Open Access Article
Xiao
Dong
*a,
Rajeev K.
Brahma
b,
Chao
Fang
c and
Shao Q.
Yao
*b
aDepartment of Pharmacy, School of Medicine, Shanghai University, Shanghai 200444, China. E-mail: dong-xiao@shu.edu.cn
bDepartment of Chemistry, National University of Singapore, Singapore 117543, Singapore. E-mail: chmyaosq@nus.edu.sg
cState Key Laboratory of Oncogenes and Related Genes, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
First published on 18th March 2022
Small-molecule prodrugs have become the main toolbox to improve the unfavorable physicochemical properties of potential therapeutic compounds in contemporary anti-cancer drug development. Many approved small-molecule prodrugs, however, still face key challenges in their pharmacokinetic (PK) and pharmacodynamic (PD) properties, thus severely restricting their further clinical applications. Self-assembled prodrugs thus emerged as they could take advantage of key benefits in both prodrug design and nanomedicine, so as to maximize drug loading, reduce premature leakage, and improve PK/PD parameters and targeting ability. Notably, temporally and spatially controlled release of drugs at cancerous sites could be achieved by encoding various activable linkers that are sensitive to chemical or biological stimuli in the tumor microenvironment (TME). In this review, we have comprehensively summarized the recent progress made in the development of single/multiple-stimulus-responsive self-assembled prodrugs for mono- and combinatorial therapy. A special focus was placed on various prodrug conjugation strategies (polymer–drug conjugates, drug–drug conjugates, etc.) that facilitated the engineering of self-assembled prodrugs, and various linker chemistries that enabled selective controlled release of active drugs at tumor sites. Furthermore, some polymeric nano-prodrugs that entered clinical trials have also been elaborated here. Finally, we have discussed the bottlenecks in the field of prodrug nanoassembly and offered potential solutions to overcome them. We believe that this review will provide a comprehensive reference for the rational design of effective prodrug nanoassemblies that have clinic translation potential.
Various strategies, including the use of prodrugs and physical encapsulation of drugs within nanostructures, have been exploited to address the aforementioned challenges associated with conventional small-molecule drugs (Fig. 1A and B).12–15 Following its introduction in 1958 by Albert, the concept of prodrugs remained the dominant approach to ameliorate the limitations in the ADMET properties of a parent drug without compromising its pharmacological activities (Fig. 1A).16–18 Between 2008 and 2017, prodrugs accounted for approximately 12% of all approved small-molecule drugs on the market.19 Despite the great progress that has been made in small-molecule prodrugs, they still suffer from challenges mainly associated with inherent small-molecule characteristics, including hepatic metabolism, short blood circulation time, fast renal clearance, premature activation, etc. Thus, the development of more innovative strategies is urgently needed.
Physical encapsulation of small-molecule (pro)-drugs within various organic nanocarriers (e.g. liposomes, micelles, polymers, nanogels, and dendrimers) or inorganic nanocarriers (e.g. silica nanocapsules) has recently emerged as a highly viable strategy to improve the ADMET properties of small-molecule (pro)-drugs in tumor therapy (Fig. 1B).20–24 Such nanocarriers effectively shield the small-molecule (pro)-drugs, resulting in an improvement in their stability, solubility, bioavailability and biodistribution.25 Notably, nanometer-sized vehicles can cause small-molecule (pro)-drugs to selectively accumulate near tumor tissues through the well-known effect of enhanced permeability and retention (EPR), which works by passively targeting the leaky tumor vascular system.26–28 These nanocarriers can additionally achieve active tumor targeting by surface modifications of the nanoparticle (NP) with suitable cancer-targeting ligands.29–31 Despite the many obvious benefits of these drug-loaded nanosystems, there still exist significant obstacles hindering their widespread clinical application, including low drug-loading capacity (<10% in most cases), premature drug release during blood circulation, bursting or poor drug release at target sites and carrier material-related biosafety issues.32–34
Fortunately with new advances in materials science and conjugation chemistry, self-assembled nanostructures of (poly)-prodrug amphiphiles or amphiphilized homo/heterodimers have emerged as a new paradigm for tumor-specific drug delivery.35–37 In recent years, various versatile stimulus-responsive prodrug nanoassemblies have been successfully developed for chemotherapy, phototherapy, immunotherapy, theranostics, and combinatorial therapy (Table 1). This review starts by introducing the concept of prodrugs, the design of prodrug nanoassemblies and commonly used methods in nano-prodrug preparation. Next, we summarize the rational design of self-assembled prodrugs on the basis of different conjugation approaches (i.e. polymer–drug conjugates, drug–drug conjugates, etc.) for tumor treatments. We emphasize the various approaches for controlled release of drugs based on innovative linker designs that exploit various tumor-associated stimuli (pH, ROS, GSH, enzyme, hypoxia, etc.; see Table 2). Finally, we discuss the various emerging challenges and future prospects of self-assembled prodrugs in relation to cancer therapy. Metal-catalysed nano-prodrug and prodrug-based hydrogels in tumor therapy, which have received increasing attention in recent years, are beyond the scope of this review. We expect that the inimitable and clear logical framework in this review will provide a comprehensive reference for readers, especially novice readers.
| Drug | Linkage | Therapy | Ref. |
|---|---|---|---|
| DOX | Hydrazone | Chemotherapy | 66, 68, 137, 138 and 148 |
| Chemotherapy & immunotherapy | 67 | ||
| Imine bond | Chemotherapy | 70 and 71 | |
| Enamine | Chemotherapy | 72 | |
| Dihydrazide | Chemotherapy | 82 | |
| Carbamate | Chemotherapy | 83 | |
| Anhydride | Chemotherapy | 84 | |
| Ketal | Chemotherapy & photodynamic therapy | 95 | |
| Disulfide | Chemotherapy | 109 | |
| Thioether/disulfide/trisulfide | Chemotherapy | 115 | |
| Peptide | Chemotherapy, photodynamic therapy & immunotherapy | 126 | |
| Epacadostat | Peptide | Photothermo-immunotherapy | 125 |
| Epothilone B | Ketal | Chemotherapy | 93 |
| Paclitaxel | Ketal | Chemotherapy | 74 |
| Thioether | Chemotherapy & photodynamic therapy | 91 and 99 | |
| Chemotherapy & oxidation therapy | 92 and 100 | ||
| Disulfide | Chemotherapy & photodynamic therapy | 113 and 114 | |
| Azobenzene | Chemotherapy & photodynamic therapy | 132 | |
| Boronate ester | Chemotherapy & photodynamic therapy | 97 | |
| Aminoacrylate | Chemotherapy & photodynamic therapy | 99 | |
| Peroxalate ester | Chemotherapy, photodynamic therapy & photothermal therapy | 100 | |
| Peptide | Chemotherapy & photodynamic therapy | 124 | |
| Dithiomaleimide (DTM) | Chemotherapy & gene therapy | 139 | |
| Thioether/Disulfide/Selenoether/Diselenide | Chemotherapy | 113 | |
| Selenoether/Te/Thioether | Chemotherapy | 140 | |
| Gemcitabine | Ketal | Chemotherapy | 75 |
| Acetal | Chemotherapy | 76 | |
| Indomethacin | Ortho-ester | Chemotherapy | 85 |
| Capecitabine | Boronate esters | Theranostics | 93 |
| Bortezomib | Boronate esters | Chemotherapy | 79 and 81 |
| Boronic acid–salicylhydroxamate | Chemotherapy | 80 | |
| Camptothecin | Thioketal | Chemotherapy & photodynamic therapy | 90 and 103 |
| Chemotherapy & oxidation therapy | 94 | ||
| Disulfide | Chemotherapy | 110 and 135 | |
| Chemotherapy & photothermal therapy | 111 | ||
| Aspirin | p-Boronabenzyl ester | Immunotherapy | 96 |
| Gefitinib | Disulfide | Chemotherapy | 116 |
| Pt | Polyphenol–metal coordination | Chemotherapy | 136 |
| Cabazitaxel | Thioether | Chemotherapy & photodynamic therapy | 101 |
| Imidazoquinoline | Ester bond | Immunotherapy | 122 |
| IPM-Br | 2-Nitroimidazole | Chemotherapy & photodynamic therapy | 131 |
