Back to Journals » International Journal of Nanomedicine » Volume 17

Advances in Antitumor Nano-Drug Delivery Systems of 10-Hydroxycamptothecin

Authors Chen Y, Wang Z, Wang X, Su M, Xu F, Yang L, Jia L , Zhang Z

Received 2 June 2022

Accepted for publication 25 August 2022

Published 14 September 2022 Volume 2022:17 Pages 4227—4259

DOI https://doi.org/10.2147/IJN.S377149

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Dr Yan Shen



Yukun Chen,1 Zhenzhi Wang,2 Xiaofan Wang,3 Mingliang Su,1 Fan Xu,1 Lian Yang,1 Lijun Jia,1 Zhanxia Zhang1

1Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China; 2Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China; 3Department of Oncology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China

Correspondence: Lijun Jia; Zhanxia Zhang, Email [email protected]; [email protected]

Abstract: 10-Hydroxycamptothecin (HCPT) is a natural plant alkaloid from Camptotheca that shows potent antitumor activity by targeting intracellular topoisomerase I. However, factors such as instability of the lactone ring and insolubility in water have limited the clinical application of this drug. In recent years, unprecedented advances in biomedical nanotechnology have facilitated the development of nano drug delivery systems. It has been found that nanomedicine can significantly improve the stability and water solubility of HCPT. NanoMedicines with different diagnostic and therapeutic functions have been developed to significantly improve the anticancer effect of HCPT. In this paper, we collected reports on HCPT nanomedicines against tumors in the past decade. Based on current research advances, we dissected the current status and limitations of HCPT nanomedicines development and looked forward to future research directions.

Keywords: 10-hydroxycamptothecin, antitumor, nanomedicine, nano-drug delivery systems

Background

A series of multiple processes, including the rapid spread and uncontrollable multiplication of abnormal cells, lead to the formation of malignant tumors with the potential for metastasis.1 There are many different approaches to cancer treatment, such as conventional radiotherapy, chemotherapy, etc. However, some therapies are not as effective and some have side effects.2 Natural compounds are a valuable resource for discovering and developing novel drugs, primarily for cancer treatment, that can serve as safe alternatives to the various synthetic drugs used in existing clinical therapies.3,4 Evidence suggests that alkaloids are essential for the development of antitumor botanicals. Alkaloids are commonly found in plants, and new alkaloids have been found to have potent apoptotic and antitumor abilities against various cancer cells during the screening process of new drugs.5,6 HCPT, a natural alkaloid, is the most potent anticancer compound among more than 20 monomers isolated from Camptotheca, with an anticancer activity equivalent to 30 times that of Camptothecin (CPT).7 Numerous studies have shown that HCPT has the advantages of high efficiency, low toxicity and broad anticancer spectrum and is clinically used to treat colorectal cancer, liver cancer, gastric cancer, ovarian cancer, leukemia, head and neck tumors, etc.8–11 However, its clinical application is limited by poor water solubility and low bioavailability. Current formulation research is focused on improving the solubility of lactone HCPT in water and improving the slow and controlled release of the drug to enhance its clinical application.12 One promising approach to overcome these challenges is the incorporation of HCPT into different nano-sized delivery vehicles.13 The use of nano-drug delivery systems for tumor-targeted therapy is an effective way to overcome the lack of specificity of conventional chemotherapeutic agents and the shortcomings of clinical treatment of tumors.14,15 Nano-drug delivery systems can effectively enhance the in vivo distribution of chemotherapeutic drugs after systemic administration and improve the balance of efficacy and toxicity in systemic chemotherapeutic interventions.16,17 Nanomedicines show potential therapeutic advantages over free drugs in vitro and in vivo.18,19 However, although HCPT nanomedicines have been very much researched, there has not been significant progress in the clinical application of HCPT nanomedicines so far.20 Studies have shown that inefficient accumulation and penetration in human tumors is the main reason for the poor clinical translation of most nanomedicines.21 For this reason, many strategies, including enhancement of vascular permeability and optimization of nanoparticles, have been investigated. However, the results have been unsatisfactory.22,23 A comprehensive overview of recent research advances in HCPT nanomedicines is essential to study this dilemma. However, although some studies on HCPT nanomedicines in cancer have been reported, these literatures have never provided a comprehensive review of the anti-tumor capacity of HCPT nanomedicines. On this basis, this paper classifies HCPT nanomedicines into organic nanoparticles (NPs), inorganic NPs and carrier-free NPs according to the material composition of HCPT nanocarriers (Figure 1). The research progress of the HCPT drug delivery system is critically reviewed and discussed, while the limitations and future directions of nanomedicine enhanced permeability and retention (EPR) effects are explored. We hope to provide a scientific basis and reference for HCPT nanomedicines research and clinical translation.

Figure 1 Schematic diagram of the classification of HCPT nano-drug delivery systems.

Organic NPs

Organic NPs such as polyethylene glycol (PEG), poly (lactic-co-glycolic acid) (PLGA), poly (D, L-lactic acid) (PLA), proteins, lipid, gels, peptides, dendrimer, cyclodextrins, electrospun fiber, polysaccharide and cell membrane, etc. can be physically encapsulated or loaded by conjugation of molecules at the core or surface functionalization and thus show great potential in biomedical applications. Due to their specific properties, such as high cellular uptake, high biodegradability and high loading efficiency, they have been widely used in cancer therapy.24,25

PEG-Based NPs

PEG is one of the most commonly used polymers for encapsulating nanocarriers to enhance biocompatibility, hydrophilicity, stability and biodegradability.26 PEGylation increases the cycle time of nanocarriers by avoiding the absorption of the reticuloendothelial system (RES), and this Food and Drug Administration (FDA)-approved polymer is popular for its modifiable properties and well-established safety profile.27 Combination therapy has been shown to be an effective strategy for improving treatment outcomes and reducing tumor resistance.28 Diosgenin and HCPT are two typical natural anticancer drugs. Li et al prepared NPs loaded with 8armPEGylated diosgenin and HCPT. Studies have shown that NPs exhibit better tumor growth inhibition compared to free drugs.29 It has been shown that the anticancer drug methotrexate (MTX) has a targeting effect and that MTX loaded in pellets can act not only as a drug but also as a potential targeting ligand.30,31 Li et al synthesized PEG NPs loaded with HCPT and MTX. The NPs can target tumors while breaking down in the acidic microenvironment of the tumor with synergistic therapeutic effects.32,33 Dexamethasone (DEX) is a hormone drug commonly used in clinical practice.34 Meng et al reported that novel NPs consisting of HCPT and DEX could further increase the level of reactive oxygen species (ROS) in tumor cells and promote the normalization of blood vessels at tumor sites and reducing inflammation (Figure 2).35 The synergistic application of chemotherapeutic drugs and small interfering RNA (siRNA) has emerged as an innovative approach for the treatment of cancer.36 siRNA can suppress tumors by silencing oncogenes on the one hand, and improve the sensitivity of tumors to chemotherapy on the other.37,38 Li et al synthesized NPs with poly-HCPT and siBcl-2 as the core, amphiphilic lipid-PEG as the shell, and lactic acid (LA) decoration. Through the specific binding between LA and asialoglycoprotein (ASGP) receptors, the NPs can accumulate and target in tumor tissues, exerting excellent synergistic tumor suppression effects.39 Antibody-drug conjugates show great potential in cancer immunotherapy.40 Liu et al fabricated an HCPT-based PEG amphiphilic antibody-drug coupling. The NPs have active tumor targeting through a specific interaction between anti-CD20 antibody and antigen.41 The effect of linear/branched PEG chains in nanocarriers on antitumor efficacy is a little reported. Guo et al prepared HCPT NPs using the linear PEG45, brush oligo (triethylene glycol) (TEG10), and oligo (ethylene glycol) dendron (G2) as nanocarriers. All these NPs showed similar drug loading. The resultant proven in vitro and in vivo antitumor effects depended on the degree of nanocarrier branching, with G2 NPs showing the best antitumor effect compared to PEG45 NPs and TEG10 NPs.42 iRGD as a tumor-targeting and tumor penetrating agent could enhance drug penetration through specific receptor binding affinity for αvβ3 and NRP-1.43,44 Li et al prepared PEG and iRGD modified NPs. Compared with non-targeting NPs, iRGD-modified NPs significantly improved tumor therapeutic efficacy.45 Nanosuspensions are drug delivery systems formed by mixing pure drug NPs in a solvent, generally with a surfactant to improve their stability and have a high drug loading capacity.46 Researchers synthesized PEG-modified HCPT nanosuspensions have similar properties and show superior antitumor effects.47,48 Due to its excellent biocompatibility and water solubility, mPEG is commonly used to encapsulate hydrophobic drugs to increase their circulation time in vivo.49 Lv et al prepared novel prodrug micelles using water-soluble mPEG and HCPT as side chains. The prodrug micelles exhibited effective tumor cell internalization and excellent anticancer efficacy.50 Compared with conventional chemotherapy, photodynamic therapy (PDT) is considered an advanced non-invasive approach in anti-cancer treatment.51,52 Studies have shown that HCPT and photosensitizer loaded PEG NPs showed excellent anticancer efficiency under laser irradiation.53,54 In addition to light triggering, the development of photosensitizers activated by specific triggers such as enzyme-activated has received much attention.55,56 Lu et al synthesized a cathepsin B-activated photosensitizer, further PEGylated and modified with tumor cell-targeting peptide cRGD and used as a nanocarrier to load HCPT. The NPs showed cathepsin B-activated PDT and strong synergistic inhibitory effects on 4T1 cells.57 It has been shown that NPs with positively charged surfaces have a short blood circulation half-life.58 In contrast, the negatively charged NPs have a longer blood circulation time.59 TAT peptide is a positively charged cell-penetrating peptide. Jing et al prepared negatively charged TAT peptide modified PEG nanomicelles that can undergo charge inversion in the acidic environment of tumor tissue, exposing positively charged TAT peptides to promote cellular internalization and subsequent nuclear targeting, showing efficient antitumor efficacy and inhibition of lung metastasis.60 Folic acid (FA) has a high affinity for folate receptor (FR), which is overexpressed on the surface of tumor cells, thus targeting tumor cells.61 It was shown that FA-modified HCPT-loaded PEG NPs enhanced cellular uptake via FR-mediated endocytosis and exhibited higher cytotoxic and tumor suppressive effects.62,63 Recent observations suggest that physicochemical properties such as the shape of the nanocarrier can significantly affect its anticancer activity.64 Zhou et al compared the shape-regulated anticancer activity of HCPT-loaded PEG nanorods (NRs) and NPs. It was shown that NRs exhibited better anticancer efficiency than NPs in 4T1 and MCF-7 cells.65 It has been reported that shorter nanocrystals (NCs) with a high aspect ratio (AR) generally exhibit better drug delivery efficiency, but how AR will affect cellular uptake rates and biocompatibility remains unknown.66 Tian et al developed HCPT-loaded PEG NCs with four different ARs. Studies have shown that shorter NCs with AR=1 are more readily captured by the liver and have less tumor uptake than longer NCs and higher NCs.67 Similarly, Wu et al demonstrated that nanoneedle with high ARs exhibited strong shape-dependent effects on cell internalization.68 The study of pH-responsive nanomedicines has become a hotspot in the field of drug delivery.69 Researchers prepared HCPT-loaded pH-responsive PEG NPs exhibited greater tumor growth inhibition.70–72 Furthermore, Researchers have developed HCPT-loaded PEG NPs with redox-responsive73,74 and glutathione (GSH)-sensitive release.75,76 Several studies have also demonstrated that HCPT-loaded PEG NPs exhibit significant liver targeting,77 increase the rate of dissolution,78 improve endocytosis79 and enhance anti-tumor effect.80

Figure 2 Schematic diagram of (keto-)pHCPT-PEG NPs synthesis and treatment of tumors.

Notes: Reprinted from Journal of Controlled release, 340, Meng Q, Hu H, Jing X, et al. A modular ROS-responsive platform co-delivered by 10-hydroxycamptothecin and dexamethasone for cancer treatment. J Control Release. 2021;102–113, with permission from Elsevier.35

PLGA-Based NPs

PLGA is an excellent biodegradable polymer. It has been approved by the FDA to use in drug delivery systems due to its controlled and sustained-release properties, low toxicity, and biocompatibility with tissue and cells.81 Low-intensity focused ultrasound (LIFU) has been intensively studied for tumor treatment and diagnostic imaging as a non-invasive means of focusing energy on specific sites with considerable tissue penetration, which can significantly improve the efficacy of chemotherapy while preventing damage to surrounding tissues.82,83 Wang et al synthesized cetuximab (C225) and HCPT-loaded PLGA NPs. The NPs can be triggered by LIFU and exhibit epidermal growth factor (EGFR) targeting and LIFU response properties in ultrasonography and synergistic chemotherapy of anaplastic thyroid carcinoma.84 Yang et al demonstrated that dual drug PLGA nanoneedles loaded with MTX and HCPT are characterized by effective targeting, high drug loading and longer drug release, and easier access to HeLa cells.85 Some excipients can enhance the cytotoxicity and chemosensitivity of NPs by overcoming drug efflux or maximizing the internalization of cancer cells.86 Zaki showed HCPT-loaded PLGA NPs containing D-a-tocopheryl PEG 1000 succinate (TPGS 1000), pluronic P85 or chitosan were more cytotoxic to cancer cells.87 Arsenic trioxide (As2O3), commonly known as arsenic, is a highly toxic substance that can kill tumor cells.88 Wu et al prepared PLGA microspheres loaded HCPT and As2O3 using multiple emulsion solvent evaporation method. The microspheres acted on vascular-associated cells and inhibited the formation of new blood vessels, thereby suppressed tumor growth.89 Triphenylphosphonium (TPP) is a highly lipophilic polar cation that can penetrate the mitochondrial membrane.90 Li et al showed that TPP-modified PLGA NPs could target mitochondria and nuclei, deliver more ring-closed form of HCPT to tumor tissues, and significantly enhance anti-tumor activity.91 Furthermore, Ma et al demonstrated HCPT-loaded PLGA NPs with promising controlled release behavior, well stability and excellent tumor inhibition.92

PLA-Based NPs

PLA is widely used in various biomedical applications due to its biocompatible and non-toxic properties. Various methods, such as emulsion, salting and precipitation, have been used to fabricate better PLA NPs widely used as controlled drug delivery systems for therapeutic molecules.93 Hou et al fabricated HCPT-loaded PLA microbubbles by a double emulsion-solvent evaporation method for use as an ultrasound-triggered drug delivery system. The PLA microbubbles had a smooth, spherical surface and the drug was amorphously dispersed within the shell. Nearly 20% of the HCPT was released after 10 min of exposure to diagnostic ultrasound at 3.5 MHz. Cytotoxicity tests on BEL-7402 cells showed that PLA microvesicles combined with ultrasound exposure showed more cytotoxicity than microvesicles alone.94 Yang et al used a dialysis technique to prepare HCPT NPs using PEG-b-PLA and PLA, respectively. The results showed that although both exhibited slow and prolonged release profiles, the PEG-b-PLA NPs exhibited smaller particle size, faster drug release and higher cytotoxicity compared to the PLA NPs.95

Protein-Based NPs

Protein-based nanocarriers meet the requirements of low cytotoxicity, abundant renewable resources, high drug binding capacity and remarkable uptake into target cells. They are promising candidates for efficient drug and gene delivery.96

Albumin-Based NPs

Albumin is the most abundant protein in plasma and has high biocompatibility, biodegradability, non-immunogenicity and safety for clinical applications.97 Also, the chemical structure and conformation of albumin allow interaction with many different drugs, thus potentially protecting them from in vivo elimination and metabolism, with the potential to facilitate half-life extension and targeted drug delivery.98 Yang et al prepared HCPT-human serum albumin (HSA) NPs by a liquid composite method, which is relatively simple compared to the supercritical antisolvent process of emulsification and albumin coating method, which has the advantages of low toxicity, high step continuity, and less batch feeding, and the NPs can significantly inhibit tumor growth compared to free HCPT.99 Glycyrrhetinic acid (GA) is commonly used in clinical practice for the treatment of chronic hepatitis.100 Zu et al showed HCPT-GA-conjugated bovine serum albumin (BSA) NPs have hepatocyte targeting properties.101 Wang et al similarly demonstrated the tumor targeting properties of FA-modified HSA NPs.102

Other Protein-Based NPs

High-density lipoproteins (HDL) are of great interest in drug delivery due to their relatively long half-life in the circulation, small particle size, and lipid core.103 Yuan et al developed HCPT-loaded HDL NPs. Cytotoxicity studies in HT29 colon cancer cells showed that the IC50 of the NPs was approximately 3-fold lower than that of free HCPT.104 Cui et al constructed T7 and dA7R peptide dual modified ligands HCPT-loaded HDL NPs. The dual modified NPs were found to show higher glioma localization than the single ligand modified NPs or free HCPT.105 Casein (CA) is the main component of milk, and hollow casein nanospheres have the extraordinary ability to penetrate cell membranes, making them an ideal carrier.106 Gao et al demonstrated that menthol-modified casein NPs loaded with HCPT can cross the blood-brain barrier and target gliomas.107

Lipid-Based NPs

Lipid-based NPs show great potential for drug delivery by offering excellent biocompatibility while improving drug solubility, encapsulating drugs in lipid membranes to achieve desired bioavailability and mitigating adverse drug reactions. Lipid-based NPs such as liposomes, solid lipid NPs, nanostructured lipid carriers, lipid nanoemulsion and lipid-polymer hybrid NPs, have promising applications in drug delivery and tumor therapy.108–110

Liposomes

Liposomes are the most successful nanocarriers for clinical applications. The bilayer structure of liposomes can be composed of natural phospholipids, cholesterol, etc., making them ideal carriers for different drugs with different solubilities, as hydrophilic molecules can be incorporated into the core. In contrast, hydrophobic drugs can be accommodated within the lipid membrane.111 Thus, liposomes can carry water-soluble and insoluble drugs to the target site. In addition, they have low immunogenicity, toxicity and high pharmacoprotective effects.112

Li et al synthesized cell-penetrating peptide modified phase-transformation lipid NPs loaded with HCPT by thin film hydration method and phacoemulsification method. The NPs with ultrasound/PA dual-modality imaging capability could penetrate deeply into the tumor and release the drug under LIFU irradiation.113 Stearyl glycyrrhetinate (SG) is a derivative of GA. It has been demonstrated that GA and its derivatives may be used as ligands targeting the liver.114 It was shown that GA/SG-modified HCPT lipid NPs enhanced accumulated in liver tumors.115,116 NK4 protein is a specific hepatocyte growth factor (HGF) antagonist that can act as a liver-targeting ligand, actively recognizing and binding to hepatocytes.117 Zhou et al prepared NK4-HCPT liposomal NPs had excellent liver-targeting properties that could enhance the therapeutic effect of the HCPT for the treatment of hepatocellular carcinoma.118 Compared to PDT, sonodynamic therapy (SDT) has been widely explored for cancer treatment because of its superior tissue penetration, lower cost, and higher safety profile.119 Xiao et al prepared cationic HCPT liposomes by thin film method, combined with 5-aminolevulinic acid (5-ALA) by endotracheal administration for the chemotherapy and SDT treatment of metastatic lung cancer (Figure 3). The NPs showed higher anticancer effects in treating metastatic lung cancer mice.120 Chen et al prepared HCPT-encapsulated liposomes by the thin film evaporation method. It was found that because this nanoparticle could continuously release the drug, it had a strong inhibitory effect on HepG-2, A549 and SGC-7901 cancer cells.121

Figure 3 Schematic diagram of the preparation of HCPT liposomes.

