dai, J. Ruan, Y. Guo, Z. Sun, J. Liu, X. Bao, H. Zhang, C. A. Schonewolf, K. Cuneo, I. Kryczek, V. T. Ma, C. D. Lao,
Q. Li, C. Ye and X. Wang, Chem. Eng. J., 2021, 422, 130109. T. S. Lawrence, N. Ramnath, F. Wen, A. M. Chinnaiyan,
380 X. Song, J. Xu, C. Liang, Y. Chao, Q. Jin, C. Wang, M. Chen M. Cieslik, A. Alva and W. Zou, Nat. Med., 2021, 27,
and Z. Liu, Nano Lett., 2018, 18, 6360–6368. 152–164.
381 Q. Chen, J. Chen, Z. Yang, J. Xu, L. Xu, C. Liang, X. Han and 397 M. De Martino, O. Padilla, C. Daviaud, C.-C. Wu,
Z. Liu, Adv. Mater., 2019, 31, 1802228. R. D. Gartrell and C. Vanpouille-Box, Front. Oncol., 2021,
382 L. Tian, Y. Wang, L. Sun, J. Xu, Y. Chao, K. Yang, S. Wang 11, 671044.
and Z. Liu, Matter, 2019, 1, 1061–1076. 398 C. Groth, X. Hu, R. Weber, V. Fleming, P. Altevogt, J. Utikal
383 Y. Duo, Q. Liu, D. Zhu, B. Zhang, G. Luo, F. B. Wang, and V. Umansky, Brit. J. Cancer, 2019, 120, 16–25.
J. H. Chen and Y. Cao, Chem. Eng. J., 2022, 429, 132328. 399 M. Oshi, Y. Tokumaru, M. Asaoka, L. Yan, V. Satyananda,
384 Z. Yang, D. Gao, X. Guo, L. Jin, J. Zheng, Y. Wang, S. Chen, R. Matsuyama, N. Matsuhashi, M. Futamura, T. Ishikawa,
X. Zheng, L. Zeng, M. Guo, X. Zhang and Z. Tian, ACS K. Yoshida, I. Endo and K. Takabe, Sci. Rep., 2020, 10, 16554.
Nano, 2020, 14, 17442–17457. 400 H. Grégoire, L. Roncali, A. Rousseau, M. Chérel,
385 M. Shi, J. Zhang, Y. Wang, C. Peng, H. Hu, M. Qiao, X. Zhao Y. Delneste, P. Jeannin, F. Hindré and E. Garcion, Front.
and D. Chen, Biomaterials, 2022, 283, 121448. Pharmacol., 2020, 11, 368.
386 N. Hasan, J. Lee, D. Kwak, H. Kim, A. Saparbayeva, 401 Z. Li, Y. Ding, J. Liu, J. Wang, F. Mo, Y. Wang, T. J. Chen-
H. J. Ahn, I. S. Yoon, M. S. Kim, Y. Jung and J. W. Yoo, Mayfield, P. M. Sondel, S. Hong and Q. Hu, Nat. Commun.,
Carbohydr. Polym., 2021, 270, 118387. 2022, 13, 1845.
387 T. Kim, J. Suh, J. Kim and W. J. Kim, Adv. Sci., 2022, 402 X. Dai, L. Ren, M. Liu, H. Cai, H. Zhang, Q. Gong, Z. Gu
9, 2101935. and K. Luo, Nano Today, 2021, 39, 101163.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

403 N. T. Trac and E. J. Chung, Bioact. Mater., 2020, 5, 92–101. 423 Y. Zhu, B. L. Knolhoff, M. A. Meyer, T. M. Nywening,
404 C. Ngambenjawong, H. H. Gustafson and S. H. Pun, Adv. B. L. West, J. Luo, A. Wang-Gillam, S. P. Goedegebuure,
Drug Delivery Rev., 2017, 114, 206–221. D. C. Linehan and D. G. DeNardo, Cancer Res., 2014, 74,
405 L. Tian, X. Yi, Z. Dong, J. Xu, C. Liang, Y. Chao, Y. Wang, 5057–5069.
K. Yang and Z. Liu, ACS Nano, 2018, 12, 11541–11551. 424 A. Kalbasi, C. Komar, G. M. Tooker, M. Liu, J. W. Lee,
406 W. Huang, L. He, J. Ouyang, Q. Chen, C. Liu, W. Tao and W. L. Gladney, E. Ben-Josef and G. L. Beatty, Clin. Cancer
T. Chen, Matter, 2020, 3, 1725–1753. Res., 2017, 23, 137–148.
407 F. Castro, M. L. Pinto, C. L. Pereira, K. Serre, M. A. Barbosa, 425 X. Cao, J. Chen, B. Li, J. Dang, W. Zhang, X. Zhong,
K. Vermaelen, F. Gärtner, R. M. Gonçalves, O. De Wever C. Wang, M. Raoof, Z. Sun, J. Yu, M. G. Fakih and
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