| Stimulus | Linkage | Chemical structure | Ref. |
|---|---|---|---|
| pH | Hydrazone |
|
66–68, 137, 138 and 148 |
| Imine |
|
70 and 71 | |
| Enamine |
|
72 | |
| Ketal |
|
74 and 75 | |
| Acetal |
|
76 | |
| Boronate esters |
|
79 and 81 | |
| Boronic acid–salicylhydroxamate |
|
80 | |
| Dihydrazide |
|
82 | |
| Carbamate |
|
83 | |
| Anhydride |
|
84 | |
| Ortho-ester |
|
85 | |
| ROS | Thioketal |
|
90, 93–95 and 103 |
| Boronate ester |
|
96–98 | |
| Aminoacrylate |
|
99 | |
| Peroxalate ester |
|
100 | |
| GSH | Disulfide |
|
109–111, 113–116, 134 and 135 |
| Diselenide |
|
113 | |
| Selenoether |
|
113 and 140 | |
| Trisulfide |
|
115 | |
| GSH/ROS | Thioether |
|
91, 92, 101, 102, 113, 115 and 140 |
| Te |
|
140 | |
| Enzyme | Ester bond |
|
122 |
| Peptide | GL2/GFLG/PVGLLG/FRRG | 123–126 | |
| L-γ-glutamylglycine |
|
134 | |
| Hypoxia | 2-Nitroimidazole |
|
131 |
| Azobenzene |
|
132 | |
| pH/ROS | Polyphenol–metal coordination |
|
136 |
Significantly, there are several prerequisites for a successful prodrug design: (1) parent drugs should have functional groups that are suitable for chemical modifications; (2) favorable pro-moieties that can improve the deficient ADMET properties of the parent drug; (3) a reversible linkage between the parent drug and the pro-moiety that allows stimuli-responsive controlled-release of drugs without formation of cytotoxic byproducts.43 Various functional groups in a parent drug, including carboxyl, hydroxyl, amine and carbonyl, can be reversibly transformed into the corresponding ester, carbonate, carbamate, amide, and oxime, respectively. The pro-moiety generally dictates the chemical identity and can improve the ADMET properties of the parent drug.44,45 For example, lipidation is an effective approach to enhance the membrane permeability of a hydrophilic drug.46 Conversely, the aqueous solubility of a hydrophobic drug may be improved by introducing an ionizable pro-moiety.47
Due to the high content of polymers used, (poly)-prodrug amphiphile-based nanoassemblies still possess a low drug loading capacity. Although most of the currently FDA-approved nanomedicines use inert biomaterials as drug carriers, they still elicit significant adverse effects. Therefore, the development of carrier-free nano-prodrugs through the self-assembly of prodrugs themselves has become necessary. One example is shown in Fig. 1C, in which homo- or heterodimeric drug–drug conjugates were synthesized by conjugating two identical or different drug molecules via a biodegradable linker, respectively.53 The resultant drug–drug conjugate-based nanoassemblies possessed a high drug loading capacity capable of minimizing excipient-triggered adverse toxicity. Therefore, by taking advantage of the benefits in both prodrug design strategies and nanomedicine, these prodrug nanoassemblies could possess key properties including improved systemic stability, prolonged blood circulation, and passive/active targeting ability, as well as potential for application in theranostics and combination therapy. More importantly, by properly encoding suitable linker chemistries, the resulting self-assembled prodrugs could be tailored for tumor-specific temporal/spatial controlled-release of drugs in response to chemical or biological stimuli present at the tumor sites.54,55
![]() | ||
| Fig. 3 (A) Schematic illustration of the chemical structures of DOM@DOX and its self-assemblies. Reproduced from ref. 66 with permission from Royal Society of Chemistry, Copyright 2020. (B) Illustration of the fabrication of carrier-free, self-assembled prodrug (PEG@D:siRNA) to synergistically induce ICD and reverse tumor immunosuppression. Reproduced from ref. 67 with permission from Elsevier Ltd, Copyright 2020. (C) Synthesis of PEG-Schiff-DOX and the acid-sensitive intracellular drug release. Reproduced from ref. 68 with permission from Frontiers Media SA, Copyright 2021. (D) Schematic illustration of the preparation of DMMA-P-DOX and SA-P-DOX. Reproduced from ref. 70 with permission from Elsevier Ltd, Copyright 2020. (E) Synthesis of zwitterionic BCP-TPZ prodrug conjugates. Reproduced from ref. 71 with permission from American Chemical Society, Copyright 2021. (F) Chemical structure of acid-responsive DOX-ena-PPEG-ena-DOX and its drug activation mechanism. Reproduced from ref. 72 with permission from American Chemical Society, Copyright 2019. | ||
The imine bond is another promising linker for pH-triggered release of drugs due to its acid-labile properties.69 Recently, Guo et al. prepared a dual pH-responsive polymeric nano-prodrug (DMMA-P-DOX/LAP) with a charge-switchable ability for synergistic cancer therapy (Table 1).70 To synthesize 2,3-dimethylmaleic anhydride (DMMA)-P-DOX, as shown in Fig. 3D, DOX was first linked to the side chain of PEGylated ε-poly-L-lysine (EPLYS) via an imine bond to ensure a pH-triggered release of DOX (Table 2). SA-P-DOX was synthesized as a control. The CMC values of DMMA-P-DOX and SA-P-DOX were calculated to be 0.0069 mg mL−1 and 0.0057 mg mL−1, respectively. Such low CMC values enabled the corresponding nano-prodrugs to maintain an integral nanostructure during the blood circulation. The DMMA-P-DOX/LAP NPs were subsequently prepared by physical encapsulation of lapatinib (LAP), a dual tyrosine kinase inhibitor, within (DMMA-P-DOX)-based nanoassemblies. The hydrodynamic size of DMMA-P-DOX/LAP was determined to be 86.92 ± 1.82 nm, which was larger than those of DMMA-P-DOX (34.41 ± 2.97 nm) and SA-P-DOX (30.42 ± 4.11 nm). The positive charge of residual amino groups on the NP surface was shielded by modification of DMMA to further prolong its blood circulation time. Upon successful accumulation in the mildly acidic TME, the negative surface charge of the NP was reversed to positive, leading to an improved cell internalization and deep tumor penetration. The hydrolysis of imine bonds under low intracellular pH conditions could induce the disassembly of DMMA-P-DOX/LAP NPs, thus leading to the concurrent release of DOX and LAP in the cytosol for synergistic tumor therapy. Zhao et al. developed another zwitterionic block copolymer (BCP)-based micellar prodrug for pH-sensitive release of hypoxia-responsive prodrugs (Fig. 3E).71 The zwitterionic BCP-containing prodrug micelles were fabricated by self-assembly of PMPC-b-P(DEGMA-co-FPMA-g-TPZD) (BCP-TPZ), in which tirapazamine (TPZ) derivatives (named TPZD) were first conjugated onto PMPC-b-P(DEGMA-co-FPMA) through acid-labile imine bonds to facilitate pH-triggered drug release under acidic conditions (Table 2). By using pyrene as a fluorescence probe, the CMC values of BCP and BCP-TPZ were calculated to be 0.0608 and 0.0277 mg mL−1, respectively. The hydrodynamic size of BCP-TPZ micelles in PBS was determined to be 51–59 nm by DLS, which was larger than the size of BCP-only micelles (∼25 nm). The resulting micellar prodrugs maintained a good colloidal stability in PBS and DEME (with 10% FBS). In vitro TPZD release at different pH conditions of 7.4, 6.5 and 5.5 within 50 h was estimated to be 30%, 60% and 90%, respectively. As expected, the micellar BCP-TPZ exhibited at least 13.7-fold higher hypoxia-selective cytotoxicity against both HeLa and HepG2 cancer cells. In another study, Li and co-workers synthesized a pH-responsive polymeric prodrug (DOX-ena-PPEG-ena-DOX) that could self-assemble into homogeneous NPs with a good water solubility and biocompatibility (Fig. 3F).72 To obtain DOX-ena-PPEG-ena-DOX, the acid-sensitive alkynyl-terminated block polymer (A-P(PEG-alt-HMDA)-A) was synthesized by alternately connecting polyethylene glycol (PEG) with hexamethylenediamino (HMDA) via enamine bonds (-ena-). Subsequently, the corresponding polymer–prodrug conjugate (DOX-ena-PPEG-ena-DOX) was synthesized by attaching DOX to A-P(PEG-alt-HMDA)-A via amino–yne click chemistry. The resulting nano-prodrug had an average hydrodynamic size of 140 nm capable of causing ∼16.1% drug release at pH = 7.4 after 118 h of incubation. In contrast, approximately 85.0% of drug release was detected at pH = 5 (which mimicked the intracellular lysosome/endosome conditions), presumably caused by cleavage of the acid-labile enamine bond (Table 2).