Notes: Reprinted from International Journal of Pharmaceutics, 601:120572, Xiao Z, Zhuang B, Zhang G, Li M, Jin Y. Pulmonary delivery of cationic liposomal hydroxycamptothecin and 5-aminolevulinic acid for chemo-sonodynamic therapy of metastatic lung cancer. Int J Pharm. 2021, with permission from Elsevier.120

Solid Lipid NPs (SLNs)

SLNs are colloidal drug carriers with superior biocompatibility, and their main components are solid at room temperature. The main advantages of SLNs over conventional drug carriers are their controlled and sustained drug release capability and excellent stability.122,123 Xyloglcan (XG) is a natural polysaccharide with excellent biocompatibility and biodegradability to target drugs for hepatocellular carcinoma.124 Liu et al prepared HCPT-loaded SLNs, which were coated with XG. This nanoparticle was specifically recognized by ASGPR on the surface of HepG2 cell membranes, thus enabling targeting of hepatocellular carcinoma cells and accumulation of higher drug content, exhibiting superior antitumor effects.125

Nanostructured Lipid Carriers (NLCs)

NLCs are modified and improved forms of SLNs, in which lipid phase contains both solid and liquid lipids. Due to the nature of nanostructures, NLCs enhance their loading and stability to drugs.126,127 Sun et al prepared N-Arginine-N-octyl chitosan (AOCS)-modified pH-sensitive NLCs. The in vitro and in vivo antitumor activities showed that such HCPT-loaded NLCs had better therapeutic effects than free HCPT. Under physiological conditions, the nanocarriers have a negative surface charge. They are safe in normal tissues, and when in an acidic environment, they can effectively disrupt lysosomes and disassembled drugs entrapped into cytoplasm efficiently.128 Su et al prepared NLCs loaded with HCPT by the solvent evaporation method. The NLCs modified with octreotide-PEG (100) monostearate exhibited better-sustained release, the most efficient cellular uptake and cytotoxicity in SMMC-7721 overexpressed ligands of a growth inhibitor receptor (SSTR).129

Lipid Nanoemulsion

Lipid nanoemulsions are considered very attractive nanocarriers, increasing the solubility and bioavailability of drugs. They are capable of simultaneous sustained drug release while reducing drug toxicity.130 Zhao et al prepared HCPT-loaded lipid emulsions, which were found to improve the therapeutic efficiency of HCPT by comparing it with HCPT injections and the inhibition of tumor growth in mice was stronger.131

Lipid-Polymer Hybrid NPs (LPHs)

LPHs exhibit characteristics of liposomal and polymeric NPs and are ideal nanocarriers. With advantages such as simple fabrication process, tunable size and surface charge, high loading capacity of low water-soluble drugs, and sustained and controlled drug release.132,133 Yang et al synthesized HCPT-loaded LPHs by a modified emulsification technique. The LPHs consisted of a monolayer of lipid and a PEG shell with a hydrophobic polymer core. It was found that the LPHs improved cellular uptake efficiency and increased the cytotoxicity of HCPT on MDA-MB-435s cells.134

Other Lipid-Based NPs

Lipid-based NPs are widely used in drug delivery due to their unique advantages, and there are some other lipid-based NPs besides the classification mentioned above. Li et al developed a lipid microbubble carrying HCPT with high drug encapsulation and loading content while maintaining the acoustic properties as an ultrasound contrast agent. The combination of microbubbles with ultrasound exhibited significant drug accumulation and superior antitumor effects compared to microbubbles alone or HCPT injection.135 Zhao et al prepared peptide-modified lipid NPs, which could penetrate the extracellular matrix, cell membrane, and even enter the nucleus under the induction of the novel cysteine-flanked cell-penetrating peptide CG-TAT-GC. Triggered by LIFU, the NPs can also improve drug release and enhance imaging of tumor sites.136 Wang et al proposed lipid NPs loaded with HCPT and photosensitizer. The nanoparticle combined LIFU and laser with enhanced imaging, targeting and ovarian cancer treatment.137 Some HCPT-loaded lipid NPs also showed excellent targeting. MTX-functionalized NPs enhance cellular uptake.138,139 Several studies have shown that cellular sensitivity to anti-cancer drugs or radiation is significantly correlated with circadian rhythms and that the role of circadian clock genes is crucial in cancer therapy.140,141 Hou et al found that when tumor suppressor period circadian regulator 2 (PER2) overexpression combined with HCPT-loaded lipid NPs can significantly enhance the anti-tumor effect.142 Zhong et al prepared HCPT-loaded lipid nanocochleates, which can effectively improve the bioavailability of drugs. In addition, it can open cellular tight junctions and paracellular routes and showed more pronounced inhibition of tumor growth compared to normal saline, HCPT suspensions and HCPT-loaded liposomes.143 Berberine hydrochloride (BBR), a class of drugs that regulate intestinal flora, has shown strong antitumor activity, reduces HIF-1a levels and induces apoptosis.144 Qi et al prepared a novel lipid microsphere loaded with 10-HCPT-BBR that could significantly slow down tumor drug resistance.145

Gels-Based NPs

Nanogels and hydrogels have emerged as promising materials for biomedical applications due to their large surface area and tunable mechanical and chemical properties.146 Their large surface area is well suited for biocoupling, while the internal porous network can be used to transport valuable biomolecules. The use of biocompatible hydrogels and nanogels avoids undesirable side effects within biological systems and preserves the excellent water content, thus creating an environment very similar to that of the extracellular matrix.147,148

Nanogel-Based NPs

Nanogels carriers are rapidly becoming a significant delivery strategy in biology and medicine due to their small particle size, excellent solubility, high loading capacity, and controlled release.149 Studies showed positively charged nanogels with high HCPT loading efficiency, extended residence time, improved tissue penetration.150 Positively charged disulfide-bonded nanogels that can specifically release cargo in cancer cells triggered by intracellular reduction microenvironment. Avoids the potential risk of HCPT resistance associated with high-dose chemotherapy.151 Prolonged the retention period of HCPT in the rat bladder wall and enhanced tissue permeability.152 Similarly, Qin et al demonstrated that HCPT-loaded phytanetriol cubic phase gel solutions, exhibited significant cytotoxic and anticancer activities against HepG2 and SMMC7721 cells.153

Hydrogel-Based NPs

Hydrogel-based therapies are a promising option for cancer treatment because of their controllability, biocompatibility, high drug loading capacity, extended drug release time, and specific stimulus sensitivity.154 Several studies have revealed hydrogels maintain long-term sustained release of HCPT at a high accumulation rate, with optimized anticancer efficacy, satisfactory stability, injectability and recyclability.155–158 Liu et al synthesized a novel octapeptide (1-YSV) to enable its co-assembly with HCPT to obtain supramolecular hydrogels with excellent viscoelasticity and increased the water stability of HCPT (Figure 4).159 Guo et al prepared a supramolecular hydrogel composed of a self-assembling peptide, HCPT and macrocyclic polyamine cyclen can depleting cellular ATP and reversing ATP-dependent drug efflux, greatly improving cellular uptake and nuclear aggregation of HCPT.160

Figure 4 (A) Schematic illustration of co-assembly of tyroservatide-derived octapeptide (1-YSV) and HCPT for cancer therapy; (B) TEM micrograph of the 1-YSV hydrogel; and (C) TEM micrograph of the 1-YSV/HCPT hydrogel.

Notes: Used with permission of Nanoscale, from Molecular self-assembly of a tyroservatide-derived octapeptide and hydroxycamptothecin for enhanced therapeutic efficacy. Liu J, Wu C, Dai G, et al. Nanoscale. 2021;13(9):5094–5102. Permission conveyed through Copyright Clearance Center, INC.159

Peptide-Based NPs

Peptides are sequences of approximately 2–50 amino acids that have gained significant interest in therapeutic diagnostic applications in cancer research due to their better biosafety, customizability, ease of synthesis process, ability to target biological receptors by recognizing them on cancer cells.161 D-peptides consist of D-amino acids that are resistant to endogenous peptidase-catalyzed hydrolysis and are very promising nanocarriers.162 Liu et al developed D-peptide nanofibers for the controlled delivery of HCPT. The results showed that D-peptide nanofibers could improve the aqueous solubility of HCPT and exhibit better long-term stability and better cancer cell selectivity.163 Zeng et al constructed a bladder tumor-specific peptide prodrug by enzyme-assisted assembly. The prodrugs can target bladder tumors through the specific binding ability of the YSA peptide to the membrane protein EphA2. Catalyzed by the enzyme cathepsin B, they could induce small molecule prodrugs to form nanostructures, which prolong the retention time of the HCPT, thereby reducing HCPT side effects.164 The tLyP-1 peptide-modified NPs are targeted and penetrating, increasing tumor accumulation and penetrating deeply into extravascular tumor tissue.165 Zhu et al developed phase change NPs modified by tLyP-1 peptide with loaded HCPT. In combination with LIFU, NPs can phase transform into microvesicles and enhance tumor ultrasound molecular imaging for tumor diagnosis while releasing drugs.166 In addition, some experiments have shown that peptide-based NPs enhance the cellular uptake and aggregation of HCPT in the nucleus, improving the anti-tumor activity and anti-metastatic efficacy of HCPT.167–171

Dendrimer-Based NPs

Dendrimers are chemically synthesized, highly branched polymers with a highly symmetric spherical shape. They are typically made from natural or synthetic components, including sugars, nucleotides and amino acids.172 Drugs can be trapped in the core of a dendrimer through hydrogen bonding, and electrostatic or hydrophobic interactions. Hydrophobic or hydrophilic anticancer drugs can also be covalently attached to the surface of the dendrimer.173 Dendrimers are easily functionalized and have the unique advantages of high stability, excellent water solubility, low immunogenicity, and high antigenicity, making them attractive drug delivery vehicles.174 Kong et al prepared novel multifunctional dendrimer NPs, which tightly encapsulated HCPT by simple complexation. The NPs could selectively target the drug to cancer cells overexpressing integrin avb3 through high affinity interactions and shown to exhibit significantly high cytotoxicity.175 Zhang et al synthesized dendritic polymers loaded with doxorubicin (DOX) and HCPT. Studies of drug release and cellular uptake showed that the NPs were released in a pH-dependent manner and efficiently taken up by MCF-7 cells in anticancer cell therapy exhibited enhanced anticancer effects.176 Guo et al prepared HCPT-loaded NRs using fluorescently labeled low-PEG co-dendrimers (POC) as carriers and showed higher internalization of HCPT NRs in HepG2 and 4T1 cells and higher cytotoxicity compared to HCPT injections.177 Guo et al synthesized OEG dendrimer conjugated with octadecylamine (G2-C18) and further used as nanocarriers to prepare HCPT nanospheres and NRs by inverse solvent precipitation method. The NRs were shown to have significantly enhanced cytotoxicity compared to nanospheres and exhibited significantly higher antitumor activity in vivo.178

Cyclodextrin-Based NPs

Cyclodextrins are a family of cyclic oligosaccharides with a hydrophilic outer surface and a lipophilic central cavity that have the unique ability to form inclusion complexes with a variety of organic and inorganic lipophilic molecules.179 Used primarily as complexing agents to increase the aqueous solubility of insoluble drugs and to enhance their bioavailability and stability, cyclodextrins hold promise in a wide range of areas such as drug delivery, cancer therapy, gene delivery, and biosensing.180 Lignin is an amorphous polyphenol with properties such as non-cytotoxicity.181 Zhou et al grafted β-cyclodextrin (β-CD) with a hollow ring structure onto enzymatically hydrolyzed lignin to form NPs encapsulating HCPT. The results showed that the HCPT NPs were effective in inhibiting tumor cell growth, exhibiting higher specific surface area and porosity as well as better encapsulation and retarded release.182 The supramolecular assembly of cyclodextrin-based polyrotaxane is also attracting more and more researchers. Zhang et al prepared HCPT-loaded cyclodextrin polyrotaxanes NPs and found that the NPs were more readily taken up by tumor cells and could effectively inhibit tumor growth and prolong the survival time of tumor-bearing mice.183

Electrospun Fiber -Based NPs

Electrostatic spinning is an effective and versatile method for the preparation of continuous polymer nanofibers, electrospun fibers due to their high loading capacity and encapsulation efficiency, surface area and porosity, and modification potential. They have promising applications in cancer diagnosis and treatment and are treated as promising candidates for drug delivery.184 Wang et al developed HCPT-loaded fiber membranes by electrostatic spinning. They found that drug release from HCPT-loaded fiber membranes lasted for approximately seven days and well inhibited the growth of rabbit subcutaneous VX2 tumors.185 Luo et al emulsion prepared core-sheath structured fibers with core-loaded HCPT by electrostatic spinning. In vitro and in vivo antitumor efficacy assays showed that HCPT-loaded fibers showed superior in vivo antitumor activity and fewer side effects than free HCPT.186 Wei et al constructed fragmented fibers of negative HCPT by freeze-cutting neatly aligned electrospun fibers. The results showed that longer length fragment fibers indicated higher accumulation in the tumor and better retention at the injection site, and the HCPT-loaded fiber fragments showed superior in vivo antitumor activity and fewer side effects.187 Luo et al prepared electrospun fibers loaded with HCPT, which can degrade in an acidic environment and release the drug. Cytotoxicity assays showed that the electrospun fibers showed 6-fold higher inhibitory activity against HepG2 cells after incubation in pH 6.8 medium compared to pH 7.4.188 Combretastatin A-4 (CA4), a commonly used clinical vascular disruptor, inhibits the growth and metastasis of various tumors.189 Luo et al prepared electrostatic spun fibers loaded with combretastatin A-4 (CA4) and HCPT. In situ breast tumor model, fibers loaded with CA4 and HCPT showed superior antitumor efficacy and higher survival rates than fibers loaded with individual drugs.190 Phenolic derivatives, especially tea polyphenols (TP), showed significant ROS inhibitory properties.191 Li et al developed an electrospun polymer patch from an emulsion to make nanofibers loaded with HCPT and hydrophilic TP. The nanofibers were shown to significantly synergistic tumor suppression.192

Polysaccharide-Based NPs

Polysaccharide-based biomaterials (chitosan, hyaluronic acid (HA), starch, pectin, cellulose, etc.) have received significant attention in the pharmaceutical field due to their excellent biodegradability and biocompatibility, while being easily modified chemically or physically, especially in the delivery of drugs for oncology treatment.193

Chitosan-Based NPs

Chitosan is a naturally occurring linear polysaccharide cationic hydrophilic polymer produced by the alkaline decomposition of chitin.194 Chitosan and its derivatives are widely used in various biomedical applications due to their unique biocompatibility, mucosal adhesion, non-toxicity and gel-forming ability.195,196 Studies have shown that chitosan NPs prolong the duration of HCPT administration, improve oral bioavailability, and improve antitumor effects while reducing side effects.197–199 Positively charged chitosan NPs exhibited slow-release behavior, ensuring higher intracellular drug concentrations and more significant cytotoxicity.200 FA-modified chitosan nanoneedles (Figure 5) and micelles exhibit significantly enhanced cellular uptake, higher cytotoxicity against folate-receptor-positive tumor cells.201,202 Loaded with HCPT and MTX-dual drug chitosan NPs that exhibit sustained and prolonged drug release properties, as well as effective cellular internalization and enhanced cytotoxicity.203 Studies have shown that nitric oxide (NO) is an endogenous gas transmitter can overcome multidrug resistance (MDR) by making nitrification of certain tyrosine residue of P-glycoprotein (Pgp) to inhibit the pumping function.204 Niu et al formulated chitosan NPs loaded with HCPT by ionic gel binding and chemical cross-linking methods. The NPs possessed GSH-responsive NO donor release and pH-sensitive charge reversal ability, significantly enhanced cellular uptake at pH 6.5, and improved anti-tumor ability against drug-resistant tumors.205

Figure 5 (A) Illustration of the completely green method of preparing chitosan nanoneedles. (B and C) The SEM images of chitosan nanoneedles.

Notes: Reproduced from Wu S, Yang X, Zou M, Hou Z, Yan J. A New Method Without Organic Solvent to Targeted Nanodrug for Enhanced Anticancer Efficacy. Nanoscale research letters. 2017;12(1):416.201

HA-Based NPs

HA is a naturally occurring linear polysaccharide with excellent hydrophilicity, biocompatibility, biodegradability and low immunogenicity, making it one of the most attractive biopolymers for biomedical research and applications.206 Due to its intrinsic affinity for CD44 (a receptor highly expressed on various cancer cells), HA has been widely designed for targeted anti-tumor drug-loaded NPs.207 Chen et al developed a polymeric nanocomplex targeting CD44 by encapsulating HCPT into HA NPs. Compared with free HCPT, the NPs exhibited excellent biocompatibility, tumor cell targeting and specificity.208 Li et al developed HA-modified NPs based on loaded HCPT and MTX. In vivo near-infrared fluorescence and photoacoustic (PA) bimodal imaging demonstrated effective aggregation of NPs at tumor sites through passive plus active targeting, producing highly synergistic tumor cell killing and tumor growth inhibitory effects.209

Starch-Based NPs

Starch is one of the most abundant biopolymers in nature and is usually isolated from plants in the form of microgranules. Since starch is environmentally friendly, starch NPs are considered a promising biomaterial and are widely used as binders, disintegrants and fillers in drug delivery systems.210 Amphiphilic α-tocopherol branched chain starch NPs, proved to be more cytotoxic than free HCPT.211 Some experiments have also demonstrated that hydroxyethyl starch NPs loaded with HCPT have superior tumor suppressive effects and lower side effects.212,213 Li et al prepared pH/reduction/α-amylase multisensitive hydroxyethyl starch micelles for amino acid transport proteins overexpressed on the surface of hepatocellular carcinoma cells. This micelle has a regular structure, suitable particle size and excellent drug release performance, and has better in vitro anti-proliferative ability and in vivo anti-tumor effect on human hepatocellular carcinoma HepG2 cells compared with conventional micelles and HCPT injection.214

Pectin-Based NPs

Pectin is one of the few polysaccharides with biomedical activity. The prevalence of hydroxyl and carboxyl groups in pectin contributes to their hydrophilicity and therefore excellent biocompatibility, low toxicity and biodegradability, making it an essential candidate for biomedical and drug delivery applications.215 The galactose residue of the pectin molecule can be recognized by the asialoglycoprotein receptor on the surface of the liver cancer cell, promoting the rapid penetration and release of the HCPT.216 Multi-drug combinations play an important role in tumor treatment. Research reports ursolic acid (UA)-HCPT loaded pectin NPs217,218 and dihydroartemisinin (DHA)-HCPT pectin NPs219 both exhibited significant synergistic effects and enhanced antitumor effects.

