Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Aug 2.
Published in final edited form as: ACS Nano. 2023 May 1;17(10):8935–8965. doi: 10.1021/acsnano.3c02303

Advanced Nitric Oxide Generating Nanomedicine for Therapeutic Applications

Zhixiong Wang 1, Albert Jin 2, Zhen Yang 3,4, Wei Huang 5,6
PMCID: PMC10395262  NIHMSID: NIHMS1921067  PMID: 37126728

Abstract

Nitric oxide (NO), a gaseous transmitter extensively present in the human body, regulates vascular relaxation, immune response, inflammation, neurotransmission, and other crucial functions. Nitrite donors have been used clinically to treat angina, heart failure, pulmonary hypertension, and erectile dysfunction. Based on NO’s vast biological functions, it further can treat tumors, bacteria/biofilms and other infections, wound healing, eye diseases, and osteoporosis. However, delivering NO is challenging due to uncontrolled blood circulation release and a half-life of under five seconds. With advanced biotechnology and the development of nanomedicine, NO donors packaged with multifunctional nanocarriers by physically embedding or chemically conjugating have been reported to show improved therapeutic efficacy and reduced side effects. Herein, we review and discuss recent applications of NO nanomedicines, their therapeutic mechanisms, and the challenges of NO nanomedicines for future scientific studies and clinical applications. As NO enables the inhibition of the replication of DNA and RNA in infectious microbes, including COVID-19 coronaviruses and malaria parasites, we highlight the potential of NO nanomedicines for anti-pandemic efforts. This review aims to provide deep insights and practical hints into design strategies and applications of NO nanomedicines.

Keywords: Nitric oxide nanomedicine, Cancer, Bacterial, Biofilm, Wound healing, Glaucoma, Osteoporosis, Coronavirus, Biotechnology

Graphical Abstract

graphic file with name nihms-1921067-f0017.jpg

INTRODUCTION

In 1982, nitric oxide (NO) was discovered by Furchgott, Ignarro, and Murad as a signalling molecule named endothelial-derived relaxing factor (EDRF).1 Whereafter, this great discovery inaugurated extensive research on the key roles of NO in many physiological systems, earning the Nobel Prize in Physiology or Medicine for the three scientists in 1998.2 L-Arginine (L-Arg) is the endogenous NO donor, under the catalysis of NO synthase (NOS) to produce the NO required by the body. In the human body, various biological processes such as vasodilation, platelet aggregation, immune response, and neurotransmission are related to NO.36 Due to its indispensable role in physiology, defects in nitric oxide biosynthesis are related to many disease states. Despite forty years of extensive research, the complicated mechanism of NO in the human body remains uncovered. Fortunately, the strategies for the delivery of exogenous NO have been used in many life-saving biomedical applications. Inhaled NO is an FDA-approved treatment for clinical pulmonary hypertension.7,8 The direct inhalation of NO cannot yet therapy other diseases due to critical concern for its in vivo distribution and high reactivity. Thus, the design syntheses of NO donors have become more efficient at delivering NO to specific disease lesions, thereby expanding NO applications. Organic nitrates (RONO2s) (e.g., glyceryl trinitrate and isosorbide mononitrate) are widely used clinically to treat diseases including angina, heart failure, anal fissures, and pulmonary hypertension. Still, they are common side effects, such as the risk of hypotension, headaches, and evolving tolerance (Table 1).9 Nitrosothiols (RSNOs) (e.g., S-nitrosoglutathione (GSNO), S-nitrosocysteine, S-nitrosoalbumin, and Snitroso-N-acetylpenicillamine (SNAP)) can be antiplatelet agents, vasodilators, or cell cytoprotective agents for reduction of oxidative stresses. In addition, NONOates have therapeutic efficacy in pulmonary hypertension, arterial smooth muscles, and traumatic brain injury.10 However, low molecular weight species of NO donors have had limited clinical application in other diseases owing to the rapid systemic clearance.11

Table 1.

Clinical NO Donors.

NO donor Disease Half-life Working mechanism of NO Side effects
Sodium nitroprusside Hypertensive emergency, heart failure, decrease bleeding < 2 minutes NO stimulates intracellular cyclic guanosine monophosphate (cGMP) production. low blood pressure, cyanide toxicity, methemoglobinemia
Glyceryl trinitrate Angina pectoris, chronic heart failure 2 to 3 minutes Relaxation of vascular smooth muscles, arteriolar and venous dilatation. Headache, dizziness, lightheadedness, nausea, flushing, and burning/tingling under the tongue
Isosorbide dinitrate Angina pectoris, heart failure, esophageal spasms 1 hour Relaxation of vascular smooth muscle, dilatation of peripheral arteries and vein Headache, dizziness, lightheadedness, nausea, and flushing
Pentaerythritol tetranitrate Angina, heart conditions 4 to 5 hours Increased cellular cGMP concentration in vascular smooth muscle. Flushing, dizziness, nausea, headache, hypersensitivity, rash, fast heart rate, low blood pressure (hypotension),
Molsidomine Angina pectoris, myocardial infarction 1 to 2 hours Increased cGMP levels and decreased intracellular calcium ions in smooth muscle cells. Headache, low blood pressure

The efficacy of NO-based therapeutics largely depends on the concentration profile of NO in the disease region vs. the general body. At low concentrations, NO interacts with transition metal-containing proteins, such as those carrying heme groups or metal regulatory transcription factors, to regulate various biological processes and the progression of diseases. In addition to such direct effects on biomolecules, NO can generate biologically active intermediates and reactive nitrogen species (RNS) at high concentrations, such as nitrogen dioxide (NO2.). NO is further oxidized by NO2. to form dinitrogen trioxide (N2O3) and rapidly decomposes into NO+, which nitrates electrophilic compounds (e.g., thiols, phenols, secondary amines) and causes deamination of DNA bases. NO can also react with reactive oxygen species (ROS) to generate peroxynitrite (ONOO), which can modulate protein function by nitration. More importantly, these RNS can directly kill invading microbe and malignant cells.12 However, when exposed to excessive levels of ROS and RNS, normal cells also experience damaging stresses that are oxidative (hydroxylation, lipid peroxidation, DNA strand breaks) and nitrosative (nitration, nitrosylation, DNA deamination). These stresses can damage DNA/RNA, proteins (e.g., heme groups), lipids, and other molecules, leading to impaired cellular functions, enhanced inflammatory reactions, inhibition of mitochondrial respiration, cell apoptosis, and genotoxicity. Therefore, controlling these parameters (e.g., delivery sites, concentration, and release rate) of NO remains critical in the development of advanced NO therapeutics.13 Toxic side effects will increase when the NO drugs cannot be specifically applied to the disease lesion where self-tolerance may also develop. To solve this main challenge, nano-delivery systems of NO donors provide possibilities for specific targeting and control release of NO.14 For example, NO at the micromolar level can directly affect nitric/oxide mitochondria or DNA for cancer therapy. NO at the Picomolar lever contributes to the angiogenesis of cell proliferation. In addition, since the half-life of NO is extremely short (less than 5 seconds), the delivery of NO must be highly targeted and selective, and the distance of NO donors to reactants is limited to about 100 microns.15 More importantly, researchers have developed a series of activable precision NO delivery systems based on the endogenous (pH, glutathione (GSH), H2O2, enzyme) and exogenous (light, heat, X-ray, ultrasound) stimulus for the disease treatment.16 Therefore, developing multifunctional NO delivery systems is significant for disease treatment.

This review discusses primary NO-treatable diseases such as cancer, anti-bacterial/biofilm, wound healing, eye diseases, and osteoporosis. The strategies of design synthesis of nano-sized NO-delivery platforms and their therapeutic mechanism are introduced in detail (Scheme 1). We also evaluate the potential challenges of NO nanomedicine in clinical translation. Nowadays, the COVID-19 pandemic has affected our lives all over the globe. The NO has been considered a promising therapeutic gas currently undergoing COVID-19 clinical trials, due to its capability of replication inhibition of the DNA and RNA viruses, including coronaviruses.17,18 Thus, we especially highlight potential NO drug applications for COVID-19. We also note more broadly that NO nanomedicine may have important roles in fighting other forms of infections including parasitic malaria.

Scheme 1.

Scheme 1.

The treatment mechanisms of nitric oxide (NO) for various diseases. In addition to the direct effects of NO on biomolecules, NO reacts with oxygen or other reactive oxygen species (ROS) to generate reactive nitrogen species (RNS) to act on proteins, lipids, nucleosides, and metals as well as to induce transnitration, which can cause DNA strand breaks, abasic sites, enzyme activity inhibitions, mitochondrial depolarization, mitochondrial dysfunction, and DNA/RNA replication inhibitions. High-intensity focused ultrasound, HIFU; Glutathione, GSH.

CANCER

Cancer remains to be the second worst disease in the world by the fatality rate.19 The current primary treatment modalities for clinical cancer include chemotherapy, surgery, and radiotherapy. Newer therapeutic modalities for cancer include phototherapy, ultrasound therapy, gas therapy, immunotherapy, and starvation therapy. However, these treatments still have drawbacks and limitations. For example, surgery is difficult to remove the small residual tumors and tumour cells scattered in the blood vessels and lymphatics, which easily induce tumor recurrence and further metastasis. Failure of chemotherapy mainly follows the development of drug resistance by the cancer cells. Hypoxia tumor environment also hampers the therapeutic efficacy of radiotherapy, sonodynamic and photodynamic. NO has an anti-cancer effect within the concentration range of 1 μm to mm by oxidation and nitrosative stress with few adverse effects.20 Fabrication of NO prodrug achieves on-demand release of NO in the tumor microenvironment (TME) not only can inhibit the expression of mammalian HIF-1α, base-excision DNA repair enzyme and multidrug transporter P-glycoprotein (P-gp), but also normalize tumor blood vessels, even react with other ROS to generate highly reactive ONOO to sensitize chemotherapy, radiotherapy, phototherapy,21 and chemodynamic therapy.22 But NO concentration higher than 1 mM may cause NO poisoning.23 Picomolar NO has anti-apoptotic effects and promotes angiogenesis, thereby increasing nutrient delivery and promoting tumor growth.24 This section covers the recent advanced development of NO-based nanomedicine for tumor therapy (Table 2).

Table 2.

NO Nanomedicines for Tumor Therapya

Nanomedicine NO donor Release condition Working mechanism of NO Refs
RBS-T-UCNPs Roussin’s black salt (RBS) 808 nm laser irradiation Sensitizing chemotherapy by reducing tumorigenic ability: inhibiting cancer stem-like cells and mammosphere formation ability, reducing CD44+ / CD24 subsets. 184
α-CD-Ce6-NO NPs α-CD-NO GSH Sensitizing PDT: depleting intracellular GSH, relieving hypoxia at tumor sites, and ONOO generation to enhance ROS reactivity. 69
RBS-UCNPs RBS 808 nm laser irradiation Sensitizing chemotherapy: High NO concentration kills cancer cells; low NO concentration reduces P-gp level to overcome MDR. 30
NanoNO DNIC [Fe(μ-SEt)2(NO)4] Physiological condition Sensitizing chemotherapy: gradient NO generation efficiently reprograms tumor vasculature and microenvironments to improve chemotherapy 42
NMOF−SNO R-SNO 808 nm laser irradiation Sensitizing PTT: NO releasing to enhance PTT efficiency 185
DN@MSN R-SNO Natural release Sensitizing chemotherapy: NO activating MMP-1 and MMP-2, promoting DOX delivery to more deep tumour tissues 47
Peptide-HMSN-LA L-Arg ROS NO direct oxides proteins 81
photoNORM/UCNP Metal-NO 794 nm laser irradiation Low dose reduces HIF-1a, and high doses are cytotoxic 186
PTNGs R-SNO 808 nm laser irradiation (Photothermal) Sensitizing chemotherapy: NO reverses MDR by inhibiting Pgp expression 187
GCZ@M nitrosoglutathione (GSNO) Ultrasound irradiation Sensitizing SDT: ONOO generation, relieving tumour hypoxia 188
IDDHN 2-(Nitrooxy)acetic 808 nm laser irradiation (Photothermal) Sensitizing chemotherapy: NO improves the EPR effect 189
L-Arg-HMON-GOx L-Arg H2O2 Starving-like/NO for synergistic cancer therapy 190
BNN-Bi2S3 Bis-N-nitroso compounds 808 nm laser irradiation NO impairs the autophagic self-repairing ability of tumor cells in situ 76
PFTDPP-SNAP NPs R-SNO 808 nm laser irradiation (Photothermal) Sensitizing PTT: NO generation enhances PTT efficiency 191
Lip-SNAP S-nitroso-N-acetylpenicillamine GSH NO induces stromal depletion for improved nanoparticle penetration 192
S–NO NPs Aryl N-nitrosamine 808 nm laser irradiation (Photothermal) NO release activates photothermal agent for synergistic tumor treatment 193
QM-NPQ@PDHN NPQ Glutathione S-transferases π Specific, high-efficacy, and low-toxic patocellular carcinoma therapy 194
AL-SISIN-1 SISIN-1 Physiological conditions Inhibiting tumour metastasis by inducing cytotoxicity preferentially on tumour cells in lymph nodes 195
iCPDN R-SNO GSH Sensitizing chemoimmunotherapy: Reversing DOX resistance and enhancing antitumor immune responses by reprogramming the tumor microenvironments. 196
WB@hydrogel BNN6 1064 nm laser irradiation (photothermal) Anti-angiogenesis and tumor microenvironment reprogramming: activating wild type p53 expression, alternating pro-angiogenic TME to anti-angiogenic TME. 80
pPTX/pCD-pSNO R-SNO Redox conditions Sensitizing chemoimmunotherapy: Enhancing dendritic cell activation, T cell expansion, cytotoxicity, and immunogenic cell death, 197
TPE-RSNO micelles R-SNO H2O2 Reducing P-gp expression, reversing MDR, RNS 198
FZ-SS-FZ@FA NPs phenylsulfonylfuroxan GSH Upregulating p53 and cleaved caspase-3 proteins 199
Ce6/PDE5-i@FHMON-O2 _ DE5-inhibited PDE5 pathway to upregulate eNOS RNS helps ROS to evade the hypoxia-induced resistance to ROS-based antitumor 200
NPSD-IR NTC GSH Sensitizing chemotherapy: inhibiting Pgp expression to overcome MDR 25
PIH-NO R-SNO Ultrosound ONOO generation to enhance SDT, promote the maturation of dendritic cells, and increase immune cells infiltration 201
PtR/CPG L-Arg H2O2 Enhancing anticancer chemoimmunotherapy: NO can trigger immunogenic cell death to produce tumor-associated antigens 202
HFC/DTX/aPD1 L-Arg The environment of cancer cells Promoting anticancer chemoimmunotherapy 203
NO-DOX@PDA-TPGS-Gal N,N′-di-sec-butyl-N,N′-dinitroso-1,4-phenylenediamine (BNN) 808 nm laser irradiation (photothermal) Enhancing chemo−photothermal therapy: inhibiting P-gp -the related efflux of DOX 204
S1P/JS-K/Lipo JS-K Glutathione S-transferases Promoting glioblastoma multiforme cell death 20
CMH-OBN Benzofuroxan GSH ONOO generation enhances PDT/PTT/immunotherapy 205
Alb-PLP/NO NPs Diazeniumdiolate Physiological conditions Enhancing tumor penetration and inhibiting melanoma. 206
P@BDOX/β-lapachone-NO-NPs R-O-NO2 GSH Overcoming chemo-resistance and enhancing the efficacy of HIFU in combination with chemotherapy 207
SPNAPt/NO R-O-NO GSH ONOO generation down-regulates glutathione reductase (GR) and xeroderma pigmentosum group A 32
LPFe3O4 NPs L-Arg iNOS NO enhances immune therapy 208
BPNs-Arg-GOx@MnO2 L-Arg H2O2 NO activates matrix metalloproteinases to degrade the dense extracellular matrix 209
UC-ZIF/BER R-O-NO NIR irradiation to UV by upconversion NO turns on the ryanodine receptors for Ca2+ elevation to achieve Ca2+-initiated cancer therapy 210
Artificial microbots (AMBs) L-Arg iNOS and ROS Regulating vasodilation and invasion to promote drug release to solid tumors 211
HFLA-DOX L-Arg H2O2 Promoting deep drug penetration and reversal of MDR in cancer chemotherapy. 212
L-Arg@Ce6@P NPs L-Arg H2O2 Inhibiting mitochondrial respiration 213
HA@MOF/D-Arg D-Arg H2O2 Down-regulating HIF-1α to alleviate tumor hypoxia for sensitizing radiotherapy 214
ArgCCN L-Arg H2O2 High concentration NO induces cancer cell apoptosis 215
RBCm/PAAVSNO/IR1061 + 1-MT NPs R-S-NO Heat and pH NO normalizes tumor vessels 216
NO-NCPs DETA NONOates pH and photoacoustic ONOO generation to damage lysosome, mitochondria, and DNA 86
CuS-PEI/NO-TPP Diazeniumdiolate 1064 nm laser irradiation (photothermal) Inhibiting heat shock proteins expression 74
GMOF-LA L-Arg H2O2 NO sensitizes PDT 217
ZGO-Mn-RBS Roussin’s black salt X-ray excitation depth-independent NO-releasing strategy for gas-sensitized therapeutic applications. 218
Ptx@AlbSNO R-SNO GSH Enhancing immune cell infiltration into tumor microenvironments. 219
BSA-IRLA@RVs-RGD L-Arg ROS Inhibiting cancer-associated platelet activation and disrupting tumour vascular barriers 220
Au@SiO2-SNO/PEG/TPP R-SNO 808 nm laser irradiation (Photothermal) Activating MMPs to break collagen fibers to enhance the cellular internalization 221
α-CD-DOX-NO-DA NPs R-SNO GSH NO facilitates mitochondrial membrane permeabilization and downregulates ATP level and inhibits pgp to reverse MDR 222
Micellar NO@HMs NONOate pH ONOO generation sensitizes radiotherapy of hypoxia tumor 50
DM1-NO-NPs R-SNO X-ray irradiation ONOO causes DNA and lipid damage to sensitize radiotherapy. 223
SNO-HSA Dimer R-SNO Physiological conditions NO augments the EPR effect to promote drugs to the tumors. 224
DPP-NF NPs 4-Nitro-3-Trifluoromethylaniline 660 nm laser irradiation. NO directly damages DNA, and inhibits the expression of HIF-α to enhance PDT efficiency 225
Lyso-Ru-NO@FA@C-TiO2 R-NO 808 nm light irradiation Lysosome-targeted NO delivery to enhance PDT 226
PpRE@PEG-PpIX NPs R-Fe(NO)2 637 nm laser irradiation Reversing MDR and overcoming hypoxia to enhance PTT. 227
Ce6-loaded NO-mannan R-O-NO2 GSH NO prompts vessel-relaxing and hypoxia relief 228
N-GQDs@Ru-NO@Gal R-NO 808 nm light irradiation NO enhances PTT 229
CPNs R-O-NO2 GSH ONOO and NO inhibit Pgp expression to reverse MDR 33
L-Arg@PCN@Mem L-Arg ROS NO overcomes hypoxia to sensitize PDT 70
P(IR/BNN6/AIPH)@Lip-RGD BNN6 1064 nm laser irradiation (Photothermal) Synergistic NO and alkyl radical action 230
Fe(II)-BNCP BPDB GSH Synergistic NO and chemodynamic therapy 89
ADAu@CuS YSNPs L-Arg ROS Inhibiting P-gp expression to reverse MDR 29
IPO-NO R-SNO 808 nm laser irradiation (Photothermal) Low NO concentration increases the EPR effect and high concentration directly kills the tumors. 231
IMesNO/DOX@MCs R-NO HIFU irradiation Accelerating drug accumulation in tumor 232
PV-TS Sodium nitroprusside dihydrate GSH NO inhibits cellular respiration to relieve tumor hypoxia 65
NO-M@DOX R-O-NO2 GSH NO reverses MDR to enhance chemotherapy 233
N-GQDs@Ru-Cl@TPP R-NO 808 nm light illumination NO enhances PTT 78
M@BPAG L-Arg H2O2 Reprogramming the tumour immune microenvironment and significant synergistic antitumor effect 234
AI-MPHA NCs L-Arg ROS NO sensitizes PTT 235
PNOC-PDA/DOX R-SNO 808 nm laser irradiation (photothermal) NO reverses MDR to sensitize PTT and chemotherapy 34
RBS-T-SCNPs Roussin’s black salt X-ray irradiation ONOO-generation directly damages DNA and downregulates the DNA-repair enzyme 52
HMs DETA NONOate pH NO inhibits P-gp expression to reverse CPT MDR. 31
PEG-USMSs-SNO R-SNO X-ray irradiation NO sensitizes radiotherapy of hypoxia tumor 55
P-lapa-Fc L-Arg ROS ONOO generation enhances tumor therapy 64
UMNOCC-PEG R-SNO pH RNS generation enhances PDT/CDT 90
mCuMNO S-nitrosoglu-tathione Cu+ Interrupting the interaction between platelets and circulating tumor cells and enhancing CDT 236
T-NPCA/NO R-SNO GSH ONOO promotes mitochondrial membrane permeabilization 237
a