Similarly, acetals or ketals are also promising linkers for the development of acid-responsive prodrugs.73 Mu and co-workers reported pH-sensitive polymeric paclitaxel (PTX) prodrugs (named PKPs) for ovarian cancer therapy (Table 1).74 As shown in Fig. 4A, in order to synthesize PKPs, the intermediate compound 2 containing acrylate groups was synthesized by linking PTX with compound 1via the acid-sensitive acetone-based acyclic ketal linker (Table 2). After that, compound 2 was subsequently conjugated with different lengths of poly(ethylene glycol) (PEG) to obtain PKPs, which formed into uniform micelles upon nanoprecipitation. The obtained micellar prodrug remained stable in the normal physiological environment but achieved accelerated drug release upon hydrolysis of the acid-labile bonds within the mildly acidic TME. Notably, the authors found that the molecular weight of PEG could affect the CMC values and the particle size of PKPs. The shorter PEGs could endow PKPs with favourable properties including smaller NP sizes, lower CMC values, prolonged circulation time, high accumulation at the tumor site and better antitumor activities. The particle sizes of various NPs (named PKP750, PKP1000, and PKP2000) determined by DLS were about 13, 22, and 48 nm, respectively. In addition, the author investigated the hydrolysis kinetics of PKPs at different pH conditions (7.4, 6.8 and 5.0). After 24 h incubation, release of PTX from PKP750, PKP1000, and PKP2000 NPs reached about 5.4%, 10.5%, and 11.4% at pH = 7.4, respectively (Fig. 4B). In contrast, higher degrees of PTX release were observed for the same PKPs (25.5%, 37.7% and 45.3%, respectively) at pH = 6.8. Finally, nearly complete release of PTX was observed for all three types of PKPs at pH = 5.0 after only 4 h incubation. In another study, Zhong et al. synthesized acid-activable gemcitabine (Gem)–polyketal conjugates (PKGems) by means of Michael addition polymerization of dithiol and diacryl-modified Gem.75 As shown in Fig. 4C, Gem was introduced onto a polyketal backbone via an acid-labile ketal linkage (Table 2), which facilitated the pH-triggered drug release under the acidic TME and intracellular endolysosomal conditions. The resulting hydrophobic PKGems were co-polymerized with mPEG-PDLLA to form a nano-prodrug (named GKNPs) that exhibited a good stability with minimal drug leakage at physiological pH = 7.4, but achieved a rapid drug release within the acidic TME. Consequently, these GKNPs showed a better antitumor effect than free gemcitabine in the ovarian tumor animal model implanted with A2780 cancer cells. Takemoto et al. reported a GEM-conjugated polymer (PGEM) for improved drug solubility and pharmacological activity of GEM.76 As shown in Fig. 4D, PGEM was synthesized by conjugating the native drug GEM onto the side chains of a poly(amino acid) through a cyclic acetal linkage. The obtained PGEM was able to self-assemble into a nano-prodrug that had a hydrodynamic size of 9.90 ± 0.97 nm in PBS. In comparison to Doxil (ca. 80 nm), the PGEM could achieve a deep homogeneous intratumor distribution in tumor tissues due to its smaller size. The PGEM system also showed a minimal drug leakage due to the stability of the 1,3-dioxane linkage under physiological conditions (pH = 7.4), thus causing negligible GEM-derived off-target effects. Upon entering into tumor cells via endocytosis, the encapsulated GEM was rapidly released due to hydrolysis of the 1,3-dioxane linkage in response to the acidic luminal pH in the endosomes/lysosomes. In addition, this PGEM system possessed a prolonged blood circulation time and exhibited a higher antitumor effect in two distinct pancreatic tumor models.
![]() | ||
| Fig. 4 (A) Illustration of the design and self-assembly of pH-responsive PEGylated paclitaxel prodrugs (PKPs). (B) PTX release from PKP750, PKP1000 and PKP2000 NPs after 24 h incubation at pH 7.4, 6.8 and 5, respectively. Reproduced from ref. 74 with permission from Elsevier Ltd, Copyright 2020. (C) Molecular structures of pH-responsive gemcitabine (Gem)–polyketal conjugates (PKGems) and nanoparticle formation by nanoprecipitation. Reproduced from ref. 75 with permission from American Chemical Society, Copyright 2020. (D) Chemical design of polymer–GEM conjugates and acid-responsive drug release mechanism under endolysosomal conditions. Reproduced from ref. 76 with permission from Elsevier Ltd, Copyright 2020. | ||
The chelation between boronic acid and other reactive functional groups (such as hydroxyl or amino groups) could be used as a self-immolative linker for traceless release of some boric acid- or amine-containing drugs/proteins.77,78 Recently, Hu et al. designed a pH-activable dendrimeric bortezomib (BTZ) prodrug to improve the stability of BTZ during blood circulation and minimize its system toxicity.79 To prepare this dendrimeric BTZ prodrug, an amphiphilic PEGylated dendrimer linked with dopamine was synthesized by conjugating azide-functionalized polyethylene glycol with an alkyne-functionalized dendrimer, followed by attachment of dopamine (Fig. 5A). The resulting BTZ drug could be subsequently introduced onto the PEGylated dendrimer by virtue of the high reactivity between boric anhydride on BTZ and dopamine, leading to formation of reversible drug conjugates (Fig. 5B and Table 2). At low pH conditions, the conjugation between dopamine and BTZ was weakened, thus resulting in selective release of BTZ in the acidic TME. The obtained amphiphilic PEGylated dendrimer loaded with BTZ was next formulated into NPs (named c(RGDyK)-NPs/BTZ) with additional filling agents including c(RGDyK)-PEG3400-DSPE and PEG-PLGA. Transmission electron microscopy (TEM) images of the resulting NPs showed a size distribution of ∼100 nm. The encapsulation efficiency of BTZ in the c(RGDyK)-NPs/BTZ was estimated to be above 95%. The obtained c(RGDyK)-NPs/BTZ was shown to possess an improved serum stability and reduced system toxicity than the free BTZ. Due to the existence of c(RGDyK), this nano-prodrug could significantly improve the tumor-targeting ability. When compared to the PBS or free BTZ-treated group, the tumor growth in both NPs/BTZ- and c(RGDyK)-NPs/BTZ-treated groups was significantly suppressed. Notably, cRGDyK modification could further improve the therapeutic efficacy of the NPs/BTZ. More importantly, this design can be readily applied for preparation of similar nano-drugs from well-known chemotherapeutics such as ixazomib and cisplatin. In a similar study, Liu et al. developed an efficient method for preparation of pH-sensitive polymer–drug conjugates (P1-BTZ) based on the ultrafast and reversible boronic acid–salicylhydroxamate click reaction (Fig. 5C).80 To prepare P1-BTZ, the salicylhydroxamate-bearing polymer (P1) composed of salicylhyroxymate and PEG-methacrylate monomers was first synthesized through an initial RAFT co-polymerization. Due to the rapid condensation between boronic acid and salicylhydroxamate, the resultant P1 could be used for pH-responsive delivery of drugs that have an intrinsic boronic acid group. In this study, the FDA-approved bortezomib (BTZ) was used as a model drug to test the utility of such strategies. The condensation between boronic acid and salicylhydroxamate subsequently induced in situ self-assembly of the polymer–drug conjugate. The P1-BTZ conjugate, but not P1 alone, was shown to self-assemble into uniform NPs with a hydrodynamic size of about 50 nm. As expected, pH-dependent release of bortezomib from this nanoassembly was observed due to the reversibility of the boronic acid–salicylhydroxamate linkage under mildly acidic conditions (Fig. 5C). Similarly, P1 could also be applicable for drugs that were pre-linked with arylboronic acid by using a self-immolative linker. In another study, Ma and co-workers developed a biomimetic peptide (BTZ-PEP-RGD)-based nano-prodrug for pH-triggered release of BTZ selectively at tumor sites (Table 2).81 To obtain BTZ-PEP-RGD, as shown in Fig. 5D, the dye-labeled biomimetic PEP-RGD (FITC-(DOPA)4-G5-RGDS) was synthesized by inserting a non-bioactive quintuple glycine (G5) spacer between the cancer-targeting ligand RGDS and fluorescein isothiocyanate (FITC)-linked tetrapeptide (DOPA)4 bearing catechol groups. The catechol groups were used for dynamic conjugation of BTZ through formation of pH-sensitive boronic acid–catechol esters. The obtained BTZ-PEP-RGD could self-assemble into homogeneous NPs with a small size (173 ± 34 nm) and high drug-loading content (37.4 wt%). The resulting nano-prodrug showed a fast drug release in acidic endosomes/lysosomes (pH 5.0–6.0) due to the rapid dissociation of the catechol–BA linkage, and a significantly higher antitumor activity when compared to free drugs.