Cellulose-Based NPs

As a biopolymer with relatively low cost, fine cross-section, excellent hydrophilicity, biocompatibility, high strength, processability, volumetric stability and durability, cellulose is playing an increasing role as a carrier material in the biomedical field.220 Studies showed carboxymethylcellulose NPs have attractive properties, including easy preparation, suitable size, high HCPT loading capacity, well stability, rapid uptake by tumor cells, high synergistic effect and few side effects.221 With a rapid HCPT release in response to acidic intracellular stimuli while maintaining sufficient stability under normal physiological conditions.222

Other Polysaccharide-Based NPs

Ganoderma lucidum polysaccharide (GLP), as the main bioactive components of Ganoderma lucidum, are natural polysaccharides with medicinal value, safe and non-toxic, and are widely used in drug delivery.223 Zheng et al synthesized MTX and HCPT loaded GLP NPs. The NPs are irregularly spherical with uniform particle size, have high drug loading capacity and excellent biocompatibility, and can be rapidly released in the acidic microenvironment of tumor cells.224 As a natural and renewable biomolecule, dextran is not only biodegradable but also has excellent biocompatibility.225 Zhao et al constructed HCPT-loaded cinnamaldehyde (CA)-dextran polymers. The NPs not only induce cancer cell death by generating ROS, but also promote drug uptake, effectively prolonging drug circulation and increasing drug accumulation at tumor sites.226 Xylan is the main component of hemicellulose, an important polysaccharide.227 It has been shown that xylan modified NPs could be specifically recognized by ASGPR on the membrane surface of tumor cells, thus allowing the HCPT NPs to target hepatocellular carcinoma cells.125 It has excellent cytotoxic and apoptosis-inducing effects on HepG2 cells.228

Cell Membrane-Based NPs

Cell membrane-based NPs, a new class of bio-NPs, are widely used for drug delivery because of their cell-specific targeting, biocompatibility, biodegradability and long circulating half-life.229,230 Fan et al prepared a C6 cell membrane-encapsulated NPs using the ultrasonic encapsulation method. The NPs were spherical in shape, with core-shell structure, homologous cancer cell membrane-targeting mechanism and immune escape ability.231 Ye et al developed and prepared red blood cell (RBC) membrane -camouflaged NPs loaded with HCPT and indocyanine green (ICG). Stimulated by near-infrared (NIR) laser and acidic stimulation, the NPs can rapidly disintegrate and accelerate drug release. The dual stimulation of NPs achieved efficient apoptosis in cancer cells by chemotherapy and photothermal therapy (PTT) compared to treatment alone.232 Zhang et al synthesized NCs with high loading of HCPT by a mild nanoprecipitation process. Camouflaged cancer cell membranes (CMs) composed of a large number of membrane proteins conferred homotypic targeting ability of NCs at tumor sites. The photosensitizer ICG not only converts light energy into heat for PTT, but also promotes the breakdown of NCs, achieving high-performance tumor suppression in a triple-negative breast cancer model.233

Other Organic NPs

In addition to the above mentioned involved nanocarriers, there are some other organic carriers. Zwitterionic materials such as betaine are a class of superhydrophilic biomaterials that are expected to be an ideal substitute for PEG.234 Li et al designed a zwitterionic sulfobetaine surfactant TSSB as a novel surface modifier for NRs loaded with HCPT. The TSSB-modified NRs were found to have higher cellular uptake efficiency and exhibit higher cytotoxicity through caveolin-mediated endocytosis.235 Sun et al chemically synthesized NRs with 5-fluorouracil, HCPT and valine-citrulline monomethyl auristatinE (vcMMAE) primers. The NRs showed high specific affinity and improved internalization with enhanced antitumor efficacy.236 Luo et al encapsulated HCPT into matrix polymers containing acid unstable chain segments and galactose fraction by electrospray technique. Compared with other NPs preparation methods, electrosprayed NPs demonstrated improved drug delivery efficiency and prolonged HCPT release. Facilitated uptake of PGBELA NPs into HepG2 cells and accumulation in tumors of H22 tumor-bearing mice demonstrated the targeting ability of the galactose fraction.237 Chen et al synthesized novel diblock copolymers loaded with HCPT, which have a relatively high drug loading capacity, significant stability, longer blood retention time and better therapeutic efficacy compared to free HCPT.238 Sun et al developed a novel multipolyprodrug-arm hyperbranched amphiphiles (hPCM) that were efficiently internalized by MCF-7 cells and exhibited comparable cytotoxicity compared to free HCPT.239

Inorganic NPs

Compared with organic NPs, inorganic NPs such as silicon dioxide (SiO2), gold (Au), magnetic nanomaterials, graphene oxide (GO), copper (Cu), Prussian blue, platinum (Pt) and calcium carbonate (CaCO3) exhibit intrinsic structural robustness and relatively low manufacturing costs, and have great potential for cancer therapy.240

SiO2-Based NPs

Mesoporous SiO2 NPs have large specific surface area, large pore capacity, uniform and adjustable pore size, and stable skeleton, which has led to the widespread use of SiO2-based materials and their oxides for drug delivery.241 Fan et al prepared SiO2@Au NPs loaded with HCPT and DOX. The results showed that the NPs could specifically target tumor cells and rapidly internalize them, which enhanced their sensitivity to the drug.242 As a monocarboxylate transporter (MCT), MCT-4 allows lactate efflux to maintain intracellular pH stability and induces weakly acidic TME.243 Silencing the expression of MCT-4 increases the intracellular lactate content of tumor cells and induces apoptosis. Li et al prepared hollow mesoporous SiO2 NPs loaded with HCPT and siMCT-4 (Figure 6). The NPs could respond to weakly acidic TME and high levels of GSH in tumor cells. Combining inhibition of lactate efflux and chemotherapy effectively removed immunosuppressive TME, inhibited tumor growth, and suppressed lung metastasis from B16F10 cells and 4T1 cells.244

Figure 6 (A) Synthesis of SiO2 NPs and stimuli-responsive degradation. (B) The SiO2 NPs directly induces tumor cell apoptosis though HCPT and the increased intracellular lactate.

Notes: Reprinted with permission from Li K, Lin C, He Y, et al. Engineering of Cascade-Responsive Nanoplatform to Inhibit Lactate Efflux for Enhanced Tumor Chemo-Immunotherapy. ACS Nano. 2020;14(10):14164–14180. Copyright 2020 American Chemical Society.244

Magnetic Material-Based NPs

Magnetic nanomaterials can be loaded with drugs into NPs by electrostatic adsorption, encapsulation, and covalent binding and selectively transferred to focal sites by external magnetic field guidance.245 Due to their superparamagnetic properties, targeting, biocompatibility, and ease of surface modification have been widely used in biomedical applications, especially for drug delivery for cancer therapy.246 Liu et al developed a phase-change FA-targeted perfluoropentane (PFP) nanodrop containing HCPT and superparamagnetic Fe3O4. After intravenous administration to nude mice bearing SKOV3 ovarian cancer, the nanodrops exhibited enhanced magnetic resonance (MR) and PA imaging.247 The key role of integrin αVβ3 in tumor angiogenesis and metastasis has been identified as an ideal therapeutic target for tumor chemotherapy.248 Ding constructed arginine-glycine-aspartic acid-cysteine (RGDC) tetrapeptide functionalized and HCPT-encapsulated magnetic NPs for integrin αVβ3-targeted drug delivery. The NPs exhibit superior anti-cell migration activity with significantly enhanced cytotoxicity against A549 cells overexpressing αVβ3 compared to free HCPT and non-targeted micelles.249 Studies showed that the HCPT-encapsulated magnetic NPs exhibited enhanced activity in inhibiting tumor cells migration and could be rapidly and efficiently internalized into cells at the target site under external magnetic guidance, thereby selectively killing cells in the region.250,251 Yang et al fabricated HCPT-loaded iron oxide NRs having a drug loading capacity of up to 72%. In vitro studies have shown that the NRs have imaging capabilities and excellent chemical-photothermal synergistic effects for tumor ablation. More importantly, 100% in vivo tumor elimination was achieved at low laser power density with no weight loss and tumor recurrence.252 Wang et al constructed novel magnetic NPs of HA-bound iron oxide nanoparticle (IONP). Dopamine can form a stable and strong shell on the surface of the NPs, which results in excellent biocompatibility, biostability and tumor targeting of the NPs. The NPs have triple tumor targeting ability through magnetic targeting, CD44 molecular targeting and passive EPR targeting.253 Qu et al prepared HCPT-loaded chitosan-coated Fe3O4 NPs. Cytotoxicity experiments showed that the NPs showed significantly increased antitumor activity against HepG2 cells compared to free HCPT. The NPs also produced a local thermotherapeutic (42–45°C) effect on cancer when an external local alternating magnetic field was added.254

Cu-Based NPs

Cu NPs have attracted increasing interest in biomedical applications.255 Metal organic backbones (MOFs) are widely used for drug delivery due to their porous structure as well as their large specific surface area and simple synthesis process, especially in cancer therapy.256 Shi et al synthesized a hydrophilic Cu organic backbone with an amphiphilic carboxylic acid ligand as a connector, improved the solubility of HCPT in water. Cytotoxicity tests showed excellent solubilization, slow release and excellent biocompatibility of hydrophilic NPs.257

Au-Based NPs

Au NPs are increasingly used in drug delivery due to their unique physicochemical and optical properties and low toxicity.258 Compared with organic nanocarriers used for therapeutic agents, Au NPs exhibit superior properties as drug delivery carriers, including inertness, proven synthesis strategies, adjustable size, and flexible and easy surface modification with various chemical and biological molecules.259 Li et al prepared Au NPs loaded with HCPT by electrostatic deposition (Figure 7). The NPs can release drugs on demand in the NIR while exerting photothermal effects. 100% in vivo tumor elimination was achieved at a low laser irradiation power density of 1 W·cm−2 with no weight loss or tumor recurrence.260 Bao et al prepared a series of HCPT-Au NPs of different sizes and compared their cytotoxic effects in vitro and antitumor effects in vivo. Transmission electron micrographs showed that the NPs were round and regular in shape with an average diameter of about 10, 25 and 50 nm. It was found that NPs with an average diameter of 50 nm showed the strongest oncogenic activity against mouse MDA-MB-231 tumors.261 Wang et al prepared NPs with Au NPs as the core and FA-conjugated amphiphilic Zein-polydopamine (PDA) as the shell. The surface modification of FA conjugated PDA made the NPs more stable and also promoted the selective cellular internalization and enhanced endocytosis of the NPs. NPs exert superior tumor suppressive ability and low side effects compared to free HCPT and its non-targeted equivalents due to their active and passive targeted delivery in vitro and in vivo.262 Evans Blue (EB) is a non-toxic dye that is widely used because of its excellent human serum albumin (HSA) binding affinity and excellent hydrophilicity.263 Wang et al constructed truncated Evans Blue (tEB)-modified Au NRs for better tumor treatment. HSA/HCPT was further complexed with Au NRs through the high binding affinity of tEB with albumin. In vitro and in vivo studies confirmed that the resulting the NRs have excellent tumor targeting, photothermal conversion efficiency and biostability.264

Figure 7 Schematic diagram of the preparation of Au NPs loaded with HCPT.

Notes: Reproduced from Li W, Zhang X, Zhou M, et al. Functional core/shell drug nanoparticles for highly effective synergistic cancer therapy. Adv Healthc Mater. 2014;3(9):1475–1485. © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.260

GO-Based NPs

GO is widely studied as a drug nanocarrier due to its high surface area, photothermal properties, high loading capacity and efficient cellular uptake.265 Liu et al prepared starch-functionalized graphene nanosheets loaded with HCPT by the physical adsorption method. The nanosheets exhibited excellent biocompatibility and high drug loading capacity. The nanosheets exhibited high toxicity to SW-620 cells under the dual action of the acidic microenvironment and SW-620 cell amylase.266 Huang et al prepared HCPT NPs using carboxymethyl chitosan and GO modified with HA as carriers. The NPs could target human hepatocellular carcinoma cells, improve drug uptake, and enhance the efficacy of HCPT in vitro and in vivo.267

Prussian Blue-Based NPs

Prussian blue NPs have attracted increasing research interest in bioimaging, drug delivery and applications as therapeutic agents due to their large inner pore size, tunable dimensions, ease of synthesis and surface modification, excellent thermal stability and excellent biocompatibility.268 Jing et al reported hollow Prussian blue NPs loaded with HCPT and modified by HA-grafted PEG. The NPs were found to have excellent colloidal stability, prolonged circulation time, and the ability to target Hela cells overexpressing CD44 receptor. The NPs exhibited excellent photothermal efficiency and light-triggered drug release under NIR irradiation and showed significant inhibitory effects on cancer cells.269

Pt-Based NPs

Pt NPs have superior physicochemical properties and great potential in biomedical applications.270 The combination of PDT with chemotherapy is of increasing interest, and the hypoxic nature of tumors greatly hinders the efficiency of PDT.271 Fu et al prepared porous shuttle-shaped platinum methylene blue coordination polymer NPs loaded with HCPT. The NPs have a spatiotemporally controlled O2 self-supply, self-administration of singly linear oxygen (1O2) and excellent photothermal effects. Once they are taken up by tumor cells, the NPs acting as cascade catalysts can effectively catalyze the degradation of endogenous hydrogen peroxide (H2O2) into O2 to alleviate tumor hypoxia. Subsequently, stimulated by external NIR irradiation and internal lysosomal acidity, the NPs can achieve on-demand release of HCPT to increase in situ mitochondrial ROS and efficient tumor ablation by phototherapy.272

CaCO3-Based NPs

In recent years, the applications of CaCO3 NPs have gained extensive interest as targeted drug/gene delivery systems to cancerous tissues and cells due to their accessibility, low cost, safety, biocompatibility, pH-sensitivity, and slow biodegradability.273 Immunogenic cell death (ICD) can reinforce tumor immunotherapy by stimulating the auto-immune system through secreting associated signals. Qiu et al prepared pH-responsive NPs based on amorphous calcium carbonate loaded with HCPT and the photosensitizer Chlorin e6 (Ce6). Chemodynamic therapy (CDT)/PDT triggered immunogenic death for tumor eradication was achieved.274

Carrier-Free NPs

Carrier-free NPs are nanomedicine made from pure drug molecules without any organic and inorganic carriers involved or using only small amounts of organic molecules as surfactants to stabilize the nanomedicine or modulate the physical/chemical properties of the drug molecule so that it can self-assemble into it.275 Synthetic carrier-free NPs, can be formulated from one or more drugs or coupled with functional organic molecules such as photosensitizers using covalent bonding and physical methods. Carrier-free NPs have attracted increasing interest in cancer therapy because of their improved pharmacodynamics/pharmacokinetics, reduced toxicity, and high drug loading capacity.276,277 Some researchers have assembled HCPT and the photosensitizer Ce6 into stable NRs. These NRs not only circumvent the extreme hydrophobicity of HCPT, but also integrate two tumor treatment modalities into one. In vitro and in vivo antitumor studies have shown that NRs-mediated chemophotodynamic combination therapies exhibit superior antitumor efficacy compared to single chemotherapy or single PDT.278,279 Wei Li et al chose HCPT as a model hydrophobic drug and then used FA for surface functionalization. The FA-modified NPs were found to exhibit higher cytotoxicity due to enhanced cellular uptake of HCPT via folate receptor (FR)-mediated targeted delivery.280 Zhou et al prepared three widely used hydrophobic drugs MTX, HCPT, and paclitaxel (PTX) into a single NRs, which was then conjugated with PEG to improve its water dispersibility and bioenvironmental stability. It was shown that the NRs showed significantly higher cytotoxicity than the same dose of individual drugs and inhibited the resistance of MCF-7/ADR tumor cells to PTX.281 Zhao et al used a convenient self-assembly method was used to formulate carrier-free NPs, which improved the water solubility of HCPT. The NPs improved the retention of drug in drug-resistant MCF-7R cancer cells, effectively enhancing the cytotoxicity.282 Chen et al developed HCPT and DOX carrier-free NPs (Figure 8). When HCPT and DOX are assembled, they form small spherical drug NPs with a positive surface charge. The cellular uptake of HCPT was improved compared to HCPT alone, which exhibited enhanced synergistic cytotoxicity against breast cancer cells in vitro.283 Han et al prepared stable HCPT nanosuspensions with small and narrow size distribution using a precipitation-ultrasonication method. Compared with HCPT injection, the nanosuspension showed better anticancer effects than injection in H22-tumor bearing mice.284 Yang et al prepared NCs of HCPT using a modified acid-base micro-precipitation combined with a high-pressure homogenization technique. The NCs showed a sustained release pattern and were shown to have higher cellular uptake and antiproliferative activity than HCPT injection.285 Wang et al found tumor suppression by NPs with shape and polycrystal type dependence, needle-like NPs with longer blood retention time and more effective cellular uptake and higher cytotoxicity.286

Figure 8 (A) Schematic illustration (top) and SEM images (bottom) of the co-assembly of HCPT and DOX molecules into carrier-free NPs. The bar is 1 μm. (B) Schematic diagram showing the different intracellular drug accumulation of free HCPT and carrier-free NPs.