and M. J. Oliveira, Biomaterials, 2020, 257, 120218. M. Feng, Sci. Adv., 2022, 8, eabl9171.
408 P. Zhang, J. Miska, C. Lee-Chang, A. Rashidi, W. K. Panek, 426 M. S. Alghamri, K. Banerjee, A. A. Mujeeb, A. Mauser,
S. An, M. Zannikou, A. Lopez-Rosas, Y. Han, T. Xiao, A. Taher, R. Thalla, B. L. McClellan, M. L. Varela,
K. C. Pituch, D. Kanojia, I. V. Balyasnikova and S. M. Stamatovic, G. Martinez-Revollar, A. V. Andjelkovic,
M. S. Lesniak, Proc. Natl. Acad. Sci. U. S. A., 2019, 116, J. V. Gregory, P. Kadiyala, A. Calinescu, J. A. Jiménez,
23714–23723. A. A. Apfelbaum, E. R. Lawlor, S. Carney, A. Comba,
409 Y. D. Zhao, M. Muhetaerjiang, H. W. An, X. Fang, Y. Zhao S. M. Faisal, M. Barissi, M. B. Edwards, H. Appelman,
and H. Wang, Biomaterials, 2021, 268, 120552. Y. Sun, J. Gan, R. Ackermann, A. Schwendeman,
410 M. A. Sallam, C. Wyatt Shields Iv, S. Prakash, J. Kim, M. Candolfi, M. R. Olin, J. Lahann, P. R. Lowenstein and
D. C. Pan and S. Mitragotri, J. Controlled Release, 2021, M. G. Castro, ACS Nano, 2022, 16, 8729–8750.
335, 333–344. 427 D. De Ruysscher, G. Niedermann, N. G. Burnet, S. Siva,
411 S. Theivendran, Z. Gu, J. Tang, Y. Yang, H. Song, Y. Yang, A. W. M. Lee and F. Hegi-Johnson, Nat. Rev. Dis. Primers,
M. Zhang, D. Cheng and C. Yu, ACS Nano, 2022, 16, 2019, 5, 13.
10943–10957. 428 J. Constanzo, J. Faget, C. Ursino, C. Badie and J. P. Pouget,
412 W. Zhang, Y. Shi, H. Li, M. Yu, J. Zhao, H. Chen and Front. Immunol., 2021, 12, 680503.
M. Kong, Carbohydr. Polym., 2022, 288, 119418. 429 A. Cohen, K. Ioannidis, A. Ehrlich, S. Regenbaum,
413 N. Qiu, G. Wang, J. Wang, Q. Zhou, M. Guo, Y. Wang, M. Cohen, M. Ayyash, S. S. Tikva and Y. Nahmias, Sci.
X. Hu, H. Zhou, R. Bai, M. You, Z. Zhang, C. Chen, Y. Liu Transl. Med., 2021, 13, eabd6299.
and Y. Shen, Adv. Mater., 2021, 33, 2100137. 430 D. B. Johnson, K. L. Reynolds, R. J. Sullivan, J. M. Balko,
414 M. Cao, H. Yan, X. Han, L. Weng, Q. Wei, X. Sun, W. Lu, J. R. Patrinely, L. C. Cappelli, J. Naidoo and J. J. Moslehi,
Q. Wei, J. Ye, X. Cai, C. Hu, X. Yin and P. Cao, Lancet Oncol., 2020, 21, 398–404.
J. Immunother. Cancer, 2019, 7, 326. 431 L. B. Kennedy and A. K. S. Salama, CA-Cancer J. Clin., 2020,
415 K. Ni, T. Luo, A. Culbert, M. Kaufmann, X. Jiang and 70, 86–104.
W. Lin, J. Am. Chem. Soc., 2020, 142, 12579–12584. 432 J. Luo, J. A. Beattie, P. Fuentes, H. Rizvi, J. V. Egger,