![]() | ||
| Fig. 5 (A) Chemical structure of the dopamine-linked amphiphilic PEGylated dendrimer; (B) reaction between dopamine and boric acid on BTZ. Reproduced from ref. 79 with permission from Wiley-VCH, Copyright 2020. (C) Synthesis of the polymeric BTZ prodrug (P1-BTZ) and mechanism of pH-triggered drug release. Reproduced from ref. 80 with permission from Wiley-VCH, Copyright 2020. (D) Illustration of the pH-responsive dynamic conjugation between the mussel-derived cancer-targeting peptide and BTZ. Reproduced from ref. 81 with permission from American Chemical Society, Copyright 2019. | ||
:
5. The obtained D-DOXMAH-S-PEG-based nanoassemblies with a high drug-loading capacity (51.2%) exhibited low premature drug leakage (∼4.5%) at pH = 7.4 but achieved 40.6% of cumulative DOX release under acidic intracellular tumor conditions.
![]() | ||
| Fig. 6 (A) Chemical structures of pH-responsive DOX-ADH-DOX-PEG and ADH-(DOX-PEG)2. Reproduced from ref. 82 with permission from WILEY-VCH, Copyright 2019. (B) Synthesis of the homodimeric doxorubicin prodrug (D-DOXcar). Reproduced from ref. 83 with permission from Elsevier Ltd, Copyright 2020. (C) Synthesis of D-DOXMAH and D-DOXMAH-S-PEG. Reproduced from ref. 84 with permission from Elsevier Ltd, Copyright 2020. (D) Chemical structure and the nano-assembly of the proposed ortho-ester-linked indomethacin (IND) dimer. Reproduced from ref. 85 with permission from American Chemical Society, Copyright 2019. | ||
Wang et al. reported an acid-responsive indomethacin (IND) dimeric nano-prodrug (named IND-OE/DOX) with physical encapsulation of doxorubicin for breast cancer therapy (Table 1).85 The dimeric IND prodrug was synthesized by conjugating two molecules of IND via an acid-labile ortho-ester linkage. The prepared IND dimers could form into homogeneous NPs by using an O/W single-emulsion solvent volatilization method (Fig. 6D). The hydrodynamic diameters of the obtained pH-sensitive NPs (named IND-OE/DOX) and pH-insensitive NPs (named IND-C12/DOX) were about 160–170 nm. The DOX drug-loading capacity of IND-OE/DOX and IND-C12/DOX was 11.39% and 9.43%, respectively. When compared to the pH-insensitive IND-C12/DOX, IND-OE/DOX showed a rapid degradation behaviour and accelerated drug release in mildly acidic environments. More than 85% DOX was released from IND-OE/DOX at pH = 5.0 within 48 h and about 15% of DOX was released at pH = 7.4. In contrast, about 20% DOX was released from IND-C12/DOX NPs after incubation at pH 5.0 or 7.4 within 4 days. Upon internalization by tumor cells, IND-OE/DOX exhibited an accelerated drug release inside the acidic endo-lysosomal system due to hydrolysis of the ortho ester. Notably, the released IND could reduce DOX efflux by P-gp down-regulation to enhance the chemotherapeutic effects. In vivo experiments confirmed that IND-OE/DOX showed synergistic antitumor properties when compared to free DOX.
![]() | ||
| Fig. 7 (A) Chemical design of MPEG-(TK-CPT)-PPa and its self-assembly for ROS-triggered CPT release and tumor therapy under laser irradiation. Reproduced from ref. 90 with permission from WILEY-VCH, Copyright 2020. (B) Schematic illustration of engineered PTX-S-OA/PPa-PEG2k NPs with photodynamic PEG coating for self-enhanced core–shell combination therapy. Reproduced from ref. 91 with permission from Elsevier Ltd, Copyright 2019. (C) Schematic representation of exosome-cloaked sequential-bioactivating paclitaxel prodrug nanoassemblies for cascade-amplified PTX chemotherapy. (D) In vivo fluorescence imaging of DiR NPs and EM@DiR NPs in orthotopic MDA-MB-231 tumor-bearing mice at 2, 8, 12, and 24 h post i.v. injection of NPs. (E) Ex vivo fluorescence imaging of DiR and EM@DiR NPs in the major organs at 24 h after NP injection. Reproduced from ref. 92 with permission from Elsevier Ltd, Copyright 2020. | ||
In another study, Wang et al. described an exosome mimetic sequential-bioactivation paclitaxel nano-prodrug (named EMPCs) that could inhibit breast cancer metastasis by recognizing circulating tumor cells (CTCs) in the bloodstream.92 To fabricate EMPCs, paclitaxel was attached to linoleic acid by using a ROS-responsive thioether linker, and the resulting PTX-S-LA was co-encapsulated with cucurbitacin B (CuB) within PEG-PCL polymeric micelles followed by cloaking with exosome membrane (EM) to achieve a homotypic tumor targeting capability (Fig. 7C). The prepared EMPCs with a spherical shape had an average hydrodynamic size of 100 nm capable of simultaneously targeting the primary tumor and capturing the CTCs during blood circulation. Upon successful entry into tumor cells, the released CuB not only inhibited the tumor metastasis by blocking the FAK/MMP signaling pathway, but also increased endogenous ROS levels to facilitate ROS-triggered PTX release. As shown in Fig. 7D and E, the fluorescence intensity of the exosome membrane-cloaked NPs (EM@DiR) showed an obvious increase at tumor sites from 2 h to 12 h post i.v. injection in MDA-MB-231 tumor-bearing mice, when compared to NPs without EM cloaking. The author demonstrated that the excellent tumor-targeting ability was caused by CD44-based homotypic targeting and CD47-mediated prolonged blood circulation time. When compared to control groups treated with saline or PCNPs that showed remarkable micro-metastases in the lungs, negligible metastatic nodules were observed in the lungs of mice upon treatment with EMPCs. Recently, Xia et al. reported an amphiphilic peptide–drug conjugate-based nanoassembly (named RGD-TK-Epo B) for tumor-targeted therapy.93 The RGD-TK-Epo B was synthesized by conjugating hydrophilic RGD peptide with hydrophobic cytotoxin epothilone B (Epo B) through the ROS-labile thioketal (TK) linker (Fig. 8A). Due to its inherent amphiphilic structure, the resulting RGD-TK-Epo B self-assembled into NPs (RECNs) with a uniform hydrodynamic size of 85.73 nm in an aqueous solution. The resulting RECNs had a good colloidal stability in PBS supplemented with 10% FBS and exhibited a specific targeting ability to tumor cells due to the high affinity of RGD for αvβ3 integrin overexpressed in tumor cells. Upon internalization by targeted tumor cells, Epo B was rapidly released as a result of cleavage in the thioketal bond under high intracellular levels of ROS. In vivo biodistribution studies showed that these RECNs exhibited excellent tumor-targeting ability after 8 h post i.v. injection. As expected, the RECNs presented an efficient antitumor effect in PC-3 tumor-bearing mice.