RBS-T-UCNPs, Roussin’s black salt-upconversion nanoparticles; α-CD-Ce6-NONPs, α-cyclodextrin-chlorin e6-NO nanoparticles; NMOF-SNO, nanoscale metal-organic framework-S-Nitrosothiol; DN@MSN, doxorubicin-NO-Mesoporous silica nanoparticles; Peptide-HMSN-LA, Peptide-hollow mesoporous silica nanoparticles-L-Arg; photoNORM/UCNP, photochemical precursor of NO-upconversion nanoparticles; PTNGs, phototriggered NO nanogenerators; GCZ@M, GSNO/Ce6@ZIF-8@Cytomembrane; IDDHN, intelligent nanoparticle; L-Arg-HMON-Gox, L-Arg-hollow mesoporous organosilica nanoparticle-glucose oxidase; BNN-Bi2S3, bis-N-nitroso compounds-bismuth sulfide; PFTDPP-SNAP NPs, semiconducting polymer-s-nitrosothiol groups nanoparticles; Lip-SNAP, SNAP loaded liposomes; S–NO NPs, N-nitrosamine nanoparticles; QM-NPQ@PDHN, fluorogen QM-2-O2-(2,4-dinitro-5-{[2-(β-d-galactopyranosyl olean-12-en-28-oate-3-yl)-oxy-2-oxoethyl] piperazine-1-yl}- phenyl) 1-(methylethanolamino)diazen-1-ium-1,2-dilate-PEGylated disulfide-doped hybrid nanocarriers; AL-SISIN-1, N-((2-pyridin-2-yldisulfanyl)ethoxyl)carbonyl-3-morpholinosydnonimine; iCPDN, poly(amidoamine)-Doxorubicin-NO; WB@hydrogel, WO2.9-N,N′-di-sec-butyl-N,N′-dinitroso-1,4-phenylenediamine@hydrogen; BNN6, N,N′-di-sec-butyl-N,N′-dinitroso-1,4-phenylenediamine; pPTX/pCD-pSNO, polymerized paclitaxel-nitric oxide-incorporated polymerized β-cyclodextrin; TPE-RSNO micells, S-nitrosothiol-functionalized tetraphenylethene; FZ-SS-FZ@FA NPs, phenylsulfonylfuroxan nanoparticles; Ce6/PDE5-i@FHMON-O2, photocleaved O2-released nanoplatform; NPSD-IR, IR-780-Doxorubicin NO nanoparticles; PIH-NO, perfluorodecalin-IR780-human serum albumin- NO; PtR/CPG, cis-platinum-L-arginine/ Cytosine-phosphorothioate-guanine; HFC/DTX/aPD1, heparin-folate-cy5.5/l-arginine/ docetaxel/anti-PD-1; S1P/JS-K/Lipo, sphingosine-1-phosphate/ O2-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl) piperazin-1-yl] diazen-1-ium-1,2-diolate/liposome; JS-K, O2-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl) piperazin-1-yl] diazen-1-ium-1,2-diolate; CMH-OBN, chlorin e6-melanin-hyaluronic acid nanoparticles-oxidized bletilla striata polysaccharide microcapsules; Alb-PLP/NO NPs, albumin-coated poly(lactic-co-glycolic acid) (PLGA)-conjugated linear polyethylenimine diazeniumdiolate (LP/NO) nanoparticles; P@BDOX/β-lapachone-NO-NPs, peptides (pHLIPs)-poly(ethylene glycol) and nitrated gluconic acid copolymers @Doxorubicin prodrug/β-lapachone-NO; SPNAPt/NO, supramolecular prodrug nanoassemblies-platinum(IV) prodrug/NO; LPFe3O4 NPs, L-arginine-poly(acrylic acid)-hollow iron oxide nanoparticles; BPNs-Arg-GOx@MnO2, black phosphorus nanosheets-L-Arginine-glucose oxidase @MnO2 nanosheets; UC-ZIF/BER, upconversion nanoparticles-zeolitic nitro-/nitrile-imidazole framework-82-berbamine; HFLA-DOX, doxorubicin-heparin/folic acid/L-arginine; L-Arg@Ce6@P NPs, L-arginine@ chlorin e6@ poly-lactic-co-glycolic acid nanoparticle; HA@MOF/D-Arg, hyaluronic acid@ metal-organic frameworks/D-arginine; ArgCCN, poly-L-arginine modified carbon-dots-doped graphitic carbon nitride nanomaterial; RBCm/PAAVSNO/IR1061 + 1-MT NPs, red blood cell membrane/copolymer (poly(acrylamide-co-acrylonitrile-co-vinylimidazole)-S-nitrosothiols copolymer+1-methyl-tryptophan; NO-NCPs, NO-nanocapsules; Ptx@AlbSNO, paclitaxel@ NO donor-modified albumin; BSA-IRLA@RVs-RGD, BSA-L-Arginine-IR783@ red blood cells membrane derived vesicle-RGD; NO@HMs, NO-poly(lactic-co-glycolic acid) (PLGA) hollow microsphere; DPP-NF NPs, diketopyrrolopyrrole-4-nitro-3-trifluoromethylaniline nanoparticles; Lyso-Ru-NO@FA@C-TiO2, Lysosome-Ru-NO@ folic acid@ carbon-doped titanium dioxide nanoparticles; N-GQDs@Ru-NO@Gal, N-doped graphene quantum dots@ Ru-NO@ galactose derivative; CPNs, cocktail polyprodrug nanoparticles; L-Arg@PCN@Mem, L-arginine@ porous coordination network@ cancer cell membrane; P(IR/BNN6/AIPH)@Lip-RGD, IR 1061/BNN6/alkyl radical initiator@Liposome-RGD; Fe(II)-BNCP, 1,5-bis[(l-proline-1-yl)diazen-1-ium-1,2-diol-O2-yl]-2,4-dinitrobenzene nanoscale coordination polymer; ADAu@CuS YSNPs, l-arginine/Dox-loaded gold@ copper sulfide yolk–shell nanoparticls; IPO-NO, IR780-paclitaxel-NO donor-S-nitrosated human serum albumin; IMesNO/DOX@MCs, 1,3-bis-(2,4,6-trimethylphenyl)imidazolylidene nitric oxide/ Doxorubicin@ Micelles; PV-TS, polymeric nanovesicles- tetraphenylporphyrin- sodium nitroprusside; NO-M@DOX, Nitric Oxide Donor-containing polycarbonate-based micelles@ Doxorubicin; N-GQDs@Ru-Cl@TPP, N-doped graphene quantum dots@ ruthenium nitrosyl@ triphenylphosphonium; M@BPAG, macrophage membrane@ black phosphorus nanosheets-L-arginine-glucose oxidase; AI-MPHA NCs, indocyanine green/L-arginine-mesoporous core–shell structure nano-composites; PNOC-PDA/DOX, poly(L-cysteine)20-poly(ethylene oxide)45-SNO-polydopamine/ Doxorubicin; RBS-T-SCNPs, Roussin’s black salt- tocopheryl polyethylene glycol 1000 succinate-scintillating nanoparticles; HMs, hollow microsphere system; PEG-USMSs-SNO, PEG-upconversion nanotheranostic system- S-nitrosothiol; P-lapa-Fc, poly(ε-caprolactone) (PCL)-b-PArg-ferrocene; UMNOCC-PEG, copper peroxide nanodots-chlorin e6-polyethylene glycol-silicon pores; mCuMNO, S-nitrosoglutathione-copper-based metal-organic framework; T-NPCA/NO, cinnamaldehyde-NO nanoparticles.

NO enhances tumor chemotherapy

NO inhibits P-gp to reverse multi-drug resistance (MDR)

Modern chemotherapy has achieved a huge positive impact on pain relief for cancer patients and lifetime extension, but further improvements are necessary to overcome multi-drug resistance (MDR). Recently, several works related to NO) are the potential function to overcome MDR have been reported.25 Chen et al. enlisted the mechanism by which NO could reverse the chemotherapy resistance (Figure. 1A).26 NO could suppress DNA repair, reduce detoxification capacity, inhibit the activation of HIF and NF-kB, and enhance the nuclear transport of drugs.27,28 In addition, the overexpression of the drug efflux P-gp to continuously pump the chemo drug outside of cancer cells broadly hinders cancer chemotherapy.29,30 Sung et al. reported a pH-responsive NO-generating hollow microsphere (HM) system to overcome the resistance of MCF-7 breast cancer cells to the anticancer agent irinotecan (CPT-11) by inhibiting the P-gp expression of cancer cells (Figure. 1B).31 The HM simultaneously carries CPT-11 and a NO-donor (EDTA NONOate), in acidic tumor tissue, acidic protons infiltrate HMs and generate NO bubbles from the encapsulated NONOate, triggering CPT-11 release and inhibit P-gp expression to enhance chemotherapy.

Figure 1.

Figure 1.

NO inhibits P-gp expression to reverse MDR. A) Schematic mechanisms of nitric oxide (NO) against multidrug resistance (MDR). Reprinted with permission from ref 26. Copyright 2017 Elsevier. B) pH-responsive NO generating to reverse P-gp-mediated MDR. (I) Schematic composition of HMs and NO generating mechanism to responsive acid tumors environment. (II) Confocal images of P-gp expression levels after different treatments. Reprinted with permission from ref 31. Copyright 2015 WILEY-VCH. C) Polyprodrug nanoparticles (CPNs) using cisplatin and ONOO to overwhelm cisplatin-resistant cancers. (I) Mechanism of CPNs to overcome cisplatin-resistant cancers. (II) P-gp expression level for cells imaged after CPN and different treatments. Reprinted with permission from ref 33. Copyright 2020 Elsevier. D) Combined NO generation, mild photothermal, and chemotherapy to overcome MDR by PNOC-PDA/DOX. (I) Schematic mechanism. (II) Western blot detection of P-gp expression in MCF-7/ADR upon different treatments. Reprinted with permission from ref 34. Copyright 2019 American Chemical Society. P-glycoprotein 1 (permeability glycoprotein), P-gp; multidrug resistance protein 1, MDR1; Hypoxia-inducible factor, HIF-1; Photothermal Therapy, PTT.

NO is reactive with superoxide (O2.−) to generate more highly reactive ONOO.32 Hu et al. designed cocktail polyprodrug nanoparticles (CPNs) that, in response to reductive cytosolic, can release NO and generate ONOO to kill cisplatin-resistant cancers (Figure. 1C).33 Inside cancer cells, CPNs are degraded so that the released cisplatin activates nicotinamide adenine dinucleotide phosphate oxidase (NOXs) to catalyze oxygen to O2.−, which further reacts with in-situ released NO to produce ONOO. In vitro and in vivo analyses demonstrate that CPNs can efficiently inhibit P-gp expression to overcome cisplatin resistance and enhance therapy efficiency for resistant cancer.

Dong et al. reported a general approach to the integration of heat-sensitive NO donors, chemotherapeutic DOX, and photothermal conversion attributes into polypeptide nanocomposite (PNOC-PDA/DOX) for overcoming MDR (Figure 1D).34 Under NIR irradiation, PNOC-PDA/DOX induces photothermal therapy(PTT), NO, and chemotherapy (PTT-NO-CT) synergistic triple therapy, which produced a superior synergistic effect of NO release characteristics, light-to-heat conversion and chemotherapy on MDR reversal and killing MCF-7/ADR cells in vitro, and the P-gp expression level was down-regulated to about half. Notably, after tail-vein injection of the nanocomposites followed by single near-infrared laser irradiation, triple-modality therapy of mild PTT, NO therapy, and chemotherapy achieves complete ablation of MCF-7/ADR tumors without tumor recurrence and skin damage in a month.

NO promotes tumor environment normalization and enhances tumor chemotherapy

The abnormal neovessels in TME contribute to metastasis, immunosuppression, and other aspects of malignant progression, leading to resistance to chemotherapy and cancer immunotherapy.3537 Regulating TME by normalizing tumorous vascular functions is becoming a promising treatment method to combine with other traditional therapeutics to enhance overall anti-cancer efficacy.38,39 NO can normalize tumor vasculature by regulating angiogenesis and homeostasis.40,41 However, useful NO delivery schemes must overcome the limitation of a short half-life and the inability to sustain the release. Chen et al. developed a nano-scale carrier NanoNO that can continuously release NO into hepatocellular carcinoma (Figure 2A).42 Low-concentration NO treatment normalized tumor vessels, promoted tumor tissue uptake of chemotherapeutic drugs, and improved therapy effectiveness against primary tumors and metastases through tumor necrosis factors and apoptosis. In addition, low-dose NO from NanoNO reprogrammed the TME to improve the immune efficacy of cancer vaccine immunotherapy, providing a concept for NO-based cancer therapy.

Figure 2.

Figure 2.

NO promotes tumour environment normalization and enhances tumor chemotherapy. A) Schematic of the mechanism NanoNO to suppress hepatocellular carcinoma (HCC) progression in mice. The perivascular gradient produces nitric oxide (NO) and promotes the normalization of vessels and cancer suppression via apoptotic and programmed cell death ligand 1 (PD-L1) pathways in the tumor microenvironment (TME). Reprinted with permission from ref 42. Copyright 2019 Nature. B) NO inhibited collagen expression and improved chemotherapeutic penetration in tumors. (I) Mechanism of matrix metalloproteinase aka matricin (MMP) production and collagen degradation induced by ONOO. (II) NO-generating nanoparticles increased the production of MMP-1 and MMP-2 shown by Western blots. (III) Confocal imaging for detecting collagenase activity under various treatments. (IV) immunofluorescent staining of Collagen I in tumors. Reprinted with permission from ref 47. Copyright 2019 American Chemical Society.

The poor permeability of nanoparticles in solid tumors is considered the main factor limiting the clinical applications of nanomedicine.43,44 Nanoparticles can accumulate at the tumor site through the enhanced penetration and retention (EPR) effect.45,46 However, it is difficult to deliver nanoparticles into tumor parenchyma with a dense interstitial matrix containing collagen and hyaluronan. Fang et al. designed a collagen depletion strategy based on NO-active endogenous matrix metalloproteinases (MMP-1 MMP-2) to deliver chemotherapeutic into solid tumors. The NO-releasing nanoparticles (N@MSN and DN@MSN) were created by loading mesoporous silica nanoparticles (MSN) with NO donor S-nitrosothiol and DOX (Figure 2B).47 The formation of ONOO is the key to the activation of MMP, causing a nearly 2-fold increase of 3-nitrotyrosine, indicating the generation of ONOO, and up to a 3.5-fold increase of MMP-1 and −2 in tumors. Notably, nanoparticles (N@MSN and DN@MSN) significantly reduced collagen I expression and degraded collagen in the tumor TEM, leading to the enhanced penetration of chemotherapeutic DOX and antitumor efficacy without toxic side effects.

NO enhances tumor radiotherapy

Radiotherapy is conventionally used for 50–60% of tumor patients.48,49 However, selectively enhancing the radiation damage to tumor tissues over healthy tissues remains a challenge.50 Radiosensitizers are promising in targeting tumor tissue by producing free radicals and accelerating DNA damage locally.51 Zhao et al. reported a multifunctional nano-radiosensitizer by loading UV-responsive NO donors Roussin’s black salt into scintillating nanoparticles (SCNPs) (Figure 3A).52 Upon X-ray irradiation, the nanocomposite can simultaneously produce NO and O2.−, due to high X-ray-absorbing properties and the radioluminescence of SCNPs. NO and O2.− react further to form a more toxic ONOO for sensitizing tumor radiotherapy by damaging DNA, inhibiting DNA-repair enzyme, and reversing hypoxia-associated radiotherapy resistance. T-SCNPs + X-ray significantly up-regulated the production of a marker of radiotherapy, Phospho-Histone H2A.53 In most cases, poly (ADP-ribose) polymerase (PARP) can promote the repair of damaged DNA by ionizing radiation.54 These results suggested that ONOO can inhibit PARP expression by directly targeting zinc-finger motifs on PARP proteins, increasing the radiation-induced DNA damage by suppressing the PARP-related DNA repair process that decreases the efficiency of radiotherapy. Importantly, RBS-T-SCNPs under X-ray irradiation produce ONOO, leading to nitrification of PARP protein and ultimately promoting cell death. In addition, Shi et al. also proved that NO reduced the expression level of HIF-1α and enhanced the radiotherapy of hypoxic cancer (Figure 3B).55

Figure 3.

Figure 3.

NO enhances cancer radiotherapy. A) X-Ray-inducible ONOO from nanosized scintillators of LiLuF4:Ce3+for radiosensitization. (I) Schematic illustration of RBS-T-SCNPs and the X-ray-controlled ONOO generation for improving radiotherapy. (II) Detection of cell viability of A549 cells, expression of γ-H2AX, and Nitro-Tyrosine with various treatments (scale bar= 20 μm). Reprinted with permission from ref 52. Copyright 2018 WILEY-VCH. B) X-ray-triggered depth-independent on-demand NO-release for hypoxic radiosensitization. (I) Construction of PEG-USMSs-SNO. (II) X-ray-triggered NO release in zebrafish larvae from PEG-USMSs-SNO. (III) X-ray dose-dependent NO release from PEG-USMSs-SNO in one hour. (IV) Cumulative NO release during the first day from PEG-USMSs-SNO after dose-dependent X-ray irradiation. (V) Relative tumor growth and (VI) weight change curve of mice with 4T1 tumors after indicated treatments. Reprinted with permission from ref 55. Copyright 2015 WILEY-VCH.

NO enhances tumor phototherapy

NO enhances photodynamic tumor therapy

Photodynamic therapy (PDT) uses a photosensitizer and light energy to generate ROS to kill disease cells (Table 3).56 PDT is highly biocompatible, highly spatiotemporal selective, and minimally invasive. Under the irradiation of suitable wavelength, the photosensitizer can absorb photons and activate from the ground state to an unstable excited singlet state that quickly transitions into a relatively stable triplet excited state through the intersystem crossing. Excited triplet-state photosensitizers can then transfer electrons to oxygen and other substances to form free radicals, such as O2•−, hydroxyl radical (OH•), and hydroperoxyl radical (HO2) (Type-I process),57 or directly transfer energy to oxygen molecules (the ground state is the triplet state) to form singlet oxygen (1O2) (Type-II process). Thus, Oxygen is the critical factor in photodynamic therapy relying on these ROS. Unfortunately, the hypoxic environment of cancer tissues decreases the concentration of oxygen and ROS production in PDT.58,59 Interestingly, NO can provide synergistic therapeutic efficacy to other treatment modalities via the following three mechanisms. Firstly, NO and ROS can interact to yield ONOO, which is more highly reactive and induces oxidation of DNA and proteins,60 thereby sensitizing radiotherapy, PDT, and chemodynamic therapy (CDT). Secondly, NO inhibits the expression of P-glycoprotein (P-gp) and DNA repair enzymes, thereby improving the therapeutic effect of chemotherapy and radiotherapy. Finally, NO can suppress hypoxia-inducible factor-1α (HIF-1α), thereby reversing the hypoxic TME for the enhancement of PDT and radiotherapy.61 NO can reverse hypoxia by inhibiting cell respiration and metabolism by competitively binding to the oxygen-binding site of mitochondria.6264 Zhang et al. designed a PDT-specific O2 economizer by loading a NO donor and a photosensitizer into poly (D, L-lactide-co-glycolide) for PDT of hypoxia tumors (Figure 4A).65 NO donor release NO in the tumor’s reduction environment to inhibit cell respiration, allowing more O2 in tumor cells and profound enhancement of PDT.22 This O2 restoration stops hypoxia-induced tumor resistance to PDT interventions. In addition, NO can inhibit HIF-α expression to enhance PDT efficacy.66

Table 3.

The Basic Characteristics of ROS, NO, ONOO

Free radical Half life Migration distance Main activities
Superoxide anion (O2.−) 10−6 s 30 nm Reacts with Fe-S proteins, dismutases to H2O2
Hydroxyl radical (OH.) 10−9 s 1 nm Extremely reactive with DNA/RNA, lipids, and proteins
Hydrogen peroxide (H2O2) Chemical stable ≥ 1 mm Reacts with proteins, heme proteins, and DNA/RNA
Singlet oxygen (1O2) 10−6 s 30 nm Oxidases lipids, proteins, and G residues on DNA/RNA
Nitric oxide (NO) < 5 s 100 μm Regulates a variety of biological processes, nitrosative proteins
Peroxynitrite (ONOO) 1.9 s 100 μm Oxidizes and nitrifies DNA/RNA, proteins, and lipids.
Figure 4.

Figure 4.

NO enhances hypoxia tumor photodynamic therapy by inhibiting HIF-α expression and generation of ONOO. A) Schematic illustration of PDT-specific O2 economizer used to inhibit cellular respiration to combat hypoxia tumor. Reprinted with permission from ref 65. Copyright 2019 American Chemical Society. B) A cascade reaction of NO and hydrogen radicals for anti-hypoxia PDT. (I) Schematic generation of the H, O2•–/HO2, NO, ONOO, and 1O2 from DANO and GSH upon light irradiation. (II) Cell viability images with DANO, calcein-AM, and PI staining under normoxic and hypoxic conditions with LED irradiation. Scale bars, 100 μm. Reprinted with permission from ref 71. Copyright 2021 American Chemical Society.