Notes: Reprinted with permission from Chen F, Zhao Y, Pan Y, et al. Synergistically Enhanced Therapeutic Effect of a Carrier-Free HCPT/DOX Nanodrug on Breast Cancer Cells through Improved Cellular Drug Accumulation. Molecular pharmaceutics. 2015;12(7):2237–2244. Copyright 2015 American Chemical Society.283

Discussion

The introduction of nanocarriers as drug delivery systems can be traced back to a long time ago, and in recent years people have reported different functions and shapes of NPs through diffusion effect,244 nanoprecipitation method,221 self-assembly method,198 dialysis method,202 film dispersion method,116 emulsion solvent evaporation method,89 water/oil/water double emulsion process,84 high-pressure homogenization,47 solvent-exchange method252 and other methods were used to prepare HCPT-loaded NPs with different functions and shapes. To ensure the stability of nanoparticles, various modifications have been performed on the surface of the particles. From the results, most HCPT-loaded NPs have excellent water dispersion and favorable bioenvironmental stability to release in the acidic tumor microenvironment. Depending on the type of material, HCPT-loaded NPs are classified differently and have different advantages and disadvantages (Table 1). For example, PEG, PLGA and PLA-based NPs are used as HCPT carriers for tumor therapy because of their excellent biocompatibility, stability and biodegradability. However, PEG-based NPs may limit the drug uptake by tumor cells although they prolong the circulation time of the drug in vivo. The release of PLGA-based NPs is not controlled during the initial drug delivery phase, and the same problem exists for PLA-based NPs.287–289 Similarly, although protein-based NPs have the advantages of low cytotoxicity, reproducibility, and high drug binding capacity, they suffer from weak targeting and uncontrolled drug release.290 As the most FDA-approved nanomedicine type, lipid-based NPs are known for their excellent non-toxicity, flexibility, and high drug-to-lipid ratio. However, when applied to deliver drugs, it is also found to suffer from instability, short half-life in vivo, and membrane leakage before reaching tumor sites.291 To prolong the half-life of drugs, dendrimer-based NPs were chosen but were found to be time-consuming to produce, potentially cytotoxic and non-degradable.292 When encountering insoluble drugs such as HCPT, cyclodextrins have often been chosen to increase the solubility of the drug, but cyclodextrin-based NPs were found to be easily and rapidly cleared in circulation.293 Nanogels and hydrogels are favored for their excellent biocompatibility, large surface area and adjustable properties, but also have the problem of being easily cleared.294 Electrospun fibers are increasingly used as drug delivery due to their high drug loading capacity, large surface area, high porosity and modification potential, however this it is also plagued by the problems of small pore size and lack of proper cellular infiltration inside the fiber.295 Peptide-based NPs are easy to synthesize, customizable, have better biosafety, and can recognize bioreceptors on the surface of cancer cells for targeting, but they also suffer from unstable physicochemical properties, susceptible to oxidative hydrolysis, prone to agglomeration, short half-life and does not easily cross cell membranes.296 Similar to peptides, polysaccharide-based NPs have excellent biocompatibility and degradability, and they are also readily available and easily modified, however, when they are used in practice, they are also found to have problems of easy swelling and premature disintegration.297 Cell membrane-based NPs have attracted much attention because of their cell-specific targeting, long circulating half-life and immune escape function, but it is difficult to ensure that the cell membrane can be completely coated on the drug surface when they are prepared, and it is also difficult to control the inner and outer surfaces of the cell membrane.298

Table 1 Summary of Advantages, Disadvantages and Research Stages of Different Nanocarriers of HCPT

In addition to organic nanomaterials, inorganic nanomaterials are also the focus of research on HCPT carriers. Mesoporous SiO2-based NPs are often used as nanocarriers for HCPT because of their large surface area, large pore capacity, controllable pore size adjustment and stable backbone structure, however, such carriers also have the problem of easy agglomeration.241 CaCO3-based NPs are widely used for drug delivery due to their availability, low cost, safety, biocompatibility, and pH sensitivity, but they also have the same problem of agglomeration as SiO2.299 Au-based NPs have long been used for drug delivery in HCPT due to their high surface area, tunable size, high stability, and high photothermal properties. However, they are susceptible to removal in circulation, potential biotoxicity, and non-degradability.300 Magnetic materials-based NPs have received much attention for their excellent magnetic, imaging, and photothermal conversion capabilities, however it also suffers from easy scavenging.301 GO-based NPs is of interest due to its superior properties such as high surface area, photothermal properties, high drug loading, and efficient cellular uptake, however, it is limited due to the fact that large-scale production of graphene is very difficult and expensive and may have toxicity risks.302 Cu and Pt-based NPs, one in mesoporous form with large specific surface area and simple synthesis process, and one with controlled release of drugs, are thus attracting increasing interest, yet both share a common problem of possible toxicity risk.303 Prussian blue is famous for its use as a dye, and mesoporous Prussian blue-based NPs are widely used for drug delivery due to their large internal pore size, tunable size, ease of synthesis and surface modification, excellent photothermal properties and biocompatibility, but their reproducibility and controllability are not satisfactory.304,305 In addition to the above organic and inorganic nanocarriers, there is another type of HCPT nano drug delivery system that does not require any carrier, called “carrier-free NPs”. Compared with carrier-based nano-delivery systems, carrier-free can greatly improve the drug efficacy and pharmacokinetics, reduce toxicity, and have high drug loading capacity, which is of great interest. However, they also have the defects that the self-assembly process is difficult to predict and control, easily unstable, and prone to precipitation and aggregation.276 To date, most of the reported HCPT NPs, whether organic, inorganic or carrier-free, have shown superior antitumor effects compared to free HCPT. These NPs show promise in improving the aqueous solubility of drugs, enhancing cellular uptake and cytotoxicity, enhancing tumor targeting, and achieving a combination of diagnostic and therapeutic functions. These results suggest that nano-delivery systems are now a promising approach to improve the anti-tumor efficacy of HCPT and play a unique role in tumor therapy. Currently HCPT NPs are mainly used to kill tumor cells by chemotherapy or in combination with PTT or PDT, and not many are used in combination with other innovative therapies (Table 2). Meanwhile, unfortunately none of the HCPT nanomedicines have been approved by FDA for clinical use, which overshadows the development of nanocarriers for HCPT.

Table 2 Classification and Primary Treatment Strategy of the HCPT Nano-Drug Delivery System

The main potential mechanism for nanomedicine design is the enhanced permeability and retention (EPR) effect, an approach known as passive targeting approach.306 The EPR effect was first identified and named by Maeda et al in 1986 in solid tumors of mice.307 The EPR effect has been well documented in small animal models, the number of related articles have grown exponentially, and scientists have made great efforts to develop and create nanomedicines that present a variety of shapes, sizes, therapeutic and imaging functions.308 But this creative boom has failed to translate into better clinical outcomes. The number of anticancer nanomedicines currently in clinical trials has just broken into the single digits, and the vast majority are liposomal formulations, with no major innovative nanomedicines available.309 More strikingly, even at the clinical stage nanomedicines have shown very limited improvement in the therapeutic effect of tumors.310 The EPR effect as a mechanism of nanoparticle therapy for tumors has been increasingly challenged.311 The low tumor penetration rate of nanomedicines is considered the most important reason for clinical translation failure.312 Studies showed that mouse tumor models differ significantly from human cancers in many ways, including rate of progression, metabolic rate, and tumor-to-body weight ratio.313 There is heterogeneity in the EPR effect because tumors vary greatly in blood flow and vascular permeability, and nanomedicines typically exhibit a more uniform EPR effect in small tumors and a less pronounced one in large, advanced tumors.312,314

If not the EPR effect, then what is the pathway of nanomedicines entry into tumors? Studies suggest that transcytosis, rather than the classical EPR effect, is the primary mechanism of NPs entry into tumors.315,316 When there is simply not enough endothelial space in the vessel wall to support effective NPs extravasation and accumulation, active transcytosis of endothelial cells (involving active uptake, intracellular transport, and exocytosis) to be much more effective for in vivo nanoparticle delivery to solid tumors.317 Active transcytosis allows rapid delivery of macromolecules and NPs into solid tumors, not only through alveolar epithelial cells but also across the blood-brain barrier.318 Active transcytosis creating new hope for in vivo nanomedicines delivery and tumor penetration.319

Furthermore, there is research that in addition to transcytosis, nano-enhanced immunotherapy could offer a completely different approach to cancer nanomedicine.320 Rather than designing NPs to overcome the vascular endothelial barrier, it would be more effective to use immune cells to destroy tumors. This approach bypasses the long-standing problem of in vivo delivery by formulating NPs that specifically stimulate the immune system and can effectively penetrate solid tumors.321 It has also been found that after a specific dose threshold is breached, the hepatic clearance of NPs decreases and the circulation time is prolonged, which can effectively improve the drug delivery efficiency.322 Others believe that nanomedicines can also be used for oncology applications that do not require EPR effects: reducing the toxicity of chemotherapeutic agents, local delivery of anticancer drugs, and as a tool for imaging.309 In conclusion, the development of nanomedicines still has much to offer for deeper reflection and innovation, and we should reconsider our strategies and gain a deeper understanding of nanomedicine action and tumor properties with the aim of developing nanomedicines with higher clinical translation rates.

Conclusion and Outlook

This article reviews the latest research advances of HCPT nano delivery systems for antitumor. It is found that there have been many studies related to HCPT-loaded NPs, and different types of NPs with various functions have been developed. The only troubling thing is that most of the studies on HCPT nanomedicines have remained in the papers and have not been used in the clinical setting for the treatment of tumors. This may be due to various objective conditions on the one hand, and the mechanism of nanomedicines for tumor treatment other than the EPR effect, on the other hand, is still unclear. As a promising development direction to overcome tumors, nanomedicines are regarded with high expectations. In the future, in addition to the innovation related to nanomedicine carriers and functions based on the original research, we should pay more attention to the study of the mechanism by which nanomedicine exerts anti-tumor effects. It is confident that in the near future, HCPT nanomedicines with high clinical translation rate will be developed to bring the dawn for tumor treatment.

Abbreviations

HCPT, 10-Hydroxycamptothecin; CPT, Camptothecin; EPR, Enhanced permeability and retention; NPs, Nanoparticles; NRs, Nanorods; NCs, Nanocrystals; PEG, Polyethylene glycol; PLGA, Poly (lactic-co-glycolic acid); PLA, poly (D, L-lactic acid); RES, Reticuloendothelial system; FDA, Food and Drug Administration; MTX, Methotrexate; DEX, Dexamethasone; ROS, Reactive oxygen species; siRNA, Small interfering RNA; ASGP, Asialoglycoprotein; FA, Folic acid; FR, Folate receptor; AR, Aspect ratio; GSH, Glutathione; LIFU, Low-intensity focused ultrasound (LIFU); EGFR, Epidermal growth factor; TPP, Triphenylphosphonium; HAS, Human serum albumin (HSA); BSA, Bovine serum albumin; GA, Glycyrrhetinic acid; HDL, High-density lipoproteins; CA, Casein; SDT, Sonodynamic therapy; 5-ALA, 5-Aminolevulinic acid; BBR, Berberine hydrochloride (BBR); SG, Stearyl glycyrrhetinate; HGF, Hepatocyte growth factor; β-CD, β-cyclodextrin; CA4, Combretastatin A-4; TP, Tea polyphenols; HA, Hyaluronic acid; NO, Nitric oxide; MDR, Multidrug resistance; Pgp, P-glycoprotein; PA, Photoacoustic; UA, Ursolic acid; DHA, Dihydroartemisinin; GLP, Ganoderma lucidum polysaccharide; RBC, Red blood cell; ICG, Indocyanine green; NIR, Near-infrared; CMs, Cancer cell membranes; vcMMAE, Valine-citrulline monomethyl auristatinE; SiO2, Silicon dioxide; Au, gold; GO, Graphene oxide; Cu, Copper; Pt, Platinum; CaCO3, Calcium carbonate; PFP, Perfluoropentane; MR, Magnetic resonance; MOFs, Metal organic backbones; PDA, Polydopamine; EB, Evans Blue; 1O2, Singly linear oxygen; H2O2, Hydrogen peroxide; ICD, Immunogenic cell death; CDT, Chemodynamic therapy; PTX, Paclitaxel; PTT, Photothermal therapy; PDT, Photodynamic therapy.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (Grant Nos. 81625018, 81820108022, 82003297), Program of Shanghai Academic/Technology Research Leader (18XD1403800), Natural Science Foundation of Shanghai (20ZR1459100) and Biomedical Science and Technology Support Project of Shanghai (22S11900800).

Disclosure

The authors declare that they have no competing interests.

References

1. Yeung KT, Yang J. Epithelial-mesenchymal transition in tumor metastasis. Mol Oncol. 2017;11(1):28–39. doi:10.1002/1878-0261.12017

2. Jackson CM, Choi J, Lim M. Mechanisms of immunotherapy resistance: lessons from glioblastoma. Nat Immunol. 2019;20(9):1100–1109. doi:10.1038/s41590-019-0433-y

3. Al-Yozbaki M, Wilkin PJ, Gupta GK, Wilson CM. Therapeutic potential of natural compounds in lung cancer. Curr Med Chem. 2021;28(39):7988–8002. doi:10.2174/0929867328666210322103906

4. Rejhová A, Opattová A, Čumová A, Slíva D, Vodička P. Natural compounds and combination therapy in colorectal cancer treatment. Eur J Med Chem. 2018;144:582–594. doi:10.1016/j.ejmech.2017.12.039

5. Liu C, Yang S, Wang K, et al. Alkaloids from Traditional Chinese Medicine against hepatocellular carcinoma. Biomed Pharmacother. 2019;120:109543. doi:10.1016/j.biopha.2019.109543

6. Mondal A, Gandhi A, Fimognari C, Atanasov AG, Bishayee A. Alkaloids for cancer prevention and therapy: current progress and future perspectives. Eur J Pharmacol. 2019;858:172472. doi:10.1016/j.ejphar.2019.172472

7. Song M, Yin S, Zhao R, et al. (S)-10-hydroxycamptothecin inhibits esophageal squamous cell carcinoma growth in vitro and in vivo via decreasing topoisomerase I enzyme activity. Cancers. 2019;11(12):1964. doi:10.3390/cancers11121964

8. Zhang Y, Deng Q, Hu GX, Yuan K, Yuan F, Huang YQ. 羟基喜树碱 (HCPT) 对大鼠肝星状细胞增殖与凋亡的影响 [Effect of hydroxycamptothecin (HCPT) on proliferation and apoptosis of rat hepatic stellate cells]. Zhonghua Gan Zang Bing Za Zhi = Zhonghua Ganzangbing Zazhi. 2010;18(3):199–203. Chinese. doi:10.3760/cma.j.issn.1007-3418.2010.03.012

9. Wang T, Ding Y, Yang Y, et al. Synergistic antitumour effects of triptolide plus 10-hydroxycamptothecin onbladder cancer. Biomed Pharmacother. 2019;115:108899. doi:10.1016/j.biopha.2019.108899

10. Chai LP, Su ZZ, Xian ZX. 羟基喜树碱对喉鳞癌细胞株抑制作用的研究 [Inhibition of hydroxycamptothecin on laryngeal squamous carcinoma cell line]. Ai zheng = Aizheng. 2003;22(4):372–375. Chinese.

11. Wei Y, Li C, Zhang Y, et al. Hydroxycamptothecin mediates antiproliferative effects through apoptosis and autophagy in A549 cells. Oncol Lett. 2018;15(5):6322–6328. doi:10.3892/ol.2018.8107

12. Guo XM, Guo B, Zhang Q, Sun X. Absorption of 10-hydroxycamptothecin on Fe3O4 magnetite nanoparticles with layer-by-layer self-assembly and drug release response. Dalton Trans. 2011;40(12):3039–3046. doi:10.1039/c0dt01455a

13. Duan L, Yang L, Jin J, et al. Micro/nano-bubble-assisted ultrasound to enhance the EPR effect and potential theranostic applications. Theranostics. 2020;10(2):462–483. doi:10.7150/thno.37593

14. Zhou F, Teng F, Deng P, Meng N, Song Z, Feng R. Recent progress of nano-drug delivery system for liver cancer treatment. Anticancer Agents Med Chem. 2018;17(14):1884–1897. doi:10.2174/1871520617666170713151149

15. Patra JK, Das G, Fraceto LF, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology. 2018;16(1):71. doi:10.1186/s12951-018-0392-8

16. Liu J, Huang Y, Kumar A, et al. pH-sensitive nano-systems for drug delivery in cancer therapy. Biotechnol Adv. 2014;32(4):693–710. doi:10.1016/j.biotechadv.2013.11.009

17. George A, Shah PA, Shrivastav PS. Natural biodegradable polymers based nano-formulations for drug delivery: a review. Int J Pharm. 2019;561:244–264. doi:10.1016/j.ijpharm.2019.03.011

18. Singh AK, Pandey A, Tewari M, et al. Prospects of nano-material in breast cancer management. Pathol Oncol Res. 2013;19(2):155–165. doi:10.1007/s12253-013-9609-1

19. Zhou S, Zhong Q, Wang Y, et al. Chemically engineered mesoporous silica nanoparticles-based intelligent delivery systems for theranostic applications in multiple cancerous/non-cancerous diseases. Coord Chem Rev. 2022;2022:452.