![]() | ||
| Fig. 8 (A) Chemical structure of an RGD-tk-Epo B conjugate and its self-assembly for tumor-targeted drug delivery. (a and b) RGD-mediated tumor targeting and cell internalization of RECNs; (c) ROS-triggered release of Epo B from RECNs upon cleavage of the thioketal linker; (d) Epo B-induced cell apoptosis. Reproduced from ref. 93 with permission from American Chemical Society, Copyright 2020. (B) Schematic illustration of amphiphilic-block-copolymer Lapa@NPs with encapsulation of β-lapachone, which could catalyze the NAD(P)H-dependent increase in intracellular oxidative stress, thus enabling ROS-responsive drug release in tumor cells. Reproduced from ref. 94 with permission from Elsevier Ltd, Copyright 2019. (C) Chemical structure of PPE-TK-DOX and its self-assembly with encapsulation of photosensitizer Ce6 to enable ROS-triggered drug release under laser irradiation. Reproduced from ref. 95 with permission from Elsevier Ltd, Copyright 2019. | ||
Yin et al. synthesized an amphiphilic block copolymer prodrug (PEG-b-PTCPT) by conjugating camptothecin (CPT) to the backbones of PEG-linked polymerized methacrylate monomers via ROS-labile thioketal bonds (Table 2).94 The nano-prodrugs (Lapa@NPs) were prepared by encapsulation of β-lapachone within PEG-b-PTCPT-based micelles (Fig. 8B). The resulting Lapa@NPs with a hydrodynamic size of 48 nm exhibited good colloidal stability in serum-containing media for over 3 days. High drug-loading capacity (9%) and high encapsulation efficiency (98.5%) of Lapa in Lapa@NPs could be achieved when the weight ratio of polymer to Lapa was set at 10
:
1 during NP formation. Upon internalization to targeted tumor cells, β-lapachone could catalyze NAD(P)H to remarkably increase the intracellular ROS level, which concurrently amplified tumor oxidative stress to induce tumor cell apoptosis and ensure ROS-triggered drug release without additional laser irradiation.
In vitro drug release experiments showed that about 75% of the encapsulated CPT was released from the obtained nano-prodrugs under the conditions of 1 mM H2O2 in the presence of Fe2+ within 80 h. As expected, in vivo experiments showed that the Lapa@NP-treated group exhibited better antitumor effects than other groups, which may be attributed to combinatorial oxidation/chemotherapy. In another study, Pei and co-workers synthesized a polyphosphoester–doxorubicin conjugate (PPE-TK-DOX) by linking doxorubicin (DOX) to the side chains of polyphosphoesters (PPEs) through ROS-sensitive thioketal linkers (Fig. 8C).95 Upon co-self-assembly with photosensitizer Ce6, the resulting PPE-TK-DOX could form into NPs (Ce6@PPE-TK-DOX) in aqueous solution capable of avoiding premature drug leakage during blood circulation. The prepared Ce6@PPE-TK-DOX NPs had a hydrodynamic size of 73 nm and maintained a good colloidal stability with insignificant size changes in PBS supplemented with 10% FBS for over 7 days. Notably, DOX was successfully activated and locally released at tumor sites upon laser irradiation, resulting in minimized systemic side effects. Approximately 71.3% of the thioketal bond was cleaved under 660 nm laser irradiation for 40 min. The in vivo imaging data showed that the Ce6 fluorescence intensity at tumor sites reached a plateau at 4 h post i.v. injection of Ce6@PPE-TK-DOX. Ma et al. synthesized a ROS-activable aspirin–dextran (DEX) conjugate that could self-assemble into a polymeric prodrug (P3C-Asp) for TME regulation and enhanced tumor immunotherapy (Table 1).96 As shown in Fig. 9A, aspirin was attached to DEX via ROS-cleavable p-boronabenzyl ester. The resulting P3C-Asp with an average hydrodynamic size of 40 nm had a high drug-loading content (10 wt%). Upon successful entry into tumor cells, P3C-Asp NPs could be hydrolysed under elevated intracellular levels of ROS leading to the rapid release of deacetylated active aspirin (which could act as a COX-2 inhibitor) to inhibit the PGE2 secretion. Moreover, P3C-Asp could also enhance the infiltration of CD8+ T cells and M1 macrophages while inhibiting the infiltration of myeloid-derived suppressor cells (MDSC) in the TME. Consequently, this polymeric prodrug was capable of eradicating tumor cells in a murine colorectal cancer model upon combination treatment with anti-PD-1 antibodies.
![]() | ||
| Fig. 9 (A) Chemical structures of the ROS-responsive P3C-Asp prodrug and its self-assembly for TME regulation and enhanced tumor immunotherapy. Reproduced from ref. 96 with permission from Chinese Chemical Society, Copyright 2020. (B) Schematic representation of engineered PTX prodrug-based micelles (PEG-B-PTX) for ROS-triggered drug release in tumor therapy. Reproduced from ref. 97 with permission from Elsevier Ltd, Copyright 2020. (C) Chemical structure of PMPC-b-P[MPA(Cap)-co-TPMA]–PAEMA (PMMTAb-Cap) and its self-assembly for ROS-responsive drug release. Reproduced from ref. 98 with permission from American Chemical Society, Copyright 2019. (D) Chemical structures of PEG–PGA–β-CD, Ada–BODIPY, Ada–PTX and their nanoassemblies at an optimized ratio for combined chemo-dynamic therapy. Reproduced from ref. 99 with permission from Wiley-VCH, Copyright 2019. (E) Chemical design of PEG-PO-PTX and its drug release mechanism in the presence of H2O2. Reproduced from ref. 100 with permission from Elsevier Ltd, Copyright 2019. | ||
Dong and co-workers developed an amphiphilic paclitaxel (PTX) prodrug (PEG-B-PTX) by conjugating PTX to polyethylene glycol (PEG) via a p-(boronic ester) benzyl-based linker (Table 2).97 As shown in Fig. 9B, both ROS-sensitive PEG-B-PTX and ROS-insensitive PEG-H-PTX could form into micellar nano-prodrugs (∼50 nm) by self-assembly, providing NPs with a high PTX loading (13.3 wt%). The PEG-B-PTX micelles exhibited a good stability in physiological environments and exhibited a prolonged blood circulation time. In vitro experiments showed that negligible PTX (<5%) was released from PEG-B-PTX under physiological conditions (pH 7.4) even under acidic conditions (pH 5) without the addition of H2O2. In contrast, approximately 65% encapsulated PTX was released within 96 h upon addition of 200 μM of H2O2 at pH 7.4. Notably, accelerated drug release was observed in the presence of H2O2 (200 μM) at pH 5.0 which was approximately due to a stronger oxidation potential of H2O2 under acidic conditions. In another study, Ma et al. synthesized a hierarchical pH and ROS dual-responsive triblock polymeric prodrug (PMMTAb-Cap) that could self-assemble into homogeneous prodrug micelles for efficient cancer theranostics (Fig. 9C).98 The resulting nano-prodrugs possessed a core–shell structure and the outer block poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC) was conjugated with a two-photon fluorophore (TP)-linked poly(2-azepane ethyl methacrylate) (PAEMA) through a pH-sensitive benzoyl imide linker (Table 2), and the Capecitabine (Cap) was introduced onto PAEMA via an ROS-responsive boron ester bond. The resulting PMMTAb-Cap prodrug micelles with a hydrodynamic size of 69 nm exhibited an excellent colloidal stability in PBS supplemented with 10% FBS. Upon accumulation in the mildly acidic TME (pH 6.8), the PMPC could be detached from the prodrug micelles due to the cleavage of imide bonds. Meanwhile, the inner PAEMA became electropositive and hydrophilic (from the original hydrophobic form), thus enabling a decrease in the micellar size and subsequently leading to the eventual enhancement in tissue penetration and endocytosis of these prodrug micelles. Upon successful internalization by targeted tumor cells, rapid drug release was observed under overexpressed intracellular ROS conditions. Notably, the two-photon fluorophore loading enabled these prodrug micelles to possess two-photon AIE bioimaging ability and therefore could be used as a potential theranostic drug delivery system, in which the co-delivery of photosensitizers and prodrugs at a precise ratio might have potential utilities to improve combination therapy.