More importantly, Type-I PDT can generate O2•−, which further reacts with nitric oxide to generate more toxic ONOO to improve the PDT of hypoxia tumors.6770 Wang et al. reported an organic molecule, DANO, which possesses a functional π-conjugated donor-acceptor (D-A) backbone, an N-nitrosamine substituent, and two amphiphilic triphenylphosphines (TPP) ligands (Figure 4B).71 The DANO can specifically target to mitochondria of tumor cells. Under LED light irradiation, it can simultaneously produce hydrogen radicals (H), O2•-, and 1O2 through the dual PDT process. In addition, the DANO releases NO in response to the glutathione (GSH), which further reacts with O2•− to generate ONOO. This cascade reaction by NO and H can effectively improve the photodynamic therapy of hypoxia tumors.

NO enhances tumor photothermal therapy

Photothermal therapy (PTT) is an extension of the PDT, converting light energy into heat through photothermal agents (PTA).72,73 To completely ablate the tumor, it usually was heated to more than 50 oC by high-power lasers, thus severely damaging normal tissues.74,75 Zhao et al. reported multifunctional nanoparticles composited from bismuth sulfide (Bi2S3) and thermal-responsive NO donor (bis-N-nitroso compounds, BNN) for NO-sensitized mild PTT (Figure 5A).76 Upon near-infrared (NIR) irradiation, Bi2S3 can absorb light energy and transform it into heat to increase the temperature of the tumor site and realize mild PTT, and simultaneously trigger-releasing of NO that could not only damage the mild PTT-induced autophagic of tumor cells in situ but also can directly kill surrounding cells by NO. In the meantime, Liu et al. also designed NIR light-controlled and organelles-targeted NO delivery nanoplatform for enhancing PTT.77,78 NIR-II (1000–1700 nm) laser possesses lower photon scattering, tissue absorption, and deeper penetration for tissue; thus, it is better spatial resolution and is more suitable for the treatment of deep tissues diseases, compared to the NIR-I (650–950 nm).79 Liu et al. further presented NIR-II responsive hydrogel angiogenesis inhibition agents (WB@hydrogel) based on NO-releasing donors (Figure 5B).80 The wild-type p53 protein was activated by NO generation, to alternate pro-angiogenic to anti-angiogenic TEM. This method completely inhibited tumor growth and achieve an anti-recurrence purpose.

Figure 5.

Figure 5.

NO enhances photothermal therapy for cancer. A) NIR light-triggered NO release for sensitizing mild photothermal therapy (PTT). (I) Schematic illustration of the synthesis of multifunctional BNN-Bi2S3 and NIR triggered NO and mild PTT in cancer therapy. (II) LC3-II, LC3-I, and p62 expression in BEL-7402 cells with different treatments by Western blot. Reprinted with permission from ref 76. Copyright 2018 WILEY-VCH. B) Schematic illustration of NIR-II -responsive NO-release anti-angiogenesis hydrogel. (I) Construction of WB@hydrogel and NIR-II laser-triggered anti-angiogenesis therapy of cancer. (II) Anti-angiogenesis mechanism of WB@hydrogel under laser irradiation. Reprinted with permission from ref 80, Copyright 2021 WILEY-VCH. Vascular endothelial growth factor, VEGF; Basic fibroblast growth factor, bFGF; Thrombospondins 1, TSP-1; Prolyl 4-hydroxylase subunit alpha-2, P4HA2.

NO sensitizes tumor photoacoustic therapy

High-intensity focused ultrasound (HIFU) is being used to treat disease by itself or in combination with other therapies. Chen et al. demonstrated ultrasound could activate H2O2 to generate more reactivity ROS for oxidizing L-Arg to generate NO for suppressing one of the most aggressive and lethal cancers.81 Recently, pulsed laser-triggered ultrasound wave for disease treatment has received widespread attention, which is utilized the probe’s PA effects (shockwave) to target selectively disease tissue with minimal regular cell damage and therapy resistance.8285 Wang et al. constructed a multi-functional nanocapsule (NO-NCPs) by loading a NO donor, EDTA NONOate into an acid-responsive NIR-absorbing polymer (Figure 6).86 Upon entering tumor cells by lysosome-mediated endocytosis, NO-NCPs release NO responsively to the acidic environment. Pulsed laser irradiation generates photoacoustic (PA) cavitation and splits water into H and OH•, in which H further reacts with O2 to generate hydroperoxyl radical (HO2) and then dissociating to form O2•−. In addition, the photoacoustic effect accelerates NO-releasing, which recombines with in situ generated O2•− to produce ONOO, thereby significantly promoting mitochondrial depolarization, apoptosis-related proteins expression, and DNA fragmentation to initiate cancer cell death. PA signal of the NO-NCPs also can provide precise guidance for pulsed laser treatment during diagnosis and treatments. In vivo anti-tumor experiments demonstrated that NO-NCPs had superior tumor inhibition capability and even complete tumor ablation after treatment of 18 days. Precise control of ROS and RNS generation in tumor tissues was achieved and visualized by PA and fluorescence imaging.

Figure 6.

Figure 6.

NO sensitizes photoacoustic therapy by generating ONOO. A) Schematic illustration of photoacoustic (PA) cavitation-triggered ONOO generation for cancer therapy. B) In vitro NO-releasing of NO-NCPs under different treatments. C) Western blotting analysis of apoptosis-related proteins (cleaved-caspase 3 and Nitro-Tyrosine proteins). D) Gel electrophoresis detected DNA fragmentation of EMT6 cells after different treatments. E) In vivo PA imaging of tumor-bearing mice following intravenous injection of NO-NCPs. F) Tumor response to various treatment processes. Reprinted with permission from ref 86. Copyright 2021 WILEY-VCH.

NO enhances tumor chemodynamic therapy

Chemodynamic therapy (CDT) is a developing cancer treatment that utilizes advanced nanotechnology to catalytic hydrogen peroxide in TME into •OH through Fenton and Fenton-like reactions, inducing cell apoptosis and necrosis.87,88 Ding et al. designed a nanoscale coordination polymer (Fe(II)-BNCP) that a glutathione(GSH)-sensitive NO donor (1,5-bis[(L-proline-1-yl)diazen-1-ium-1,2-diol-O2-yl]-2,4-dinitrobenzene, BPDB) was co-coordinate to irons by precipitation and partial ion exchange process ( Figure 7).89 The NO of Fe (II)-BNCP was not leaked in the blood circulation, only the high concentration of GSH in cancer cells led to the specific release of NO, reducing the damage to normal tissues. An overproduction of H2O2 reacts with Fe2+ ions to generate •OH to realize the CDT of the tumor. In addition, the Haber-Weiss reaction of Fe2+ ions with H2O2 generates O2•−, which can further react with NO to produce ONOO. The combined therapy effect of CDT and NO and biocompatibility of the Fe (II)-BNCP was proven in Heps xenograft ICR mouse models. In the meantime, Zhao et al. reported a versatile Cu2+ nanocomposite (UMNOCC-PEG) for luminescence (UCL), CT, and MRI trimodal imaging-guided synergistic CDT/PDT/NO therapy.90 As a result, specifically generated ROS/RNS (NO, •OH, ONOO) to respond TME by the Fenton-like catalytic reaction to cause DNA damage, improving the therapeutic effect.

Figure 7.

Figure 7.

NO enhances chemodynamic therapy for liver tumors. A) Illustration of Fe (III)-BNCP and Fe (II)-BNCP synthesis as nanoscale coordination polymers. B) The mechanism of NO-CDT synergistic therapy using Fe (II)-BNCP for tumor cells. C) NO release at different content of GSH. D) Tumor growth curves during various treatment processes: (a) saline, (b) Zn (II)-DNCP, (c) Fe (II)-DNCP, (d) Zn (II)-BNCP, and (e) Fe (II)-BNCP. Reprinted with permission from ref 89. Copyright 2019 American Chemical Society.

BACTERIA AND BACTERIAL BIOFILMS

Bacterial infections are a major threat to human health due to their high pathogenicity and fatality, causing increasing economic losses worldwide.91 Although the emergence of antibiotics has effectively suppressed bacterial infections, the emergence of bacterial resistance has exacerbated the deterioration of the situation.92 Developing a new generation of antimicrobial drugs and methods to combat bacterial infections is an urgent priority. NO is an excellent antibacterial candidate because of its critical role in the mammalian immune response to pathogens (Table 4).12,93,94 More importantly, NO has a broad spectrum of activity, inducing oxidative and nitrosative damage to DNA, metabolic enzymes, microbial proteins, and membrane structures.95,96 Chemical alteration of DNA by RNS is the main mechanism of NO-mediated antimicrobial action, which is demonstrated by three mechanisms: 1, RNS directly reacts with DNA to inhibit the DNA repair capacity and increase the neration of genotoxic alkylating agents and H2O2. 2, N2O3 can deaminate cytosine, adenine, and guanine. 3, ONOO and NO2. cause DNA strand breaks and abasic sites. Schoenfisch et al. reported a variety of NO carriers based on macromolecules to combat bacterial infections.97100 NO can also sensitize PTT,101 PDT, and CDT to bacteria and enhance their efficacy.102,103 For example, Hu et al. synthesized NO/ formaldehyde (FA) -releasing polymer PEO-b-PNNBM (PNOFA) and self-assembled it into nanoparticles (Figure 8A).104 The nanoparticle simultaneously released NO and FA under irradiation of visible light and could combat both Gram-negative and Gram-positive bacteria without side effects. Xue et al. constructed another nanocomposite by combining photothermal and NO release to achieve a more effective antibacterial effect (Figure 8B).105

Table 4.

NO Nanomedicines for Anti-Bacterial/Biofilma

Nanomedicine NO donor Release condition Working mechanism of NO Refs
AuNC@NO N-Hydroxy-N-nitrosamine NO donor NIR irradiation (heat) NO/PTT synergistically degrades MRSA biofilms 238
PGalNO CouN(NO)-R NIR irradiation High concentration of NO kills bacteria 115
pH@MSN-CaP-NO diazeniumdiolate pH 5.0 NO promotes cornea wound healing 147
BP1 Vesicles oNBN, pNBN, BN UV 365 nm irradiation NO promotes cell migration and viability 149
PU/PPEG-OH-MPS-NO R-SNO Physiological conditions. NO has a broad-spectrum antibacterial property 239
PNOFA R-N-NO Visible light irradiation NO has broad-spectrum antibacterial performance with low drug resistance 104
CoFe2O4@MnFe2O4 nanoparticles R-SNO Magnetothermal Efficiently killing sessile bacteria by rapidly releasing nitric oxide (NO) inside biofilms 240
nbi/NO film N-Diazeniumdiolate Cu2+ to accelerate NO release Nitrosative stress partly causes DNA deamination; Oxidative stress causes membrane destruction through lipid peroxidation, tyrosine nitrosation, and DNA cleavage 241
SNP@MOF@Au-Mal Sodium nitroprusside Photothermal High concentration of NO and derivatives to combat bacteria 101
AI-MPDA L-Arg ROS NO enhances PDT and mild PTT for biofilm elimination 120
α-CD-Ce6-NO-DA nanocarriers R-SNO GSH NO greatly improves the PDT efficiency by releasing ONOO 121
PNO NONOate Physiological conditions NO for synergistic eradication of bacterial biofilms 242
PNBNPs N-Diazeniumdiolate pH High concentration of NO kills bacteria 243
PDA-NO HNP NONOate Physiological conditions NO has a broad-spectrum antibacterial effect 244
UKON-2jNO R-SNO Sunlight Highly reactive ONOO enhances antibacterial effect 94
GEN-NO NONOate Physiological conditions Synergistic NO and antibiotic for biofilm eradication 245
ZnTPyP@NO R-NO Sunlight irradiation Highly reactive ONOO generation enhances PDT 67
UCNP@PCN@LA-PVDF L-Arg ROS Highly reactive ONOO generation enhances PDT 103
PEO-b-PCouNO R-N-NO 410 nm light irradiation NO has a broad-spectrum antibacterial effect 113
Fe3O4@PDA@PAMAM@NONOate NONOate 808 nm laser irradiation (Photothermal) NO damages DNA and kills bacteria 105
TG-NO-B R-SNO 808 nm laser irradiation (Photothermal) Synergistic NO/PTT to overcome MDR Gram-negative bacteria and their biofilms 102
Ti-RP/PCP/RSNO R-SNO 808 nm laser irradiation (Photothermal) Upregulating Opn and Ocn genes and TNF-α 122
PdTPTBP/CouN(NO)-NO2 R-N-NO Red light NO eradiates C. acnes pathogens, with antibacterial, anti-inflammatory, and anti osteoclastogenesis effects. 246
B/MA-GSNO R-SNO Heat Rapid NO releasing for antibacterial efficiency 247
DMAH R-SNO Heat Destroying bacterial nitrogen and respiratory metabolisms 248
a

AuNC@NO, nitric oxide (NO)-releasing gold nanocage; PGalNO; pH@MSN-CaP-NO, Ph@ mesoporous silica nanoparticles-calcium phosphate-NO; BP Vesicles, PEO45-b-PoNBNn; PU/PPEG-OH-MPS-NO, ozone-pretreated polyurethane-PPEG-OH-mercapto-silane-RSNO; PNOFA, PEO-b-PNNBM-NO-formaldehyde; nbi/NO film, branched polyethyleneimine-alginate-NO film; AI-MPDA,L-arginine-ICG-mesoporous polydopamine; PNO, NO-loaded polymer; PNBNPs, surface charge switchable nitric oxide (NO)-releasing nanoparticles; PDA-NO HNP, Polydopamine-NO hollow nanoparticle; UKON-2JNO, NO-Mesoporous organosilica; GEN-NO, gentamicin-NONOate; ZnTPyP@NO, zinc meso-tetra(4-pyridyl)porphyrin@NO; UCNP@PCN@LA-PVDF, upconversion nanoparticle-porphyrinic MOFs@ L-arginine-polyvinylidene fluoride; PEO-b-PCouNO, poly(ethylene oxide)-b-polyCouNO; TG-NO-B, S-nitrosothiols-thiolated graphene-4-mercaptophenylboronic acid; Ti-RP/PCP/RSNO, red phosphorus nanofilm deposited on a titanium implant; B/MA-GSNO, magnetothermal aerogel-S-nitrosoglutathione; DMAH, dual-mode antibacterial hydrogel.

Figure 8.

Figure 8.

NO for antibacterial applications. A) Illustration of visible light-triggered simultaneous release NO and formaldehyde as a broad-spectrum antibacterial. Reprinted with permission from ref 104. Copyright 2021 American Chemical Society. B) Synergistic photothermal and NO antibacterial research based on dendritic Fe3O4@Poly(dopamine)@PAMAM nanocomposite for NO-delivery. Reprinted with permission from ref 105. Copyright 2018 WILEY-VCH. C) Synergistic NO and carbon monoxide (CO) for combating methicillin-resistant Staphylococcus aureus (MRSA) infections. (I) Synthetic routes of PCNO diblock copolymers and their mechanism for combating bacteria. (II) SEM images of E. coli (top) and S. aureus (bottom) incubated with PCNO micelles after irradiation (410 nm light). (III) Quantitative analysis of MRSA infection wound healing after receiving various treatments. (IV) Quantitative analysis of bacteria on days 1, 3, and 5 in wound tissues of MRSA-infected mice receiving different treatments. Reprinted with permission from ref 109. Copyright 2021 WILEY-VCH.

Other gaseous signalling molecules (GSMs), including carbon monoxide (CO) and hydrogen sulfide (H2S), have shown promising therapeutic potential.106,107 Hu et al. synthesized a NO/CO-releasing donor by grafting the NO-releasing N-nitrosamine group108 onto the CO donor 3-hydroxyflavone (3-HF) derivatives (Figure 8C).109 The co-release NO and CO under irradiation of visible light showed excellent antibacterial effects on Gram-positive bacteria, with a combination index of 0.053. Moreover, their design efficiently eradicated methicillin-resistant Staphylococcus aureus (MRSA) infection.

The formation of biofilm makes bacteria less susceptible to antibiotics.110 Studies have shown that the dose of antibiotics used to kill bacteria in biofilms is 1,000 times higher than in the suspension of cells.111 Interestingly, Schoenfisch et al. proved microbial cells could be killed effectively by NO-releasing silica nanoparticles inside P. aeruginosa and E. coli biofilms, S. aureus, S. epidermidis biofilms, and fungus C. Albicans biofilm.112 Hu et al. reported amphiphiles polymer with NO-releasing property, poly(ethylene oxide)-b-polyCouNO (PEO-b-PCouNO), in which CouNO is a NO donor with a coumarin chromophore, featuring visible-light-triggered NO-release (Figure 9A).113 The amphiphilic polymer could load the antibiotic ciprofloxacin (Cip) to achieve co-delivery of NO and Cip by self-assembly into nanoparticles. NO specifically generated in biofilm under light irradiation can efficiently eradicate the biofilm of Pseudomonas aeruginosa, and Cip release in lesions to further strengthen biofilm eradication. Light is a promising excitation source that can be precisely fixed to the treatment site without causing normal tissue damage due to its high spatiotemporal resolution. However, most photo-sensitive NO donors are responsive to UV light, which shows low penetration and high toxicity to tissues,114 so Hu and co-workers developed a red-light responsive NO donor delivery micelle (Figure 9B).115 The micelles could efficiently load Cip to collaborate with NO to combat Pseudomonas aeruginosa (P. aeruginosa) infections, completely eradicated bacteria, and promoted wound healing.

Figure 9.

Figure 9.

NO for antibiofilm eradication. A) Visible-light-triggered NO releasing to eradicate biofilm. (I) Illustration of PEO-b-PCouNO nanoparticle preparation and synergistic therapeutic mechanism of NO and antibiotic against P. aeruginosa biofilm. (II) Two-dimensional and 3D confocal laser scanning microscopy (CLSM) images for detecting P. aeruginosa biofilms eradiation. (III) Quantitative analysis of biofilm viability. (IV) ATP assay for analysis of Planktonic bacteria. Reprinted with permission from ref 113. Copyright 2019 American Chemical Society. B) Red-light triggered NO release for efficient antibacterial treatment. (I) Illustration of the formulation of red-light triggered micelles and the NO-releasing mechanism of CouN(NO)-R derivatives responding to red light. (II) Representative images of the abscess during the treatment process in vivo. Reprinted with permission from ref 115. Copyright 2021 WILEY-VCH.

Phototherapy based on photosensitizers and photothermal agents has received more and more attention in antibacterial applications due to its spatiotemporal controllability, non-invasiveness, and avoidance of drug-resistant bacteria.116118 However, during phototherapy therapy, high local temperatures not only kill bacteria of the lesion tissues but also would destroy the normal cells. In addition, the uneven heat distribution in the biofilm limits its ablation.119 Therefore, the use of low-power laser-triggered phototherapy to eradicate biofilms remains an urgent problem. Cai et al. reported an NO/phototherapeutic nanoplatform (AI-MPDA) by mesoporous polydopamine (MPDA) to load L-arginine (L-Arg) and indocyanine green (ICG) to eliminate established biofilm (Figure 10A).120 Under NIR irradiation, AI-MPDA can generate mild heat (≤45 °C) and ROS, catalyze L-Arg to release NO, and finally achieve severe destruction of the bacterial members. The multifunctional phototherapeutic platform could efficiently eliminate biofilm in a model of the abscess. Ji et al. designed a charge switchable supramolecular nanocarrier with loading NO and a photosensitizer to synergistic NO and PDT to eradicate MRSA formation biofilm (Figure 10B).121

Figure 10.

Figure 10.

NO enhances phototherapy against biofilms. A) NIR-mediated NO-enhanced photodynamic therapy (PDT) and mild photothermal therapy (PTT) for biofilm elimination. (I) Schematic illustration of the mechanism by which NIR triggers NO release to enhance PDT and PTT for biofilm ablation. (II) Scanning electron microscopy (SEM) images of biofilm after various treatments. (III) Live/dead stained 3D confocal laser scanning microscopy (CLSM) of biofilms challenged with other treatments. Reprinted with permission from ref 120. Copyright 2020 American Chemical Society. B) supramolecular nanocarriers with a switchable surface charge for synergistic NO and PDT destruction of biofilms. Reprinted with permission from ref 121. Copyright 2020 American Chemical Society. C) Illustration of the mechanism of MRSA biofilm eradication via NO-triggered gene downregulation. Reprinted with permission from ref 122. Copyright 2020 American Chemical Society.

NO can cooperate with phototherapy to combat biofilms; NO could react with O2.− to generate more reactive ONOO to completely eradicate biofilm infection. Wu et al. designed a multifunctional titanium implant (Ti-RP/PCP/RSNO) for co-delivery of a hydrophilic and viscous hydrogel of poly (vinyl alcohol) modified chitosan, polydopamine, and a red phosphorus nanofilm (Figure 10C).122 Under the irradiation of NIR light, NO and ROS were generated for eradicating MRSA biofilms. The results confirmed that synergistic phototherapy, immunotherapy, and NO therapy have excellent osteogenic and biofilm clearance effects.