20. Bobo D, Robinson KJ, Islam J, Thurecht KJ, Corrie SR. Nanoparticle-based medicines: a review of FDA-approved materials and clinical trials to date. Pharm Res. 2016;33(10):2373–2387. doi:10.1007/s11095-016-1958-5

21. Souri M, Soltani M, Moradi Kashkooli F, Kiani shahvandi M. Engineered strategies to enhance tumor penetration of drug-loaded nanoparticles. J Control Release. 2022;341:227–246. doi:10.1016/j.jconrel.2021.11.024

22. Zi Y, Yang K, He J, Wu Z, Liu J, Zhang W. Strategies to enhance drug delivery to solid tumors by harnessing the EPR effects and alternative targeting mechanisms. Adv Drug Deliv Rev. 2022;188:114449. doi:10.1016/j.addr.2022.114449

23. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20(2):101–124. doi:10.1038/s41573-020-0090-8

24. Kumar S, Singhal A, Narang U, Mishra S, Kumari P. Recent progresses in organic-inorganic nano technological platforms for cancer therapeutics. Curr Med Chem. 2020;27(35):6015–6056. doi:10.2174/0929867326666181224143734

25. Hussein Kamareddine M, Ghosn Y, Tawk A, et al. Organic nanoparticles as drug delivery systems and their potential role in the treatment of chronic myeloid leukemia. Technol Cancer Res Treat. 2019;18:1533033819879902. doi:10.1177/1533033819879902

26. Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev. 2016;99(Pt A):28–51. doi:10.1016/j.addr.2015.09.012

27. D’Souza AA, Shegokar R. Polyethylene glycol (PEG): a versatile polymer for pharmaceutical applications. Expert Opin Drug Deliv. 2016;13(9):1257–1275. doi:10.1080/17425247.2016.1182485

28. Haider T, Pandey V, Banjare N, Gupta PN, Soni V. Drug resistance in cancer: mechanisms and tackling strategies. Pharmacol Rep. 2020;72(5):1125–1151. doi:10.1007/s43440-020-00138-7

29. Li C, Dai J, Zheng D, et al. An efficient prodrug-based nanoscale delivery platform constructed by water soluble eight-arm-polyethylene glycol-diosgenin conjugate. Mater Sci Eng C Mater Biol Appl. 2019;98:153–160. doi:10.1016/j.msec.2018.12.078

30. Jia M, Li Y, Yang X, et al. Development of both methotrexate and mitomycin C loaded PEGylated chitosan nanoparticles for targeted drug codelivery and synergistic anticancer effect. ACS Appl Mater Interfaces. 2014;6(14):11413–11423. doi:10.1021/am501932s

31. Yücel O, Şengelen A, Emik S, Önay-uçar E, Arda N, Gürdağ G. Folic acid-modified methotrexate-conjugated gold nanoparticles as nano-sized trojans for drug delivery to folate receptor-positive cancer cells. Nanotechnology. 2020;31(35):355101. doi:10.1088/1361-6528/ab9395

32. Li Y, Lin J, Cai Z, et al. Tumor microenvironment-activated self-recognizing nanodrug through directly tailored assembly of small-molecules for targeted synergistic chemotherapy. J Control Release. 2020;321:222–235. doi:10.1016/j.jconrel.2020.02.025

33. Li Y, Lin J, Liu G, et al. Dual-acting, function-responsive, and high drug payload nanospheres for combining simplicity and efficacy in both self-targeted multi-drug co-delivery and synergistic anticancer effect. Int J Pharm. 2016;512(1):194–203. doi:10.1016/j.ijpharm.2016.08.035

34. Black R, Grodzinsky AJ. Dexamethasone: chondroprotective corticosteroid or catabolic killer? Eur Cell Mater. 2019;38:246–263. doi:10.22203/eCM.v038a17

35. Meng Q, Hu H, Jing X, et al. A modular ROS-responsive platform co-delivered by 10-hydroxycamptothecin and dexamethasone for cancer treatment. J Control Release. 2021;340:102–113. doi:10.1016/j.jconrel.2021.10.027

36. Zhao J, Feng SS. Nanocarriers for delivery of siRNA and co-delivery of siRNA and other therapeutic agents. Nanomedicine. 2015;10(14):2199–2228. doi:10.2217/nnm.15.61

37. Singh A, Trivedi P, Jain NK. Advances in siRNA delivery in cancer therapy. Artif Cells, Nanomed Biotechnol. 2018;46(2):274–283. doi:10.1080/21691401.2017.1307210

38. Zhang MM, Bahal R, Rasmussen TP, Manautou JE, Zhong XB. The growth of siRNA-based therapeutics: updated clinical studies. Biochem Pharmacol. 2021;189:114432. doi:10.1016/j.bcp.2021.114432

39. Li S, Saw PE, Lin C, et al. Redox-responsive polyprodrug nanoparticles for targeted siRNA delivery and synergistic liver cancer therapy. Biomaterials. 2020;234:119760. doi:10.1016/j.biomaterials.2020.119760

40. Hafeez U, Parakh S, Gan HK, Scott AM. Antibody-drug conjugates for cancer therapy. Molecules. 2020;25(20):4764. doi:10.3390/molecules25204764

41. Liu KF, Liu YX, Dai L, et al. A novel self-assembled pH-sensitive targeted nanoparticle platform based on antibody-4arm-polyethylene glycol-pterostilbene conjugates for co-delivery of anticancer drugs. J Mater Chem B. 2018;6(4):656–665. doi:10.1039/C7TB02485A

42. Guo Y, Wang T, Qiu H, et al. Hydroxycamptothecin nanoparticles based on poly/oligo (ethylene glycol): architecture effects of nanocarriers on antitumor efficacy. Eur J Pharmaceut Biopharma. 2019;134:178–184. doi:10.1016/j.ejpb.2018.12.003

43. Kang S, Lee S, Park S. iRGD peptide as a tumor-penetrating enhancer for tumor-targeted drug delivery. Polymers. 2020;12(9):1906.

44. Yin H, Yang J, Zhang Q, et al. iRGD as a tumor‑penetrating peptide for cancer therapy (Review). Mol Med Rep. 2017;15(5):2925–2930. doi:10.3892/mmr.2017.6419

45. Li C, Chen Z, Zheng D, Zhao J, Lei J. Targeted delivery of dual anticancer drugs based on self-assembled iRGD-modified soluble drug-polymer pattern conjugate nanoparticles. ACS Appl Bio Mater. 2021;4(2):1499–1507. doi:10.1021/acsabm.0c01388

46. Jacob S, Nair AB, Shah J. Emerging role of nanosuspensions in drug delivery systems. Biomater Res. 2020;24:3. doi:10.1186/s40824-020-0184-8

47. Yang L, Jiang J, Hong J, et al. High drug payload 10-hydroxycamptothecin nanosuspensions stabilized by cholesterol-PEG: in vitro and in vivo investigation. J Biomed Nanotechnol. 2015;11(4):711–721. doi:10.1166/jbn.2015.2050

48. Yang L, Hong J, Di J, et al. 10-Hydroxycamptothecin (HCPT) nanosuspensions stabilized by mPEG(1000)-HCPT conjugate: high stabilizing efficiency and improved antitumor efficacy. Int J Nanomedicine. 2017;12:3681–3695. doi:10.2147/IJN.S134005

49. Wang J, Asghar S, Yang L, et al. Chitosan hydrochloride/hyaluronic acid nanoparticles coated by mPEG as long-circulating nanocarriers for systemic delivery of mitoxantrone. Int J Biol Macromol. 2018;113:345–353. doi:10.1016/j.ijbiomac.2018.02.128

50. Lv F, Liu D, Cong H, Shen Y, Yu B. Synthesis, self-assembly and drug release behaviors of a bottlebrush polymer-HCPT prodrug for tumor chemotherapy. Colloids Surf B Biointerfaces. 2019;181:278–284. doi:10.1016/j.colsurfb.2019.05.045

51. Zhang Q, Li L. Photodynamic combinational therapy in cancer treatment. J BUON. 2018;23(3):561–567.

52. Ji B, Wei M, Yang B. Recent advances in nanomedicines for photodynamic therapy (PDT)-driven cancer immunotherapy. Theranostics. 2022;12(1):434–458. doi:10.7150/thno.67300

53. Ma Q, Zhao Y, Guan Q, et al. Amphiphilic block polymer-based self-assembly of high payload nanoparticles for efficient combinatorial chemo-photodynamic therapy. Drug Deliv. 2020;27(1):1656–1666. doi:10.1080/10717544.2020.1850921

54. Zhu YX, Jia HR, Pan GY, Ulrich NW, Chen Z, Wu FG. Development of a light-controlled nanoplatform for direct nuclear delivery of molecular and nanoscale materials. J Am Chem Soc. 2018;140(11):4062–4070. doi:10.1021/jacs.7b13672

55. Kwiatkowski S, Knap B, Przystupski D, et al. Photodynamic therapy - mechanisms, photosensitizers and combinations. Biomed Pharmacother. 2018;106:1098–1107. doi:10.1016/j.biopha.2018.07.049

56. Abrahamse H, Hamblin MR. New photosensitizers for photodynamic therapy. Biochem J. 2016;473(4):347–364. doi:10.1042/BJ20150942

57. Lu S, Lei X, Ren H, et al. PEGylated dimeric BODIPY photosensitizers as nanocarriers for combined chemotherapy and cathepsin B-activated photodynamic therapy in 3D tumor spheroids. ACS Appl Bio Mater. 2020;3(6):3835–3845. doi:10.1021/acsabm.0c00394

58. Liu S, Wang L, Zhang M, et al. Tumor microenvironment-responsive nanoshuttles with sodium citrate modification for hierarchical targeting and improved tumor theranostics. ACS Appl Mater Interfaces. 2019;11(29):25730–25739. doi:10.1021/acsami.9b07957

59. Fu Y, Jang MS, Wang N, et al. Dual activatable self-assembled nanotheranostics for bioimaging and photodynamic therapy. J Control Release. 2020;327:129–139. doi:10.1016/j.jconrel.2020.07.045

60. Jing Y, Xiong X, Ming Y, et al. A multifunctional micellar nanoplatform with pH-triggered cell penetration and nuclear targeting for effective cancer therapy and inhibition to lung metastasis. Adv Healthc Mater. 2018;7(7):e1700974. doi:10.1002/adhm.201700974

61. Narmani A, Rezvani M, Farhood B, et al. Folic acid functionalized nanoparticles as pharmaceutical carriers in drug delivery systems. Drug Dev Res. 2019;80(4):404–424. doi:10.1002/ddr.21545

62. You H, Fu S, Qin X, et al. A study of the synergistic effect of folate-decorated polymeric micelles incorporating Hydroxycamptothecin with radiotherapy on xenografted human cervical carcinoma. Colloids Surf B Biointerfaces. 2016;140:150–160. doi:10.1016/j.colsurfb.2015.12.034

63. Liu K, Zheng D, Lei H, et al. Development of novel lignin-based targeted polymeric nanoparticle platform for efficient delivery of anticancer drugs. ACS Biomater Sci Eng. 2018;4(5):1730–1737. doi:10.1021/acsbiomaterials.8b00260

64. Dong P, Rakesh KP, Manukumar HM, et al. Innovative nano-carriers in anticancer drug delivery-a comprehensive review. Bioorg Chem. 2019;85:325–336. doi:10.1016/j.bioorg.2019.01.019

65. Zhou M, Zhang X, Yu C, Nan X, Chen X, Zhang X. Shape regulated anticancer activities and systematic toxicities of drug nanocrystals in vivo. Nanomedicine. 2016;12(1):181–189. doi:10.1016/j.nano.2015.09.006

66. Higgins SG, Becce M, Belessiotis-Richards A, Seong H, Sero JE, Stevens MM. High-aspect-ratio nanostructured surfaces as biological metamaterials. Adv Mater. 2020;32(9):e1903862. doi:10.1002/adma.201903862

67. Tian B, Zhang X, Yu C, Zhou M, Zhang X. The aspect ratio effect of drug nanocrystals on cellular internalization efficiency, uptake mechanisms, and in vitro and in vivo anticancer efficiencies. Nanoscale. 2015;7(8):3588–3593. doi:10.1039/C4NR06743F

68. Wu S, Yang X, Li Y, et al. Preparation of HCPT-loaded nanoneedles with pointed ends for highly efficient cancer chemotherapy. Nanoscale Res Lett. 2016;11(1):294. doi:10.1186/s11671-016-1491-9

69. Tang H, Zhao W, Yu J, Li Y, Zhao C. Recent development of pH-responsive polymers for cancer nanomedicine. Molecules. 2018;24(1):4. doi:10.3390/molecules24010004

70. Liu Y, Li J, Li Z, Tang X, Zhang Z. Pharmacokinetics of a ternary conjugate based pH-responsive 10-HCPT prodrug nano-micelle delivery system. Asian J Pharm Sci. 2017;12(6):542–549. doi:10.1016/j.ajps.2017.05.005

71. Liu Y, Li D, Guo X, et al. A pH-responsive prodrug delivery system of 10-HCPT for controlled release and tumor targeting. Int J Nanomedicine. 2017;12:2227–2242. doi:10.2147/IJN.S125849

72. Pu X, Zhao L, Li J, et al. A polymeric micelle with an endosomal pH-sensitivity for intracellular delivery and enhanced antitumor efficacy of hydroxycamptothecin. Acta Biomater. 2019;88:357–369. doi:10.1016/j.actbio.2019.02.039

73. Xu XD, Cheng YJ, Wu J, et al. Smart and hyper-fast responsive polyprodrug nanoplatform for targeted cancer therapy. Biomaterials. 2016;76:238–249. doi:10.1016/j.biomaterials.2015.10.056

74. Zong L, Wang Y, Qiao P, et al. Reduction-sensitive poly(ethylene glycol)-polypeptide conjugate micelles for highly efficient intracellular delivery and enhanced antitumor efficacy of hydroxycamptothecin. Nanotechnology. 2020;31(16):165102. doi:10.1088/1361-6528/ab6749

75. Zhang X, Zhang M, Wang M, et al. Facile fabrication of 10-hydroxycamptothecin-backboned amphiphilic polyprodrug with precisely tailored drug loading content for controlled release. Bioconjug Chem. 2018;29(7):2239–2247. doi:10.1021/acs.bioconjchem.8b00238

76. Zong L, Wang H, Hou X, et al. A novel GSH-triggered polymeric nanomicelles for reversing MDR and enhancing antitumor efficiency of hydroxycamptothecin. Int J Pharm. 2021;600:120528. doi:10.1016/j.ijpharm.2021.120528

77. Guo Y, Gao T, Fang F, et al. A novel polymer micelle as a targeted drug delivery system for 10-hydroxycamptothecin with high drug-loading properties and anti-tumor efficacy. Biophys Chem. 2021;279:106679. doi:10.1016/j.bpc.2021.106679

78. Li R, Wang Y, Yang Q, Lai B, Zhou X, Feng M. Enhanced stability of the pharmacologically active lactone form of 10-hydroxycamptothecin by self-microemulsifying drug delivery systems. AAPS PharmSciTech. 2020;21(8):324. doi:10.1208/s12249-020-01860-4

79. Guo Y, Wang T, Zhao S, et al. Effect of alkyl chain on cellular uptake and antitumor activity of hydroxycamptothecin nanoparticles based on amphiphilic linear molecules. Eur J Pharm Sci. 2018;124:266–272. doi:10.1016/j.ejps.2018.08.043

80. Mozhi A, Ahmad I, Kaleem QM, et al. Nrp-1 receptor targeting peptide-functionalized TPGS micellar nanosystems to deliver 10-hydroxycampothecin for enhanced cancer chemotherapy. Int J Pharm. 2018;547(1–2):582–592. doi:10.1016/j.ijpharm.2018.05.074

81. Sadat Tabatabaei Mirakabad F, Nejati-Koshki K, Akbarzadeh A, et al. PLGA-based nanoparticles as cancer drug delivery systems. Asian Pacific J Cancer Prev. 2014;15(2):517–535. doi:10.7314/APJCP.2014.15.2.517

82. Zhang Q, Wang W, Shen H, et al. Low-intensity focused ultrasound-augmented multifunctional nanoparticles for integrating ultrasound imaging and synergistic therapy of metastatic breast cancer. Nanoscale Res Lett. 2021;16(1):73. doi:10.1186/s11671-021-03532-z

83. Wang M, Yang Q, Li M, et al. Multifunctional nanoparticles for multimodal imaging-guided low-intensity focused ultrasound/immunosynergistic retinoblastoma therapy. ACS Appl Mater Interfaces. 2020;12(5):5642–5657. doi:10.1021/acsami.9b22072

84. Wang Y, Sui G, Teng D, et al. Low intensity focused ultrasound (LIFU) triggered drug release from cetuximab-conjugated phase-changeable nanoparticles for precision theranostics against anaplastic thyroid carcinoma. Biomater Sci. 2018;7(1):196–210. doi:10.1039/C8BM00970H

85. Yang X, Wu S, Xie W, et al. Dual-drug loaded nanoneedles with targeting property for efficient cancer therapy. J Nanobiotechnology. 2017;15(1):91. doi:10.1186/s12951-017-0326-x

86. Jarak I, Varela CL, Tavares da silva E, Roleira FFM, Veiga F, Figueiras A. Pluronic-based nanovehicles: recent advances in anticancer therapeutic applications. Eur J Med Chem. 2020;206:112526. doi:10.1016/j.ejmech.2020.112526

87. Zaki NM. Augmented cytotoxicity of hydroxycamptothecin-loaded nanoparticles in lung and colon cancer cells by chemosensitizing pharmaceutical excipients. Drug Deliv. 2014;21(4):265–275. doi:10.3109/10717544.2013.838808

88. Fang Y, Zhang Z. Arsenic trioxide as a novel anti-glioma drug: a review. Cell Mol Biol Lett. 2020;25:44. doi:10.1186/s11658-020-00236-7

89. Wu Y, Du D, Chen J, Liu C. Preparation of PLGA microspheres loaded with 10-hydroxycamptothecin and arsenic trioxide and their treatment for rabbit hepatocellular carcinoma. Biomed Papers Med Faculty Univ Palacky. 2021;165(1):57–63.

90. Zielonka J, Joseph J, Sikora A, et al. Mitochondria-targeted triphenylphosphonium-based compounds: syntheses, mechanisms of action, and therapeutic and diagnostic applications. Chem Rev. 2017;117(15):10043–10120. doi:10.1021/acs.chemrev.7b00042

91. Li HQ, Ye WL, Huan ML, et al. Mitochondria and nucleus delivery of active form of 10-hydroxycamptothecin with dual shell to precisely treat colorectal cancer. Nanomedicine. 2019;14(8):1011–1032. doi:10.2217/nnm-2018-0227

92. Ma Z, Liu J, Li X, et al. Hydroxycamptothecin (HCPT)-loaded PEGlated lipid-polymer hybrid nanoparticles for effective delivery of HCPT: qbD-based development and evaluation. Drug Deliv Transl Res. 2022;12(1):306–324. doi:10.1007/s13346-021-00939-0

93. Lee BK, Yun Y, Park K. PLA micro- and nano-particles. Adv Drug Deliv Rev. 2016;107:176–191. doi:10.1016/j.addr.2016.05.020

94. Hou Z, Li L, Zhan C, et al. Preparation and in vitro evaluation of an ultrasound-triggered drug delivery system: 10-hydroxycamptothecin loaded PLA microbubbles. Ultrasonics. 2012;52(7):836–841. doi:10.1016/j.ultras.2011.10.009

95. Yang X, Wu S, Wang Y, et al. Evaluation of self-assembled HCPT-loaded PEG-b-PLA nanoparticles by comparing with HCPT-loaded PLA nanoparticles. Nanoscale Res Lett. 2014;9(1):2408. doi:10.1186/1556-276X-9-687

96. Elzoghby AO, Samy WM, Elgindy NA. Protein-based nanocarriers as promising drug and gene delivery systems. J Control Release. 2012;161(1):38–49. doi:10.1016/j.jconrel.2012.04.036

97. Sleep D. Albumin and its application in drug delivery. Expert Opin Drug Deliv. 2015;12(5):793–812. doi:10.1517/17425247.2015.993313

98. Spada A, Emami J, Tuszynski JA, Lavasanifar A. The uniqueness of albumin as a carrier in nanodrug delivery. Mol Pharm. 2021;18(5):1862–1894.

99. Yang Z, Gong W, Wang Z, et al. A novel drug-polyethylene glycol liquid compound method to prepare 10-hydroxycamptothecin loaded human serum albumin nanoparticle. Int J Pharm. 2015;490(1–2):412–428.