416 D. W. Ho, Y. M. Tsui, L. K. Chan, K. M. Sze, X. Zhang, J. A. Kern, D. Y. M. Leung, M. E. Lacouture, M. G. Kris,
J. W. Cheu, Y. T. Chiu, J. M. Lee, A. C. Chan, E. T. Cheung, M. Gambarin, B. D. Santomasso, D. M. Faleck and
D. T. Yau, N. H. Chia, I. L. Lo, P. C. Sham, T. T. Cheung, M. D. Hellmann, J. Thorac. Oncol., 2021, 16, 1759–1764.
C. C. Wong and I. O. Ng, Nat. Commun., 2021, 12, 3684. 433 P. D. Brown, V. Gondi, S. Pugh, W. A. Tome, J. S. Wefel,
417 X. Li, P. Ramadori, D. Pfister, M. Seehawer, L. Zender and T. S. Armstrong, J. A. Bovi, C. Robinson, A. Konski,
M. Heikenwalder, Nat. Rev. Cancer, 2021, 21, 541–557. D. Khuntia, D. Grosshans, T. L. S. Benzinger, D. Bruner,
418 M. Mondini, P. L. Loyher, P. Hamon, M. Gerbé de Thoré, M. R. Gilbert, D. Roberge, V. Kundapur, K. Devisetty,
M. Laviron, K. Berthelot, C. Clémenson, B. L. Salomon, S. Shah, K. Usuki, B. M. Anderson, B. Stea, H. Yoon, J. Li,
C. Combadière, E. Deutsch and A. Boissonnas, Cancer N. N. Laack, T. J. Kruser, S. J. Chmura, W. Shi,
Immunol. Res., 2019, 7, 376–387. S. Deshmukh, M. P. Mehta and L. A. Kachnic, J. Clin.
419 Y. Lan, D. Zhang, C. Xu, K. W. Hance, B. Marelli, J. Qi, Oncol., 2020, 38, 1019–1029.
H. Yu, G. Qin, A. Sircar, V. M. Hernández, M. H. Jenkins, 434 M. Karami, S. Asri-Rezaei, B. Dormanesh and
R. E. Fontana, A. Deshpande, G. Locke, H. Sabzevari, A. Nazarizadeh, Int. J. Radiat. Biol., 2018, 94, 17–27.
L. Radvanyi and K. M. Lo, Sci. Transl. Med., 2018, 435 J. B. Maxhimer, D. R. Soto-Pantoja, L. A. Ridnour, H. B. Shih,
10, eaan5488. W. G. Degraff, M. Tsokos, D. A. Wink, J. S. Isenberg and
420 C. Mao, S. Yeh, J. Fu, M. Porosnicu, A. Thomas, D. D. Roberts, Sci. Transl. Med., 2009, 1, 3ra7.
G. L. Kucera, K. I. Votanopoulos, S. Tian and X. Ming, 436 S. Gerassy-Vainberg, A. Blatt, Y. Danin-Poleg,
Sci. Transl. Med., 2022, 14, eabh1261. K. Gershovich, E. Sabo, A. Nevelsky, S. Daniel, A. Dahan,
421 B. G. Kim, E. Malek, S. H. Choi, J. J. Ignatz-Hoover and O. Ziv, R. Dheer, M. T. Abreu, O. Koren, Y. Kashi and
J. J. Driscoll, J. Hematol. Oncol., 2021, 14, 55. Y. Chowers, Gut, 2018, 67, 97–107.
422 M. Lin, Z. Yang, Y. Yang, Y. Peng, J. Li, Y. Du, Q. Sun, 437 W. Lu, Y. Xie, B. Huang, T. Ma, H. Wang, B. Deng, S. Zou,
D. Gao, Q. Yuan, Y. Zhou, X. Chen and X. Qi, Nano Today, W. Wang, Q. Tang, Z. Yang, X. Li, L. Wang and L. Fang, Sci.
2022, 42, 101359. Transl. Med., 2021, 13, eabc2344.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022
View Article Online