WOUND HEALING

The wound-healing process is an integration of these overlapping stages: hemostasis, inflammation, proliferation, and tissue remodeling leading to wound resolution.123,124 NO is a promising wound healing agent,125,126 attributed to regulating various biological processes during wound healing, such as inflammatory response,127 cell proliferation, angiogenesis, microbial elimination, and collagen formation (Table 5).128,129 Shen et al. prepared the biocomposite mats by electrospinning S-nitrosated keratin (KSNO) with polyurethane (PU) and gelatine (Gel) (Figure 11A).130 The mats released NO that promoted the proliferation of L929 murine fibroblasts and human umbilical vein endothelial cells (HUVECs), accelerated the cell adhesion and growth along the random arrangement of electrospun fibers as ECM-mimicking scaffolds for cells. These results showed that the NO-releasing mats could accelerate wound healing without stimulating inflammation. Liu et al. also proved NO released can accelerate the scarless repair of burned skin by inhibiting microorganisms and pro-vascularization activities.131 NO can combine other antibacterial methods to accelerate wound healing.132 For example, Cai et al. reported a ROS-responsive NO generation system (Ce6@Arg-ADP) for synergistic combat bacteria, eventually accelerating wound healing (Figure 11B).133 NO is a vasodilator that stimulates the development of new blood vessels and improves collagen deposition. Yeh et al. reported a NO-loaded Prussian blue nanocube (PBNO) (Figure 11C).134 Blood perfusion improved in a controllable fashion following multi-delivery PBNO colloids to the wound and NIR light irradiation, effectively enhancing the angiogenesis and collagen deposition. Bacterial infection is one of the most limitations of wound healing.135

Table 5.

NO Nanomedicines for Wound Healinga

Nanomedicine NO donor Release condition Working mechanism of NO Refs
SNP@UCM Sodium nitroprusside NIR irradiation Promoting HIF-1α expression, VEGF secretion, and endothelial cell proliferation and migration. 249
PU/Gelatin/KSNO R-SNO Physiological conditions Promoting cell proliferation and adhesion to accelerate wound healing 130
TP-Por CON BNN6 635 nm irradiation (Photothermal) NO/PTT/PDT synergistically kill Gram-positive/negative bacteria via ONOO 250
pH@MSN-CaP-NO diazeniumdiolate pH 5.0 NO promotes cornea wound healing 147
BP1 Vesicles oNBN, pNBN, BN UV 365 nm irradiation NO increases cell migration and viability 149
HA-NO Diazeniumdiolate Physiological conditions NO as an antibacterial agent for promoting wound healing 98
GelMA/HA-DA/GO-βCD-BNN6 BNN6 808 nm laser irradiation (Photothermal) Promoting new blood vessels and collagen deposition to accelerate wound healing 134
MoS2-BNN6 BNN6 808 nm laser irradiation (Photothermal) NO kills the bacteria by damaging DNA, promotes the formation of collagen fibers, and reduces inflammation during wound tissue reconstruction 251
Ch/PAs-Cu R-SNO Cu2+ NO combats bacteria and accelerates wound healing 131
PB-NO R-NO 808 nm laser irradiation (Photothermal) Promoting new blood vessels and collagen deposition to accelerate wound healing 132
Arg-ADP L-Arg H2O2 NO/PDT synergistically overcomes bacterial infections and promotes wound healing 135
NO@HKUST1/PCL/Gel NONOate Physiological conditions Promoting endothelial cell growth, significantly improving angiogenesis and collagen deposition, and reducing inflammatory effects in the wound 138
FBN/PEG NONOate Physiological conditions NO enhances reepithelialisation, collagen deposition, and blood vessel formation 252
AhCeO2 NPs L-Arg H2O2 NO promotes cellular proliferation 253
MA-HA-(MEDN)-NONOates NONOate Physiological conditions Effectively promoting proliferation and migration of fibroblasts 254
CS-PAMAM/NONOate NONOate Physiological conditions NO antibacterial to accelerate wound healing 255
a

SNP@UCM, SNP@MOF-UCNP@ssPDA-Cy7/IR786s; PU/Gel/KSNO, polyurethane/gelatin/S-nitrosated keratin; TP-Por CON, porphyrin-based COF nanosheets; BP Vesicles, PEO45-b-PoNBNn Vesicles; HA-NO, hyaluronic acid-NO; GelMA/HA-DA/GO-βCD-BNN6, methacrylate-gelatin/hyaluronic acid-dopamine hydrogel/ graphene oxide-β-cyclodextrin-BNN6; Ch/PAs-Cu, chitin sponges-proanthocyanidins-Cu2+; PB-NO, Prussian blue-NO; Arg-ADP, L-Arginine-rich amphiphilic dendritic peptide; NO@HKUST1/PCL/Gel, NO@ copper-based metal–organic framework/hydrophobic polycaprolactone/gelatin; FBN/PEG, Pluronic F127-branched polyethylenimine-1-substituted diazen-1-ium-1,2-diolates/PEG; AhCeO2 NPS, hollow CeO2-L-arginine nanoparticles; MA-HA-(MEDN)-NONOates, methacrylate-hyaluronic acid-N-diazeniumdiolate; CS-PAMAM/NONOate, Polyamidoamine dendrimer-grafted chitosan/NONOate.

Figure 11.

Figure 11.

NO accelerates wound healing. A) Keratin composite mats release NO based on S-nitrosated to accelerate wound healing. Reprinted with permission from ref 130. Copyright 2020 Elsevier. B) ROS-triggered NO-releasing for synergistically combat bacterial infection and accelerate wound healing based on L-Arg-enriched amphiphilic peptide. Reprinted with permission from ref 133. Copyright 2021 WILEY-VCH. C) Photon-mediated NO-releasing from hemin-derived colloids to promote angiogenesis, microcirculation, and collagen deposition during wound healing. (I) Illustration of a NO-carrying Prussian blue (PB-NO) nanocubes for NIR-responsive NO-release healing of incisional wounds. (II) In vivo assessment of NO release for incisional wound healing. Reprinted with permission from ref 134. Copyright 2019 American Chemical Society.

Accelerating wound healing is critical for the diabetic.136 Cu2+ generation in the lesion can develop wound healing.137 Xu et al. designed synergistically released NO and Cu2+ nanoplatforms to accelerate diabetic wound healing.138 The prepared nanocomposites accelerated the diabetic wound healing, owing to NO releasing at the wound site promoting angiogenesis, collagen deposition, and endothelial cell growth without stimulating inflammation.139

EYE DISEASES

Glaucoma is a leading cause of blindness in over 70 million people worldwide.140 Ocular hypertension is the primary risk factor, and reduction of intraocular pressure (IOP) is the most effective clinical method to prevent glaucoma vision loss.141 NO could reduce outflow resistance and IOP in animal models and human patients by relaxing trabecular meshwork (TM) containing Schlemm’s cells (Table 6).142 Stevens et al. developed targeting the conventional efflux pathway NO nanoparticle to therapy for glaucoma by specifically controlling NO release in the lesion site (Figure 12A).143 These results have shown that the as-prepared platform can target delivery of NO to the lesion tissue, and achieve a control NO-releasing. Therefore, endogenous stimuli-responsive NO nanomedicine is promising for glaucoma treatment. Chen et al. designed an endogenous NO nanotherapeutic (HOS-JRLO) by biodegradable hollow SiO2 nanoparticles for efficient co-delivery of JS-K and L-Arg for inducing appreciable IOP reduction. This nanotherapeutic was dual stimuli-responsive in that releasing NO by hydrophobic JS-K in response to ascorbic acid, and by L-Arg catalyzed by endothelial NO synthase (Figure 12B).144

Table 6.

NO Nanomedicines for Eye Disease Therapya

Nanomedicine NO donor Release condition Working mechanism of NO Refs
β-galactosidase-loaded PMA capsules β-gal-NONOate β-galactosidase NO-mediated IOP-lowering therapeutics 143
HOS-JRLO JS-K/L-Arginine ascorbic acid/iNOS HOS-JRLO generates more NO to induce a larger IOP reduction 144
PEG-PAspTETA-SNO R-SNO GSH Alleviating high intraocular pressure in mice with glaucoma 256
pH@MSN-CaP-NO diazeniumdiolate pH 5.0 NO promotes cornea wound healing 147
BP1 Vesicles NBN, pNBN, BN UV 365 nm irradiation NO stimulates cornea wound healing 149
BPEI-NO NPs NONOate Physiological conditions NO improves ocular wound recovery 257
a

HOS-JRLO, hollow mesoporous organosilica-JS-K-L-Arginine; PEG-PAspTETA-SNO, PEG-poly(2-acetamido-N-triethylenetetramine-3-nitrosothiol-3-methylbutanamide) aspartamide-S-nitrosothiols; BP Vesicles, PEO45-b-PoNBNn Vesicles; BPEI-NO NPs, silica nanoparticles-branched polyethylene imine-N-diazeniumdiolates.

Figure 12.

Figure 12.

NO for treatment of eye diseases. A) Localized and controlled delivery of NO for glaucoma therapy. Reprinted with permission from ref 143. Copyright 2017 WILEY-VCH. B) NO releasing in intraocular pressure (IOP) reduction pathway for precision glaucoma therapy. Reprinted with permission from ref 144. Copyright 2021 Elsevier. C) Smart NO delivery for corneal wound healing by light-induced acid generation from pH@MSN-CaP-NO. Reprinted with permission from ref 147. Copyright 2016 American Chemical Society. D) Light-triggered NO release from polymersomes for corneal wound healing. Reprinted with permission from ref 149. Copyright 2019 Royal Society of Chemical.

NO participates in the process of wound healing of the cornea.145,146 Kim et al. designed a light-responsive gatekeeper (pH@MSN-CaP-NO) for spatiotemporal-controlled NO delivery (Figure 12C).147 The developed gatekeeper system can store NO stably prior and can promote cornea wound healing. Polymersomes have attracted extensive attention to transport both hydrophilic and hydrophobic drugs.148 Hu et al. constructed NO-releasing vesicles (BP Vesicles) via the self-assembly of NO donor-anchored amphiphiles polymer (Figure 12D).149 The photo-mediated NO release increased cell migration and viability, synergizing the faster corneal healing process.

OSTEOPOROSIS

Osteoporosis can make bones more prone to sudden and unexpected fractures. NO can alleviate osteoporosis by promoting the proliferation and differentiation of osteoblasts.150,151 Sung et al. formulated an injectable microparticle (MP) system to deliver NO donor (NONOate) by integrating phase-change materials capric acid (CA) and octadecane (Figure 13A).152 The CA/OD MPs undergo a solid-to-liquid phase transition, resulting in leakage of the NONOate encapsulated inside. CA could provide an acid environment by deprotonating to promote NONOate decomposition to generate NO. Deprotonated CA captures the generation bubble to form stable micellar. This system can efficiently prevent NO bubbles from collapsing, prolong their half-life, exert a durable therapeutic function, and reverse ovariectomized-induced osteoporosis. Wang et al. developed a bone-targeting NIR photosensitizing NO-generating nanoplatform (UCPA) in response to NIR light (Figure 13B).153 The UV-light responsive NO donor (BNN) could release NO due to the upconversion property of the nanoparticle to convert an irradiating NIR light (808 nm) into UV/blue light. Experiments both in vitro and in vivo showed that UCPA has a good affinity for the bone to directly release NO in bone and reverse osteoporosis. To achieve these targeting and therapeutic properties, upconversion nanoparticles (UCNPs) were carefully designed first with a core–shell–shell structure: (1) a monodisperse spherical core of NaYF4:Yb/Tm with a diameter of ∼26 nm from thermal decomposition, (2) a shell layer of ~2nm thickness containing Nd (NaYF4:Nd/Yb) to enable excitation by the deep penetrating 808 nm NIR light, (3) a further shell about 1 nm thickness of NaYF4 to enhance its upconversion luminescence (UCL) performance. Next these uniform UCNPs were coated with mesoporous silica (MSN) layer of ∼15 nm thickness to make the surface hydrophilic and loadable of the NO donor BNN. And finally, the UCM surface was modified by an amino group to load the NO donor BNN and coated with the outer most layer of PAA-Ald, poly(acrylic acid) (PAA) covalently bond to Ald with a high bone affinity, to form the nanomedicine UCPA for effectively targeting to the bone. The NIR light can reach UCPA accumulated deep in the bone while avoiding the potential damage caused by the UV/blue light, so the release of NO could be precisely controlled to promote osteoblast differentiation and to reverse osteoporosis. Therefore, strategically designed smart NO nanoplatforms are promising in treating osteoporosis.

Figure 13.

Figure 13.

Smart microparticles (MPs) release NO to reverse osteoporosis. A) After subcutaneous administration, MPs were converted into small micelles through a phase transition to generate NO to alleviate osteoporosis. (I) Illustration of fabrication, structure, and functional mechanism of MPs. (II) Micro-CT images and H&E staining images of bones from test rats under varying treatments. (III) Serum biomarker levels of alkaline phosphatase (ALP) and osteocalcin after multiple treatments. Reprinted with permission from ref 152. Copyright 2017 WILEY-VCH. B) NIR-induced NO therapy for osteoporosis mediated by upconversion nanoparticle (UCPA-BNN). (I) Schematic illustration of NIR-triggered NO therapy for Osteoporosis based on UCPN-BNN. (II) Alizarin red staining photos of calcium nodules after varying treatments. (III-V) The expression of Col-1, Runx2, and ALP of MC3T3-E1 cells after varying treatments. Reprinted with permission from ref 153. Copyright 2021 American Chemical Society.

INFECTIOUS DISEASES: VIRUSES AND PARASITES

NO is utilized directly and through the immune system to eliminate many types of infectious agents without unduly harming the host cells and the hosts.154 COVID-19, which immerged in 2019 and was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2),155,156 is responsible for more than 6.5 million deaths according to the World Health Organization (WHO) globally. This pandemic also has resulted in the loss of individual livelihoods and a global economic depression due to prolonged shutdowns. Despite significant progress in clinical vaccines against COVID-19 and a much better understanding of SARS-CoV-2 and COVID-19 prevention and management, the emergence of new SARS-CoV-2 variants likely will overturn the major progress achieved so far in limiting the spread of the virus and its damage.157 Notably, NO can inhibit the replication of several DNA and RNA viruses owing to its high reactivity.158,159

Application of NO in the therapy of COVID-19

SARS-CoV-2 is the latest of the RNA viruses to cause the ongoing pandemic, whereas the therapeutic potential of NO delivery systems against infectious is unlimited based on fundamental mechanisms (Figure 14A).159 NO has been demonstrated to inhibit the replication of SARS-CoV-2 by two distinct mechanisms.160 Firstly, NO or its derivatives cause a reduction in the palmitoylation of nascent expressed spike (S) protein which affects the fusion between the S protein and its cognate receptor, angiotensin-converting enzyme 2. Secondly, NO and its derivatives cause a reduction in viral RNA production in the early steps of viral replication, and this may attribute to an effect on one or both cysteine proteases encoded in Orf1a of SARS-CoV-2. But peroxynitrite does not affect the replication cycle of AARS-CoV-2.

Figure 14.

Figure 14.

Application of NO in the therapy of COVID-19. A) The mechanisms of nitric oxide (NO) antiviral. Reprinted with permission from ref 159. Copyright 2021 Elsevier. B) NO can efficiently inhibit viral RNA. Reprinted with permission from ref 161. Copyright 2005 American Society for Microbiology. C) Mitigation of the replication of SARS-CoV-2 by NO in vitro. (I) Comparison of the cytopathic effect development between cells treated with SNAP (a NO donor) and untreated controls. (II, III) Effect of NO generation on the activity of recombinant SARS-CoV-2 protease. Reprinted with permission from ref 164. Copyright 2020 Elsevier.

Mirazimi et al. found that NO can effectively curb the replication of SARS-CoV-2 in a manner proportional to its concentration. This was achieved by suppressing the production of both viral proteins and RNA (Figure 14B)161. Moreover, inhaling NO has been shown to improve oxygen levels in patients with severe acute respiratory distress syndrome and decrease pulmonary vascular resistance. 162,163 Lundkvist et al. also explored NO’s antiviral properties against SARS-CoV-2 in vitro and found that SNAP (NO donor) could successfully suppress the virus’ replication in Vero E6 cells in a dose-dependent manner. Additionally, the authors observed that NO could decrease the activity of the SARS-CoV-2 protease by nitrosation of its active site cysteine. Although the virus was not fully eradicated at concentrations of 200 μM and 400 μM, SNAP treatment delayed the onset of the viral cytopathic effect in the treated cells, and the protective effect was positively correlated with the level of inhibition of virus replication. These findings provide a theoretical foundation for the potential use of NO in the treatment of COVID-19 in a clinical setting (Figure 14C).164

Tandon et al. conducted a phase III clinical trial to assess the efficacy of NO in eliminating SARS-CoV-2 RNA from the nasal passages.165 The study involved 306 adults (aged 18 to 70) with mild symptomatic COVID-19, with a focus on patients at a high risk of illness progression. Participants self-administered NO through a nasal spray (NONS), using two sprays in each nostril (0.45 mL solution/dose) six times daily for 7 days. The results showed that the high-risk population, defined as unvaccinated individuals over 45 years of age or with one or more comorbidities, experienced a significant reduction in SARS-CoV-2 RNA burden of 93.7% at 24 hours and 99.0% at 48 hours with use of NONS. Inhaling NO was crucial for alleviating COVID-19 symptoms, especially in light of the shortage of ventilators, and was granted emergency authorization by the US FDA to save COVID-19 patients.166 Furthermore, phase 2 clinical trials have been ongoing to evaluate NO for the specific treatment or prevention of COVID-19 (NCT04305457, NCT04306393, NCT04312243). Such approaches hold great promise, particularly for correcting nitric oxide deficiencies caused by infections.167169

Guidelines for NO nanomedicines in infectious diseases

Beyond bacteria and viruses, the important role of NO is known in parasitic infections where malaria is the biggest human burden.170 Interestingly several anti-malaria drugs including Hydroxychloroquine (HCQ) and Artemisinin-based combination therapies (ACTs) were front-runner candidate drugs to combat COVID-19 at least during its onset.171,172 The mechanisms of action again all infectious agents include the direct killing of microbes and their growth inhibition via the immune system.173,174 Strong evidence has been accumulated that NO has many beneficial effects on hosts’ defense against parasites including malaria protozoans but also requires tight control to limit damage to the body’s cells either by cytotoxicity or by inflammation (Figure 15).175

Figure 15.

Figure 15.

Physiological and Immunological mechanism of NO action. Reprinted with permission from ref 175. Copyright 2001 Elsevier. Nitric oxide synthase enzymes, NOS; lipopolysaccharide, LPS; interleukin-1, IL-1; Tumor necrosis factor alpha, TNF-α; Interferon, IFN; messenger RNA, mRNA.

Fighting infectious diseases like COVID-19 and malaria requires faster and more dynamic application of NO gas or NO donor systems. Inhaled NO content is in the mM range, orders of magnitude higher than endogenously produced NO (μM–nM). However, such high NO concentrations (> 1 mm) may cause NO poisoning especially if utilized in emergency rooms or intensive care units. Developing smart NO nanomedicine loaded with NO donors via FDA-approved liposomes or polymersomes, administered as a nasal spray or injection, facilitates the sustained release of NO in the lungs and other targeted organs for effective treatment under life-threatening situations of patients with COVID-19, severe malaria, and other dangerous infections. However, careful guidelines must be developed to ensure the safe and effective use of NO nanomedicine in this setting. (1) Determining the optimal amount of NO to effectively treat COVID-19: The optimal level of NO required to effectively treat COVID-19 is not yet known and will likely vary based on the severity of the illness. It is important to determine the minimum effective concentration of NO for the treatment of COVID-19 while minimizing potential toxicities. The concentrations and durations of NO undergoing clinical or preclinical trials of COVID-19 treatment are summarized in Table 7. (2) Delivery method and system: The delivery method and system for NO nanomedicine will depend on the severity of the illness and the stage of the disease. For example, inhaled NO may be appropriate for mild cases of COVID-19, while intravenous administration may be necessary for severe cases. (3) Potential toxicities: As with any therapeutic agent, it is important to minimize the risk of toxicity while delivering effective therapeutic concentrations of NO to the infection site. (4) Preclinical and clinical testing: Before NO nanomedicine can be used in humans to treat COVID-19, it must undergo rigorous preclinical testing and clinical trials to determine its safety and validate its efficacy. (5) Interactions with other therapies: It is important to consider the potential interactions between NO nanomedicines and other therapeutics used to treat COVID-19, such as antiviral drugs and immunomodulatory agents.

Table 7.

NO for SARS-CoV-2 Infection and COVID-19

NO source Dose Duration Follow-up / d Indications/Results Refs
Inhaled NO gas 20 PPM+2 L/min O2 12–14 h/day 0–6 Improved Covid-19 patient symptoms and full recovery 258
10 PPM+2 L/min O2 7–9
10 PPM+2 L/min O2 10–12

SNAP 200 μM In vitro - Inhibited the replication cycle of SARS-CoV-2 162
400 μM

NONS 2 sprays per nostril (0.45 mL / dose) 6 times daily 18 Accelerated virus clearance (153 patients) 165

SNAP 200 μM In vitro - Reduced SARS-CoV-2 protease activity 164
400 μM

CHALLENGES IN NO NANOMEDICINE

The number of NO nanomedicines is rapidly increasing, but the efficiency of clinical translation is barely satisfactory. All NO nanomedicines face the same set of challenges. First, the safety issues of nanomedicine are the biggest for clinical application. The synthesis of nanocarriers is complex and uses many potentially hazardous chemicals that have yet to receive clinical backing for human use. Even the FDA-approved liposomes, which have been extensively studied, still face complex toxicity issues when combined with different clinical drugs. In addition, the physicochemical properties of nanomedicines, including dimension, shape, surface area, and aggregation, can impact biodistribution and interactions with cells and biomolecules at larger scales, adding to safety concerns. Second, NO is highly reactive and can damage healthy cells and tissues, thus carefully controlling the dosage to minimize its toxicity is crucial. Determining the minimum effective concentration of NO needed to treat the disease while minimizing potential toxicities must remain a key research and development focus. For example, Acute kidney injury (AKI) is a high-burden global disease partially caused by reactive oxygen/nitrogen species (RONS), so NO generating nanomedicines for disease therapy must prevent AKI and other side effects by various smart strategies such as co-development of NO scavengers. Finally, the optimal amount of NO for a specific disease varies with the disease state and severity. Thus, it is highly desirable to image the process of spatiotemporal controllable release of NO by the guidance of multi-model imaging (e.g., MRI, PET, CT, fluorescence imaging) from in vitro to in vivo, favoring the development of NO nanomedicines in the forms of multifunctional theranostics.