100. Wu F, Li X, Jiang B, et al. Glycyrrhetinic acid functionalized nanoparticles for drug delivery to liver cancer. J Biomed Nanotechnol. 2018;14(11):1837–1852. doi:10.1166/jbn.2018.2638

101. Zu Y, Meng L, Zhao X, et al. Preparation of 10-hydroxycamptothecin-loaded glycyrrhizic acid-conjugated bovine serum albumin nanoparticles for hepatocellular carcinoma-targeted drug delivery. Int J Nanomedicine. 2013;8:1207–1222. doi:10.2147/IJN.S40493

102. Wang W, Liang H, Sun B, et al. Pharmacokinetics and tissue distribution of folate-decorated human serum albumin loaded with nano-hydroxycamptothecin for tumor targeting. J Pharm Sci. 2016;105(6):1874–1880. doi:10.1016/j.xphs.2016.03.016

103. Kuai R, Li D, Chen YE, Moon JJ, Schwendeman A. High-density lipoproteins: nature’s multifunctional nanoparticles. ACS Nano. 2016;10(3):3015–3041. doi:10.1021/acsnano.5b07522

104. Yuan Y, Wen J, Tang J, et al. Synthetic high-density lipoproteins for delivery of 10-hydroxycamptothecin. Int J Nanomedicine. 2016;11:6229–6238. doi:10.2147/IJN.S112835

105. Cui L, Wang Y, Liang M, et al. Dual-modified natural high density lipoprotein particles for systemic glioma-targeting drug delivery. Drug Deliv. 2018;25(1):1865–1876. doi:10.1080/10717544.2018.1519002

106. Rehan F, Ahemad N, Gupta M. Casein nanomicelle as an emerging biomaterial-A comprehensive review. Colloids Surf B Biointerfaces. 2019;179:280–292. doi:10.1016/j.colsurfb.2019.03.051

107. Gao C, Liang J, Zhu Y, et al. Menthol-modified casein nanoparticles loading 10-hydroxycamptothecin for glioma targeting therapy. Acta pharmaceutica Sinica B. 2019;9(4):843–857. doi:10.1016/j.apsb.2019.01.006

108. Battaglia L, Gallarate M. Lipid nanoparticles: state of the art, new preparation methods and challenges in drug delivery. Expert Opin Drug Deliv. 2012;9(5):497–508. doi:10.1517/17425247.2012.673278

109. Lucia M. Lipid-based nanoparticles as carriers for dermal delivery of antioxidants. Curr Drug Metab. 2017;18(5):469–480. doi:10.2174/1389200218666170222152038

110. Harshita MA, Das SS, Pottoo FH, Beg S, Rahman Z, Rahman Z. Lipid-based nanosystem as intelligent carriers for versatile drug delivery applications. Curr Pharm Des. 2020;26(11):1167–1180. doi:10.2174/1381612826666200206094529

111. Fan Y, Marioli M, Zhang K. Analytical characterization of liposomes and other lipid nanoparticles for drug delivery. J Pharm Biomed Anal. 2021;192:113642. doi:10.1016/j.jpba.2020.113642

112. Guimarães D, Cavaco-Paulo A, Nogueira E. Design of liposomes as drug delivery system for therapeutic applications. Int J Pharm. 2021;601:120571. doi:10.1016/j.ijpharm.2021.120571

113. Li H, Shi S, Wu M, et al. iRGD peptide-mediated liposomal nanoparticles with photoacoustic/ultrasound dual-modality imaging for precision theranostics against hepatocellular carcinoma. Int J Nanomedicine. 2021;16:6455–6475. doi:10.2147/IJN.S325891

114. Zhu J, Zhang W, Wang D, Li S, Wu W. Preparation and characterization of norcantharidin liposomes modified with stearyl glycyrrhetinate. Exp Ther Med. 2018;16(3):1639–1646. doi:10.3892/etm.2018.6416

115. Huang S, Ren D, Wu X, et al. Glycyrrhetinic acid and TAT peptide modified dual-functional liposomes for treatment of hepatocellular cancer. Curr Top Med Chem. 2020;20(27):2493–2505. doi:10.2174/1568026620666200722110244

116. Zhou T, Tang X, Zhang W, Feng J, Wu W. Preparation and in vitro and in vivo evaluations of 10-hydroxycamptothecin liposomes modified with stearyl glycyrrhetinate. Drug Deliv. 2019;26(1):673–679. doi:10.1080/10717544.2019.1636422

117. Matsumoto K, Nakamura T. NK4 (HGF-antagonist/angiogenesis inhibitor) in cancer biology and therapeutics. Cancer Sci. 2003;94(4):321–327. doi:10.1111/j.1349-7006.2003.tb01440.x

118. Zhou T, Zhang W, Cheng D, Tang X, Feng J, Wu W. Preparation, characterization, and in vivo evaluation of NK4-conjugated hydroxycamptothecin-loaded liposomes. Int J Nanomedicine. 2020;15:2277–2286. doi:10.2147/IJN.S243746

119. Xu M, Zhou L, Zheng L, et al. Sonodynamic therapy-derived multimodal synergistic cancer therapy. Cancer Lett. 2021;497:229–242. doi:10.1016/j.canlet.2020.10.037

120. Xiao Z, Zhuang B, Zhang G, Li M, Jin Y. Pulmonary delivery of cationic liposomal hydroxycamptothecin and 5-aminolevulinic acid for chemo-sonodynamic therapy of metastatic lung cancer. Int J Pharm. 2021;601:120572. doi:10.1016/j.ijpharm.2021.120572

121. Chen Y, Chen C, Xiao Y, Zhang X, Chen Y. Liposomes encapsulating 10-hydroxycamptothecin-cyclodextrin complexes and their in vitro anti-tumor activities. J Nanosci Nanotechnol. 2015;15(5):3786–3795. doi:10.1166/jnn.2015.9495

122. Mu H, Holm R. Solid lipid nanocarriers in drug delivery: characterization and design. Expert Opin Drug Deliv. 2018;15(8):771–785. doi:10.1080/17425247.2018.1504018

123. Attama AA. SLN, NLC, LDC: state of the art in drug and active delivery. Recent Pat Drug Deliv Formul. 2011;5(3):178–187. doi:10.2174/187221111797200524

124. Kulkarni AD, Joshi AA, Patil CL, et al. Xyloglucan: a functional biomacromolecule for drug delivery applications. Int J Biol Macromol. 2017;104(Pt A):799–812. doi:10.1016/j.ijbiomac.2017.06.088

125. Liu M, Chen D, Wang C, et al. Intracellular target delivery of 10-hydroxycamptothecin with solid lipid nanoparticles against multidrug resistance. J Drug Target. 2015;23(9):800–805. doi:10.3109/1061186X.2015.1020427

126. Gaba B, Fazil M, Ali A, Baboota S, Sahni JK, Ali J. Nanostructured lipid (NLCs) carriers as a bioavailability enhancement tool for oral administration. Drug Deliv. 2015;22(6):691–700. doi:10.3109/10717544.2014.898110

127. Elmowafy M, Al-Sanea MM. Nanostructured lipid carriers (NLCs) as drug delivery platform: advances in formulation and delivery strategies. Saudi Pharma J. 2021;29(9):999–1012. doi:10.1016/j.jsps.2021.07.015

128. Sun M, Li J, Zhang C, et al. Arginine-modified nanostructured lipid carriers with charge-reversal and ph-sensitive membranolytic properties for anticancer drug delivery. Adv Healthc Mater. 2017;6(8):1600693. doi:10.1002/adhm.201600693

129. Su Z, Shi Y, Xiao Y, et al. Effect of octreotide surface density on receptor-mediated endocytosis in vitro and anticancer efficacy of modified nanocarrier in vivo after optimization. Int J Pharm. 2013;447(1–2):281–292. doi:10.1016/j.ijpharm.2013.01.068

130. Kanwal U, Irfan bukhari N, Ovais M, Abass N, Hussain K, Raza A. Advances in nano-delivery systems for doxorubicin: an updated insight. J Drug Target. 2018;26(4):296–310. doi:10.1080/1061186X.2017.1380655

131. Zhao YX, Liu DX, Liang WQ, Ye ZW. In-vivo pharmacokinetics, tissue distribution and anti-tumour effect of hydroxycamptothecin delivered in oil-in-water submicron emulsions. J Pharm Pharmacol. 2012;64(6):783–791. doi:10.1111/j.2042-7158.2012.01484.x

132. Hadinoto K, Sundaresan A, Cheow WS. Lipid-polymer hybrid nanoparticles as a new generation therapeutic delivery platform: a review. Eur J Pharmaceut Biopharma. 2013;85(3 Pt A):427–443. doi:10.1016/j.ejpb.2013.07.002

133. Dave V, Tak K, Sohgaura A, Gupta A, Sadhu V, Reddy KR. Lipid-polymer hybrid nanoparticles: synthesis strategies and biomedical applications. J Microbiol Methods. 2019;160:130–142. doi:10.1016/j.mimet.2019.03.017

134. Yang Z, Luo X, Zhang X, Liu J, Jiang Q. Targeted delivery of 10-hydroxycamptothecin to human breast cancers by cyclic RGD-modified lipid-polymer hybrid nanoparticles. Biomed Mater. 2013;8(2):025012. doi:10.1088/1748-6041/8/2/025012

135. Li P, Zheng Y, Ran H, et al. Ultrasound triggered drug release from 10-hydroxycamptothecin-loaded phospholipid microbubbles for targeted tumor therapy in mice. J Control Release. 2012;162(2):349–354. doi:10.1016/j.jconrel.2012.07.009

136. Zhao H, Wu M, Zhu L, et al. Cell-penetrating peptide-modified targeted drug-loaded phase-transformation lipid nanoparticles combined with low-intensity focused ultrasound for precision theranostics against hepatocellular carcinoma. Theranostics. 2018;8(7):1892–1910. doi:10.7150/thno.22386

137. Wang HY, Hou L, Li HL, et al. A nanosystem loaded with perfluorohexane and rose bengal coupled upconversion nanoparticles for multimodal imaging and synergetic chemo-photodynamic therapy of cancer. Biomater Sci. 2020;8(9):2488–2506. doi:10.1039/C9BM02081K

138. Guo F, Fan Z, Yang J, et al. A comparative evaluation of hydroxycamptothecin drug nanorods with and without methotrexate prodrug functionalization for drug delivery. Nanoscale Res Lett. 2016;11(1):384. doi:10.1186/s11671-016-1599-y

139. Li Y, Lin J, Huang Y, et al. Self-targeted, shape-assisted, and controlled-release self-delivery nanodrug for synergistic targeting/anticancer effect of cytoplasm and nucleus of cancer cells. ACS Appl Mater Interfaces. 2015;7(46):25553–25559. doi:10.1021/acsami.5b07348

140. Sulli G, Lam MTY, Panda S. Interplay between circadian clock and cancer: new frontiers for cancer treatment. Trends Cancer. 2019;5(8):475–494. doi:10.1016/j.trecan.2019.07.002

141. Kelleher FC, Rao A, Maguire A. Circadian molecular clocks and cancer. Cancer Lett. 2014;342(1):9–18. doi:10.1016/j.canlet.2013.09.040

142. Hou L, Li H, Wang H, et al. The circadian clock gene PER2 enhances chemotherapeutic efficacy in nasopharyngeal carcinoma when combined with a targeted nanosystem. J Mater Chem B. 2020;8(24):5336–5350. doi:10.1039/D0TB00595A

143. Zhong X, Chen B, Yang Z. Nanocochleates as the potential delivery systems for oral antitumor of hydroxycamptothecin. J Biomed Nanotechnol. 2018;14(7):1339–1346. doi:10.1166/jbn.2018.2572

144. Tan W, Li Y, Chen M, Wang Y. Berberine hydrochloride: anticancer activity and nanoparticulate delivery system. Int J Nanomedicine. 2011;6:1773–1777. doi:10.2147/IJN.S22683

145. Qi Y, Liu G. Berberine-10-hydroxy camptothecine-loaded lipid microsphere for the synergistic treatment of liver cancer by inhibiting topoisomerase and HIF-1α. Drug Deliv. 2021;28(1):171–182. doi:10.1080/10717544.2020.1870020

146. Zhang Y, Chen Q, Dai Z, Dai Y, Xia F, Zhang X. Nanocomposite adhesive hydrogels: from design to application. J Mater Chem B. 2021;9(3):585–593. doi:10.1039/D0TB02000A

147. Ahmed S, Alhareth K, Mignet N. Advancement in nanogel formulations provides controlled drug release. Int J Pharm. 2020;584:119435. doi:10.1016/j.ijpharm.2020.119435

148. Li C, Obireddy SR, Lai WF. Preparation and use of nanogels as carriers of drugs. Drug Deliv. 2021;28(1):1594–1602. doi:10.1080/10717544.2021.1955042

149. Zhang Z, Hao G, Liu C, et al. Recent progress in the preparation, chemical interactions and applications of biocompatible polysaccharide-protein nanogel carriers. Food Res Int. 2021;147:110564. doi:10.1016/j.foodres.2021.110564

150. Guo H, Xu W, Chen J, et al. Positively charged polypeptide nanogel enhances mucoadhesion and penetrability of 10-hydroxycamptothecin in orthotopic bladder carcinoma. J Control Release. 2017;259:136–148. doi:10.1016/j.jconrel.2016.12.041

151. Guo H, Li F, Qiu H, et al. Synergistically enhanced mucoadhesive and penetrable polypeptide nanogel for efficient drug delivery to orthotopic bladder cancer. Research. 2020;2020:8970135. doi:10.34133/2020/8970135

152. Guo H, Li F, Xu W, et al. Mucoadhesive cationic polypeptide nanogel with enhanced penetration for efficient intravesical chemotherapy of bladder cancer. Adv Sci. 2018;5(6):1800004. doi:10.1002/advs.201800004

153. Qin L, Mei L, Shan Z, et al. Phytantriol based liquid crystal provide sustained release of anticancer drug as a novel embolic agent. Drug Dev Ind Pharm. 2016;42(2):307–316. doi:10.3109/03639045.2015.1052079

154. Tan B, Huang L, Wu Y, Liao J. Advances and trends of hydrogel therapy platform in localized tumor treatment: a review. J Biomed Mater Res A. 2021;109(4):404–425. doi:10.1002/jbm.a.37062

155. Li R, Shu C, Wang W, et al. Encapsulation of 10-hydroxy camptothecin in supramolecular hydrogel as an injectable drug delivery system. J Pharm Sci. 2015;104(7):2266–2275. doi:10.1002/jps.24481

156. Ci T, Shen Y, Cui S, Liu R, Yu L, Ding J. Achieving high drug loading and sustained release of hydrophobic drugs in hydrogels through in situ crystallization. Macromol Biosci. 2017;17(3):1600299. doi:10.1002/mabi.201600299

157. Wu C, Li R, Yin Y, Wang J, Zhang L, Zhong W. Redox-responsive supramolecular hydrogel based on 10-hydroxy camptothecin-peptide covalent conjugates with high loading capacity for drug delivery. Mater Sci Eng C Mater Biol Appl. 2017;76:196–202. doi:10.1016/j.msec.2017.03.103

158. Dai L, Liu K, Wang L, et al. Injectable and thermosensitive supramolecular hydrogels by inclusion complexation between binary-drug loaded micelles and α-cyclodextrin. Mater Sci Eng C Mater Biol Appl. 2017;76:966–974. doi:10.1016/j.msec.2017.03.151

159. Liu J, Wu C, Dai G, et al. Molecular self-assembly of a tyroservatide-derived octapeptide and hydroxycamptothecin for enhanced therapeutic efficacy. Nanoscale. 2021;13(9):5094–5102. doi:10.1039/D0NR08741F

160. Guo Q, Liu Y, Wang Z, et al. Supramolecular nanofibers increase the efficacy of 10-hydroxycamptothecin by enhancing nuclear accumulation and depleting cellular ATP. Acta Biomater. 2021;122:343–353. doi:10.1016/j.actbio.2020.12.052

161. Gaurav I, Wang X, Thakur A, et al. Peptide-conjugated nano delivery systems for therapy and diagnosis of cancer. Pharmaceutics. 2021;13(9):1433. doi:10.3390/pharmaceutics13091433

162. Wang H, Feng Z, Xu B. D-amino acid-containing supramolecular nanofibers for potential cancer therapeutics. Adv Drug Deliv Rev. 2017;110–111:102–111. doi:10.1016/j.addr.2016.04.008

163. Liu J, Liu J, Chu L, et al. Self-assembling peptide of D-amino acids boosts selectivity and antitumor efficacy of 10-hydroxycamptothecin. ACS Appl Mater Interfaces. 2014;6(8):5558–5565. doi:10.1021/am406007g

164. Zeng S, Ou H, Gao Z, et al. HCPT-peptide prodrug with tumor microenvironment -responsive morphology transformable characteristic for boosted bladder tumor chemotherapy. J Control Release. 2021;330:715–725. doi:10.1016/j.jconrel.2020.12.042

165. Zhong Y, Meng F, Deng C, Zhong Z. Ligand-directed active tumor-targeting polymeric nanoparticles for cancer chemotherapy. Biomacromolecules. 2014;15(6):1955–1969. doi:10.1021/bm5003009

166. Zhu L, Zhao H, Zhou Z, et al. Peptide-functionalized phase-transformation nanoparticles for low intensity focused ultrasound-assisted tumor imaging and therapy. Nano Lett. 2018;18(3):1831–1841. doi:10.1021/acs.nanolett.7b05087

167. Zhang Q, Zhang P, Jian S, et al. Drug-bearing peptide-based nanospheres for the inhibition of metastasis and growth of cancer. Mol Pharm. 2020;17(9):3165–3176. doi:10.1021/acs.molpharmaceut.0c00118

168. Wu J, Ding W, Han G, et al. Nuclear delivery of dual anti-cancer drugs by molecular self-assembly. Biomater Sci. 2021;9(1):116–123. doi:10.1039/D0BM00971G

169. Cai Y, Shen H, Zhan J, et al. Supramolecular “Trojan Horse” for nuclear delivery of dual anticancer drugs. J Am Chem Soc. 2017;139(8):2876–2879. doi:10.1021/jacs.6b12322

170. Zhan J, Wang Y, Ma S, et al. Organelle-inspired supramolecular nanomedicine to precisely abolish liver tumor growth and metastasis. Bioactive Mater. 2022;9:120–133. doi:10.1016/j.bioactmat.2021.07.021

171. Zheng D, Liu J, Ding Y, et al. Tandem molecular self-assembly for selective lung cancer therapy with an increase in efficiency by two orders of magnitude. Nanoscale. 2021;13(24):10891–10897. doi:10.1039/D1NR01174J

172. Mittal P, Saharan A, Verma R, et al. Dendrimers: a new race of pharmaceutical nanocarriers. Biomed Res Int. 2021;2021:8844030. doi:10.1155/2021/8844030

173. Saluja V, Mishra Y, Mishra V, Giri N, Nayak P. Dendrimers based cancer nanotheranostics: an overview. Int J Pharm. 2021;600:120485. doi:10.1016/j.ijpharm.2021.120485