Chem Soc Rev Review Article

438 Y. Zhao, Y. Q. Xie, S. Van Herck, S. Nassiri, M. Gao, Y. Guo 459 C. Tang, S. I. Sherman, M. Price, J. Weng, S. E. Davis,
and L. Tang, Sci. Adv., 2021, 7, eabg7291. D. S. Hong, J. C. Yao, A. Buzdar, G. Wilding and J. J. Lee,
439 W. J. Smith, G. Wang, H. Gaikwad, V. P. Vu, E. Groman, Clin. Cancer Res., 2017, 23, 1414–1421.
D. W. A. Bourne and D. Simberg, ACS Nano, 2018, 12, 460 M. Souri, M. Soltani, F. Moradi Kashkooli, M. Kiani Shah-
12523–12532. vandi, M. Chiani, F. S. Shariati, M. R. Mehrabi and
440 R. Yudistiro, H. Hanaoka, N. Katsumata, A. Yamaguchi L. L. Munn, Mater. Today Bio., 2022, 13, 100208.
and Y. Tsushima, Mol. Pharm., 2018, 15, 2165–2173. 461 Y. Min, J. M. Caster, M. J. Eblan and A. Z. Wang, Chem.
441 S. M. Cheal, M. Patel, G. Yang, D. Veach, H. Xu, H. F. Guo, Rev., 2015, 115, 11147–11190.
P. B. Zanzonico, D. B. Axworthy, N. V. Cheung, O. Ouerfelli 462 G. Sriram, L. E. Milling, J. K. Chen, Y. W. Kong,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

and S. M. Larson, Bioconjugate Chem., 2020, 31, 501–506. B. A. Joughin, W. Abraham, S. Swartwout, E. D. Handly,
442 S. M. Sarrett, O. Keinänen, E. J. Dayts, G. Dewaele-Le Roi, D. J. Irvine and M. B. Yaffe, Sci. Signal., 2021, 14, eabc4764.
C. Rodriguez, K. E. Carnazza and B. M. Zeglis, Nat. Protoc., 463 B. Olson, Y. Li, Y. Lin, E. T. Liu and A. Patnaik, Cancer
2021, 16, 3348–3381. Discovery, 2018, 8, 1358–1365.
443 G. Parisi, J. D. Saco, F. B. Salazar, J. Tsoi, P. Krystofinski, 464 S. V. Paffenholz, C. Salvagno, Y. J. Ho, M. Limjoco, T. Baslan,
C. Puig-Saus, R. Zhang, J. Zhou, G. C. Cheung-Lau, A. J. S. Tian, A. Kulick, E. de Stanchina, J. E. Wilkinson, F. M.
Garcia, C. S. Grasso, R. Tavaré, S. Hu-Lieskovan, S. Mackay, Barriga, D. Zamarin, J. R. Cubillos-Ruiz, J. Leibold and S. W.
J. Zalevsky, C. Bernatchez, A. Diab, A. M. Wu, B. Comin- Lowe, Proc. Natl. Acad. Sci. U. S. A., 2022, 119, e2117754119.
Anduix, D. Charych and A. Ribas, Nat. Commun., 2020, 465 C. Verry, S. Dufort, B. Lemasson, S. Grand, J. Pietras,
11, 660. I. Troprès, Y. Crémillieux, F. Lux, S. Mériaux, B. Larrat,
444 C. Stavnsbjerg, E. Christensen, R. Münter, J. R. Henriksen, J. Balosso, G. Le Duc, E. L. Barbier and O. Tillement, Sci.
M. Fach, L. Parhamifar, C. Christensen, A. Kjaer, A. E. Hansen Adv., 2020, 6, eaay5279.
and T. L. Andresen, J. Controlled Release, 2021, 342, 337–344. 466 T. I. Janjua, Y. Cao, C. Yu and A. Popat, Nat. Rev. Mater.,
445 T. Kordbacheh, J. Honeychurch, F. Blackhall, C. Faivre- 2021, 6, 1072–1074.
Finn and T. Illidge, Ann. Oncol., 2018, 29, 301–310. 467 C. Liu, X. Liu, X. Xiang, X. Pang, S. Chen, Y. Zhang, E. Ren,
446 B. E. Lippitz and R. A. Harris, Radiother. Oncol., 2019, 140, L. Zhang, X. Liu, P. Lv, X. Wang, W. Luo, N. Xia, X. Chen
116–124. and G. Liu, Nat. Nanotechnol., 2022, 17, 531–540.
447 C. Pérez-Medina, A. J. P. Teunissen, E. Kluza, W. J. M. 468 Z. Chen, H. Pan, Y. Luo, T. Yin, B. Zhang, J. Liao, M. Wang,
Mulder and R. van der Meel, Adv. Drug Delivery Rev., 2020, X. Tang, G. Huang and G. Deng, Small, 2021, 17, 2007494.
154–155, 123–141. 469 B. H. Santich, S. M. Cheal, M. Ahmed, M. R. McDevitt,
448 A. A. Gabizon, R. T. M. de Rosales and N. M. La-Beck, Adv. O. Ouerfelli, G. Yang, D. R. Veach, E. K. Fung, M. Patel,
Drug Delivery Rev., 2020, 158, 140–157. D. Burnes Vargas, A. A. Malik, H. F. Guo, P. B. Zanzonico,
449 S. N. Bhatia, X. Chen, M. A. Dobrovolskaia and T. S. Monette, A. O. Michel, C. M. Rudin, S. M. Larson and
Lammers, Nat. Rev. Cancer, 2022, 22, 550–556. N. K. Cheung, Clin. Cancer Res., 2021, 27, 532–541.
450 L. Yin, J. Xue, R. Li, L. Zhou, L. Deng, L. Chen, Y. Zhang, 470 X. Wen, X. Zeng, X. Cheng, X. Zeng, J. Liu, Y. Zhang, Y. Li,
Y. Li, X. Zhang, W. Xiu, R. Tong, Y. Gong, M. Huang, Y. Xu, H. Chen, J. Huang, Z. Guo, X. Chen and X. Zhang, Mol.
J. Zhu, M. Yu, M. Li, J. Lan, J. Wang, X. Mo, Y. Wei, Pharm., 2022, 19, 3612–3622.
G. Niedermann and Y. Lu, Int. J. Radiat. Oncol. Biol. Phys., 471 U. Eberlein, M. Cremonesi and M. Lassmann, J. Nucl. Med.,
2020, 108, 212–224. 2017, 58, 97s–103s.
451 J. Lin, M. Yin, X. Liu, F. Meng and L. Luo, Macromol. Rapid 472 S. M. Qaim, B. Scholten and B. Neumaier, J. Radioanal.
Commun., 2022, 43, 2200194. Nucl. Chem., 2018, 318, 1493–1509.
452 B. Cabanillas, N. Novak and C. A. Akdis, Allergy, 2022, 77, 473 F. Man, P. J. Gawne and R. T. M. de Rosales, Adv. Drug
1658–1660. Delivery Rev., 2019, 143, 134–160.
453 G. T. Kozma, T. Shimizu, T. Ishida and J. Szebeni, Adv. 474 H. Deng, W. Yang, Z. Zhou, R. Tian, L. Lin, Y. Ma, J. Song
Drug Delivery Rev., 2020, 154–155, 163–175. and X. Chen, Nat. Commun., 2020, 11, 4951.
454 J. Zhao, G. Hu, Y. Huang, Y. Huang, X. Wei and J. Shi, Chin. 475 L. M. Coussens and Z. Werb, Nature, 2002, 420, 860–867.
Chem. Lett., 2021, 32, 1331–1340. 476 M. McLaughlin, E. C. Patin, M. Pedersen, A. Wilkins,
455 D. Zhai, J. Huang, Y. Hu, C. Wan, Y. Sun, J. Meng, H. Zi, M. T. Dillon, A. A. Melcher and K. J. Harrington, Nat.
L. Lu, Q. He, Y. Hu, H. Jin and K. Yang, Int. J. Radiat. Oncol. Rev. Cancer, 2020, 20, 203–217.
Biol. Phys., 2022, 114, 502–515. 477 L. Shi, J. Wang, N. Ding, Y. Zhang, Y. Zhu, S. Dong,
456 Y. Wang, Q. Zhao, B. Zhao, Y. Zheng, Q. Zhuang, N. Liao, X. Wang, C. Peng, C. Zhou, L. Zhou, X. Li, H. Shi,
P. Wang, Z. Cai, D. Zhang, Y. Zeng and X. Liu, Adv. Sci., W. Wu, X. Long, C. Wu and W. Liao, Nat. Commun.,
2022, 9, 2105631. 2019, 10, 5421.
457 J. Zhou, A. V. Kroll, M. Holay, R. H. Fang and L. Zhang, Adv. 478 M. M. Wattenberg and G. L. Beatty, Semin. Cancer Biol.,
Mater., 2020, 32, 1901255. 2020, 65, 38–50.
458 D. C. St Germain and W. McCaskill-Stevens, Cancer, 2021, 479 R. Weissleder and M. J. Pittet, Nat. Biomed. Eng., 2020, 4,
127, 1630–1637. 489–498.