OUTLOOK AND SUMMARY

NO is powerful and broad-spectrum in biology and medicine, so its relevance and application are seemingly limitless, and we touch upon just a few additional examples in this outlook below. Vascular stent implantation has become the central therapy to treat cardiovascular diseases, owing to the immediate reopening of acute vessel closure.176 But the long-term clinical success of stenting is limited by in-stent restenosis (i.e., thrombosis and intimal hyperplasia around the implants).177 NO has antiplatelet aggregation and SMC inhibition capabilities.178 Therefore, organic nitrates and nitrites have been used as cardiovascular therapeutic agents to improve the dilation of vascular smooth muscle.179 Indeed, NO not only has been developed for decades for effective cardiovascular disease therapy but also to improve thromboresistance, anti-restenosis, and promote re-endothelialisation after stent implantation.180182 With active ongoing research on the biological mechanism of NO, scientists will surely report more and more NO nanomedicine candidates for the treatment of a variety of additional diseases.183

NO is powerful and broad-spectrum in medicine because its prime targets are sulfhydryl and iron which are central in the physiology and biochemistry of all life and because it is small and lipophilic few cells or microbes could block its entry. But the therapeutic effect of any NO-based drug is dependent on the concentration and duration of NO released.6 Therefore, controlling NO concentration and release rate remains critical for developing helpful therapeutics. New biotechnology and nanomedicine have greatly improved the spatiotemporally controlled release of NO. Physically entrapping, adsorbing, or covalently linking small NO donors with liposomes, polymers, hydrogels, and inorganic/organic hybrid nanoparticles exhibits the following advantages: (1) Extended blood circulation time and enhanced permeability and retention in disease tissue, concurrent delivery of multiple drugs to the disease site for synergistic therapy, (3) the integration of diagnosis and treatment for precise disease therapy.

The development of multifunctional NO nanomedicines and further research on NO therapeutic mechanisms will provide theoretical guidance for clinical translation of next-generation NO drug delivery systems. Although NO’s role in disease treatment and development will remain multi-facet and not wholly understood, NO therapy is an emerging research frontier and deserves much further exploration. In this review, we have detailed the significant recent progress in NO nanomedicines and their relevant biomedical mechanisms for the treatment of a variety of diseases such as cancer, bacterial biofilm, wound healing, eye disease, osteoporosis, and infectious diseases including COVID-19 and malaria. While NO nanomedicines still require much research and development to complete clinical translation, many NO nanomedicines will eventually become life-saving precision therapies.

ACKNOWLEDGMENTS

This research was supported in part by the Intramural Research Program of the NIH, including NIBIB (No. 1ZIAEB000015), the Fujian Normal University Start-up Funds (Y0720302K13), Fujian China, and Innovation team of Photoelectric functional materials and devices for biomedical theranostics of Fujian Normal University (Y07204080K13).

VOCABULARY

nitric oxide nanomedicine

the use of nanocarriers to deliver nitric oxide (NO) to specific areas of the body in a targeted and controlled manner

coronavirus

a virus class typified by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes the current COVID-19 pandemic disease in humans

wound healing

a process by which the body repairs damage to tissues caused by injury or disease, including three main phases of wound healing: inflammation, proliferation, and remodeling

glaucoma

a type of eye disease with high intraocular pressure, which can damage the optic nerve and lead to loss of vision

osteoporosis

a bone disease in which bones become weak and brittle due to loss of tissue, increasing the risk of fractures

Footnotes

The authors declare no conflict of interest.

Contributor Information

Zhixiong Wang, Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA..

Albert Jin, Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA..

Zhen Yang, Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China; Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian, 350117, China.

Wei Huang, Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China; Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian, 350117, China.