In another study, Chen et al. reported a supramolecular nano-prodrug to co-deliver photosensitizers and ROS-responsive prodrugs at optimized ratios by using a novel host–guest strategy (Fig. 9D).99 To construct this nanosystem, a block copolymer (PEG–PGA–β-CD) was synthesized by conjugating β-CD to polyethylene glycol (PEG)-linked poly-L-glutamic acid (PGA). Aza-BODIPY and paclitaxel were modified with adamantane to serve as the PS and prodrug guest molecules, respectively. By virtue of the stable host–guest inclusion complex made by β-CD and the adamantane unit, the prodrug and PS could be easily grafted to PEG–PGA–β-CD with an optimized ratio. TEM images showed that Ada–PTX(60%)–BODIPY(40%)–PNs had a uniform and polymersome-like morphology with a hydrodynamic size of 100 nm. Upon NIR light irradiation, the ROS generated by the PS could enhance the release of active paclitaxel. Notably, a smaller NP size (∼68 nm) was observed after NIR irradiation for 30 min, indicating successful PTX release. IVIS imaging data showed that the control groups of Ada–BODIPY and Ada–BODIPY(100%)–PNs exhibited a similar tumor accumulation in tumor-bearing mice at 3 h post i.v. injection. However, the fluorescence intensity at tumor sites in the group treated with Ada–BODIPY(100%)–PNs was 6.5-fold higher in comparison to the Ada–BODIPY treatment group at 24 h post injection. These results indicated that the Ada–BODIPY(100%)–PNs with a prolonged blood circulation time could accumulate selectively in tumor regions. Notably, the mice treated with Ada–PTX(60%)–BODIPY(40%)–PNs exhibited a higher therapeutic efficiency with a lower dose of chemotherapeutic drug than other groups. Li et al. reported ROS-sensitive prodrug micelles for imaging-guided chemo-phototherapy (Fig. 9E).100 The prodrug micelles were fabricated by encapsulating free PTX into the co-self-assemblies of PEG-peroxalate ester-PTX (PEG-PO-PTX) and folate-PEG-Cypate. To achieve ROS-triggered drug release in tumor cells, PEG-PO-PTX was synthesized by conjugating PEG with PTX via an H2O2-labile peroxalate ester bond (Table 1). Folate was utilized to enable a specific tumor-targeting ability of the nano-prodrugs, as folate receptors (FR) are overexpressed in many cancer cells. The prepared nano-prodrugs had a hydrodynamic size of ∼117.6 nm capable of causing 74% PTX release in the presence of 100 μM H2O2 within 24 h. In contrast, less than 20% PTX was released in the absence of H2O2. Notably, the prodrug micelles could maintain a good colloidal stability in PBS with no obvious changes in size for over 1 week. Upon successful internalization into tumor cells which had high levels of intracellular H2O2, the micelles underwent successful disassembly to release both free and conjugated PTX as a result of cleavage in the H2O2-labile peroxalate ester bond. Cypate was used as a photosensitive/photothermal agent to enable optical imaging-guided combinatorial phototherapy under laser irradiation.
![]() | ||
| Fig. 10 (A) Schematic illustration of PEGylated PPa-driven nanoassembly of CTX-S-CTX. (B) CTX activation and release mechanism from CTX-S-CTX. Reproduced from ref. 101 with permission from Ivyspring International Publisher, Copyright 2021. (C) Schematic illustration of the construction of PPa-S-PTX-based nanoassemblies for synergistic photo-chemotherapy. Reproduced from ref. 102 with permission from Elsevier Ltd, Copyright 2019. (D) Molecular structures of the ROS-responsive HRC prodrug and the formation of nanoparticles by encapsulating HRC into F127 polymeric micelles for cancer imaging and combinatorial chemo-photodynamic therapy. Reproduced from ref. 103 with permission from American Chemical Society, Copyright 2020. | ||
In another study, Jiang et al. developed a ROS-activable nanoprodrug (HRC@F127) for combined chemo-photodynamic therapy (Table 1).103 As shown in Fig. 10D, the HRC@F127 was prepared by encapsulating the heterodimeric prodrug (HRC) in the F127-based nanoassemblies. To achieve ROS triggered drug release, the HRC was synthesized by linking camptothecin (CPT) with photosensitizer 2-(1-hexyloxyethyl)-2-devinylpyropheophorbide-a (HPPH) through a thioketal linker (Table 2). The obtained HRC@F127 (23 ± 5 nm) with a high drug loading efficiency (>95%) could keep a good colloidal stability with minimal premature drug leakage during the blood circulation. Notably, the fluorescence of HPPH in HRC@F127 could be quenched due to the existence of strong π–π stacking of HPPH. Upon successful entry into tumor cells, the HPPH and CPT could be rapidly released from HRC@F127 NPs in the presence of high endogenous ROS levels, thus lighting up the tumor cells and achieving an efficient combinatorial chemo-photodynamic therapy. High fluorescence intensity was observed in HRC@F127 NP-treated HCT116 cells, but the ROS-unresponsive control group (HCC@F127)-treated tumor cells showed a negligible fluorescence, indicating the ROS-triggered thioketal bond cleavage and HPPH release from HRC@F127 NPs. To study the in vivo biodistribution of this nano-prodrug, the control NPs (HPPH@F127) were prepared by encapsulating HPPH into the F127 NPs. Both HPPH@F127 and HRC@F127 NPs could specifically accumulate in tumor tissues via the EPR effect, while negligible PET signals were detected in free HPPH molecule-treated mice, which was probably due to the fast clearance of free HPPH during blood circulation. The HRC@F127 treated group exhibited higher PET signals than the control group, which could be probably attributed to lower premature drug leakage during blood circulation due to the higher hydrophobicity of the HRC prodrug.
![]() | ||
| Fig. 11 (A) Chemical structure of mitochondrion-targeting prodrug–peptide conjugates. Reproduced from ref. 109 with permission from John Wiley & Sons, Inc., Copyright 2021. (B) Illustration of the engineered redox-sensitive polyHCPT-based NPs and redox-responsive drug release mechanism. Reproduced from ref. 110 with permission from Elsevier Ltd, Copyright 2020. (C) Chemical structure of CPT-S-S-PEG-iRGD. Reproduced from ref. 111 with permission from Elsevier Ltd, Copyright 2020. (D) Schematic illustration of PTX–CIT conjugates linked by different chemical bonds (sulfur/selenium/carbon). Reproduced from ref. 113 with permission from Springer Nature, Copyright 2019. (E) Chemical structure of the polymeric PMPT prodrug and its self-assembly with encapsulation of two AIE photosensitizers (PyTPE and TB) for imaging-guided PDT and drug release. Reproduced from ref. 114 with permission from American Chemical Society, Copyright 2021. | ||
The diselenide bond (Se–Se), a ROS-responsive linker, was also used in the redox-responsive drug delivery system. The bond dissociation energy (172 kJ mol−1) of Se–Se is lower than the disulfide bond (240 kJ mol−1).112 Thus, the diselenide linker has a greater potential than the disulfide linker for redox-responsive drug release in tumor cells. In a recent study, Sun et al. synthesized six paclitaxel–citronellol (PTX–CIT) conjugates to investigate the effects of various chemical bonds (sulfur/selenium/carbon) and bond angles/dihedral angles on self-assembly performance, drug release, cytotoxicity, stability, and pharmacokinetics (Fig. 11D).113 The hydrophobic PTX–CIT conjugates could form into uniform nano-prodrugs through one-step nanoprecipitation followed by PEGylation with DSPE-PEG2K to obtain better colloidal stability and pharmacokinetic behavior. The obtained PEGylated nano-prodrugs had an average hydrodynamic size of ∼90 nm and high drug loading content (>50 wt%). In this study, the authors found that the thioether bonds and selenoether bonds showed much higher oxidation sensitivity than reduction sensitivity. As expected, the diselenide bond-based nano-prodrugs exhibited the most promising antitumor activity compared to other responsive linkers. The superior antitumor effect was probably attributed to several advantages of these nano-prodrugs including the good colloidal stability, prolonged blood circulation time, preferential tumor distribution, and efficient drug release. The selenoether/diselenide bonds enabled reactive oxygen species production, which could boost the cytotoxicity of these prodrugs. This study provides a strategy for rational design of redox-responsive prodrug delivery systems. In another study, Yi et al. reported a self-guiding prodrug micelle (TB@PMPT) for improved chemo-photodynamic therapy (Table 1).114 As shown in Fig. 11E, TB@PMPT was prepared by co-self-assembly of a polymeric prodrug (PEG-b-PMPMC-g-PTX-g-PyTPE, PMPT) with physical encapsulation of another AIE photosensitizer TPA-BDTP (TB). The PMPT was synthesized by simultaneously conjugating the AIE photosensitizer PyTPE and reduction-sensitive PTX prodrug (PTXSS-N3) onto the backbone of an amphiphilic polycarbonate. The obtained TB@PMPT micelles could emit yellow and red fluorescence from PyTPE and TB, respectively. Upon accumulation in tumor tissues, the PyTPE and TB generated ROS on the first light irradiation and subsequently induced lipid peroxidation and increased the permeability of the cell membrane, leading to enhanced cell internalization of TB@PMPT micelles. The cleavage of the disulfide bond under the conditions of high intracellular GSH leads to the rapid release of PTX. Notably, PTX release could improve the hydrophilicity of the residual amphiphilic polymer and increase the dispersion of PyTPE in an aqueous solution capable of causing fluorescence ratio transformation as TPA-BDTP (TB) remained aggregated after PTX release. The fluorescence ratio transformation could be employed as a ratiometric fluorescence probe to guide the occasion for second irradiation to achieve PDT, as well as for drug release monitoring.