This journal is © The Royal Society of Chemistry 2022 Chem. Soc. Rev.
View Article Online

Review Article Chem Soc Rev

480 L. M. Coussens, L. Zitvogel and A. K. Palucka, Science, 488 X. Kong, R. Cheng, J. Wang, Y. Fang and K. C. Hwang,
2013, 339, 286–291. Nano Today, 2021, 36, 101004.
481 R. J. Pickering and C. E. Bryant, Science, 2020, 369, 489 Y. Liu, W. N. Crowe, L. Wang, Y. Lu, W. J. Petty, A. A. Habib
1564–1565. and D. Zhao, Nat. Commun., 2019, 10, 5108.
482 X. Wang, P. Hua, C. He and M. Chen, Acta Pharm. Sin. B, 490 Y. Li, X. Zhao, X. Liu, K. Cheng, X. Han, Y. Zhang, H. Min,
2022, 12, 3567–3593. G. Liu, J. Xu, J. Shi, H. Qin, H. Fan, L. Ren and G. Nie, Adv.
483 Y. Liu, W. Zhen, Y. Wang, S. Song and H. Zhang, J. Am. Mater., 2020, 32, 1906799.
Chem. Soc., 2020, 142, 21751–21757. 491 M. Li, D. Liu, D. Lee, S. Kapoor, K. N. Gibson-Corley,
484 B. Ma, A. Wells, L. Wei and J. Zheng, Semin. Cancer Biol., T. P. Quinn, E. A. Sagastume, S. L. Mott, S. A. Walsh,
Published on 25 November 2022. Downloaded by Chongqing University on 11/30/2022 7:55:36 AM.

2021, 71, 2–9. M. R. Acevedo, F. L. Johnson and M. K. Schultz, Mol.


485 M. E. Giuliani, C. G. Robinson, J. K. Salama and M. E. Daly, Pharm., 2019, 16, 3904–3915.
Int. J. Radiat. Oncol. Biol. Phys., 2020, 106, 455–459. 492 M. Singhal, N. Gengenbacher, A. A. Abdul Pari,
486 M. Guckenberger, Y. Lievens, A. B. Bouma, L. Collette, M. Kamiyama, L. Hai, B. J. Kuhn, D. M. Kallenberg,
A. Dekker, N. M. deSouza, A. C. Dingemans, B. Fournier, S. R. Kulkarni, C. Camilli, S. F. Preuß, B. Leuchs,
C. Hurkmans, F. E. Lecouvet, I. Meattini, A. Méndez C. Mogler, E. Espinet, E. Besemfelder, D. Heide,
Romero, U. Ricardi, N. S. Russell, D. H. Schanne, M. Heikenwalder, M. R. Sprick, A. Trumpp, J. Krijgsveld,
M. Scorsetti, B. Tombal, D. Verellen, C. Verfaillie and M. Schlesner, J. Hu, S. E. Moss, J. Greenwood and
P. Ost, Lancet Oncol., 2020, 21, 18–28. H. G. Augustin, Sci. Transl. Med., 2021, 13, eabe6805.
487 W. Zhang, F. Wang, C. Hu, Y. Zhou, H. Gao and J. Hu, Acta 493 G. Bergers and S. M. Fendt, Nat. Rev. Cancer, 2021, 21,
Pharm. Sin. B, 2020, 10, 2037–2053. 162–180.

Chem. Soc. Rev. This journal is © The Royal Society of Chemistry 2022

You might also like