REFERENCES

  • (1).Ignarro LJ Nitric Oxide: A Unique Endogenous Signaling Molecule in Vascular Biology (Nobel Lecture). Angew. Chem. Int. Ed 1999, 38, 1882–1892. [DOI] [PubMed] [Google Scholar]
  • (2).Riccio DA; Schoenfisch MH Nitric Oxide Release: Part I. Macromolecular Scaffolds. Chem. Soc. Rev 2012, 41, 3731–3741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (3).Walford G; Loscalzo J. Nitric Oxide in Vascular Biology. J. Thromb. Haemostasis 2003, 1, 2112–2118. [DOI] [PubMed] [Google Scholar]
  • (4).Fang FC Perspectives Series: Host/Pathogen Interactions. Mechanisms of Nitric Oxide-Related Antimicrobial Activity. J. Clin. Investig 1997, 99, 2818–2825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (5).Luo J.-d.; Chen AF Nitric Oxide: A Newly Discovered Function on Wound Healing. Acta Pharmacol. Sin 2005, 26, 259–264. [DOI] [PubMed] [Google Scholar]
  • (6).Mocellin S; Bronte V; Nitti D. Nitric Oxide, a Double Edged Sword in Cancer Biology: Searching for Therapeutic Opportunities. Med. Res. Rev 2007, 27, 317–352. [DOI] [PubMed] [Google Scholar]
  • (7).Roberts JD Jr; Fineman JR; Morin FC; Shaul PW; Rimar S; Schreiber MD; Polin RA; Zwass MS; Zayek MM, Gross I. Inhaled Nitric Oxide and Persistent Pulmonary Hypertension of the Newborn. N. Engl. J. Med 1997, 336, 605–610. [DOI] [PubMed] [Google Scholar]
  • (8).Bloch KD; Ichinose F; Roberts JD Jr; Zapol WM Inhaled No as a Therapeutic Agent. Cardiovasc. Res 2007, 75, 339–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (9).Miller M; Megson I. Recent Developments in Nitric Oxide Donor Drugs. Br. J. Pharmacol 2007, 151, 305–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (10).Wang PG; Xian M; Tang X; Wu X; Wen Z; Cai T; Janczuk AJ Nitric Oxide Donors:  Chemical Activities and Biological Applications. Chem. Rev 2002, 102, 1091–1134. [DOI] [PubMed] [Google Scholar]
  • (11).Carpenter AW; Schoenfisch MH Nitric Oxide Release: Part II. Therapeutic Applications. Chem. Soc. Rev 2012, 41, 3742–3752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (12).Schairer DO; Chouake JS; Nosanchuk JD; Friedman AJ The Potential of Nitric Oxide Releasing Therapies as Antimicrobial Agents. Virulence 2012, 3, 271–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (13).Fukumura D; Kashiwagi S; Jain RK The Role of Nitric Oxide in Tumour Progression. Nat. Rev. Cancer 2006, 6, 521–534. [DOI] [PubMed] [Google Scholar]
  • (14).Hou J; Pan Y; Zhu D; Fan Y; Feng G; Wei Y; Wang H; Qin K; Zhao T; Yang Q; Zhu Y; Che Y; Liu Y; Cheng J; Kong D; Wang PG; Shen J; Zhao Q. Targeted Delivery of Nitric Oxide Via a ‘Bump-and-Hole’-Based Enzyme–Prodrug Pair. Nat. Chem. Biol 2019, 15, 151–160. [DOI] [PubMed] [Google Scholar]
  • (15).Ignarro LJ Nitric Oxide: Biology and Pathobiology. Academic press: 2000. [Google Scholar]
  • (16).Yang T; Zelikin AN; Chandrawati R. Progress and Promise of Nitric Oxide‐Releasing Platforms. Adv. Sci 2018, 5, 1701043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (17).Lisi F; Zelikin AN; Chandrawati R. Nitric Oxide to Fight Viral Infections. Adv. Sci 2021, 8, 2003895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (18).Martel J; Ko Y-F; Young JD; Ojcius DM Could Nasal Nitric Oxide Help to Mitigate the Severity of Covid-19? Microbes Infect. 2020, 22, 168–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (19).Sung H; Ferlay J; Siegel RL; Laversanne M; Soerjomataram I; Jemal A; Bray F. Global Cancer Statistics 2020: Globocan Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin 2021, 71, 209–249. [DOI] [PubMed] [Google Scholar]
  • (20).Liu Y; Wang X, Li J, Tang J, Li B; Zhang Y; Gu N; Yang F. Sphingosine 1-Phosphate Liposomes for Targeted Nitric Oxide Delivery to Mediate Anticancer Effects against Brain Glioma Tumors. Adv. Mater 2021, 33, 2101701. [DOI] [PubMed] [Google Scholar]
  • (21).Wan M; Chen H; Wang Q; Niu Q; Xu P; Yu Y; Zhu T; Mao C; Shen J. Bio-Inspired Nitric-Oxide-Driven Nanomotor. Nat. Commun 2019, 10, 966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (22).Zheng D-W; Chen Y; Li Z-H; Xu L; Li C-X; Li B; Fan J-X; Cheng S-X; Zhang X-Z Optically-Controlled Bacterial Metabolite for Cancer Therapy. Nat. Commun 2018, 9, 1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (23).Ridnour LA; Thomas DD; Donzelli S; Espey MG; Roberts DD; Wink DA; Isenberg JS The Biphasic Nature of Nitric Oxide Responses in Tumor Biology. Antioxid. Redox Signaling 2006, 8, 1329–1337. [DOI] [PubMed] [Google Scholar]
  • (24).Hirst D; Robson T. Nitric Oxide in Cancer Therapeutics: Interaction with Cytotoxic Chemotherapy. Curr. Pharm. Des 2010, 16, 411–420. [DOI] [PubMed] [Google Scholar]
  • (25).Liu Z; Zhong Y; Zhou X; Huang X; Zhou J; Huang D; Li Y; Wang Z; Dong B; Qiao H; Chen W. Inherently Nitric Oxide Containing Polymersomes Remotely Regulated by Nir for Improving Multi-Modal Therapy on Drug Resistant Cancer. Biomaterials 2021, 277, 121118. [DOI] [PubMed] [Google Scholar]
  • (26).Kim J; Yung BC; Kim WJ; Chen X. Combination of Nitric Oxide and Drug Delivery Systems: Tools for Overcoming Drug Resistance in Chemotherapy. J. Control. Release 2017, 263, 223–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (27).Callapina M; Zhou J; Schmid T; Köhl R; Brüne B. No Restores Hif-1α Hydroxylation During Hypoxia: Role of Reactive Oxygen Species. Free Radical Biol. Med 2005, 39, 925–936. [DOI] [PubMed] [Google Scholar]
  • (28).Marshall HE; Stamler JS Inhibition of Nf-Κb by S-Nitrosylation. Biochemistry 2001, 40, 1688–1693. [DOI] [PubMed] [Google Scholar]
  • (29).Wang L; Chang Y; Feng Y; Li X; Cheng Y; Jian H; Ma X; Zheng R; Wu X; Xu K; Zhang H. Nitric Oxide Stimulated Programmable Drug Release of Nanosystem for Multidrug Resistance Cancer Therapy. Nano Lett. 2019, 19, 6800–6811. [DOI] [PubMed] [Google Scholar]
  • (30).Zhang X; Tian G; Yin W; Wang L; Zheng X; Yan L; Li J; Su H; Chen C; Gu Z; Zhao Y. Controllable Generation of Nitric Oxide by Near-Infrared-Sensitized Upconversion Nanoparticles for Tumor Therapy. Adv. Funct. Mater 2015, 25, 3049–3056. [Google Scholar]
  • (31).Chung M-F; Liu H-Y; Lin K-J; Chia W-T; Sung H-W A PH-Responsive Carrier System That Generates No Bubbles to Trigger Drug Release and Reverse P-Glycoprotein-Mediated Multidrug Resistance. Angew. Chem. Int. Ed 2015, 54, 9890–9893. [DOI] [PubMed] [Google Scholar]
  • (32).Deng Y; Wang Y; Jia F; Liu W; Zhou D; Jin Q; Ji J. Tailoring Supramolecular Prodrug Nanoassemblies for Reactive Nitrogen Species-Potentiated Chemotherapy of Liver Cancer. ACS Nano 2021, 15, 8663–8675. [DOI] [PubMed] [Google Scholar]
  • (33).Chu C; Lyu X; Wang Z; Jin H; Lu S; Xing D; Hu X. Cocktail Polyprodrug Nanoparticles Concurrently Release Cisplatin and Peroxynitrite-Generating Nitric Oxide in Cisplatin-Resistant Cancers. Chem. Eng. J 2020, 402, 126125. [Google Scholar]
  • (34).Ding Y; Du C; Qian J; Dong C-M NIR-Responsive Polypeptide Nanocomposite Generates No Gas, Mild Photothermia, and Chemotherapy to Reverse Multidrug-Resistant Cancer. Nano Lett. 2019, 19, 4362–4370. [DOI] [PubMed] [Google Scholar]
  • (35).Gilkes DM; Semenza GL; Wirtz D. Hypoxia and the Extracellular Matrix: Drivers of Tumour Metastasis. Nat. Rev. Cancer 2014, 14, 430–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (36).Jain RK; Munn LL; Fukumura D. Dissecting Tumour Pathophysiology Using Intravital Microscopy. Nat. Rev. Cancer 2002, 2, 266–276. [DOI] [PubMed] [Google Scholar]
  • (37).Schaaf MB; Garg AD; Agostinis P. Defining the Role of the Tumor Vasculature in Antitumor Immunity and Immunotherapy. Cell Death Dis. 2018, 9, 115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (38).Jain RK Normalization of Tumor Vasculature: An Emerging Concept in Antiangiogenic Therapy. Science 2005, 307, 58–62. [DOI] [PubMed] [Google Scholar]
  • (39).Jain RK Normalizing Tumor Microenvironment to Treat Cancer: Bench to Bedside to Biomarkers. J. Clin. Oncol 2013, 31, 2205–2218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (40).Huang Z; Fu J; Zhang Y. Nitric Oxide Donor-Based Cancer Therapy: Advances and Prospects. J. Med. Chem 2017, 60, 7617–7635. [DOI] [PubMed] [Google Scholar]
  • (41).Kashiwagi S; Tsukada K; Xu L; Miyazaki J; Kozin SV; Tyrrell JA; Sessa WC; Gerweck LE; Jain RK; Fukumura D. Perivascular Nitric Oxide Gradients Normalize Tumor Vasculature. Nat. Med 2008, 14, 255–257. [DOI] [PubMed] [Google Scholar]
  • (42).Sung Y-C; Jin P-R; Chu L-A; Hsu F-F; Wang M-R; Chang C-C; Chiou S-J; Qiu JT; Gao D-Y; Lin C-C; Chen Y-S; Hsu Y-C; Wang J; Wang F-N; Yu P-L; Chiang A-S; Wu AY-T; Ko JJ-S; Lai CP-K; Lu T-T; Chen Y. Delivery of Nitric Oxide with a Nanocarrier Promotes Tumour Vessel Normalization and Potentiates Anti-Cancer Therapies. Nat. Nanotechnol 2019, 14, 1160–1169. [DOI] [PubMed] [Google Scholar]
  • (43).Wong C; Stylianopoulos T; Cui J; Martin J; Chauhan VP; Jiang W; Popović Z; Jain RK; Bawendi MG; Fukumura D. Multistage Nanoparticle Delivery System for Deep Penetration into Tumor Tissue. Proc. Natl. Acad. Sci. U.S.A 2011, 108, 2426–2431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (44).Han X; Li Y; Xu Y; Zhao X; Zhang Y; Yang X; Wang Y; Zhao R; Anderson GJ; Zhao Y; Nie G. Reversal of Pancreatic Desmoplasia by Re-Educating Stellate Cells with a Tumour Microenvironment-Activated Nanosystem. Nat. Commun 2018, 9, 3390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (45).Maeda H. Toward a Full Understanding of the Epr Effect in Primary and Metastatic Tumors as Well as Issues Related to Its Heterogeneity. Adv. Drug Deliv. Rev 2015, 91, 3–6. [DOI] [PubMed] [Google Scholar]
  • (46).Ekdawi SN; Jaffray DA; Allen C. Nanomedicine and Tumor Heterogeneity: Concept and Complex Reality. Nano Today 2016, 11, 402–414. [Google Scholar]
  • (47).Dong X; Liu H-J; Feng H-Y; Yang S-C; Liu X-L; Lai X; Lu Q; Lovell JF; Chen H-Z; Fang C. Enhanced Drug Delivery by Nanoscale Integration of a Nitric Oxide Donor to Induce Tumor Collagen Depletion. Nano Lett. 2019, 19, 997–1008. [DOI] [PubMed] [Google Scholar]
  • (48).Palacios Eito A; Cabezas SG; Ugalde PF; Del Campo ER; Romero AO; Martín MDMP; Arjona JMR; Paredes MM Characterization and Adequacy of the Use of Radiotherapy and Its Trend in Time. Radiother. Oncol 2013, 106, 260–265. [DOI] [PubMed] [Google Scholar]
  • (49).DuRoss AN; Neufeld MJ; Rana S; Thomas CR; Sun C. Integrating Nanomedicine into Clinical Radiotherapy Regimens. Adv. Drug Deliv. Rev 2019, 144, 35–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (50).Lin Y-J; Chen C-C; Nguyen D; Su H-R; Lin K-J; Chen H-L; Hu Y-J; Lai P-L; Sung H-W Biomimetic Engineering of a Scavenger-Free Nitric Oxide-Generating/Delivering System to Enhance Radiation Therapy. Small 2020, 16, 2000655. [DOI] [PubMed] [Google Scholar]
  • (51).Wang H; Mu X; He H; Zhang X-D Cancer Radiosensitizers. Trends Pharmacol. Sci 2018, 39, 24–48. [DOI] [PubMed] [Google Scholar]
  • (52).Du Z; Zhang X; Guo Z; Xie J; Dong X; Zhu S; Du J; Gu Z; Zhao Y. X‐Ray‐Controlled Generation of Peroxynitrite Based on Nanosized Liluf4: Ce3+ Scintillators and Their Applications for Radiosensitization. Adv. Mater 2018, 30, 1804046. [DOI] [PubMed] [Google Scholar]
  • (53).Rogakou EP; Pilch DR; Orr AH; Ivanova VS; Bonner WM DNA Double-Stranded Breaks Induce Histone H2ax Phosphorylation on Serine 139*. J. Biol. Chem 1998, 273, 5858–5868. [DOI] [PubMed] [Google Scholar]
  • (54).Pieper AA; Verma A; Zhang J; Snyder SH Poly (Adp-Ribose) Polymerase, Nitric Oxide and Cell Death. Trends Pharmacol. Sci 1999, 20, 171–181. [DOI] [PubMed] [Google Scholar]
  • (55).Fan W; Bu W; Zhang Z; Shen B; Zhang H; He Q; Ni D; Cui Z; Zhao K; Bu J; Du J; Liu J; Shi J. X‐Ray Radiation‐Controlled NO‐Release for on‐Demand Depth‐Independent Hypoxic Radiosensitization. Angew. Chem. Int. Ed 2015, 54, 14026–14030. [DOI] [PubMed] [Google Scholar]
  • (56).Dolmans DEJGJ; Fukumura D; Jain RK Photodynamic Therapy for Cancer. Nat. Rev. Cancer 2003, 3, 380–387. [DOI] [PubMed] [Google Scholar]
  • (57).Castano AP; Demidova TN; Hamblin MR Mechanisms in Photodynamic Therapy: Part One—Photosensitizers, Photochemistry and Cellular Localization. Photodiagn. Photodyn. Ther 2004, 1, 279–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (58).Ji Z; Yang G; Shahzidi S; Tkacz-Stachowska K; Suo Z; Nesland JM; Peng Q. Induction of Hypoxia-Inducible Factor-1α Overexpression by Cobalt Chloride Enhances Cellular Resistance to Photodynamic Therapy. Cancer Lett. 2006, 244, 182–189. [DOI] [PubMed] [Google Scholar]
  • (59).Zheng D-W; Li B; Li C-X; Fan J-X; Lei Q; Li C; Xu Z; Zhang X-Z Carbon-Dot-Decorated Carbon Nitride Nanoparticles for Enhanced Photodynamic Therapy against Hypoxic Tumor Via Water Splitting. ACS Nano 2016, 10, 8715–8722. [DOI] [PubMed] [Google Scholar]
  • (60).Szabó C; Ischiropoulos H; Radi R. Peroxynitrite: Biochemistry, Pathophysiology and Development of Therapeutics. Nat. Rev. Drug Discov 2007, 6, 662–680. [DOI] [PubMed] [Google Scholar]
  • (61).Zhang X; Yang S; Wang Q; Ye W; Liu S; Wang X; Zhang Z; Cao L; Jiang X. Tailored Theranostic Nanoparticles Cause Efficient Ferroptosis in Head and Neck Squamous Cell Carcinoma through a Reactive Oxygen Species “Butterfly Effect”. Chem. Eng. J 2021, 423, 130083. [Google Scholar]
  • (62).Shiva S; Brookes PS; Patel RP; Anderson PG; Darley-Usmar VM Nitric Oxide Partitioning into Mitochondrial Membranes and the Control of Respiration at Cytochrome <Em>C</Em> Oxidase. Proc. Natl. Acad. Sci. U.S.A 2001, 98, 7212–7217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (63).Beltrán B; Quintero M; García-Zaragozá E; O’Connor E; Esplugues JV; Moncada S. Inhibition of Mitochondrial Respiration by Endogenous Nitric Oxide: A Critical Step in Fas Signaling. Proc. Natl. Acad. Sci. U.S.A 2002, 99, 8892–8897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (64).Cao Y; Liu M; Cheng J; Yin J; Huang C; Cui H; Zhang X; Zhao G. Acidity-Triggered Tumor-Targeted Nanosystem for Synergistic Therapy Via a Cascade of Ros Generation and No Release. ACS Appl. Mater. Interfaces 2020, 12, 28975–28984. [DOI] [PubMed] [Google Scholar]
  • (65).Yu W; Liu T; Zhang M; Wang Z; Ye J; Li C-X; Liu W; Li R; Feng J; Zhang X-Z O2 Economizer for Inhibiting Cell Respiration to Combat the Hypoxia Obstacle in Tumor Treatments. ACS Nano 2019, 13, 1784–1794. [DOI] [PubMed] [Google Scholar]
  • (66).Hagen T; Taylor CT; Lam F; Moncada S. Redistribution of Intracellular Oxygen in Hypoxia by Nitric Oxide: Effect on HIF1α. Science 2003, 302, 1975–1978. [DOI] [PubMed] [Google Scholar]
  • (67).Wang D; Niu L; Qiao Z-Y; Cheng D-B; Wang J; Zhong Y; Bai F; Wang H; Fan H. Synthesis of Self-Assembled Porphyrin Nanoparticle Photosensitizers. ACS Nano 2018, 12, 3796–3803. [DOI] [PubMed] [Google Scholar]
  • (68).Parisi C; Failla M; Fraix A; Menilli L; Moret F; Reddi E; Rolando B; Spyrakis F; Lazzarato L; Fruttero R; Gasco A; Sortino S. A Generator of Peroxynitrite Activatable with Red Light. Chem. Sci 2021, 12, 4740–4746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (69).Deng Y; Jia F; Chen S; Shen Z; Jin Q; Fu G; Ji J. Nitric Oxide as an All-Rounder for Enhanced Photodynamic Therapy: Hypoxia Relief, Glutathione Depletion and Reactive Nitrogen Species Generation. Biomaterials 2018, 187, 55–65. [DOI] [PubMed] [Google Scholar]
  • (70).Wan S-S; Zeng J-Y; Cheng H; Zhang X-Z Ros-Induced No Generation for Gas Therapy and Sensitizing Photodynamic Therapy of Tumor. Biomaterials 2018, 185, 51–62. [DOI] [PubMed] [Google Scholar]
  • (71).Sun J; Cai X; Wang C; Du K; Chen W; Feng F; Wang S. Cascade Reactions by Nitric Oxide and Hydrogen Radical for Anti-Hypoxia Photodynamic Therapy Using an Activatable Photosensitizer. J. Am. Chem. Soc 2021, 143, 868–878. [DOI] [PubMed] [Google Scholar]
  • (72).Hirsch LR; Stafford RJ; Bankson JA; Sershen SR; Rivera B; Price RE; Hazle JD; Halas NJ; West JL Nanoshell-Mediated near-Infrared Thermal Therapy of Tumors under Magnetic Resonance Guidance. Proc. Natl. Acad. Sci. U.S.A 2003, 100, 13549–13554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (73).Cheng L; Wang C; Feng L; Yang K; Liu Z. Functional Nanomaterials for Phototherapies of Cancer. Chem. Rev 2014, 114, 10869–10939. [DOI] [PubMed] [Google Scholar]
  • (74).Wu C; Wang D; Cen M; Cao L; Ding Y; Wang J; Yuan X; Wang Y; Chen T; Yao Y. Mitochondria-Targeting No Gas Nanogenerator for Augmenting Mild Photothermal Therapy in the Nir-Ii Biowindow. ChemComm 2020, 56, 14491–14494. [DOI] [PubMed] [Google Scholar]
  • (75).Wen L; Liu H; Hu C; Wei Z; Meng Y; Lu C; Su Y; Lu L; Liang H; Xu Q; Zhan M. Thermoacoustic Imaging-Guided Thermo-Chemotherapy for Hepatocellular Carcinoma Sensitized by a Microwave-Responsive Nitric Oxide Nanogenerator. ACS Appl. Mater. Interfaces 2023, 15, 10477–10491. [DOI] [PubMed] [Google Scholar]
  • (76).Zhang X; Du J; Guo Z; Yu J; Gao Q; Yin W; Zhu S; Gu Z; Zhao Y. Efficient near Infrared Light Triggered Nitric Oxide Release Nanocomposites for Sensitizing Mild Photothermal Therapy. Adv. Sci 2019, 6, 1801122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (77).Xiang H-J; Guo M; An L; Yang S-P; Zhang Q-L; Liu J-G A Multifunctional Nanoplatform for Lysosome Targeted Delivery of Nitric Oxide and Photothermal Therapy under 808 Nm near-Infrared Light. J Mater Chem B 2016, 4, 4667–4674. [DOI] [PubMed] [Google Scholar]
  • (78).Guo M; Xiang H-J; Wang Y; Zhang Q-L; An L; Yang S-P; Ma Y; Wang Y; Liu J-G Ruthenium Nitrosyl Functionalized Graphene Quantum Dots as an Efficient Nanoplatform for Nir-Light-Controlled and Mitochondria-Targeted Delivery of Nitric Oxide Combined with Photothermal Therapy. ChemComm 2017, 53, 3253–3256. [DOI] [PubMed] [Google Scholar]
  • (79).Yang Z; Li L; Jin AJ; Huang W; Chen X. Rational Design of Semiconducting Polymer Brushes as Cancer Theranostics. Mater. Horiz 2020, 7, 1474–1494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (80).Zhao S; Zhang L; Deng L; Ouyang J; Xu Q; Gao X; Zeng Z; Liu Y-N NIR-II Responsive Hydrogel as an Angiogenesis Inhibition Agent for Tumor Microenvironment Reprogramming. Small 2021, 17, 2103003. [DOI] [PubMed] [Google Scholar]
  • (81).Zhang K; Xu H; Jia X; Chen Y; Ma M; Sun L; Chen H. Ultrasound-Triggered Nitric Oxide Release Platform Based on Energy Transformation for Targeted Inhibition of Pancreatic Tumor. ACS Nano 2016, 10, 10816–10828. [DOI] [PubMed] [Google Scholar]
  • (82).Zang Y; Wei Y; Shi Y; Chen Q; Xing D. Chemo/Photoacoustic Dual Therapy with Mrna-Triggered Dox Release and Photoinduced Shockwave Based on a DNA-Gold Nanoplatform. Small 2016, 12, 756–769. [DOI] [PubMed] [Google Scholar]
  • (83).Shi Y; Yang S; Xing D. New Insight into Photoacoustic Conversion Efficiency by Plasmon-Mediated Nanocavitation: Implications for Precision Theranostics. Nano Res. 2017, 10, 2800–2809. [Google Scholar]
  • (84).Lukianova-Hleb EY; Kim Y-S; Belatsarkouski I; Gillenwater AM; O’Neill BE; Lapotko DO Intraoperative Diagnostics and Elimination of Residual Microtumours with Plasmonic Nanobubbles. Nat. Nanotechnol 2016, 11, 525–532. [DOI] [PubMed] [Google Scholar]
  • (85).Teirlinck E; Xiong R; Brans T; Forier K; Fraire J; Van Acker H; Matthijs N; De Rycke R; De Smedt SC; Coenye T; Braeckmans K. Laser-Induced Vapour Nanobubbles Improve Drug Diffusion and Efficiency in Bacterial Biofilms. Nat. Commun 2018, 9, 4518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (86).Wang Z; Zhan M; Li W; Chu C; Xing D; Lu S; Hu X. Photoacoustic Cavitation-Ignited Reactive Oxygen Species to Amplify Peroxynitrite Burst by Photosensitization-Free Polymeric Nanocapsules. Angew. Chem. Int. Ed 2021, 60, 4720–4731. [DOI] [PubMed] [Google Scholar]
  • (87).Tang Z; Liu Y; He M; Bu W. Chemodynamic Therapy: Tumour Microenvironment-Mediated Fenton and Fenton-Like Reactions. Angew. Chem. Int. Ed 2019, 58, 946–956. [DOI] [PubMed] [Google Scholar]
  • (88).Wang X; Zhong X; Liu Z; Cheng L. Recent Progress of Chemodynamic Therapy-Induced Combination Cancer Therapy. Nano Today 2020, 35, 100946. [Google Scholar]
  • (89).Hu Y; Lv T; Ma Y; Xu J; Zhang Y; Hou Y; Huang Z; Ding Y. Nanoscale Coordination Polymers for Synergistic No and Chemodynamic Therapy of Liver Cancer. Nano Lett. 2019, 19, 2731–2738. [DOI] [PubMed] [Google Scholar]
  • (90).Liu S; Li W; Dong S; Zhang F; Dong Y; Tian B; He F; Gai S; Yang P. An All-in-One Theranostic Nanoplatform Based on Upconversion Dendritic Mesoporous Silica Nanocomposites for Synergistic Chemodynamic/Photodynamic/Gas Therapy. Nanoscale 2020, 12, 24146–24161. [DOI] [PubMed] [Google Scholar]
  • (91).Engler AC; Wiradharma N; Ong ZY; Coady DJ; Hedrick JL; Yang Y-Y Emerging Trends in Macromolecular Antimicrobials to Fight Multi-Drug-Resistant Infections. Nano Today 2012, 7, 201–222. [Google Scholar]