![]() | ||
| Fig. 12 (A) Nano-prodrug constructed by self-assembly of DSD, DSSD and DSSSD with or without DSPE-PEG2K. In vitro DOX-SH release from DSSD and DSSSD in (B) 0.5 mM or (C) 1 mM GSH. Reproduced from ref. 115 with permission from American Association for the Advancement of Science, Copyright 2020. (D) Chemical structure of a gefitinib linked near-infrared dye and its self-assembly with encapsulation of celastrol for tumor therapy. Reproduced from ref. 116 with permission from Wiley-VCH, Copyright 2019. | ||
![]() | ||
| Fig. 13 (A) Chemical design of the amphiphilic imidazoquinoline (IMDQ) prodrug and its self-assembly into vesicular nanostructures. Reproduced from ref. 122 with permission from American Chemical Society, Copyright 2020. (B) Illustration of the F-integrated ASO-b-PEG-b-PCL triblock copolymer and its self-assembly to reverse chemoresistance. Reproduced from ref. 123 with permission from American Chemical Society, Copyright 2021. (C) Preparation of TME-responsive nano-prodrugs by co-self-assembly of a photothermal agent and IDO inhibitor for cancer immunotherapy. Reproduced from ref. 125 with permission from Elsevier Ltd, Copyright 2020. (D) Chemical structures of a cathepsin B-responsive prodrug and its self-assemblies for combinatorial chemo-photodynamic therapy to potentiate the effective checkpoint blockade-based tumor immunotherapy. Reproduced from ref. 126 with permission from American Chemical Society, Copyright 2021. | ||
Luo et al. synthesized another enzyme-responsive dendritic polymer–paclitaxel prodrug conjugate (POEGMA-GFLG-PTX), in which PTX was conjugated onto poly[oligo(ethylene glycol) methyl ether methacrylate] (polyOEGMA) via a cathepsin B-responsive tetra-peptide GFLG linkage (Table 2).124 The resulting POEGMA-GFLG-PTX had a low CMC value of 0.2 μg mL−1, which could self-assemble into NPs with a hydrophobic core for encapsulation of T1 (an imidazole derivative) and pyropheophorbide a. The average hydrodynamic size of the obtained nano-prodrug was about 163.1 ± 13.0 nm. Both cargoes (T1 and PPa) in this nanoassembly had a high encapsulation efficiency of over 60%. In this study, the T1 with two-photon (TP) absorption and high energy-transfer efficiency was exploited to enhance the two-photon photodynamic therapy. Upon successful internalization by tumor cells, PTX was rapidly released due to the cleavage of GFLG peptide in the presence of intracellular cathepsin B. The resulting nano-prodrug could achieve enhanced antitumor efficiency in 4T1 xenograft mice through combinatorial chemotherapy and two-photon photodynamic therapy. In another study, Liu and co-workers reported a tumor microenvironment-responsive nano-prodrug for improved photo-immunotherapy (Table 1).125 As shown in Fig. 13C, the MMP-2 responsive peptide sequence PVGLIG was introduced as a linker to conjugate the indoleamine 2,3-dioxygenase (IDO) inhibitor (IDOi, Epacadostat) with PEG (Table 2). The PEGylated IDOi and photosensitizer (ICG) could co-self-assemble into uniform NPs (mPEG-Pep-IDOi/ICG NPs) via intermolecular interactions. As a result of the peptide cleavage by MMP-2 in the TME, the resulting nano-prodrug with a large initial size (∼140 nm) could transform to smaller-sized (<40 nm) drug–drug complexes that could enhance the drug penetration in tumor tissues. The mPEG-Pep-IDOi/ICG NPs exhibited an enhanced release of both IDOi and ICG under conditions of MMP-2 when compared to the mPEG-IDOi/ICG NPs under the same conditions. Upon NIR laser irradiation, the ICG mediated phototherapy could kill tumor cells and evoke an in situ antitumor immune response to facilitate IDO-mediated immunosuppression. The author found that the phototherapy mediated by mPEG-Pep-IDOi/ICG NPs could improve the expression of the co-stimulatory molecules (CD80 and CD86) on dendritic cells (DCs). Moreover, the mPEG-Pep-IDOi/ICG NPs could promote DC maturation in the tumor site probably due to their improved tumor specific accumulation and tumor cell internalization ability. Notably, this MMP-2-responsive nano-prodrug could synergistically enhance the tumor immunotherapy in combination with the PD-L1 checkpoint blockade, leading to a substantial antitumor effect on both primary and abscopal tumors.
![]() | ||
| Fig. 14 (A) Schematic illustration of a hypoxia-activable semiconducting polymeric nano-prodrug for synergistic chemo-photodynamic therapy. Reproduced from ref. 131 with permission from Wiley-VCH, Copyright 2018. (B) Chemical structures of a hypoxia-responsive nano-prodrug (Ce6/PTX2-Azo) and its self-assembly for synergistic photodynamic-chemotherapy. Reproduced from ref. 132 with permission from Wiley-VCH., Copyright 2020. | ||
![]() | ||
| Fig. 15 (A) Chemical design of GGT-activable cationizing PBEAGA-CPT and the non-GGT-activable PEAGA-CPT, and the mechanism for GGT-catalysed γ-glutamylamide hydrolysis. Reproduced from ref. 134 with permission from Springer Nature, Copyright 2019. (B) Molecular structure of the redox/pH dual-responsive CPT-ss-poly(BYP-hyd-DOX-co-EEP). Reproduced from ref. 135 with permission from American Chemical Society, Copyright 2019. (C) Schematic illustration of the PEG-GAx/Pt formation through coordination of the polyphenol and CDDP, and the mechanism for intracellular dual-responsive drug release. Reproduced from ref. 136 with permission from Elsevier Ltd, Copyright 2020. (D) Molecular structures of mPEG-Phe-TK-Phe-hyd-DOX and its self-assemblies for tumor therapy. Reproduced from ref. 137 with permission from Elsevier Ltd, Copyright 2021. (E) Chemical structure of diblock pDHPMA-DOX. Reproduced from ref. 138 with permission from Elsevier Ltd, Copyright 2019. | ||
In another study, Xiang et al. developed a novel polyphenol–cisplatin complexation-based core–shell structured nano-prodrug (PEG-GAx/Pt) to achieve pH and ROS-responsive drug delivery (Table 1).136 To prepare the PEG-GAx/Pt, methoxyl-PEG terminated with one (PEG-GA) or two (PEG-GA2) gallic acid moieties was complexed with cisplatin (CDDP) by using the polyphenol–metal coordination method (Fig. 15C). When the molar ratio of CDDP to GA (MPt
:
MGA) was set to 1
:
1 and 10
:
1, the resulting PEG-GA/Pt and PEG-GA2/Pt could self-assemble into homogeneous NPs that have a volume size of 80–100 nm and 110–130 nm, respectively. The obtained NPs had a good colloidal stability with no obvious size changes in PBS supplemented with 5%, 10%, or 50% FBS for 72 h at 37 °C. The loading content of CDDP in PEG-GA and PEG-GA2 was determined to be about 17.7% to 29.8%, respectively. In vitro drug release experiments showed that about 47% and 59% of Pt was released from PEG-GA/Pt and PEG-GA2/Pt NPs at pH 5.0 within 48 h, respectively. However, <20% Pt drug was released from both PEG-GA/Pt and PEG-GA2/Pt at pH 7.4. The pH-triggered Pt release was attributed to the weakened coordination between polyphenols and Pt(II) under acidic conditions.
Notably, the addition of H2O2 could promote drug release due to the oxidation of galloyl groups. The PEG-GAx/Pt NPs exhibited prolonged blood circulation time and enhanced tumor accumulation, thus increasing antitumor efficiency, and minimizing toxicity. In another study, Xu et al. synthesized a ROS/pH dual-responsive polymer–DOX conjugate (mPEG-Phe-TK-Phe-hyd-DOX) for efficient drug release and improved antitumor efficiency (Fig. 15D).137 In this study, the acylhydrazine linker (ahy-Phe-TK-Phe-ahy) containing ROS-sensitive thioketal (TK) bonds was inserted between the mPEG and DOX, in which DOX was conjugated onto the ahy-Phe-TK-Phe-ahy via acid-sensitive hydrazone bonds (Table 2). The CMC value of mPEG-Phe-TK-Phe-hyd-DOX was calculated to be 3.79 μg mL−1. The hydrophobic phenylalanine moieties (Phe-TK-Phe) could facilitate the self-assembly of the mPEG-Phe-TK-Phe-hyd-DOX to uniform nanostructures with a hydrodynamic size of 41.32 nm. The resulting prodrug micelles had a high drug loading content (11.2%) and good colloidal stability due to the π–π interactions of prodrug molecules, which could minimize the drug leakages during blood circulation. In vitro cytotoxicity experiments showed that the IC50 of mPEG-Phe-TK-Phe-hyd-DOX was about 5.50 μg mL−1 against HeLa cells, which was lower than those of mPEG-hyd-DOX (19.2 μg mL−1) and DOX/mPEG-ahy-Phe-TK-Phe-ahy (35.4 μg mL−1). As expected, the ROS/pH-responsive nano-prodrugs exhibited a better antitumor efficacy than the ROS-inert control nano-prodrug. In a similar study, Chen et al. reported an enzyme/pH dual sensitive polymer–doxorubicin conjugate (diblock pDHPMA-DOX) for improved antitumor efficiency of the chemotherapeutic drugs and minimized systemic side effects (Table 1).138 To obtain the diblock pDHPMA-DOX, the DOX was attached onto the GFLG peptide-functionalized diblock copolymer through acid-labile hydrazone bonds (Table 2), in which the N-(1,3-dihydroxypropan-2-yl) methacrylamide (DHPMA) copolymer was synthesized by RAFT polymerization (Fig. 15E). The diblock pDHPMA-DOX could self-aggregate into nanoassemblies that had an average hydrodynamic size of 21 ± 2.2 nm capable of causing ∼82% and ∼80% DOX release at pH 5.0 within 10 h in the presence of cathepsin B or no cathepsin B, respectively. In contrast, a negligible amount of DOX (∼5%) was released from the nano-prodrug at pH 7.4 with or without addition of cathepsin B. The diblock pDHPMA-DOX NPs could maintain a good colloidal stability with no significant aggregation/degradation within 24 h in PBS supplemented with 50% FBS. Upon successful internalization by tumor cells, the diblock copolymer-based NPs (90 kDa) were degraded into small fragments that had smaller molecular weight (45 kDa) as a result of the cleavage of GFLG peptide under conditions of lysosomal cathepsin B. In vivo pharmacokinetics experiments revealed that the diblock pDHPMA-DOX NPs had a prolonged blood circulation time of 9.8 h, which was longer than that of free DOX (3.7 h). As expected, the diblock pDHPMA-DOX NPs displayed an enhanced antitumor efficiency for 4T1 xenograft tumors.
![]() | ||
| Fig. 16 (A) Chemical design and synthetic routes of the PTX-chemogene conjugate and its self-assembly for combination therapy to overcome drug resistance. Reproduced from ref. 139 with permission from Wiley-VCH, Copyright 2020. (B) Schematic representation of sulfur/selenium/tellurium-bridged homodimeric prodrugs and their intracellular ROS/GSH dual-responsive drug release. (C) HPLC analysis of PTX2-S, PTX2-Se and PTX2-Te dimers in the presence of 1 mM H2O2 or 10 mM DTT for 24 h at 37 °C. Reproduced from ref. 140 with permission from Elsevier Ltd, Copyright 2020. | ||
NC-6004, a polymeric cisplatin nanoparticle (∼30 nm), was constructed by encapsulating cisplatin into the micelles through the formation of a polymer–metal complex between polyethylene glycol-poly(glutamic acid) block copolymers (PEG-P(Glu)) and cisplatin (Table 2).144,145 Compared to free cisplatin that could be excreted rapidly from the human body, NC-6004 could significantly extend the blood circulation time of cisplatin as the outer PEG shell on the surface of the micelles could minimize its clearance by the reticuloendothelial systems.146 Furthermore, NC-6004 with a sustained drug release ability has lower toxicity than the native cisplatin.147 In a phase Ib/II trial study, NC-6004 plus gemcitabine exhibited a greater anti-tumor activity and no clinically significant neuro-, oto-, or nephrotoxicity. NC-6300, another clinical candidate, was prepared by conjugating epirubicin to PEG-b-poly(aspartic acid) through hydrazone linkages.148 The resulting polymer–epirubicin conjugates could form into micelles that could keep stable during blood circulation but achieve fast release of drug in acidic environments (e.g. endosomal and lysosomal).149 Notably, NC-6300 could significantly minimize the system side-effects so that higher doses of drug could be used through micellar therapy.148 Similarly, NK911 is a doxorubicin-loaded PEG-b-poly(aspartic acid) copolymer-based prodrug micelle (∼40 nm), in which the doxorubicin is both physically encapsulated in the micelle and conjugated to the PEG-b-poly(aspartic acid) copolymer via amide bonds.150 NK911 exhibited a prolonged blood circulation time and a larger area under the plasma drug concentration–time curve (AUC) than native doxorubicin in a phase I clinical trial.150 CriPec® Docetaxel (CPC634) consists of core-cross linked micellar nanoparticles with covalently entrapped docetaxel. CPC634 entered the first-in-human study and finished its phase I testing in 2018. The CPC634 exhibited an improved pharmacokinetics and therapeutic index in patients with advanced solid tumors.151 The polymeric nanoassemblies CRLX101 and CRLX301 are constructed by self-assembly of cyclodextrin–drug conjugates, which were synthesized by covalently conjugating cyclodextrin-PEG copolymer with camptothecin and docetaxel, respectively.152,153 CRLX301 exhibited a controlled drug release ability and slower clearance than free docetaxel in a phase I/IIa study.154 In a randomized phase II trial, CRLX101 plus bevacizumab had no discernible increase in progression-free survival (PFS) when compared to the standard of care (SOC) in metastatic renal cell carcinoma.155
Despite obvious benefits afforded by self-assembled prodrug strategies, there are still several challenges that need to be circumvented before their successful clinical translation. First, the CMC value of amphiphilic prodrugs should be as low as possible, as it is an important factor that affects the systemic stability of prodrug micelles during blood circulation. Second, the non-homogeneous nano-prodrug activation and distribution still exists due to the heterogeneous nature of tumor tissues, thereby leading to the suboptimal therapeutic efficacy. Therefore, exogenous physical stimuli (light, temperature, ultrasound, X-ray, etc.) plus endogenous stimuli (pH, GSH, ROS, enzyme, and hypoxia) could thus be exploited for development of multi-responsive drug delivery systems. Third, the potential systemic cytotoxicity of the carrier materials still poses challenges that restrict the clinical translation of the polymeric nanoparticles. Thus, materials with high biodegradability should be well explored, or carrier-free drug delivery systems could be developed. Fourth, it is difficult to avoid the batch-to-batch variations of polymer–drug conjugation limiting its clinical translation. That is why the design of prodrug nanoassemblies should be as simple as possible to facilitate easy preparation. Finally, there is an urgent need to develop novel prodrug chemistry that allows more efficient and specific drug release at tumor sites. For example, we can try to develop self-immolative linker-based combinatorial chemistry with a cascade reaction ability to improve the stimulus-responsive drug release ability. Nevertheless, self-assembled prodrug designs have great potential in tumor-targeted drug delivery systems, and we look forward to seeing more clinical translation in the coming years.
| This journal is © The Royal Society of Chemistry 2022 |