174. Chauhan AS. Dendrimers for drug delivery. Molecules. 2018;23(4):938. doi:10.3390/molecules23040938

175. Kong X, Yu K, Yu M, et al. A novel multifunctional poly(amidoamine) dendrimeric delivery system with superior encapsulation capacity for targeted delivery of the chemotherapy drug 10-hydroxycamptothecin. Int J Pharm. 2014;465(1–2):378–387. doi:10.1016/j.ijpharm.2014.02.022

176. Zhang Y, Xiao C, Li M, et al. Co-delivery of 10-hydroxycamptothecin with doxorubicin conjugated prodrugs for enhanced anticancer efficacy. Macromol Biosci. 2013;13(5):584–594. doi:10.1002/mabi.201200441

177. Guo Y, Zhao Y, Wang T, et al. Hydroxycamptothecin nanorods prepared by fluorescently labeled oligoethylene glycols (OEG) codendrimer: antitumor efficacy in vitro and in vivo. Bioconjug Chem. 2017;28(2):390–399. doi:10.1021/acs.bioconjchem.6b00536

178. Guo Y, Wang T, Zhao S, et al. Amphiphilic hybrid dendritic-linear molecules as nanocarriers for shape-dependent antitumor drug delivery. Mol Pharm. 2018;15(7):2665–2673. doi:10.1021/acs.molpharmaceut.8b00190

179. Liu J, Ding X, Fu Y, et al. Cyclodextrins based delivery systems for macro biomolecules. Eur J Med Chem. 2021;212:113105. doi:10.1016/j.ejmech.2020.113105

180. Shelley H, Babu RJ. Role of cyclodextrins in nanoparticle-based drug delivery systems. J Pharm Sci. 2018;107(7):1741–1753. doi:10.1016/j.xphs.2018.03.021

181. Vanholme R, De Meester B, Ralph J, Boerjan W. Lignin biosynthesis and its integration into metabolism. Curr Opin Biotechnol. 2019;56:230–239. doi:10.1016/j.copbio.2019.02.018

182. Zhou Y, Han Y, Li G, Yang S, Chu F. Lignin-based hollow nanoparticles for controlled drug delivery: grafting preparation using β-cyclodextrin/enzymatic-hydrolysis lignin. Nanomaterials. 2019;9(7):997. doi:10.3390/nano9070997

183. Zhang Y, Zhou Q, Jia S, et al. Specific modification with TPGS and drug loading of cyclodextrin polyrotaxanes and the enhanced antitumor activity study in vitro and in vivo. ACS Appl Mater Interfaces. 2019;11(50):46427–46436. doi:10.1021/acsami.9b14075

184. Liu M, Zhang Y, Sun S, et al. Recent advances in electrospun for drug delivery purpose. J Drug Target. 2019;27(3):270–282. doi:10.1080/1061186X.2018.1481413

185. Wang L, Chang J, Qu Y, Qiu R. Combination therapy comprising irreversible electroporation and hydroxycamptothecin loaded electrospun membranes to treat rabbit VX2 subcutaneous cancer. Biomed Microdevices. 2018;20(4):88. doi:10.1007/s10544-018-0336-y

186. Luo X, Xie C, Wang H, Liu C, Yan S, Li X. Antitumor activities of emulsion electrospun fibers with core loading of hydroxycamptothecin via intratumoral implantation. Int J Pharm. 2012;425(1–2):19–28. doi:10.1016/j.ijpharm.2012.01.012

187. Wei J, Luo X, Chen M, Lu J, Li X. Spatial distribution and antitumor activities after intratumoral injection of fragmented fibers with loaded hydroxycamptothecin. Acta Biomater. 2015;23:189–200. doi:10.1016/j.actbio.2015.05.020

188. Luo X, Xu G, Song H, et al. Promoted antitumor activities of acid-labile electrospun fibers loaded with hydroxycamptothecin via intratumoral implantation. Eur J Pharmaceut Biopharma. 2012;82(3):545–553. doi:10.1016/j.ejpb.2012.08.012

189. Shan Y, Zhang J, Liu Z, Wang M, Dong Y. Developments of combretastatin A-4 derivatives as anticancer agents. Curr Med Chem. 2011;18(4):523–538. doi:10.2174/092986711794480221

190. Luo X, Zhang H, Chen M, Wei J, Zhang Y, Li X. Antimetastasis and antitumor efficacy promoted by sequential release of vascular disrupting and chemotherapeutic agents from electrospun fibers. Int J Pharm. 2014;475(1–2):438–449. doi:10.1016/j.ijpharm.2014.09.006

191. Miyata Y, Shida Y, Hakariya T, Sakai H. Anti-cancer effects of green tea polyphenols against prostate cancer. Molecules. 2019;24(1):193. doi:10.3390/molecules24010193

192. Li J, Xu W, Li D, et al. Locally deployable nanofiber patch for sequential drug delivery in treatment of primary and advanced orthotopic hepatomas. ACS Nano. 2018;12(7):6685–6699. doi:10.1021/acsnano.8b01729

193. Li M, Zhao Y, Zhang W, Zhang S, Zhang S. Multiple-therapy strategies via polysaccharides-based nano-systems in fighting cancer. Carbohydr Polym. 2021;269:118323. doi:10.1016/j.carbpol.2021.118323

194. Ma J, Zhong L, Peng X, Xu Y, Sun R. Functional chitosan-based materials for biological applications. Curr Med Chem. 2020;27(28):4660–4672. doi:10.2174/0929867327666200420091312

195. Shariatinia Z. Carboxymethyl chitosan: properties and biomedical applications. Int J Biol Macromol. 2018;120(Pt B):1406–1419. doi:10.1016/j.ijbiomac.2018.09.131

196. Mushtaq A, Li L, Grøndahl L. Chitosan nanomedicine in cancer therapy: targeted delivery and cellular uptake. Macromol Biosci. 2021;21(5):e2100005. doi:10.1002/mabi.202100005

197. Han J, Hou ZQ, Wang YG, Guo X. Synthesis and evaluation of hydroxycamptothecin-encapsulated chitosan nanospheres for the treatment of liver cancer. Technol Cancer Res Treat. 2015;14(1):111–117. doi:10.7785/tcrt.2012.500404

198. Tian Y, Shi C, Zhang X, et al. Nanomicelle based peroral delivery system for enhanced absorption and sustained release of 10-hydrocamptothecin. J Biomed Nanotechnol. 2015;11(2):262–273. doi:10.1166/jbn.2015.1909

199. Wang Y, Xuan J, Zhao G, Wang D, Ying N, Zhuang J. Improving stability and oral bioavailability of hydroxycamptothecin via nanocrystals in microparticles (NCs/MPs) technology. Int J Pharm. 2021;604:120729. doi:10.1016/j.ijpharm.2021.120729

200. Guo H, Li F, Qiu H, et al. Preparation and characterization of chitosan nanoparticles for chemotherapy of melanoma through enhancing tumor penetration. Front Pharmacol. 2020;11:317. doi:10.3389/fphar.2020.00317

201. Wu S, Yang X, Zou M, Hou Z, Yan J. A new method without organic solvent to targeted nanodrug for enhanced anticancer efficacy. Nanoscale Res Lett. 2017;12(1):416. doi:10.1186/s11671-017-2174-x

202. Zhu H, Cao J, Cui S, Qian Z, Gu Y. Enhanced tumor targeting and antitumor efficacy via hydroxycamptothecin-encapsulated folate-modified N-succinyl-N’-octyl chitosan micelles. J Pharm Sci. 2013;102(4):1318–1332. doi:10.1002/jps.23470

203. Wu S, Yang X, Lu Y, et al. A green approach to dual-drug nanoformulations with targeting and synergistic effects for cancer therapy. Drug Deliv. 2017;24(1):51–60. doi:10.1080/10717544.2016.1228716

204. 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:10.1016/j.jconrel.2016.12.026

205. Niu X, Cao J, Zhang Y, et al. A glutathione responsive nitric oxide release system based on charge-reversal chitosan nanoparticles for enhancing synergistic effect against multidrug resistance tumor. Nanomedicine. 2019;20:102015. doi:10.1016/j.nano.2019.102015

206. Chaudhry GE, Akim A, Naveed Zafar M, Safdar N, Sung YY, Muhammad TST. Understanding hyaluronan receptor (CD44) interaction, HA-CD44 activated potential targets in cancer therapeutics. Adv Pharma Bull. 2021;11(3):426–438. doi:10.34172/apb.2021.050

207. Gao M, Deng H, Zhang W. Hyaluronan-based multifunctional nano-carriers for combination cancer therapy. Curr Top Med Chem. 2021;21(2):126–139. doi:10.2174/1568026620666200922113846

208. Chen M, Zhang W, Yuan K, et al. Preclinical evaluation and monitoring of the therapeutic response of a dual targeted hyaluronic acid nanodrug. Contrast Media Mol Imaging. 2017;2017:4972701. doi:10.1155/2017/4972701

209. Li Y, Zhang H, Chen Y, et al. Integration of phospholipid-hyaluronic acid-methotrexate nanocarrier assembly and amphiphilic drug-drug conjugate for synergistic targeted delivery and combinational tumor therapy. Biomater Sci. 2018;6(7):1818–1833. doi:10.1039/C8BM00009C

210. Odeniyi MA, Omoteso OA, Adepoju AO, Jaiyeoba KT. Starch nanoparticles in drug delivery: a review. Polim Med. 2018;48(1):41–45. doi:10.17219/pim/99993

211. Wang J, Cui S, Bao Y, Xing J, Hao W. Tocopheryl pullulan-based self assembling nanomicelles for anti-cancer drug delivery. Mater Sci Eng C Mater Biol Appl. 2014;43:614–621. doi:10.1016/j.msec.2014.07.066

212. Li G, Zhao M, Zhao L. Well-defined hydroxyethyl starch-10-hydroxy camptothecin super macromolecule conjugate: cytotoxicity, pharmacodynamics research, tissue distribution test and intravenous injection safety assessment. Drug Deliv. 2016;23(8):2860–2868. doi:10.3109/10717544.2015.1110844

213. Li G, Li Y, Tang Y, et al. Hydroxyethyl starch conjugates for improving the stability, pharmacokinetic behavior and antitumor activity of 10-hydroxy camptothecin. Int J Pharm. 2014;471(1–2):234–244. doi:10.1016/j.ijpharm.2014.05.038

214. Li G, Zhao M, Zhao L. Lysine-mediated hydroxyethyl starch-10-hydroxy camptothecin micelles for the treatment of liver cancer. Drug Deliv. 2020;27(1):519–529. doi:10.1080/10717544.2020.1745329

215. Thakur BR, Singh RK, Handa AK, Rao MA. Chemistry and uses of pectin–a review. Crit Rev Food Sci Nutr. 1997;37(1):47–73. doi:10.1080/10408399709527767

216. Li DQ, Li J, Dong HL, et al. Pectin in biomedical and drug delivery applications: a review. Int J Biol Macromol. 2021;185:49–65. doi:10.1016/j.ijbiomac.2021.06.088

217. Liu Y, Kong T, Yang Z, Zhang Y, Lei J, Zhao P. Self-assembled folic acid-targeted pectin-multi-arm polyethylene glycol nanoparticles for tumor intracellular chemotherapy. ACS Omega. 2021;6(2):1223–1234. doi:10.1021/acsomega.0c04350

218. Liu Y, Liu K, Li X, et al. A novel self-assembled nanoparticle platform based on pectin-eight-arm polyethylene glycol-drug conjugates for co-delivery of anticancer drugs. Mater Sci Eng C Mater Biol Appl. 2018;86:28–41. doi:10.1016/j.msec.2017.12.018

219. Liu Y, Zheng D, Ma Y, et al. Self-assembled nanoparticles platform based on pectin-dihydroartemisinin conjugates for codelivery of anticancer drugs. ACS Biomater Sci Eng. 2018;4(5):1641–1650. doi:10.1021/acsbiomaterials.8b00920

220. Dai L, Si C. Recent advances on cellulose-based nano-drug delivery systems: design of prodrugs and nanoparticles. Curr Med Chem. 2019;26(14):2410–2429. doi:10.2174/0929867324666170711131353

221. Dai L, Liu KF, Si CL, He J, Lei JD, Guo LQ. A novel self-assembled targeted nanoparticle platform based on carboxymethylcellulose co-delivery of anticancer drugs. J Mater Chem B. 2015;3(32):6605–6617. doi:10.1039/C5TB00900F

222. Liu KF, Liu YX, Li CX, Wang LY, Liu J, Lei JD. Self-assembled ph and redox dual responsive carboxymethylcellulose-based polymeric nanoparticles for efficient anticancer drug codelivery. ACS Biomater Sci Eng. 2018;4(12):4200–4207.

223. Sohretoglu D, Huang S. Ganoderma lucidum polysaccharides as an anti-cancer agent. Anticancer Agents Med Chem. 2018;18(5):667–674. doi:10.2174/1871520617666171113121246

224. Zheng D, Zhao J, Li Y, et al. Self-assembled ph-sensitive nanoparticles based on ganoderma lucidum polysaccharide-methotrexate conjugates for the co-delivery of anti-tumor drugs. ACS Biomater Sci Eng. 2021;7(8):3764–3773. doi:10.1021/acsbiomaterials.1c00663

225. Chen F, Huang G, Huang H. Preparation and application of dextran and its derivatives as carriers. Int J Biol Macromol. 2020;145:827–834. doi:10.1016/j.ijbiomac.2019.11.151

226. Zhao C, Cao W, Zheng H, et al. Acid-responsive nanoparticles as a novel oxidative stress-inducing anticancer therapeutic agent for colon cancer. Int J Nanomedicine. 2019;14:1597–1618. doi:10.2147/IJN.S189923

227. Cartaxo da Costa Urtiga S, Rodrigues Marcelino H, Sócrates Tabosa Do Egito E, Eleamen Oliveira E. Xylan in drug delivery: a review of its engineered structures and biomedical applications. Eur J Pharmaceut Biopharma. 2020;151:199–208. doi:10.1016/j.ejpb.2020.04.016

228. Zhang Q, Su C, Lu Z, et al. Preparation, physicochemical and pharmacological study of 10-hydroxycamptothecin solid dispersion with complexation agent - xylan-nonanoic acid amphiphilic conjugates. Int J Biol Macromol. 2022;204:224–233. doi:10.1016/j.ijbiomac.2022.01.104

229. Luk BT, Zhang L. Cell membrane-camouflaged nanoparticles for drug delivery. J Control Release. 2015;220(Pt B):600–607. doi:10.1016/j.jconrel.2015.07.019

230. Oroojalian F, Beygi M, Baradaran B, Mokhtarzadeh A, Shahbazi MA. Immune cell membrane-coated biomimetic nanoparticles for targeted cancer therapy. Small. 2021;17(12):e2006484. doi:10.1002/smll.202006484

231. Fan Y, Hao W, Cui Y, et al. Cancer cell membrane-coated nanosuspensions for enhanced chemotherapeutic treatment of glioma. Molecules. 2021;26(16):5103. doi:10.3390/molecules26165103

232. Ye S, Wang F, Fan Z, et al. Light/pH-triggered biomimetic red blood cell membranes camouflaged small molecular drug assemblies for imaging-guided combinational chemo-photothermal therapy. ACS Appl Mater Interfaces. 2019;11(17):15262–15275. doi:10.1021/acsami.9b00897

233. Zhang L, Zhang X, Lu G, et al. Cell membrane camouflaged hydrophobic drug nanoflake sandwiched with photosensitizer for orchestration of chemo-photothermal combination therapy. Small. 2019;15(20):e1805544. doi:10.1002/smll.201805544

234. Arumugam MK, Paal MC, Donohue TM, Ganesan M, Osna NA, Kharbanda KK. Beneficial effects of betaine: a comprehensive review. Biology. 2021;10(6):456. doi:10.3390/biology10060456

235. Li G, Sun B, Zheng S, et al. Zwitterion-driven shape program of prodrug nanoassemblies with high stability, High tumor accumulation, and high antitumor activity. Adv Healthc Mater. 2021;10(23):e2101407. doi:10.1002/adhm.202101407

236. Sun Y, Gao F, Yang C, et al. Construction of bispecific aptamer-drug conjugate by a hybrid chemical and biological approach. Bioconjug Chem. 2020;31(5):1289–1294. doi:10.1021/acs.bioconjchem.0c00071

237. Luo X, Jia G, Song H, Liu C, Wu G, Li X. Promoting antitumor activities of hydroxycamptothecin by encapsulation into acid-labile nanoparticles using electrospraying. Pharm Res. 2014;31(1):46–59. doi:10.1007/s11095-013-1130-4

238. Chen D, Huang Y, Xu S, et al. Self-assembled polyprodrug amphiphile for subcutaneous xenograft tumor inhibition with prolonged acting time in vivo. Macromol Biosci. 2017;17(11):1700174. doi:10.1002/mabi.201700174

239. Sun P, Chen D, Deng H, et al. “Bottom-up” construction of multi-polyprodrug-arm hyperbranched amphiphiles for cancer therapy. Bioconjug Chem. 2017;28(5):1470–1480. doi:10.1021/acs.bioconjchem.7b00146

240. Naz S, Shamoon M, Wang R, Zhang L, Zhou J, Chen J. Advances in therapeutic implications of inorganic drug delivery nano-platforms for cancer. Int J Mol Sci. 2019;20(4):965. doi:10.3390/ijms20040965

241. Li Z, Zhang Y, Feng N. Mesoporous silica nanoparticles: synthesis, classification, drug loading, pharmacokinetics, biocompatibility, and application in drug delivery. Expert Opin Drug Deliv. 2019;16(3):219–237. doi:10.1080/17425247.2019.1575806

242. Fan L, Zhang Y, Wang F, et al. Multifunctional all-in-one drug delivery systems for tumor targeting and sequential release of three different anti-tumor drugs. Biomaterials. 2016;76:399–407. doi:10.1016/j.biomaterials.2015.10.069

243. Contreras-Baeza Y, Sandoval PY, Alarcón R, et al. Monocarboxylate transporter 4 (MCT4) is a high affinity transporter capable of exporting lactate in high-lactate microenvironments. J Biol Chem. 2019;294(52):20135–20147. doi:10.1074/jbc.RA119.009093

244. Li K, Lin C, He Y, et al. Engineering of cascade-responsive nanoplatform to inhibit lactate efflux for enhanced tumor chemo-immunotherapy. ACS Nano. 2020;14(10):14164–14180. doi:10.1021/acsnano.0c07071

245. Vangijzegem T, Stanicki D, Laurent S. Magnetic iron oxide nanoparticles for drug delivery: applications and characteristics. Expert Opin Drug Deliv. 2019;16(1):69–78. doi:10.1080/17425247.2019.1554647

246. Wu K, Su D, Liu J, Saha R, Wang JP. Magnetic nanoparticles in nanomedicine: a review of recent advances. Nanotechnology. 2019;30(50):502003. doi:10.1088/1361-6528/ab4241

247. Liu J, Xu F, Huang J, et al. Low-intensity focused ultrasound (LIFU)-activated nanodroplets as a theranostic agent for noninvasive cancer molecular imaging and drug delivery. Biomater Sci. 2018;6(11):2838–2849. doi:10.1039/C8BM00726H

248. Cheng TM, Chang WJ, Chu HY, et al. Nano-strategies targeting the integrin αvβ3 network for cancer therapy. Cells. 2021;10(7):1684. doi:10.3390/cells10071684

249. Ding GB, Wang Y, Guo Y, Xu L. Integrin α(V)β(3)-targeted magnetic nanohybrids with enhanced antitumor efficacy, cell cycle arrest ability, and encouraging anti-cell-migration activity. ACS Appl Mater Interfaces. 2014;6(19):16643–16652. doi:10.1021/am503359g

250. Ding GB, Liu HY, Lv YY, et al. Enhanced in vitro antitumor efficacy and strong anti-cell-migration activity of a hydroxycamptothecin-encapsulated magnetic nanovehicle. Chemistry. 2012;18(44):14037–14046. doi:10.1002/chem.201200765

251. Zhu HM, Gu JH, Xie Y, Xie B, Ling JJ. Hydroxycamptothecin liposomes based on thermal and magnetic dual-responsive system: preparation, in vitro and in vivo antitumor activity, microdialysis-based tumor pharmacokinetics. J Drug Target. 2018;26(4):345–356. doi:10.1080/1061186X.2017.1380654

252. Yang Y, Zhang X, Yu C, et al. Smart nanorods for highly effective cancer theranostic applications. Adv Healthc Mater. 2014;3(6):906–915. doi:10.1002/adhm.201300463

253. Wang G, Gao S, Tian R, et al. Theranostic hyaluronic acid-iron micellar nanoparticles for magnetic-field-enhanced in vivo cancer chemotherapy. ChemMedChem. 2018;13(1):78–86. doi:10.1002/cmdc.201700515

254. Qu JB, Shao HH, Jing GL, Huang F. PEG-chitosan-coated iron oxide nanoparticles with high saturated magnetization as carriers of 10-hydroxycamptothecin: preparation, characterization and cytotoxicity studies. Colloids Surf B Biointerfaces. 2013;102:37–44. doi:10.1016/j.colsurfb.2012.08.004

255. Naikoo G, Al-Mashali F, Arshad F, et al. An overview of copper nanoparticles: synthesis, characterisation and anticancer activity. Curr Pharm Des. 2021;27(43):4416–4432. doi:10.2174/1381612827666210804100303

256. Luo Z, Fan S, Gu C, et al. Metal-organic framework (MOF)-based nanomaterials for biomedical applications. Curr Med Chem. 2019;26(18):3341–3369. doi:10.2174/0929867325666180214123500

257. Shi Y, Liu W, Wu X, Zhu J, Zhou D, Liu X. A water-soluble polyacid polymer based on hydrophilic metal-organic frameworks using amphoteric carboxylic acid ligands as linkers for hydroxycamptothecin loading and release in vitro. Nanomaterials. 2021;11(11):2854. doi:10.3390/nano11112854

258. Li W, Cao Z, Liu R, et al. AuNPs as an important inorganic nanoparticle applied in drug carrier systems. Artif Cells, Nanomed Biotechnol. 2019;47(1):4222–4233. doi:10.1080/21691401.2019.1687501

259. Sztandera K, Gorzkiewicz M, Klajnert-Maculewicz B. Gold nanoparticles in cancer treatment. Mol Pharm. 2019;16(1):1–23. doi:10.1021/acs.molpharmaceut.8b00810

260. Li W, Zhang X, Zhou M, et al. Functional core/shell drug nanoparticles for highly effective synergistic cancer therapy. Adv Healthc Mater. 2014;3(9):1475–1485. doi:10.1002/adhm.201300577

261. Bao H, Zhang Q, Xu H, Yan Z. Effects of nanoparticle size on antitumor activity of 10-hydroxycamptothecin-conjugated gold nanoparticles: in vitro and in vivo studies. Int J Nanomedicine. 2016;11:929–940. doi:10.2147/IJN.S96422

262. Wang X, Gao S, Qin Z, et al. Evans blue derivative-functionalized gold nanorods for photothermal therapy-enhanced tumor chemotherapy. ACS Appl Mater Interfaces. 2018;10(17):15140–15149. doi:10.1021/acsami.8b02195

263. Yao L, Xue X, Yu P, Ni Y, Chen F. Evans blue dye: a revisit of its applications in biomedicine. Contrast Media Mol Imaging. 2018;2018:7628037. doi:10.1155/2018/7628037

264. Wang H, Zhu W, Huang Y, Li Z, Jiang Y, Xie Q. Facile encapsulation of hydroxycamptothecin nanocrystals into zein-based nanocomplexes for active targeting in drug delivery and cell imaging. Acta Biomater. 2017;61:88–100. doi:10.1016/j.actbio.2017.04.017

265. Sattari S, Adeli M, Beyranvand S, Nemati M. Functionalized graphene platforms for anticancer drug delivery. Int J Nanomedicine. 2021;16:5955–5980. doi:10.2147/IJN.S249712

266. Liu K, Wang Y, Li H, Duan Y. A facile one-pot synthesis of starch functionalized graphene as nano-carrier for pH sensitive and starch-mediated drug delivery. Colloids Surf B Biointerfaces. 2015;128:86–93. doi:10.1016/j.colsurfb.2015.02.010

267. Huang X, Zhang J, Song Y, Zhang T, Wang B. Combating liver cancer through GO-targeted biomaterials. Biomed Mater. 2021;16(6):065003. doi:10.1088/1748-605X/ac1f72

268. Gao X, Wang Q, Cheng C, et al. The application of prussian blue nanoparticles in tumor diagnosis and treatment. Sensors. 2020;20(23):6905. doi:10.3390/s20236905

269. Jing L, Shao S, Wang Y, Yang Y, Yue X, Dai Z. Hyaluronic acid modified hollow prussian blue nanoparticles loading 10-hydroxycamptothecin for targeting thermochemotherapy of cancer. Theranostics. 2016;6(1):40–53. doi:10.7150/thno.13250

270. Fahmy SA, Preis E, Bakowsky U, Azzazy HME. Platinum nanoparticles: green synthesis and biomedical applications. Molecules. 2020;25(21):4981. doi:10.3390/molecules25214981

271. Larue L, Myrzakhmetov B, Ben-Mihoub A, et al. Fighting hypoxia to improve PDT. Pharmaceuticals. 2019;12(4):163. doi:10.3390/ph12040163

272. Fu X, Yin W, Shi D, et al. Shuttle-shape carrier-free platinum-coordinated nanoreactors with O(2) self-supply and ROS augment for enhanced phototherapy of hypoxic tumor. ACS Appl Mater Interfaces. 2021;13(28):32690–32702. doi:10.1021/acsami.1c06668

273. Maleki Dizaj S, Sharifi S, Ahmadian E, Eftekhari A, Adibkia K, Lotfipour F. An update on calcium carbonate nanoparticles as cancer drug/gene delivery system. Expert Opin Drug Deliv. 2019;16(4):331–345. doi:10.1080/17425247.2019.1587408

274. Qiu W, Liang M, Gao Y, et al. Polyamino acid calcified nanohybrids induce immunogenic cell death for augmented chemotherapy and chemo-photodynamic synergistic therapy. Theranostics. 2021;11(19):9652–9666. doi:10.7150/thno.64354

275. Zhang X, Li N, Zhang S, et al. Emerging carrier-free nanosystems based on molecular self-assembly of pure drugs for cancer therapy. Med Res Rev. 2020;40(5):1754–1775. doi:10.1002/med.21669

276. Huang L, Zhao S, Fang F, Xu T, Lan M, Zhang J. Advances and perspectives in carrier-free nanodrugs for cancer chemo-monotherapy and combination therapy. Biomaterials. 2021;268:120557. doi:10.1016/j.biomaterials.2020.120557

277. Karaosmanoglu S, Zhou M, Shi B, Zhang X, Williams GR, Chen X. Carrier-free nanodrugs for safe and effective cancer treatment. J Control Release. 2021;329:805–832. doi:10.1016/j.jconrel.2020.10.014

278. Wen Y, Zhang W, Gong N, et al. Carrier-free, self-assembled pure drug nanorods composed of 10-hydroxycamptothecin and chlorin e6 for combinatorial chemo-photodynamic antitumor therapy in vivo. Nanoscale. 2017;9(38):14347–14356. doi:10.1039/C7NR03129G

279. Zhao Y, Zhao Y, Ma Q, et al. Carrier-free, dual-functional nanorods via self-assembly of pure drug molecules for synergistic chemo-photodynamic therapy. Int J Nanomedicine. 2019;14:8665–8683. doi:10.2147/IJN.S224704

280. Li W, Yang Y, Wang C, et al. Carrier-free, functionalized drug nanoparticles for targeted drug delivery. Chem Commun. 2012;48(65):8120–8122. doi:10.1039/c2cc33214k

281. Zhou M, Zhang X, Yang Y, et al. Carrier-free functionalized multidrug nanorods for synergistic cancer therapy. Biomaterials. 2013;34(35):8960–8967. doi:10.1016/j.biomaterials.2013.07.080

282. Zhao Y, Chen F, Pan Y, et al. Nanodrug formed by coassembly of dual anticancer drugs to inhibit cancer cell drug resistance. ACS Appl Mater Interfaces. 2015;7(34):19295–19305. doi:10.1021/acsami.5b05347

283. Chen F, Zhao Y, Pan Y, et al. Synergistically enhanced therapeutic effect of a carrier-free HCPT/DOX nanodrug on breast cancer cells through improved cellular drug accumulation. Mol Pharm. 2015;12(7):2237–2244. doi:10.1021/mp500744m

284. Han M, Liu X, Guo Y, Wang Y, Wang X. Preparation, characterization, biodistribution and antitumor efficacy of hydroxycamptothecin nanosuspensions. Int J Pharm. 2013;455(1–2):85–92. doi:10.1016/j.ijpharm.2013.07.056

285. Yang X, Liu Y, Zhao Y, et al. A stabilizer-free and organic solvent-free method to prepare 10-hydroxycamptothecin nanocrystals: in vitro and in vivo evaluation. Int J Nanomedicine. 2016;11:2979–2994. doi:10.2147/IJN.S102726

286. Wang H, Feng J, Liu G, Chen B, Jiang Y, Xie Q. In vitro and in vivo anti-tumor efficacy of 10-hydroxycamptothecin polymorphic nanoparticle dispersions: shape- and polymorph-dependent cytotoxicity and delivery of 10-hydroxycamptothecin to cancer cells. Nanomedicine. 2016;12(4):881–891. doi:10.1016/j.nano.2015.12.373

287. Kong L, Campbell F, Kros A. DePEGylation strategies to increase cancer nanomedicine efficacy. Nanoscale Horizons. 2019;4(2):378–387. doi:10.1039/C8NH00417J

288. Yoo J, Won YY. Phenomenology of the initial burst release of drugs from PLGA microparticles. ACS Biomater Sci Eng. 2020;6(11):6053–6062. doi:10.1021/acsbiomaterials.0c01228

289. Singhvi MS, Zinjarde SS, Gokhale DV. Polylactic acid: synthesis and biomedical applications. J Appl Microbiol. 2019;127(6):1612–1626. doi:10.1111/jam.14290

290. Wang M, Zhang L, Cai Y, et al. Bioengineered human serum albumin fusion protein as target/enzyme/ph three-stage propulsive drug vehicle for tumor therapy. ACS Nano. 2020;14:17405–17418. doi:10.1021/acsnano.0c07610

291. Bukhari SZ, Zeth K, Iftikhar M, et al. Supramolecular lipid nanoparticles as delivery carriers for non-invasive cancer theranostics. Curr Res Pharmacol Drug Discov. 2021;2:100067. doi:10.1016/j.crphar.2021.100067

292. Liu L, Han L, Wu Q, et al. Multifunctional DNA dendrimer nanostructures for biomedical applications. J Mater Chem B. 2021;9(25):4991–5007. doi:10.1039/D1TB00689D

293. Mura P. Advantages of the combined use of cyclodextrins and nanocarriers in drug delivery: a review. Int J Pharm. 2020;579:119181. doi:10.1016/j.ijpharm.2020.119181

294. Mauri E, Giannitelli SM, Trombetta M, Rainer A. Synthesis of nanogels: current trends and future outlook. Gels. 2021;7(2):36. doi:10.3390/gels7020036

295. Bhardwaj N, Kundu SC. Electrospinning: a fascinating fiber fabrication technique. Biotechnol Adv. 2010;28(3):325–347. doi:10.1016/j.biotechadv.2010.01.004

296. Berillo D, Yeskendir A, Zharkinbekov Z, Raziyeva K, Saparov A. Peptide-based drug delivery systems. Medicina. 2021;57(11):1209. doi:10.3390/medicina57111209

297. Plucinski A, Lyu Z, Schmidt B. Polysaccharide nanoparticles: from fabrication to applications. J Mater Chem B. 2021;9(35):7030–7062. doi:10.1039/d1tb00628b

298. Sun L, Xiong Z, Shen F, Wang Z, Liu Z. Biological membrane derived nanomedicines for cancer therapy. Sci China Chem. 2021;64(5):719–733. doi:10.1007/s11426-020-9943-9

299. Sharma S, Verma A, Teja BV, et al. An insight into functionalized calcium based inorganic nanomaterials in biomedicine: trends and transitions. Colloids Surf B Biointerfaces. 2015;133:120–139. doi:10.1016/j.colsurfb.2015.05.014

300. Ding X, Li D, Jiang J. Gold-based inorganic nanohybrids for nanomedicine applications. Theranostics. 2020;10(18):8061–8079. doi:10.7150/thno.42284

301. Farzin A, Etesami SA, Quint J, Memic A, Tamayol A. Magnetic nanoparticles in cancer therapy and diagnosis. Adv Healthc Mater. 2020;9(9):e1901058. doi:10.1002/adhm.201901058

302. Rhazouani A, Gamrani H, El Achaby M, et al. Synthesis and toxicity of graphene oxide nanoparticles: a literature review of in vitro and in vivo studies. Biomed Res Int. 2021;2021:5518999. doi:10.1155/2021/5518999

303. Ingle AP, Duran N, Rai M. Bioactivity, mechanism of action, and cytotoxicity of copper-based nanoparticles: a review. Appl Microbiol Biotechnol. 2014;98(3):1001–1009. doi:10.1007/s00253-013-5422-8

304. Busquets MA, Estelrich J. Prussian blue nanoparticles: synthesis, surface modification, and biomedical applications. Drug Discov Today. 2020;25(8):1431–1443. doi:10.1016/j.drudis.2020.05.014

305. Qin Z, Li Y, Gu N. Progress in applications of prussian blue nanoparticles in biomedicine. Adv Healthc Mater. 2018;7(20):e1800347. doi:10.1002/adhm.201800347

306. Das RP, Gandhi VV, Singh BG, Kunwar A. Passive and active drug targeting: role of nanocarriers in rational design of anticancer formulations. Curr Pharm Des. 2019;25(28):3034–3056. doi:10.2174/1381612825666190830155319

307. Ikeda-Imafuku M, Wang LL, Rodrigues D, Shaha S, Zhao Z, Mitragotri S. Strategies to improve the EPR effect: a mechanistic perspective and clinical translation. J Control Release. 2022;345:512–536. doi:10.1016/j.jconrel.2022.03.043

308. de Lázaro I, Mooney DJ. A nanoparticle’s pathway into tumours. Nat Mater. 2020;19(5):486–487. doi:10.1038/s41563-020-0669-9

309. Danhier F. To exploit the tumor microenvironment: since the EPR effect fails in the clinic, what is the future of nanomedicine? J Control Release. 2016;244(Pt A):108–121. doi:10.1016/j.jconrel.2016.11.015

310. Sharifi M, Cho WC, Ansariesfahani A, et al. An updated review on EPR-based solid tumor targeting nanocarriers for cancer treatment. Cancers. 2022;14(12):2868. doi:10.3390/cancers14122868

311. Challenging paradigms in tumour drug delivery. Nat Mater. 2020;19(5):477. doi:10.1038/s41563-020-0676-x

312. Fang J, Islam W, Maeda H. Exploiting the dynamics of the EPR effect and strategies to improve the therapeutic effects of nanomedicines by using EPR effect enhancers. Adv Drug Deliv Rev. 2020;157:142–160. doi:10.1016/j.addr.2020.06.005

313. Subhan MA, Yalamarty SSK, Filipczak N, Parveen F, Torchilin VP. Recent advances in tumor targeting via EPR effect for cancer treatment. J Personal Med. 2021;11(6):571. doi:10.3390/jpm11060571

314. 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:10.1016/j.addr.2015.01.002

315. Zhou Q, Dong C, Fan W, et al. Tumor extravasation and infiltration as barriers of nanomedicine for high efficacy: the current status and transcytosis strategy. Biomaterials. 2020;240:119902. doi:10.1016/j.biomaterials.2020.119902

316. Liu Y, Huo Y, Yao L, et al. Transcytosis of nanomedicine for tumor penetration. Nano Lett. 2019;19(11):8010–8020. doi:10.1021/acs.nanolett.9b03211

317. Liu X, Jiang J, Nel AE, Meng H. Major effect of transcytosis on nano drug delivery to pancreatic cancer. Mol Cell Oncol. 2017;4(4):e1335273. doi:10.1080/23723556.2017.1335273

318. Liu X, Jiang J, Meng H. Transcytosis - An effective targeting strategy that is complementary to “EPR effect” for pancreatic cancer nano drug delivery. Theranostics. 2019;9(26):8018–8025. doi:10.7150/thno.38587

319. Pandit S, Dutta D, Nie S. Active transcytosis and new opportunities for cancer nanomedicine. Nat Mater. 2020;19(5):478–480. doi:10.1038/s41563-020-0672-1

320. Bockamp E, Rosigkeit S, Siegl D, Schuppan D. Nano-enhanced cancer immunotherapy: immunology encounters nanotechnology. Cells. 2020;9(9):2102. doi:10.3390/cells9092102

321. Martin JD, Cabral H, Stylianopoulos T, Jain RK. Improving cancer immunotherapy using nanomedicines: progress, opportunities and challenges. Nat Rev Clin Oncol. 2020;17(4):251–266. doi:10.1038/s41571-019-0308-z

322. Ouyang B, Poon W, Zhang YN, et al. The dose threshold for nanoparticle tumour delivery. Nat Mater. 2020;19(12):1362–1371. doi:10.1038/s41563-020-0755-z

Creative Commons License © 2022 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.