  • (92).Coates AR; Halls G; Hu Y. Novel Classes of Antibiotics or More of the Same? Br. J. Pharmacol 2011, 163, 184–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (93).Yang L; Feura ES; Ahonen MJR; Schoenfisch MH Nitric Oxide–Releasing Macromolecular Scaffolds for Antibacterial Applications. Adv. Healthc. Mater 2018, 7, 1800155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (94).Gehring J; Trepka B; Klinkenberg N; Bronner H; Schleheck D; Polarz S. Sunlight-Triggered Nanoparticle Synergy: Teamwork of Reactive Oxygen Species and Nitric Oxide Released from Mesoporous Organosilica with Advanced Antibacterial Activity. J. Am. Chem. Soc 2016, 138, 3076–3084. [DOI] [PubMed] [Google Scholar]
  • (95).Privett BJ; Broadnax AD; Bauman SJ; Riccio DA; Schoenfisch MH Examination of Bacterial Resistance to Exogenous Nitric Oxide. Nitric Oxide 2012, 26, 169–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (96).Callari FL; Sortino S. Amplified Nitric Oxide Photorelease in DNA Proximity. ChemComm 2008, 1971–1973. [DOI] [PubMed] [Google Scholar]
  • (97).Lu Y; Slomberg DL; Schoenfisch MH Nitric Oxide-Releasing Chitosan Oligosaccharides as Antibacterial Agents. Biomaterials 2014, 35, 1716–1724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (98).Maloney SE; McGrath KV; Ahonen MJR; Soliman DS; Feura ES; Hall HR; Wallet SM; Maile R; Schoenfisch MH Nitric Oxide-Releasing Hyaluronic Acid as an Antibacterial Agent for Wound Therapy. Biomacromolecules 2021, 22, 867–879. [DOI] [PubMed] [Google Scholar]
  • (99).Yang L; Teles F; Gong W; Dua SA; Martin L; Schoenfisch MH Antibacterial Action of Nitric Oxide-Releasing Hyperbranched Polymers against Ex Vivo Dental Biofilms. Dent. Mater 2020, 36, 635–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (100).Yang L; Feura ES; Ahonen MJR; Schoenfisch MH Nitric Oxide-Releasing Macromolecular Scaffolds for Antibacterial Applications. Adv. Healthc. Mater 2018, 7, e1800155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (101).Wu Y; Deng G; Jiang K; Wang H; Song Z; Han H. Photothermally Triggered Nitric Oxide Nanogenerator Targeting Type Iv Pili for Precise Therapy of Bacterial Infections. Biomaterials 2021, 268, 120588. [DOI] [PubMed] [Google Scholar]
  • (102).Zhao B; Wang H; Dong W; Cheng S; Li H; Tan J; Zhou J; He W; Li L; Zhang J; Luo G; Qian W. A Multifunctional Platform with Single-Nir-Laser-Triggered Photothermal and No Release for Synergistic Therapy against Multidrug-Resistant Gram-Negative Bacteria and Their Biofilms. J. Nanobiotechnol 2020, 18, 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (103).Sun J; Fan Y; Ye W; Tian L; Niu S; Ming W; Zhao J; Ren L. Near-Infrared Light Triggered Photodynamic and Nitric Oxide Synergistic Antibacterial Nanocomposite Membrane. Chem. Eng. J 2021, 417, 128049. [Google Scholar]
  • (104).Duan Y; He K; Zhang G; Hu J. Photoresponsive Micelles Enabling Codelivery of Nitric Oxide and Formaldehyde for Combinatorial Antibacterial Applications. Biomacromolecules 2021, 22, 2160–2170. [DOI] [PubMed] [Google Scholar]
  • (105).Yu S;Li G; Liu R; Ma D; Xue W. Dendritic Fe3O4@Poly(Dopamine)@Pamam Nanocomposite as Controllable No-Releasing Material: A Synergistic Photothermal and No Antibacterial Study. Adv. Funct. Mater 2018, 28, 1707440. [Google Scholar]
  • (106).Hu J; Fang Y; Huang X; Qiao R; Quinn JF; Davis TP Engineering Macromolecular Nanocarriers for Local Delivery of Gaseous Signaling Molecules. Adv. Drug Deliv. Rev 2021, 179, 114005. [DOI] [PubMed] [Google Scholar]
  • (107).Chen L; Zhou S-F; Su L; Song J. Gas-Mediated Cancer Bioimaging and Therapy. ACS Nano 2019, 13, 10887–10917. [DOI] [PubMed] [Google Scholar]
  • (108).Zhou EY; Knox HJ; Reinhardt CJ; Partipilo G; Nilges MJ; Chan J. Near-Infrared Photoactivatable Nitric Oxide Donors with Integrated Photoacoustic Monitoring. J. Am. Chem. Soc 2018, 140, 11686–11697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (109).Gao L; Cheng J; Shen Z; Zhang G; Liu S; Hu J. Orchestrating Nitric Oxide and Carbon Monoxide Signaling Molecules for Synergistic Treatment of Mrsa Infections. Angew. Chem. Int. Ed 2022, 61, e202112782. [DOI] [PubMed] [Google Scholar]
  • (110).Lebeaux D; Ghigo J-M; Beloin C. Biofilm-Related Infections: Bridging the Gap between Clinical Management and Fundamental Aspects of Recalcitrance toward Antibiotics. Microbiol. Mol. Biol. Rev 2014, 78, 510–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (111).Smith AW Biofilms and Antibiotic Therapy: Is There a Role for Combating Bacterial Resistance by the Use of Novel Drug Delivery Systems? Adv. Drug Delivery Rev 2005, 57, 1539–1550. [DOI] [PubMed] [Google Scholar]
  • (112).Hetrick EM; Shin JH; Paul HS; Schoenfisch MH Anti-Biofilm Efficacy of Nitric Oxide-Releasing Silica Nanoparticles. Biomaterials 2009, 30, 2782–2789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (113).Shen Z; He K; Ding Z; Zhang M; Yu Y; Hu J. Visible-Light-Triggered Self-Reporting Release of Nitric Oxide (NO) for Bacterial Biofilm Dispersal. Macromolecules 2019, 52, 7668–7677. [Google Scholar]
  • (114).Sortino S. Light-Controlled Nitric Oxide Delivering Molecular Assemblies. Chem. Soc. Rev 2010, 39, 2903–2913. [DOI] [PubMed] [Google Scholar]
  • (115).Shen Z; Zheng S; Xiao S; Shen R; Liu S; Hu J. Red-Light-Mediated Photoredox Catalysis Enables Self-Reporting Nitric Oxide Release for Efficient Antibacterial Treatment. Angew. Chem. Int. Ed 2021, 60, 20452–20460. [DOI] [PubMed] [Google Scholar]
  • (116).Wu M-C; Deokar AR; Liao J-H; Shih P-Y; Ling Y-C Graphene-Based Photothermal Agent for Rapid and Effective Killing of Bacteria. ACS Nano 2013, 7, 1281–1290. [DOI] [PubMed] [Google Scholar]
  • (117).Wang C; Wang Y; Zhang L; Miron RJ; Liang J; Shi M; Mo W; Zheng S; Zhao Y; Zhang Y. Pretreated Macrophage-Membrane-Coated Gold Nanocages for Precise Drug Delivery for Treatment of Bacterial Infections. Adv. Mater 2018, 30, 1804023. [DOI] [PubMed] [Google Scholar]
  • (118).Ray PC; Khan SA; Singh AK; Senapati D; Fan Z. Nanomaterials for Targeted Detection and Photothermal Killing of Bacteria. Chem. Soc. Rev 2012, 41, 3193–3209. [DOI] [PubMed] [Google Scholar]
  • (119).Li M; Li L; Su K; Liu X; Zhang T; Liang Y; Jing D; Yang X; Zheng D; Cui Z; Li Z; Zhu S; Yeung KWK; Zheng Y; Wang X; Wu S. Highly Effective and Noninvasive near-Infrared Eradication of a Staphylococcus Aureus Biofilm on Implants by a Photoresponsive Coating within 20 Min. Adv. Sci 2019, 6, 1900599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (120).Yuan Z; Lin C; He Y; Tao B; Chen M; Zhang J; Liu P; Cai K. Near-Infrared Light-Triggered Nitric-Oxide-Enhanced Photodynamic Therapy and Low-Temperature Photothermal Therapy for Biofilm Elimination. ACS Nano 2020, 14, 3546–3562. [DOI] [PubMed] [Google Scholar]
  • (121).Hu D; Deng Y; Jia F; Jin Q; Ji J. Surface Charge Switchable Supramolecular Nanocarriers for Nitric Oxide Synergistic Photodynamic Eradication of Biofilms. ACS Nano 2020, 14, 347–359. [DOI] [PubMed] [Google Scholar]
  • (122).Li Y; Liu X; Li B; Zheng Y; Han Y; Chen D.-f.; Yeung KWK; Cui Z; Liang Y; Li Z; Zhu S; Wang X; Wu S. Near-Infrared Light Triggered Phototherapy and Immunotherapy for Elimination of Methicillin-Resistant Staphylococcus Aureus Biofilm Infection on Bone Implant. ACS Nano 2020, 14, 8157–8170. [DOI] [PubMed] [Google Scholar]
  • (123).Guo S; DiPietro LA Factors Affecting Wound Healing. J. Dent. Res 2010, 89, 219–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (124).Bjarnsholt T; Kirketerp-Møller K; Jensen PØ; Madsen KG; Phipps R; Krogfelt K; Høiby N; Givskov M. Why Chronic Wounds Will Not Heal: A Novel Hypothesis. Wound Repair Regen 2008, 16, 2–10. [DOI] [PubMed] [Google Scholar]
  • (125).Powers JG; Higham C; Broussard K; Phillips TJ Wound Healing and Treating Wounds: Chronic Wound Care and Management. J. Am. Acad. Dermatol 2016, 74, 607–625. [DOI] [PubMed] [Google Scholar]
  • (126).Xu Y; Li H; Xu S; Liu X; Lin J; Chen H; Yuan Z. Light-Triggered Fluorescence Self-Reporting Nitric Oxide Release from Coumarin Analogues for Accelerating Wound Healing and Synergistic Antimicrobial Applications. J. Biol. Chem 2022, 65, 424–435. [DOI] [PubMed] [Google Scholar]
  • (127).Afzali H; Khaksari M; Norouzirad R; Jeddi S; Kashfi K; Ghasemi A. Acidified Nitrite Improves Wound Healing in Type 2 Diabetic Rats: Role of Oxidative Stress and Inflammation. Nitric Oxide 2020, 103, 20–28. [DOI] [PubMed] [Google Scholar]
  • (128).Zhao Q; Zhang J; Song L; Ji Q; Yao Y; Cui Y; Shen J; Wang PG; Kong D. Polysaccharide-Based Biomaterials with on-Demand Nitric Oxide Releasing Property Regulated by Enzyme Catalysis. Biomaterials 2013, 34, 8450–8458. [DOI] [PubMed] [Google Scholar]
  • (129).Yang C; Hwang HH; Jeong S; Seo D; Jeong Y; Lee DY; Lee K. Inducing Angiogenesis with the Controlled Release of Nitric Oxide from Biodegradable and Biocompatible Copolymeric Nanoparticles. Int. J. Nanomed 2018, 13, 6517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (130).Wan X; Liu S; Xin X; Li P; Dou J; Han X; Kang I-K; Yuan J; Chi B; Shen J. S-Nitrosated Keratin Composite Mats with No Release Capacity for Wound Healing. Chem. Eng. J 2020, 400, 125964. [Google Scholar]
  • (131).Li Z; Huang X; Lin L; Jiao Y; Zhou C; Liu Z. Polyphenol and Cu2+ Surface-Modified Chitin Sponge Synergizes with Antibacterial, Antioxidant and Pro-Vascularization Activities for Effective Scarless Regeneration of Burned Skin. Chem. Eng. J 2021, 419, 129488. [Google Scholar]
  • (132).Huang S; Liu H; Liao K; Hu Q; Guo R; Deng K. Functionalized Go Nanovehicles with Nitric Oxide Release and Photothermal Activity-Based Hydrogels for Bacteria-Infected Wound Healing. ACS Appl. Mater. Interfaces 2020, 12, 28952–28964. [DOI] [PubMed] [Google Scholar]
  • (133).Zhu J; Tian J; Yang C; Chen J; Wu L; Fan M; Cai X. L-Arg-Rich Amphiphilic Dendritic Peptide as a Versatile No Donor for No/Photodynamic Synergistic Treatment of Bacterial Infections and Promoting Wound Healing. Small 2021, 17, 2101495. [DOI] [PubMed] [Google Scholar]
  • (134).Su C-H; Li W-P; Tsao L-C; Wang L-C; Hsu Y-P; Wang W-J; Liao M-C; Lee C-L; Yeh C-S Enhancing Microcirculation on Multitriggering Manner Facilitates Angiogenesis and Collagen Deposition on Wound Healing by Photoreleased No from Hemin-Derivatized Colloids. ACS Nano 2019, 13, 4290–4301. [DOI] [PubMed] [Google Scholar]
  • (135).Gurtner GC; Werner S; Barrandon Y; Longaker MT Wound Repair and Regeneration. Nature 2008, 453, 314–321. [DOI] [PubMed] [Google Scholar]
  • (136).Xiao J; Zhu Y; Huddleston S; Li P; Xiao B; Farha OK; Ameer GA Copper Metal–Organic Framework Nanoparticles Stabilized with Folic Acid Improve Wound Healing in Diabetes. ACS Nano 2018, 12, 1023–1032. [DOI] [PubMed] [Google Scholar]
  • (137).Kornblatt AP; Nicoletti VG; Travaglia A. The Neglected Role of Copper Ions in Wound Healing. J. Inorg. Biochem 2016, 161, 1–8. [DOI] [PubMed] [Google Scholar]
  • (138).Zhang P; Li Y; Tang Y; Shen H; Li J; Yi Z; Ke Q; Xu H. Copper-Based Metal–Organic Framework as a Controllable Nitric Oxide-Releasing Vehicle for Enhanced Diabetic Wound Healing. ACS Appl. Mater. Interfaces 2020, 12, 18319–18331. [DOI] [PubMed] [Google Scholar]
  • (139).Malone-Povolny MJ; Maloney SE; Schoenfisch MH Nitric Oxide Therapy for Diabetic Wound Healing. Adv. Healthc. Mater 2019, 8, 1801210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (140).Weinreb RN; Khaw PT Primary Open-Angle Glaucoma. The Lancet 2004, 363, 1711–1720. [DOI] [PubMed] [Google Scholar]
  • (141).Zhang K; Zhang L; Weinreb RN Ophthalmic Drug Discovery: Novel Targets and Mechanisms for Retinal Diseases and Glaucoma. Nat. Rev. Drug Discovery 2012, 11, 541–559. [DOI] [PubMed] [Google Scholar]
  • (142).Garhöfer G; Schmetterer L. Nitric Oxide: A Drug Target for Glaucoma Revisited. Drug Discov. Today 2019, 24, 1614–1620. [DOI] [PubMed] [Google Scholar]
  • (143).Chandrawati R; Chang JYH; Reina-Torres E; Jumeaux C; Sherwood JM; Stamer WD; Zelikin AN; Overby DR; Stevens MM Localized and Controlled Delivery of Nitric Oxide to the Conventional Outflow Pathway Via Enzyme Biocatalysis: Toward Therapy for Glaucoma. Adv. Mater 2017, 29, 1604932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (144).Fan W; Song M; Li L; Niu L; Chen Y; Han B; Sun X; Yang Z; Lei Y; Chen X. Endogenous Dual Stimuli-Activated No Generation in the Conventional Outflow Pathway for Precision Glaucoma Therapy. Biomaterials 2021, 277, 121074. [DOI] [PubMed] [Google Scholar]
  • (145).Ziche M; Morbidelli L; Choudhuri R; Zhang HT; Donnini S; Granger HJ; Bicknell R. Nitric Oxide Synthase Lies Downstream from Vascular Endothelial Growth Factor-Induced but Not Basic Fibroblast Growth Factor-Induced Angiogenesis. J. Clin. Investig 1997, 99, 2625–2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (146).Bonfiglio V; Camillieri G; Avitabile T; Leggio GM; Drago F. Effects of the Cooh-Terminal Tripeptide Α-Msh11–13 on Corneal Epithelial Wound Healing: Role of Nitric Oxide. Exp. Eye Res 2006, 83, 1366–1372. [DOI] [PubMed] [Google Scholar]
  • (147).Choi HW; Kim J; Kim J; Kim Y; Song HB; Kim JH; Kim K; Kim WJ Light-Induced Acid Generation on a Gatekeeper for Smart Nitric Oxide Delivery. ACS Nano 2016, 10, 4199–4208. [DOI] [PubMed] [Google Scholar]
  • (148).Rideau E; Dimova R; Schwille P; Wurm FR; Landfester K. Liposomes and Polymersomes: A Comparative Review Towards Cell Mimicking. Chem. Soc. Rev 2018, 47, 8572–8610. [DOI] [PubMed] [Google Scholar]
  • (149).Duan Y; Wang Y; Li X; Zhang G; Zhang G; Hu J. Light-Triggered Nitric Oxide (NO) Release from Photoresponsive Polymersomes for Corneal Wound Healing. Chem. Sci 2020, 11, 186–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (150).Zuin M; Rigatelli G; Scaranello F; Roncon L. Nitrates and Osteoporosis: Which Relationship? Eur. J. Intern. Med 2017, 43, e22–e23. [DOI] [PubMed] [Google Scholar]
  • (151).Bogdan C. Nitric Oxide and the Immune Response. Nat. Immunol 2001, 2, 907–916. [DOI] [PubMed] [Google Scholar]
  • (152).Lin YJ; Chen CC; Chi NW; Nguyen T; Lu HY; Nguyen D; Lai PL; Sung HW In Situ Self‐Assembling Micellar Depots That Can Actively Trap and Passively Release No with Long‐Lasting Activity to Reverse Osteoporosis. Adv. Mater 2018, 30, 1705605. [DOI] [PubMed] [Google Scholar]
  • (153).Ye J; Jiang J; Zhou Z; Weng Z; Xu Y; Liu L; Zhang W; Yang Y; Luo J; Wang X. Near-Infrared Light and Upconversion Nanoparticle Defined Nitric Oxide-Based Osteoporosis Targeting Therapy. ACS Nano 2021, 15, 13692–13702. [DOI] [PubMed] [Google Scholar]
  • (154).MacMicking J; Xie Q -w.; Nathan, C. Nitric Oxide and Macrophage Function. Annu. Rev. Immunol 1997, 15, 323–350. [DOI] [PubMed] [Google Scholar]
  • (155).Laaribi I; Kachmar S; Bouayed Z; Mimouni H; Mekkaoui I; El Rhalete A; El Mouhib A; Bkiyar H; Housni B. Plasmapherisis in Covid-19 Patient under Ecmo: A Moroccan Case Report Experience. Ann. Med. Surg 2022, 74, 103250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (156).Zhou F; Yu T; Du R; Fan G; Liu Y; Liu Z; Xiang J; Wang Y; Song B; Gu X; Guan L; Wei Y; Li H; Wu X; Xu J; Tu S; Zhang Y; Chen H; Cao B. Clinical Course and Risk Factors for Mortality of Adult Inpatients with Covid-19 in Wuhan, China: A Retrospective Cohort Study. The Lancet 2020, 395, 1054–1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (157).Cascella M; Rajnik M; Aleem A; Dulebohn, Napoli SC; Napoli RD Features, Evaluation, and Treatment of Coronavirus (Covid-19). StatPearls Publishing LLC.: Treasure Island (FL), 2022. [PubMed] [Google Scholar]
  • (158).Chen L; Liu P; Gao H; Sun B; Chao D; Wang F; Zhu Y; Hedenstierna G; Wang CG Inhalation of Nitric Oxide in the Treatment of Severe Acute Respiratory Syndrome: A Rescue Trial in Beijing. Clin. Infect. Dis 2004, 39, 1531–1535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (159).Pieretti JC; Rubilar O; Weller RB; Tortella GR; Seabra AB Nitric Oxide (NO) and Nanoparticles – Potential Small Tools for the War against Covid-19 and Other Human Coronavirus Infections. Virus Res. 2021, 291, 198202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (160).Garren MR; Ashcraft M; Qian Y; Douglass M; Brisbois EJ; Handa H. Nitric Oxide and Viral Infection: Recent Developments in Antiviral Therapies and Platforms. Appl. Mater. Today 2021, 22, 100887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (161).Åkerström S; Mousavi-Jazi M; Klingström J; Leijon M; Lundkvist Å; Mirazimi A. Nitric Oxide Inhibits the Replication Cycle of Severe Acute Respiratory Syndrome Coronavirus. J. Virol 2005, 79, 1966–1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (162).Åkerström S; Gunalan V; Keng CT; Tan Y-J; Mirazimi A. Dual Effect of Nitric Oxide on Sars-Cov Replication: Viral Rna Production and Palmitoylation of the S Protein Are Affected. Virology 2009, 395, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (163).Alvarez RA; Berra L; Gladwin MT Home Nitric Oxide Therapy for Covid-19. Am. J. Respir. Crit. Care Med 2020, 202, 16–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (164).Akaberi D; Krambrich J; Ling J; Luni C; Hedenstierna G; Järhult JD; Lennerstrand J; Lundkvist Å Mitigation of the Replication of Sars-Cov-2 by Nitric Oxide in Vitro. Redox Biol. 2020, 37, 101734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (165).Tandon M; Wu W; Moore K; Winchester S; Tu Y-P; Miller C; Kodgule R; Pendse A; Rangwala S; Joshi S. Sars-Cov-2 Accelerated Clearance Using a Novel Nitric Oxide Nasal Spray (Nons) Treatment: A Randomized Trial. Lancet Reg. Health - Southeast Asia 2022, 3, 100036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (166).Wu D; Lu J; Liu Y; Zhang Z; Luo L. Positive Effects of Covid-19 Control Measures on Influenza Prevention. Int. J. Infect. Dis 2020, 95, 345–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (167).Adusumilli NC; Zhang D; Friedman JM; Friedman AJ Harnessing Nitric Oxide for Preventing, Limiting and Treating the Severe Pulmonary Consequences of Covid-19. Nitric Oxide 2020, 103, 4–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (168).Green SJ Covid-19 Accelerates Endothelial Dysfunction and Nitric Oxide Deficiency. Microbes Infect. 2020, 22, 149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (169).Ranjbar S; Fatahi Y; Atyabi F. The Quest for a Better Fight: How Can Nanomaterials Address the Current Therapeutic and Diagnostic Obstacles in the Fight against Covid-19? J Drug Deliv. Sci. Technol 2022, 67, 102899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (170).Brunet LR Nitric Oxide in Parasitic Infections. Int. Immunopharmacol 2001, 1, 1457–1467. [DOI] [PubMed] [Google Scholar]
  • (171).Krishna S; Augustin Y; Wang J; Xu C; Staines HM; Platteeuw H; Kamarulzaman A; Sall A; Kremsner P. Repurposing Antimalarials to Tackle the Covid-19 Pandemic. Trends Parasitol. 2021, 37, 8–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (172).Pahan P; Pahan K. Smooth or Risky Revisit of an Old Malaria Drug for Covid-19? J Neuroimmune Pharmacol. 2020, 15, 174–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (173).Sobolewski P; Gramaglia I; Frangos J; Intaglietta M; van der Heyde HC Nitric Oxide Bioavailability in Malaria. Trends Parasitol. 2005, 21, 415–422. [DOI] [PubMed] [Google Scholar]
  • (174).Weinberg JB; Lopansri BK; Mwaikambo E; Granger DL Arginine, Nitric Oxide, Carbon Monoxide, and Endothelial Function in Severe Malaria. Curr. Opin. Infect. Dis 2008, 21, 468–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (175).Coleman JW Nitric Oxide in Immunity and Inflammation. Int. Immunopharmacol 2001, 1, 1397–1406. [DOI] [PubMed] [Google Scholar]
  • (176).Dangas GD; Claessen BE; Caixeta A; Sanidas EA; Mintz GS; Mehran R. In-Stent Restenosis in the Drug-Eluting Stent Era. J. Am. Coll. Cardiol 2010, 56, 1897–1907. [DOI] [PubMed] [Google Scholar]
  • (177).Lowe HC; Oesterle SN; Khachigian LM Coronary in-Stent Restenosis: Current Status and Future Strategies. J. Am. Coll. Cardiol 2002, 39, 183–193. [DOI] [PubMed] [Google Scholar]
  • (178).Naghavi N; De Mel A; Alavijeh OS; Cousins BG; Seifalian AM Nitric Oxide Donors for Cardiovascular Implant Applications. Small 2013, 9, 22–35. [DOI] [PubMed] [Google Scholar]
  • (179).Zhu D; Hou J; Qian M; Jin D; Hao T; Pan Y;Wang H; Wu S; Liu S; Wang F; Wu L;Zhong Y; Yang Z;Che Y; Shen J. Kong D; Yin M; Zhao Q. Nitrate-Functionalized Patch Confers Cardioprotection and Improves Heart Repair after Myocardial Infarction Via Local Nitric Oxide Delivery. Nat. Commun 2021, 12, 4501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (180).Yang Z; Zhao X; Hao R; Tu Q;Tian X; Xiao Y; Xiong K; Wang M; Feng Y; Huang N; Pan G. Bioclickable and Mussel Adhesive Peptide Mimics for Engineering Vascular Stent Surfaces. Proc. Natl. Acad. Sci. U.S.A 2020, 117, 16127–16137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (181).Gao P; Qiu H; Xiong K; Li X; Tu Q; Wang H; Lyu N; Chen X; Huang N; Yang Z. Metal-Catechol-(Amine) Networks for Surface Synergistic Catalytic Modification: Therapeutic Gas Generation and Biomolecule Grafting. Biomaterials 2020, 248, 119981. [DOI] [PubMed] [Google Scholar]
  • (182).Jeong H; Choi D; Oh Y; Heo J; Hong J. A Nanocoating Co-Localizing Nitric Oxide and Growth Factor onto Individual Endothelial Cells Reveals Synergistic Effects on Angiogenesis. Adv. Healthc. Mater 2022, 2102095. [DOI] [PubMed] [Google Scholar]
  • (183).Yasuda H. Solid Tumor Physiology and Hypoxia-Induced Chemo/Radio-Resistance: Novel Strategy for Cancer Therapy: Nitric Oxide Donor as a Therapeutic Enhancer. Nitric Oxide 2008, 19, 205–216. [DOI] [PubMed] [Google Scholar]
  • (184).Zhang X; Guo Z; Liu J; Tian G; Chen K; Yu S; Gu Z. Near Infrared Light Triggered Nitric Oxide Releasing Platform Based on Upconversion Nanoparticles for Synergistic Therapy of Cancer Stem-Like Cells. Sci. Bull 2017, 62, 985–996. [DOI] [PubMed] [Google Scholar]
  • (185).Zhang H; Tian X-T; Shang Y; Li Y-H; Yin X-B Theranostic Mn-Porphyrin Metal–Organic Frameworks for Magnetic Resonance Imaging-Guided Nitric Oxide and Photothermal Synergistic Therapy. ACS Appl. Mater. Interfaces 2018, 10, 28390–28398. [DOI] [PubMed] [Google Scholar]
  • (186).Evans MA; Huang P-J; Iwamoto Y; Ibsen Kelly N.; Chan EM; Hitomi Y; Ford PC; Mitragotri S. Macrophage-Mediated Delivery of Light Activated Nitric Oxide Prodrugs with Spatial, Temporal and Concentration Control. Chem. Sci 2018, 9, 3729–3741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (187).Guo R; Tian Y; Wang Y; Yang W. Near-Infrared Laser-Triggered Nitric Oxide Nanogenerators for the Reversal of Multidrug Resistance in Cancer. Adv. Funct. Mater 2017, 27, 1606398. [Google Scholar]
  • (188).An J; Hu Y-G; Li C; Hou X-L; Cheng K; Zhang B; Zhang R-Y; Li D-Y; Liu S-J; Liu B; Zhu D; Zhao Y-D A Ph/Ultrasound Dual-Response Biomimetic Nanoplatform for Nitric Oxide Gas-Sonodynamic Combined Therapy and Repeated Ultrasound for Relieving Hypoxia. Biomaterials 2020, 230, 119636. [DOI] [PubMed] [Google Scholar]
  • (189).Hu C; Cun X; Ruan S; Liu R; Xiao W; Yang X; Yang Y; Yang C; Gao H. Enzyme-Triggered Size Shrink and Laser-Enhanced No Release Nanoparticles for Deep Tumor Penetration and Combination Therapy. Biomaterials 2018, 168, 64–75. [DOI] [PubMed] [Google Scholar]
  • (190).Fan W; Lu N; Huang P; Liu Y; Yang Z; Wang S; Yu G; Liu Y; Hu J; He Q. Glucose‐Responsive Sequential Generation of Hydrogen Peroxide and Nitric Oxide for Synergistic Cancer Starving‐Like/Gas Therapy. Angew. Chem. Int. Ed 2017, 129, 1249–1253. [DOI] [PubMed] [Google Scholar]
  • (191).Li J; Jiang R; Wang Q; Li X; Hu X; Yuan Y; Lu X; Wang W; Huang W; Fan Q. Semiconducting Polymer Nanotheranostics for Nir-Ii/Photoacoustic Imaging-Guided Photothermal Initiated Nitric Oxide/Photothermal Therapy. Biomaterials 2019, 217, 119304. [DOI] [PubMed] [Google Scholar]
  • (192).Chen X; Jia F; Li Y; Deng Y; Huang Y; Liu W; Jin Q; Ji J. Nitric Oxide-Induced Stromal Depletion for Improved Nanoparticle Penetration in Pancreatic Cancer Treatment. Biomaterials 2020, 246, 119999. [DOI] [PubMed] [Google Scholar]
  • (193).Xu Y; Wang S; Chen Z; Hu R; Zhao Y; Wang K; Qu J; Liu L. Nitric Oxide Release Activated near-Infrared Photothermal Agent for Synergistic Tumor Treatment. Biomaterials 2021, 276, 121017. [DOI] [PubMed] [Google Scholar]
  • (194).Jia X; Zhang Y; Zou Y; Wang Y; Niu D; He Q; Huang Z; Zhu W; Tian H; Shi J; Li Y. Dual Intratumoral Redox/Enzyme-Responsive No-Releasing Nanomedicine for the Specific, High-Efficacy, and Low-Toxic Cancer Therapy. Adv. Mater 2018, 30, 1704490. [DOI] [PubMed] [Google Scholar]
  • (195).Kim T; Suh J; Kim J; Kim WJ Lymph-Directed Self-Immolative Nitric Oxide Prodrug for Inhibition of Intractable Metastatic Cancer. Adv. Sci 2022, 9, 2101935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (196).Wang K; Jiang M; Zhou J; Liu Y; Zong Q; Yuan Y. Tumor-Acidity and Bioorthogonal Chemistry-Mediated on-Site Size Transformation Clustered Nanosystem to Overcome Hypoxic Resistance and Enhance Chemoimmunotherapy. ACS Nano 2022, 16, 721–735. [DOI] [PubMed] [Google Scholar]
  • (197).Kim J; Sestito LF; Im S; Kim WJ; Thomas SN Poly(Cyclodextrin)-Polydrug Nanocomplexes as Synthetic Oncolytic Virus for Locoregional Melanoma Chemoimmunotherapy. Adv. Funct. Mater 2020, 30, 1908788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (198).Fan Y; Wu W; Luo T; Hu Y; Zhang Q; Zhang J; Xia X. Cross-Linking of S-Nitrosothiolated Aiegens inside Cancer Cells to Monitor No Release and Reverse Chemo-Resistance. ChemComm 2021, 57, 12520–12523. [DOI] [PubMed] [Google Scholar]
  • (199).Gu G; Chen C; Zhang S; Yin B; Wang J. Self-Assembly Dual-Responsive No Donor Nanoparticles for Effective Cancer Therapy. ACS Appl. Mater. Interfaces 2021, 13, 50682–50694. [DOI] [PubMed] [Google Scholar]
  • (200).Luo T; Wang D; Liu L; Zhang Y; Han C; Xie Y; Liu Y; Liang J; Qiu G; Li H; Su D; Liu J; Zhang K. Switching Reactive Oxygen Species into Reactive Nitrogen Species by Photocleaved O2-Released Nanoplatforms Favors Hypoxic Tumor Repression. Adv. Sci 2021, 8, 2101065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (201).Ji C; Si J; Xu Y; Zhang W; Yang Y; He X; Xu H; Mou X; Ren H; Guo H. Mitochondria-Targeted and Ultrasound-Responsive Nanoparticles for Oxygen and Nitric Oxide Codelivery to Reverse Immunosuppression and Enhance Sonodynamic Therapy for Immune Activation. Theranostics 2021, 11, 8587–8604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (202).Wang H; Liu Y; Zhu X; Chen C; Fu Z; Wang M; Lin D; Chen Z; Lu C; Yang H. Multistage Cooperative Nanodrug Combined with Pd-L1 for Enhancing Antitumor Chemoimmunotherapy. Adv. Healthc. Mater 2021, 10, 2101199. [DOI] [PubMed] [Google Scholar]
  • (203).Chen H; Shi T; Wang Y; Liu Z; Liu F; Zhang H; Wang X; Miao Z; Liu B; Wan M; Mao C; Wei J. Deep Penetration of Nanolevel Drugs and Micrometer-Level T Cells Promoted by Nanomotors for Cancer Immunochemotherapy. J. Am. Chem. Soc 2021, 143, 12025–12037. [DOI] [PubMed] [Google Scholar]
  • (204).Du Z; Mao Y; Zhang P; Hu J; Fu J; You Q; Yin J. Tpgs–Galactose-Modified Polydopamine Co-Delivery Nanoparticles of Nitric Oxide Donor and Doxorubicin for Targeted Chemo–Photothermal Therapy against Drug-Resistant Hepatocellular Carcinoma. ACS Appl. Mater. Interfaces 2021, 13, 35518–35532. [DOI] [PubMed] [Google Scholar]
  • (205).Ding M; Shao K; Wu L; Jiang Y; Cheng B; Wang L; Shi J; Kong X. A NO/ROS/RNS Cascaded-Releasing Nano-Platform for Gas/Pdt/Ptt/Immunotherapy of Tumors. Biomater. Sci 2021, 9, 5824–5840. [DOI] [PubMed] [Google Scholar]
  • (206).Lee J; Hlaing SP; Hasan N; Kwak D; Kim H; Cao J; Yoon I-S; Yun H; Jung Y; Yoo J-W Tumor-Penetrable Nitric Oxide-Releasing Nanoparticles Potentiate Local Antimelanoma Therapy. ACS Appl. Mater. Interfaces 2021, 13, 30383–30396. [DOI] [PubMed] [Google Scholar]
  • (207).Li Q; Zhang J; Li J; Ye H; Li M; Hou W; Li H; Wang Z. Glutathione-Activated No-/Ros-Generation Nanoparticles to Modulate the Tumor Hypoxic Microenvironment for Enhancing the Effect of Hifu-Combined Chemotherapy. ACS Appl. Mater. Interfaces 2021, 13, 26808–26823. [DOI] [PubMed] [Google Scholar]
  • (208).Wu X; Cheng Y; Zheng R; Xu K; Yan J; Song P; Wang Y; Rauf A; Pan Y; Zhang H. Immunomodulation of Tumor Microenvironment by Arginine-Loaded Iron Oxide Nanoparticles for Gaseous Immunotherapy. ACS Appl. Mater. Interfaces 2021, 13, 19825–19835. [DOI] [PubMed] [Google Scholar]
  • (209).Su Y; Zhang X; Lei L; Liu B; Wu S; Shen J. Tumor Microenvironment-Activatable Cyclic Cascade Reaction to Reinforce Multimodal Combination Therapy by Destroying the Extracellular Matrix. ACS Appl. Mater. Interfaces 2021, 13, 12960–12971. [DOI] [PubMed] [Google Scholar]
  • (210).Chu X; Jiang X; Liu Y; Zhai S; Jiang Y; Chen Y; Wu J; Wang Y; Wu Y; Tao X; He X; Bu W. Nitric Oxide Modulating Calcium Store for Ca2+-Initiated Cancer Therapy. Adv. Funct. Mater 2021, 31, 2008507. [Google Scholar]
  • (211).Wu H; Zhong D; Zhang Z; Wu Y; Li Y; Mao H; Luo K; Kong D; Gong Q; Gu Z. A Bacteria-Inspired Morphology Genetic Biomedical Material: Self-Propelled Artificial Microbots for Metastatic Triple Negative Breast Cancer Treatment. ACS Nano 2021, 15, 4845–4860. [DOI] [PubMed] [Google Scholar]
  • (212).Wan MM; Chen H; Da Wang Z; Liu ZY; Yu YQ; Li L; Miao ZY; Wang XW; Wang Q; Mao C; Shen J; Wei J. Nitric Oxide-Driven Nanomotor for Deep Tissue Penetration and Multidrug Resistance Reversal in Cancer Therapy. Adv. Sci 2021, 8, 2002525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (213).Xiang Q; Qiao B; Luo Y; Cao J; Fan K; Hu X; Hao L; Cao Y; Zhang Q; Wang Z. Increased Photodynamic Therapy Sensitization in Tumors Using a Nitric Oxide-Based Nanoplatform with Atp-Production Blocking Capability. Theranostics 2021, 11, 1953–1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (214).Du C; Zhou M; Jia F; Ruan L; Lu H; Zhang J; Zhu B; Liu X; Chen J; Chai Z; Hu Y. D-Arginine-Loaded Metal-Organic Frameworks Nanoparticles Sensitize Osteosarcoma to Radiotherapy. Biomaterials 2021, 269, 120642. [DOI] [PubMed] [Google Scholar]
  • (215).Fang X; Cai S; Wang M; Chen Z; Lu C; Yang H. Photogenerated Holes Mediated Nitric Oxide Production for Hypoxic Tumor Treatment. Angew. Chem. Int. Ed 2021, 60, 7046–7050. [DOI] [PubMed] [Google Scholar]
  • (216).Yang Z; Gao D; Guo X; Jin L; Zheng J; Wang Y; Chen S; Zheng X; Zeng L; Guo M; Zhang X; Tian Z. Fighting Immune Cold and Reprogramming Immunosuppressive Tumor Microenvironment with Red Blood Cell Membrane-Camouflaged Nanobullets. ACS Nano 2020, 14, 17442–17457. [DOI] [PubMed] [Google Scholar]
  • (217).Mu J; He L; Fan W; Tang W; Wang Z; Jiang C; Zhang D; Liu Y; Deng H; Zou J; Jacobson O; Qu J; Huang P; Chen X. Cascade Reactions Catalyzed by Planar Metal–Organic Framework Hybrid Architecture for Combined Cancer Therapy. Small 2020, 16, 2004016. [DOI] [PubMed] [Google Scholar]
  • (218).Xue Z; Jiang M; Liu H; Zeng S; Hao J. Low Dose Soft X-Ray-Controlled Deep-Tissue Long-Lasting No Release of Persistent Luminescence Nanoplatform for Gas-Sensitized Anticancer Therapy. Biomaterials 2020, 263, 120384. [DOI] [PubMed] [Google Scholar]
  • (219).Xu Y; Liu J; Liu Z; Ren H; Yong J; Li W; Wang H; Yang Z; Wang Y; Chen G; Li X. Blockade of Platelets Using Tumor-Specific No-Releasing Nanoparticles Prevents Tumor Metastasis and Reverses Tumor Immunosuppression. ACS Nano 2020, 14, 9780–9795. [DOI] [PubMed] [Google Scholar]
  • (220).Ma Z; Liu S; Ke Y; Wang H; Chen R; Xiang Z; Xie Z; Shi Q; Yin J. Biomimetic Nano-Nos Mediated Local No Release for Inhibiting Cancer-Associated Platelet Activation and Disrupting Tumor Vascular Barriers. Biomaterials 2020, 255, 120141. [DOI] [PubMed] [Google Scholar]
  • (221).Liu P; Wang Y; Liu Y; Tan F; Li J; Li N. S-Nitrosothiols Loaded Mini-Sized Au@Silica Nanorod Elicits Collagen Depletion and Mitochondrial Damage in Solid Tumor Treatment. Theranostics 2020, 10, 6774–6789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (222).Deng Y; Jia F; Chen X; Jin Q; Ji J. Atp Suppression by Ph-Activated Mitochondria-Targeted Delivery of Nitric Oxide Nanoplatform for Drug Resistance Reversal and Metastasis Inhibition. Small 2020, 16, 2001747. [DOI] [PubMed] [Google Scholar]
  • (223).Gao S; Zhang W; Wang R; Hopkins SP; Spagnoli JC; Racin M; Bai L; Li L; Jiang W; Yang X; Lee C; Nagata K; Howerth EW; Handa H; Xie J; Ma Q; Kumar A. Nanoparticles Encapsulating Nitrosylated Maytansine to Enhance Radiation Therapy. ACS Nano 2020, 14, 1468–1481. [DOI] [PubMed] [Google Scholar]
  • (224).Kinoshita R; Ishima Y; Chuang VTG; Nakamura H; Fang J; Watanabe H; Shimizu T; Okuhira K; Ishida T; Maeda H; Otagiri M; Maruyama T. Improved Anticancer Effects of Albumin-Bound Paclitaxel Nanoparticle Via Augmentation of Epr Effect and Albumin-Protein Interactions Using S-Nitrosated Human Serum Albumin Dimer. Biomaterials 2017, 140, 162–169. [DOI] [PubMed] [Google Scholar]
  • (225).Wang Y; Huang X; Tang Y; Zou J; Wang P; Zhang Y; Si W; Huang W; Dong X. A Light-Induced Nitric Oxide Controllable Release Nano-Platform Based on Diketopyrrolopyrrole Derivatives for Ph-Responsive Photodynamic/Photothermal Synergistic Cancer Therapy. Chem. Sci 2018, 9, 8103–8109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (226).Xiang H-J; Deng Q; An L; Guo M; Yang S-P; Liu J-G Tumor Cell Specific and Lysosome-Targeted Delivery of Nitric Oxide for Enhanced Photodynamic Therapy Triggered by 808 Nm near-Infrared Light. ChemComm 2016, 52, 148–151. [DOI] [PubMed] [Google Scholar]
  • (227).Huang X; Xu F; Hou H; Hou J; Wang Y; Zhou S. Stimuli-Responsive Nitric Oxide Generator for Light-Triggered Synergistic Cancer Photothermal/Gas Therapy. Nano Res. 2019, 12, 1361–1370. [Google Scholar]
  • (228).Liu F; Gong S; Shen M; He T; Liang X; Shu Y; Wang X; Ma S; Li X; Zhang M; Wu Q; Gong C. A Glutathione-Activatable Nanoplatform for Enhanced Photodynamic Therapy with Simultaneous Hypoxia Relief and Glutathione Depletion. Chem. Eng. J 2021, 403, 126305. [Google Scholar]
  • (229).Li Y-H; Guo M; Shi S-W; Zhang Q-L; Yang S-P; Liu J-G A Ruthenium-Nitrosyl-Functionalized Nanoplatform for the Targeting of Liver Cancer Cells and Nir-Light-Controlled Delivery of Nitric Oxide Combined with Photothermal Therapy. J Mater Chem B 2017, 5, 7831–7838. [DOI] [PubMed] [Google Scholar]
  • (230).Wu W; Yang Y; Liang Z; Song X; Huang Y; Qiu L; Qiu X; Yu S; Xue W. Near Infrared Ii Laser Controlled Free Radical Releasing Nanogenerator for Synergistic Nitric Oxide and Alkyl Radical Therapy of Breast Cancer. Nanoscale 2021, 13, 11169–11187. [DOI] [PubMed] [Google Scholar]
  • (231).Xu Y; Ren H; Liu J; Wang Y; Meng Z; He Z; Miao W; Chen G; Li X. A Switchable No-Releasing Nanomedicine for Enhanced Cancer Therapy and Inhibition of Metastasis. Nanoscale 2019, 11, 5474–5488. [DOI] [PubMed] [Google Scholar]
  • (232).Kang Y; Kim J; Park J; Lee YM; Saravanakumar G; Park KM; Choi W; Kim K; Lee E; Kim C; Kim WJ Tumor Vasodilation by N-Heterocyclic Carbene-Based Nitric Oxide Delivery Triggered by High-Intensity Focused Ultrasound and Enhanced Drug Homing to Tumor Sites for Anti-Cancer Therapy. Biomaterials 2019, 217, 119297. [DOI] [PubMed] [Google Scholar]
  • (233).Gao L; Dong B; Zhang J; Chen Y; Qiao H; Liu Z; Chen E; Dong Y; Cao C; Huang D; Chen W. Functional Biodegradable Nitric Oxide Donor-Containing Polycarbonate-Based Micelles for Reduction-Triggered Drug Release and Overcoming Multidrug Resistance. ACS Macro. Lett 2019, 8, 1552–1558. [DOI] [PubMed] [Google Scholar]
  • (234).Huang X; Ren K; Chang Z; Ye Y; Huang D; Zhao W; Yang L; Dong Y; Cao Z; Qiao H. Glucose Oxidase and L-Arginine Functionalized Black Phosphorus Nanosheets for Multimodal Targeted Therapy of Glioblastoma. Chem. Eng. J 2022, 430, 132898. [Google Scholar]
  • (235).Wang Y-C; Dai H-L; Li Z-H; Meng Z-Y; Xiao Y; Zhao Z. Mesoporous Polydopamine-Coated Hydroxyapatite Nano-Composites for Ros-Triggered Nitric Oxide-Enhanced Photothermal Therapy of Osteosarcoma. J Mater. Chem. B 2021, 9, 7401–7408. [DOI] [PubMed] [Google Scholar]
  • (236).Ren D; Cheng Y; Xu W; Qin W; Hao T; Wang F; Hu Y; Ma L; Zhang C. Copper-Based Metal-Organic Framework Induces No Generation for Synergistic Tumor Therapy and Antimetastasis Activity. Small 2023, 19, 2205772. [DOI] [PubMed] [Google Scholar]
  • (237).Dai W; Deng Y; Chen X; Huang Y; Hu H; Jin Q; Tang Z; Ji J. A Mitochondria-Targeted Supramolecular Nanoplatform for Peroxynitrite-Potentiated Oxidative Therapy of Orthotopic Hepatoma. Biomaterials 2022, 290, 121854. [DOI] [PubMed] [Google Scholar]
  • (238).Tang Y; Wang T; Feng J; Rong F; Wang K; Li P; Huang W. Photoactivatable Nitric Oxide-Releasing Gold Nanocages for Enhanced Hyperthermia Treatment of Biofilm-Associated Infections. ACS Appl. Mater. Interfaces 2021, 13, 50668–50681. [DOI] [PubMed] [Google Scholar]
  • (239).Wang L; Hou Z; Pranantyo D; Kang E-T; Chan-Park M. High-Density Three-Dimensional Network of Covalently Linked Nitric Oxide Donors to Achieve Antibacterial and Antibiofilm Surfaces. ACS Appl. Mater. Interfaces 2021, 13, 33745–33755. [DOI] [PubMed] [Google Scholar]
  • (240).Wang J; Wang L; Pan J; Zhao J; Tang J; Jiang D; Hu P; Jia W; Shi J. Magneto-Based Synergetic Therapy for Implant-Associated Infections Via Biofilm Disruption and Innate Immunity Regulation. Adv. Sci 2021, 8, 2004010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (241).Jeong H; Kim T; Earmme T; Hong J. Acceleration of Nitric Oxide Release in Multilayer Nanofilms through Cu(Ii) Ion Intercalation for Antibacterial Applications. Biomacromolecules 2021, 22, 1312–1322. [DOI] [PubMed] [Google Scholar]
  • (242).Namivandi-Zangeneh R; Sadrearhami Z; Bagheri A; Sauvage-Nguyen M; Ho KKK; Kumar N; Wong EHH; Boyer C. Nitric Oxide-Loaded Antimicrobial Polymer for the Synergistic Eradication of Bacterial Biofilm. ACS Macro Lett. 2018, 7, 592–597. [DOI] [PubMed] [Google Scholar]
  • (243).Liu T; Wei J; Fu G; Zhang P; Zhang Z; Guo D-S; Yang X. Surface Charge Switchable Nanoparticles Capable of Controlled Nitric Oxide Release for the Treatment of Acidity-Associated Bacterial Infections. Polym. Chem 2021, 12, 1023–1029. [Google Scholar]
  • (244).Park D; Kim J; Lee YM; Park J; Kim WJ Polydopamine Hollow Nanoparticle Functionalized with N-Diazeniumdiolates as a Nitric Oxide Delivery Carrier for Antibacterial Therapy. Adv. Healthcare Mater 2016, 5, 2019–2024. [DOI] [PubMed] [Google Scholar]
  • (245).Nguyen T-K; Selvanayagam R; Ho KKK; Chen R; Kutty SK; Rice SA; Kumar N; Barraud N; Duong HTT; Boyer C. Co-Delivery of Nitric Oxide and Antibiotic Using Polymeric Nanoparticles. Chem. Sci 2016, 7, 1016–1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (246).Tao S; Shen Z; Chen J; Shan Z; Huang B; Zhang X; Zheng L; Liu J; You T; Zhao F; Hu J. Red Light-Mediated Photoredox Catalysis Triggers Nitric Oxide Release for Treatment of Cutibacterium Acne Induced Intervertebral Disc Degeneration. ACS Nano 2022, 16, 20376–20388. [DOI] [PubMed] [Google Scholar]
  • (247).Yang Y; Bu H; Xu Y; Li S; Xu J; Xia X; Yin Z; Chen L; Chen Z; Tan W. Heat Confinement Aerogel Enables Supramagnetothermal Effect for Triggering Nitric Oxide Generation. Nano Lett. 2022, 22, 8339–8345. [DOI] [PubMed] [Google Scholar]
  • (248).Gong C; Guan W; Liu X; Zheng Y; Li Z; Zhang Y; Zhu S; Jiang H; Cui Z; Wu S. Biomimetic Bacteriophage-Like Particles Formed from Probiotic Extracts and No Donors for Eradicating Multidrug-Resistant Staphylococcus Aureus. Adv. Mater 2022, 34, 2206134. [DOI] [PubMed] [Google Scholar]
  • (249).Yang Y; Huang K; Wang M; Wang Q; Chang H; Liang Y; Wang Q; Zhao J; Tang T; Yang S. Ubiquitination Flow Repressors: Enhancing Wound Healing of Infectious Diabetic Ulcers through Stabilization of Polyubiquitinated Hypoxia-Inducible Factor-1α by Theranostic Nitric Oxide Nanogenerators. Adv. Sci 2021, 33, 2103593. [DOI] [PubMed] [Google Scholar]
  • (250).Sun B; Ye Z; Zhang M; Song Q; Chu X; Gao S; Zhang Q; Jiang C; Zhou N; Yao C; Shen J. Light-Activated Biodegradable Covalent Organic Framework-Integrated Heterojunction for Photodynamic, Photothermal, and Gaseous Therapy of Chronic Wound Infection. ACS Appl. Mater. Interfaces 2021, 13, 42396–42410. [DOI] [PubMed] [Google Scholar]
  • (251).Gao Q; Zhang X; Yin W; Ma D; Xie C; Zheng L; Dong X; Mei L; Yu J; Wang C; Gu Z; Zhao Y. Functionalized Mos2 Nanovehicle with near-Infrared Laser-Mediated Nitric Oxide Release and Photothermal Activities for Advanced Bacteria-Infected Wound Therapy. Small 2018, 14, 1802290. [DOI] [PubMed] [Google Scholar]
  • (252).Kang Y; Kim J; Lee YM; Im S; Park H; Kim WJ Nitric Oxide-Releasing Polymer Incorporated Ointment for Cutaneous Wound Healing. J. Control. Release 2015, 220, 624–630. [DOI] [PubMed] [Google Scholar]
  • (253).Ma X; Cheng Y; Jian H; Feng Y; Chang Y; Zheng R; Wu X; Wang L; Li X; Zhang H. Hollow, Rough, and Nitric Oxide-Releasing Cerium Oxide Nanoparticles for Promoting Multiple Stages of Wound Healing. Adv. Healthc. Mater 2019, 8, 1900256. [DOI] [PubMed] [Google Scholar]
  • (254).Gwon K; Choi WI; Lee S; Lee JS; Shin JH Biodegradable Hyaluronic Acid-Based, Nitric Oxide-Releasing Nanofibers for Potential Wound Healing Applications. Biomater. Sci 2021, 9, 8160–8170. [DOI] [PubMed] [Google Scholar]
  • (255).Li G; Yu S; Xue W; Ma D; Zhang W. Chitosan-Graft-Pamam Loading Nitric Oxide for Efficient Antibacterial Application. Chem. Eng. J 2018, 347, 923–931. [Google Scholar]
  • (256).Jia F; Li L; Fang Y; Song M; Man J; Jin Q; Lei Y; Ji J. Macromolecular Platform with Super-Cation Enhanced Trans-Cornea Infiltration for Noninvasive Nitric Oxide Delivery in Ocular Therapy. ACS Nano 2020, 14, 16929–16938. [DOI] [PubMed] [Google Scholar]
  • (257).Jeong H; Park J-H; Shin JH; Yoo J-C; Park CY; Hong J. Prolonged Release Period of Nitric Oxide Gas for Treatment of Bacterial Keratitis by Amine-Rich Polymer Decoration of Nanoparticles. Chem. Mater 2018, 30, 8528–8537. [Google Scholar]
  • (258).Zamanian RT; Charles V. Pollack J; Gentile MA; Rashid M; Fox JC; Mahaffey KW; Perez V. d. J. Outpatient Inhaled Nitric Oxide in a Patient with Vasoreactive Idiopathic Pulmonary Arterial Hypertension and Covid-19 Infection. Am. J. Respir. Crit. Care Med 2020, 202, 130–132. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES