Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Biomacromolecules ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829675

ABSTRACT

Despite great progress in the hydrogel hemostats and dressings, they generally lack resistant vascular bursting pressure and intrinsic bioactivity to meet arterial massive hemorrhage and proheal wounds. To address the problems, we design a kind of biomimetic and wound microenvironment-modulating PEGylated glycopolypeptide hydrogels that can be easily injected and gelled in ∼10 s. Those glycopolypeptide hydrogels have suitable tissue adhesion of ∼20 kPa, high resistant bursting pressure of ∼150 mmHg, large microporosity of ∼15 µm, and excellent biocompatibility with ∼1% hemolysis ratio and negligible inflammation. They performed better hemostasis in rat liver and rat and rabbit femoral artery bleeding models than Fibrin glue, Gauze, and other hydrogels, achieving fast arterial hemostasis of <20 s and lower blood loss of 5-13%. As confirmed by in vivo wound healing, immunofluorescent imaging, and immunohistochemical and histological analyses, the mannose-modified hydrogels could highly boost the polarization of anti-inflammatory M2 phenotype and downregulate pro-inflammatory tumor necrosis factor-α to relieve inflammation, achieving complete full-thickness healing with thick dermis, dense hair follicles, and 90% collagen deposition. Importantly, this study provides a versatile strategy to construct biomimetic glycopolypeptide hydrogels that can not only resist vascular bursting pressure for arterial massive hemorrhage but also modulate inflammatory microenvironment for wound prohealing.

2.
Int J Biol Macromol ; 272(Pt 1): 132736, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38830494

ABSTRACT

Fatal massive hemorrhage and diabetic wound healing are world widely challenging in surgical managements, and uncontrolled bleeding, chronic inflammation and damaged remodeling heavily hinder the whole healing processes. Considering hemostasis, inflammation and wound microenvironment cooperatively affect the healing progression, we design all-in-one beta-glucan (BG) hybrid hydrogels reinforced with laponite nanoclay that demonstrate tunable tissue adhesion, resistant vascular burst pressure and cooperative wound microenvironment regulation for arterial hemostasis and diabetic wound prohealing. Those hydrogels had honeycomb-like porous microstructure with average pore size of 7-19 µm, tissue adhesion strength of 18-46 kPa, and vascular burst pressure of 58-174 mmHg to achieve superior hemostasis in rat liver and femoral artery models. They could effectively scavenge reactive oxygen species, transform macrophages from proinflammatory M1 into prohealing M2, and shorten the inflammation duration via synergistic actions of BG and nitric oxide (NO). Single treatment of NO-releasing BG hybrid hydrogels attained complete closure of diabetic wounds within 14 days, orchestrated to accelerate the epithelization and dermis growth, and restored normal vascularization, achieving high performance healing with optimal collagen deposition and hair follicle regeneration. Consequently, this work opens up a new avenue to design all-in-one polysaccharide hydrogels for applications in massive bleeding hemostats and diabetic wound dressings.


Subject(s)
Hemorrhage , Hydrogels , Wound Healing , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Wound Healing/drug effects , Rats , Hemorrhage/drug therapy , Diabetes Mellitus, Experimental/complications , Male , Nitric Oxide/metabolism , beta-Glucans/chemistry , beta-Glucans/pharmacology , Mice , Rats, Sprague-Dawley , Polysaccharides/pharmacology , Polysaccharides/chemistry
3.
Biomater Sci ; 12(5): 1211-1227, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38240342

ABSTRACT

Regulating the wound microenvironment to promote proliferation, vascularization, and wound healing is challenging for hemostats and wound dressings. Herein, polypeptide composite hydrogels have been simply fabricated by mixing a smaller amount of metal ion-coordinated nanoparticles into dopamine-modified poly(L-glutamic acid) (PGA), which had a microporous size of 10-16 µm, photothermal conversion ability, good biocompatibility, and multiple biological activities. In vitro scratch healing of fibroblast L929 cells and the tube formation of HUVECs provide evidence that the PGA composite hydrogels could promote cell proliferation, migration, and angiogenesis with the assistance of mild photothermia. Moreover, these composite hydrogels plus mild photothermia could effectively eliminate reactive oxygen species (ROS), alleviate inflammation, and polarize the pro-inflammatory M1 macrophage phenotype into the pro-healing M2 phenotype to accelerate wound healing, as assessed by means of fluorescent microscopy, flow cytometry, and quantitative real-time polymerase chain reaction (qRT-PCR). Meanwhile, a rat liver bleeding model illustrates that the composite hydrogels reduced the blood loss ratio to about 10% and shortened the hemostasis time to about 25 s better than commercial chitosan-based hemostats. Furthermore, the full-thickness rat skin defect models showcase that the composite hydrogels plus mild photothermia could proheal wounds completely with a fast healing rate, optimal neovascularization, and collagen deposition. Therefore, the biodegradable polypeptide PGA composite hydrogels are promising as potent wound hemostats and dressings.


Subject(s)
Glutamic Acid , Nanoparticles , Rats , Animals , Glutamic Acid/pharmacology , Hydrogels/pharmacology , Wound Healing , Hemostasis , Peptides/pharmacology , Anti-Bacterial Agents/pharmacology
4.
Macromol Biosci ; 23(11): e2300215, 2023 11.
Article in English | MEDLINE | ID: mdl-37363952

ABSTRACT

Ferroptosis is a novel type of iron-dependent non-apoptotic pathway that regulates cell death and shows unique mechanisms including causing lipid peroxide accumulation, sensitizing drug-resistant cancers, priming immunity by immunogenic cell death, and cooperatively acting with other anticancer modalities for eradicating aggressive malignancies and tumor relapse. Recently, there has been a great deal of effort to design and develop anticancer biocompatible polymeric nanoplatforms including polypeptide and PEGylated ones to achieve effective ferroptosis therapy (FT) and synergistic combination therapies including chemotherapy (CT), photodynamic therapy (PDT), sonodynamic therapy (SDT), photothermal therapy (PTT), gas therapy (GT) including nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2 S), and immunotherapy (IT). To be noted, the combo therapies such as FT-CT, FT-PTT, FT-GT, and FT-IT are attracting much efforts to fight against intractable and metastatic tumors as they can generate synergistic antitumor effects and immunogenic cell death (ICD) effects or modulate immunosuppressive tumor microenvironments to initiate strong antitumor immunity and memory effects. The polymeric Fenton nano-agents with good biosafety and high anticancer efficacy will provide a guarantee for their applications. In this review, various biocompatible polymer-modified nanoplatforms designed for FT and combo treatments are summarized for anticancer therapies and discussed for potential clinical transitions.


Subject(s)
Ferroptosis , Nanoparticles , Neoplasms , Photochemotherapy , Humans , Combined Modality Therapy , Immunotherapy , Polymers , Cell Line, Tumor , Neoplasms/drug therapy , Nanoparticles/therapeutic use , Tumor Microenvironment
5.
Biomacromolecules ; 23(6): 2655-2666, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35583462

ABSTRACT

Owing to having a unique mechanism to kill cancer cells via the membrane accumulation of lipid peroxide (LPO) and the downregulation of glutathione peroxidase-4 (GPX-4), the ferroptosis therapy (FT) of tumors based on the Fenton reaction of iron nanoparticles has been receiving much attention in the past decade; however, there are some hurdles including the uncontrollable release of iron ions, slower kinetics of the intracellular Fenton reaction, and poor efficacy of FT that need to be overcome. Considering cooperative coordination of a multivalent thiol-pendant polypeptide ligand with iron ions, we put forward a facile strategy for constructing the iron-coordinated nanohybrid of methacryloyloxyethyl phosphorylcholine-grafted polycysteine/iron ions/tannic acid (i.e., PCFT), which could deliver a higher concentration of iron ions into cells. The dynamic and unsaturated coordination in PCFT is favorable for the intracellular stimuli-triggered release and fast Fenton reaction to realize efficient FT, while its intrinsic photothermia would boost the Fenton reaction to induce a synergistic effect between FT and photothermal therapy (PTT). Both immunofluorescence analyses of reactive oxygen species (ROS) and LPO confirmed that the intracellular Fenton reaction resulted in efficient FT, during which process the photothermia greatly boosted ferroptosis, and the Western blot assay corroborated that the expression level of GPX-4 was downregulated by FT and highly degraded by the photothermia to induce synergistic PTT-FT in vitro. Excitingly, by a single intravenous dose of PCFT plus one NIR irradiation, in vivo PTT-FT treatment completely eradicated 4T1 tumors without skin scar and tumor recurrence for 16 days, demonstrating prominent antitumor efficacy, as evidenced by the GPX-4, H&E, and TUNEL assays.


Subject(s)
Ferroptosis , Hyperthermia, Induced , Nanoparticles , Neoplasms , Cell Line, Tumor , Humans , Iron , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Peptides/therapeutic use , Photothermal Therapy , Tannins
6.
Acta Biomater ; 146: 396-405, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35470074

ABSTRACT

Tumor microenvironment responsive nanomedicine has drawn considerable attention for combination therapy, but still remains a significant challenge for less side effects and enhanced anti-tumor efficiency. Herein, we develop a pH/ROS dual-responsive supramolecular polypeptide nanoprodrug (PFW-DOX/GOD) by using pillar[5]arene-based host-guest strategy for combined glucose degradation, chemodynamic therapy (CDT), and chemotherapy (CT). The PFW-DOX/GOD consists of a pH-responsive ferrocene/pillar[5]arene-containing polypeptide, a ROS-responsive polyprodrug, and encapsulated glucose oxidase (GOD). Upon into intracellular acidic environment, PFW-DOX/GOD exhibits rapid pH-triggered disassembly behavior. Simultaneously, the released GOD can catalyze intratumoral glucose into massive H2O2, which are further converted into highly toxic hydroxyl radicals (•OH) by the catalysis of ferrocene via the Fenton reaction. Thereafter, induced by the ROS-responsive cleavage of thioketal linkage, the conjugated DOX prodrug was released and activated. The combined glucose degradation, chemodynamic therapy (CDT), and chemotherapy (CT) of PFW-DOX/GOD present anti-tumor effect with 96% of tumor inhibitory rate (TIR). Therefore, such tumor microenvironment-responsive supramolecular polypeptide nanoprodrugs represent a potential candidate for combination therapy with minimal side effects. STATEMENT OF SIGNIFICANCE: In this work, a tumor microenvironment-responsive supramolecular polypeptide nanoprodrug (PFW-DOX/GOD) was prepared via pillar[5]arene-based host-guest interactions, and presented low side effects and high tumor accumulation owing to the diameters of about 200 nm and surface PEG segment. After pH-responsive release of GOD in the intracellular acidic environment, the cascade catalytic reactions including GOD-catalyzed degradation of intratumoral glucose and Fenton reaction, effectively happened to generate •OH for chemodynamic therapy (CDT), which subsequently induced the cleavage of thioketal linkage to activate free DOX for chemotherapy (CT). Collectively, this supramolecular polypeptide nanoprodrugs provide a promising strategy for combination therapy with synergetic anti-tumor effect.


Subject(s)
Nanoparticles , Neoplasms , Cell Line, Tumor , Doxorubicin/chemistry , Glucose/pharmacology , Glucose Oxidase , Humans , Hydrogen Peroxide/pharmacology , Metallocenes/pharmacology , Nanoparticles/chemistry , Neoplasms/drug therapy , Peptides/pharmacology , Reactive Oxygen Species , Tumor Microenvironment
7.
Acta Biomater ; 143: 381-391, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35272024

ABSTRACT

Supramolecular nanomedicine assembly combined with polypeptide prodrug could become a powerful strategy to minimize drug leakage in blood circulation and trigger sufficient drug release at tumor tissue. Here, we developed a charge-reversal amphiphilic pillar[5]arene-modified polypeptide (P5-PLL-DMA), and reactive oxygen species (ROS)-sensitive polypeptide prodrug (P-PLL-DOX) including a ROS-cleavable thioketal (TK) linker between doxorubicin (DOX) and poly(L-lysine) (PLL), which could assemble via pillar[5]arene host-guest recognition, and further encapsulate chlorin e6 (Ce6) to obtain a supramolecular polypeptide prodrug (SPP-DOX/Ce6). The chemical conjugation to load drugs of DOX and the negatively charge of SPP-DOX/Ce6 could prevent premature drug leakage, and reduce undesirable interaction with serum proteins to enhance stability under physiological conditions (pH 7.4). Simultaneously, the carried charge of SPP-DOX/Ce6 reversed from negative to positive could effectively enhance the cellular internalization for efficient DOX delivery under acidic tumor microenvironment (pH 6.5). Upon 660 nm near-infrared light (NIR) irradiation, the ROS generated by encapsulated Ce6 rapidly cleaved the TK linker to release activated DOX, inducing the tumor-specific drug delivery. This intelligent supramolecular polypeptide prodrug based on pillar[5]arene host-guest recognition represents new avenues to develop stimulus responsive prodrug for enhanced cancer therapy with minimized the side effect. STATEMENT OF SIGNIFICANCE: In this work, a pH/ROS dual-sensitive supramolecular polypeptide prodrug (SPP-DOX/Ce6) was developed to minimize drug leakage in blood circulation and trigger sufficient drug release at tumor tissue. The chemical conjugation to load drugs of DOX via a ROS-cleavable thioketal (TK) linker and the distinctive charge-reversal capacity of SPP-DOX/Ce6 significantly enhances the stability under physiological conditions (pH 7.4), while facilitates cellular uptake at tumor site (pH 6.8). Upon 660 nm near-infrared light (NIR) irradiation, the ROS generated by encapsulated Ce6 induces the rapid cleavage of TK linker to release activated DOX, achieving a tumor-specific drug delivery. This intelligent supramolecular polypeptide prodrug SPP-DOX/Ce6 provides an effective strategy to construct stimulus responsive prodrug for enhanced cancer therapy.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Prodrugs , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Liberation , Hydrogen-Ion Concentration , Nanomedicine , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Peptides/metabolism , Peptides/pharmacology , Prodrugs/pharmacology , Reactive Oxygen Species/metabolism
8.
Adv Healthc Mater ; 11(6): e2101809, 2022 03.
Article in English | MEDLINE | ID: mdl-34865324

ABSTRACT

Polymeric hydrogels have been increasingly studied for wound sealants, adhesives, hemostats, and dressings, however, multi-component gelation, adhesion-causing tissue damage, inefficient hemostasis, and skin scarring in wound healing hamper their advances. So it is urgent to develop multifunctional single-component polymeric hydrogels with benign tissue detachment, high performance hemostasis, and scarless wound healing attributes. Herein, a dopamine-modified poly(l-glutamate) hydrogel at an ultralow concentration of 0.1 wt% is serendipitously constructed by physical treatments, in which a gelation mechanism is disclosed via oxidative catechol-crosslinking and sequential dicatechol-carboxyl hydrogen-bonding interactions. The covalent/H-bonding co-crosslinked and highly negative-charged networks enable the polypeptide hydrogels thermo-, salt-, urea-resistant, self-healing, injectable, and adhesive yet detachable. In vitro and in vivo assays demonstrate they have superior biocompatibility with ≈0.5% hemolysis and negligible inflammation. The polypeptide/graphene oxide hybrid hydrogel performs fast and efficient hemostasis of 12 s and 1.4% blood loss, surpassing some hydrogels and commercial counterparts. Remarkably, the polypeptide hydrogels achieve scarless and full wound healing and regenerate thick dermis with some embedded hair follicles within 14 days, presenting superior full-thickness wound healing and skin scar-preventing capabilities. This work provides a simple and practicable method to construct multifunctional polypeptide hemostatic and healing hydrogels that overcome some above-mentioned hurdles.


Subject(s)
Hydrogels , Wound Healing , Bandages , Hemostasis , Hydrogels/pharmacology , Peptides/pharmacology
9.
Article in English | MEDLINE | ID: mdl-34310063

ABSTRACT

Stimuli-responsive polypeptide nanoassemblies exhibit great potentials for cancer nanomedicines because of desirable biocompatibility and biodegradability, unique secondary conformations, varying functionalities, and especially the stimuli-enhanced therapeutic efficacy and reduced side effect. This review introduces the design and fabrication of stimuli-responsive polypeptide nanoassemblies that exhibit endogenous stimuli (e.g., pH, reduction, reactive oxygen species, adenosine triphosphate and enzyme, etc.) and exogenous light stimuli (e.g., UV and near-infrared light), which are biologically related or applied in the clinic. We also discuss the applications and prospects of those stimuli-responsive polypeptide nanoassemblies that might overcome the biological barriers of cancer nanomedicines for in vivo administration. Much more effort is needed to accelerate the second-generation stimuli-responsive polypeptide nanomedicines for clinical transition and applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.


Subject(s)
Nanoparticles , Neoplasms , Drug Delivery Systems , Drug Discovery , Humans , Nanomedicine , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Peptides , Reactive Oxygen Species
10.
J Biomed Mater Res A ; 110(4): 954-963, 2022 04.
Article in English | MEDLINE | ID: mdl-34913253

ABSTRACT

Stimuli-responsive prodrug-based nanoplatform with synergistic antitumor activity is of central importance to the development of promising nanomedicines for cancer therapy. Here, we describe a polydopamine-drug conjugate nanocomposite (ZP-PDA-DOX) with targeted cancer photothermal-chemotherapy (PTT-CT), which constructed by a gradual copolymerization of dopamine (DA) and pH-sensitive dopamine-derived prodrug (DA-DOX) into the porous channels of zeolite imidazolate frameworks-8 (ZIF-8), followed by PEGylation with amino-terminated folic acid-polyethylene glycol (NH2 -PEG-FA) to acquire the high biocompatibility, specificity, and excellent tumor-targeting property. The incorporation of polydopamine strengthened the stability and dispersion of ZIF-8, and also conferred photothermal conversion effect. In the tumor acidic microenvironment, the acid-labile hydrazone linker of DA-DOX and ZIF-8 promptly degraded to release activated DOX. Moreover, the generated hyperthermia due to the high photothermal conversion efficiency of PDA component could accelerate drug release, and simultaneously thermally ablate tumor tissue to maximize the DOX-induced CT, which could also assist PTT to eradicate tumor cells. This study provides a promising strategy for targeted cancer PTT-CT with synergistic anti-tumor effect.


Subject(s)
Hyperthermia, Induced , Nanocomposites , Nanoparticles , Neoplasms , Zeolites , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Liberation , Humans , Indoles , Nanocomposites/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Phototherapy , Polymers , Tumor Microenvironment
11.
ACS Appl Mater Interfaces ; 13(37): 44002-44012, 2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34494817

ABSTRACT

Ferroptosis therapy (FT) based on the Fenton reaction of ferrous nanoparticles has been becoming a unique strategy for cancer treatment; however, current ferrous nanoparticles suffer from slower Fenton reaction kinetics, lower ferroptosis efficacy, and long-term toxicity, so it is urgent to construct biocompatible ferrous nanomaterials with highly efficient Fenton reaction activity for cancer FT. Inspired by single-atom catalysis and size-determined tumor penetration, we conceived an innovative strategy for constructing ultrasmall zwitterionic polypeptide-coordinated nanohybrids of PCGA@FeNP with about 6 nm by utilizing thiol/hydroxyl-iron cooperative coordination chemistry. The ultrasmall size, unsaturated ferrous coordination, and intracellular acidic pH could accelerate the Fenton reaction, thus boosting the efficacy of ferroptosis. Moreover, those coordinated nanohybrids exhibited prominent photothermia with 59.5% conversion efficiency, further accelerating the Fenton reaction and inducing a synergistic effect between FT and photothermal therapy (PTT). In vitro and in vivo GPX-4 expression ascertained that PCGA@FeNP indeed induced effective FT and synergistic FT-PTT. Remarkably, in vivo FT-PTT completely ablated 4T1 solid tumors by one treatment, presenting outstanding and synergistic antitumor efficacy via the photothermia-boosted ferroptosis and apoptosis pathways. This work supplies a practicable strategy to fabricate ultrasmall zwitterionic coordination nanohybrids for highly efficient cancer FT and FT-PTT theranostics with potential clinical transitions.


Subject(s)
Antineoplastic Agents/therapeutic use , Ferroptosis/drug effects , Metal Nanoparticles/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/radiation effects , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/radiation effects , Coordination Complexes/therapeutic use , Female , Gallic Acid/chemistry , Gallic Acid/radiation effects , Hydroxyl Radical/metabolism , Infrared Rays , Iron/chemistry , Iron/radiation effects , Iron/therapeutic use , Metal Nanoparticles/chemistry , Metal Nanoparticles/radiation effects , Mice, Inbred BALB C , Neoplasms/metabolism , Peptides/chemistry , Peptides/radiation effects , Photoacoustic Techniques , Photothermal Therapy
12.
J Mater Chem B ; 9(27): 5484-5491, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34161406

ABSTRACT

The second near infrared photoacoustic imaging (NIR-II PAI) and photothermal therapy (NIR-II PTT) have attracted wide interest in cancer theranostics because of maximum permission exposure (MPE), deep penetration, and lower scattering and background noise compared to NIR-I counterparts; however, it is imperative to develop biocompatible nanomaterials having NIR-II response. By utilizing multivalent Au-S coordination bonds, we constructed a zwitterionic polypeptide nanocomposite of PMC@AuNP with a suitable size of 48 ± 2 nm, which possessed a strong and broad absorbance at 650-1100 nm and an excellent photothermal conversion efficiency of 49.5%. In vitro biological studies demonstrated that NIR-II PTT within MPE was more effective than NIR-I PTT beyond MPE. Along with X-ray computed tomography and photothermal imaging functions, PMC@AuNP in vivo presented unique NIR-I/II PAI with 2.6-5.9 times signal enhancement compared to the contrast. By single dose and NIR-II irradiation (1064 nm, 1 W cm-2, 10 min), NIR-II PTT within MPE completely eradicated MCF-7 tumors without tissue damage and tumor recurrence within 24 days, inducing a better antitumor efficacy than NIR-I PTT beyond MPE. Importantly, this study provides an innovative method for the fabrication of biocompatible zwitterionic polypeptide nanocomposites with unique NIR-I/II PAI and NIR-II PTT attributes, thus holding great potential for precise cancer theranostics and further clinical transitions.


Subject(s)
Breast Neoplasms/therapy , Nanocomposites/chemistry , Peptides/pharmacology , Photoacoustic Techniques , Photothermal Therapy , Animals , Breast Neoplasms/pathology , Cell Survival/drug effects , Gold/chemistry , Humans , Infrared Rays , MCF-7 Cells , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Metal Nanoparticles/chemistry , Mice , Mice, Nude , Molecular Structure , Particle Size , Peptides/chemistry , Surface Properties
13.
Acta Biomater ; 123: 335-345, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33476826

ABSTRACT

Multidrug resistance (MDR) induced by the overexpression of P-glycoprotein (P-gp) transporters mainly leads to chemotherapy (CT) failure. Herein, a NIR/pH dual-sensitive charge-reversal polypeptide nanocomposite (PDA-PLC) was developed for co-delivering a nitric oxide (NO) donor and doxorubicin (DOX). Under near-infrared (NIR) irradiation, the released high-concentration of NO gas inhibited the P-gp expression to sensitize the chemotherapeutic medicine DOX and assisted photothermal therapy (PTT) to eradicate cancer cells without skin scarring. Further, the distinctive charge-reversal capacity of PDA-PLC significantly facilitated cellular uptake in the tumor acidic microenvironment (pH 6.8) and enhanced its stability in the physiological environment (pH 7.4). This DOX-loading polypeptide nanocomposite (PDA-PLC/DOX) provides an effective strategy for the PTT-NO-CT triple-combination therapy to overcome MDR STATEMENT OF SIGNIFICANCE: Multidrug resistance (MDR) has been considered to be the paramount factor of chemotherapy (CT) failure in cancer. In this work, an NIR/pH dual-sensitive charge-reversal polypeptide nanomedicine (PDA-PLC/DOX) was developed to overcome MDR through the triple combination therapy of photothermal therapy (PTT), NO gas therapy, and CT. The distinctive charge-reversal capacity of PDA-PLC/DOX significantly facilitated cellular uptake in the tumor acidic microenvironment (pH 6.8) and enhanced its stability in the physiological environment (pH 7.4), while the NIR trigger-released NO gas greatly inhibited the expression of P-gp and synergistically enhanced PTT and CT efficacy. This polypeptide nanocomposite PDA-PLC/DOX provides an effective strategy of using the PTT-NO-CT triple combination therapy with charge-reversal property to completely eradicate the MCF-7/ADR tumor.


Subject(s)
Nanocomposites , Neoplasms , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , MCF-7 Cells , Nitric Oxide , Peptides/pharmacology
14.
J Mater Chem B ; 9(6): 1478-1490, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33427844

ABSTRACT

Over the past decades, near infrared light (NIR)-sensitive photothermal agents (PTAs) that can efficiently absorb light and generate heat have been investigated worldwide for cancer photothermal therapy (PTT) and the combination treatments, which have some peculiar advantages including spatiotemporal targeting, the ability-to-reverse multidrug resistance, the immunity-stimulating function, and the synergistic effect in combination treatments. In this review, we first focus on emerging melanin-like polymers and coordination polyphenol polymer-based PTAs that hold transition potential because of their facile synthesis and good biocompatibility/biodegradability. We briefly introduce polymeric PTAs for emerging NIR-II (1000-1700 nm) PTT in deep tumors to overcome shallow penetration depth and threshold irradiation intensity of NIR-I (700-900 nm). Then we discuss polymeric PTAs for combination PTT treatments with photodynamic therapy (PDT), ferroptosis therapy (ferrotherapy), and immunotherapy, which are intensively studied for achieving anticancer synergistic effects. Finally, we discuss those polymeric PTAs for reversing cancer multidrug resistance and for mild/low-temperature PTT (43 °C ≤ T < 50 °C) in contrast to conventional high-temperature PTT (>50 °C). The polymeric PTA-based PTT and the combination treatments are still being developed in the early stage and need much more effort before potential clinical transitions and applications.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/pharmacology , Polyphenols/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Humans , Hyperthermia, Induced , Neoplasms/pathology , Photosensitizing Agents/chemistry , Polyphenols/chemistry
15.
Biomacromolecules ; 21(12): 5345-5357, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33307698

ABSTRACT

Efficient therapeuic proteins' delivery into mammalian cells and subcellular transport (e.g., fast escape from endolysosomes into cytoplasm) are two key biological barriers that need to be overcome for antigen-based immunotherapy and related biomedical applications. For those purposes, we designed a novel kind of photoresponsive polypeptide-glycosylated poly(amidoamine) (PAMAM) dendron amphiphiles (PGDAs), and their synthesis, UV-responsive self-assembly, and triggered ovalbumin (OVA) release have been fully investigated. The highly anisotropic PGDA4 with a glycosylated second-generation PAMAM dendron self-assembled into stable polypeptide vesicles (polymersomes) within 20-50 wt % water, which exhibited UV-responsive reassembly, dynamic binding with a lectin of concanavalin A, and an accelerated OVA release in vitro. Moreover, upon 365 nm UV irradiation, the self-assembled polymersomes of those glycopolypeptides were transformed into micellar aggregates in aqueous solution at pH 7.4 but disassembled completely at pH 5. The OVA-loaded polymersomes could efficiently deliver OVA into RAW264.7 cells and achieve enhanced endolysosomes escape upon UV irradiation, as revealed by flow cytometry and confocal laser scanning microscopy (CLSM). Furthermore, the enzyme-linked immunosorbent assay (ELISA) showed that the blank sugar-coated polypeptidosomes activated a high level of tumor necrosis factor α (TNF-α) of 468 pg/mL, playing a better role of immune adjuvant for activating the macrophages. Upon the UV irradiation with a dose of 3 J/cm2, the OVA-loaded polymersomes could further stimulate RAW264.7 and enhance the TNF-α level by about 45%. Consequently, this work provides a versatile platform to construct photosensitive and sugar-coated polymersomes of glycopolypeptides that have potential applications for protein delivery, immune adjuvant, and antigen-based immunotherapy.


Subject(s)
Dendrimers , Animals , Immunity , Micelles , Ovalbumin , Polyethylene Glycols
16.
J Mater Chem B ; 7(35): 5306-5319, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31411235

ABSTRACT

Combating multidrug resistance (MDR) of tumors is still challenging for clinical chemotherapy, cocktail chemotherapy (CCT), and currently widely-studied nanodrug-based treatments. Inspired by different MDR-overcoming and antitumor mechanisms of CCT and photothermal therapy (PT), a dual drug-paired polyprodrug nanoparticle (PDCN25-CDDP) was constructed to achieve the combination therapy PT-CCT for reversing MDR and combating multidrug resistant cancers. The PT-CCT treatment can greatly downregulate the P-gp expression level and achieve utmost MDR-reversal and antitumor efficacy by both a cocktail effect of CCT and a synergistic effect of CCT with PT; meanwhile, PT can inhibit the expression of heat shock protein 90 and enhance the thermosensitivity of cancer cells. Upon NIR irradiation, PDCN25-CDDPin vivo produced a selective tumor accumulation effect and relatively deep tumor penetration, as evidenced by fluorescent and photoacoustic imaging and CLSM. The mild PT-CCT treatment completely eradicated MCF-7/ADR and OVCAR-3/DDP tumors without skin damage or tumor recurrence for 30 days, exhibiting synergistic MDR-reversal and superior antitumor efficacy in vivo. Importantly, this work provides an innovative strategy for reversing MDR and combating DOX-resistant breast and CDDP-resistant ovarian cancers.


Subject(s)
Cisplatin/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Prodrugs/pharmacology , Animals , Cell Line, Tumor , Drug Therapy, Combination/methods , Female , Humans , L Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/administration & dosage , Photochemotherapy/methods , Rats, Sprague-Dawley , Theranostic Nanomedicine/methods
17.
Nano Lett ; 19(7): 4362-4370, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31199153

ABSTRACT

Multidrug resistance (MDR) of cancers that results from overexpression of a P-glycoprotein (P-gp) transporter mainly causes chemotherapy (CT) failure and hinders clinical transitions of current polypeptide nanomedicines. Herein, a novel polypeptide nanocomposite PNOC-PDA that integrates heat-sensitive NO gas delivery and photothermal conversion attributes can overcome MDR and maximize CT; meanwhile the optimized CT and intracellular high-concentration NO gas can assist a mild photothermal therapy (PTT) to eradicate cancer cells. The triple therapies produced a superior and synergistic effect on MDR-reversal and killing MCF-7/ADR in vitro, and the P-gp expression level was downregulated to 46%, as confirmed by means of MTT, Western blot, flow cytometry, and confocal laser scanning microscopy. Significantly, by using one intravenous injection of PNOC-PDA/DOX and a single near-infrared irradiation, the triple therapies of mild PTT, NO gas therapy, and CT achieved complete MCF-7/ADR tumor ablation without skin damage, scarring, and tumor recurrence within 30 days. This work provides a versatile method for the fabrication of NIR-responsive polypeptide nanocomposite with intrinsic photothermal conversion and NO-releasing attributes, opening up a new avenue for reversing MDR in tumors.


Subject(s)
Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Hyperthermia, Induced , Nanocomposites , Neoplasms, Experimental , Nitric Oxide , Peptides , Phototherapy , Animals , Humans , Infrared Rays , MCF-7 Cells , Mice , Mice, Nude , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Nitric Oxide/chemistry , Nitric Oxide/pharmacokinetics , Nitric Oxide/pharmacology , Peptides/chemistry , Peptides/pharmacology
18.
J Mater Chem B ; 7(3): 415-432, 2019 01 21.
Article in English | MEDLINE | ID: mdl-32254729

ABSTRACT

Although photothermal therapy (PT) and photothermal-chemotherapy (PT-CT) treatments have been used to achieve complete ablation of solid tumors, they are often implemented at more than 50 °C under high intensity and using a high dose of NIR irradiation, concomitantly inducing heavy skin burning, tissue damage, and ugly scarring. Moreover, the residual tumor cells at the treated site cannot be completely eradicated, resulting in tumor recurrence and metathesis. These key obstacles have prohibited PT and PT-CT treatments from transitioning to clinical use, therefore achieving traceless ablation of solid tumors without recurrence is still a challenge for real applications. To balance hyperthermia and a high drug-loading capacity in polyprodrugs to achieve mild PT-CT, we rationally designed a novel type of intracellular pH and reduction-cleavable chlorambucil prodrug and synthesized high drug-loading polydopamine-chlorambucil conjugate nanoparticles (PDCBs). The PDCBs show good photothermal properties and demonstrate intracellular pH-, reduction-cleavable, and external near infrared (NIR)-triggered drug release profiles. Polydopamine-chlorambucil conjugate nanoparticles with 40 wt% CB (PDCB40) and mild NIR irradiation could facilitate cellular internalization and subcellular trafficking, generating an excellent and synergistic antitumor effect in vitro. Pharmacokinetics and small animal fluorescent and photoacoustic imaging demonstrate that PDCB40 has a 3.6-fold longer blood circulation time compared to free CB and attained selective tumor accumulation, simultaneously inducing a 4.1-fold stronger photoacoustic signal than the control. By using one intravenous injection of PDCB40 and a single dose of mild NIR irradiation, this simple and mild PT-CT treatment achieved a non-discerned tumor on the sixth day, and traceless and complete ablation of a solid MCF-7 tumor without recurrence within 50 days, opening up a new avenue for precise cancer therapy with the potential for real applications.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Nanoparticles/chemistry , Photochemotherapy , Prodrugs/pharmacology , Stimuli Responsive Polymers/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Chlorambucil/chemical synthesis , Chlorambucil/chemistry , Chlorambucil/pharmacology , Combined Modality Therapy , Dopamine/chemical synthesis , Dopamine/chemistry , Dopamine/pharmacology , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Infrared Rays , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Particle Size , Prodrugs/chemical synthesis , Prodrugs/chemistry , Rats , Rats, Sprague-Dawley , Surface Properties , Tumor Cells, Cultured
19.
ACS Appl Mater Interfaces ; 9(37): 31576-31588, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28838236

ABSTRACT

Some of the biomedical polymer-drug conjugates are being translated into clinical trials; however, they intrinsically lack photothermal and multi-imaging capabilities, hindering them from imaging-guided precision cancer therapy and complete tumor regression. We introduce a new concept of all-in-one biopolymer-drug conjugate nanotheranostics and prepare a kind of intracellular pH-sensitive polydopamine-doxorubicin (DOX) conjugate nanoparticles (PDCNs) under mild conditions. Significantly, this strategy integrates polymeric prodrug-induced chemotherapy (CT), near-infrared (NIR) light-mediated photothermal therapy (PT), and triple modalities including DOX self-fluorescence, photothermal, and photoacoustic (PA) imaging into one conjugate nanoparticle. The PDCNs present excellent photothermal property, dual stimuli-triggered drug release behavior, and about 12.4-fold blood circulation time compared to free DOX. Small animal fluorescent imaging technique confirms that PDCNs have preferential tumor accumulation effect in vivo, giving a 12.8-fold DOX higher than the control at 12 h postinjection. Upon NIR laser irradiation (5 min, 808 nm, and 2 W·cm-2), the PDCN-mediated photothermal effect can quickly elevate the tumor over 50 °C, exhibiting good photothermal and PA imaging functions, of which the PA amplitude is 3.6-fold greater than the control. In vitro and in vivo assays persuasively verify that intravenous photothermal-CT of PDCNs produces synergistic antitumor activity compared to single PT or CT, achieving complete tumor ablation during the evaluation period.


Subject(s)
Biopolymers/chemistry , Animals , Antineoplastic Agents , Doxorubicin , Drug Liberation , Multimodal Imaging , Nanostructures , Neoplasms , Phototherapy
20.
ACS Macro Lett ; 6(3): 292-297, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-35650905

ABSTRACT

Increasing efforts are being made on controlled photopolymerization methodologies; however, the previous polymerization systems need additional photoactive initiators or catalysts. The controlled synthesis of the hyperbranched polypeptide is still challenging, and developing a photopolymerization method to prepare a hyperbranched polypeptide is urgent for constructing biodegradable polymers and biomaterials. Without addition of any initiator/catalyst, we combine the inimer (initiator + monomer) ring-opening polymerization (ROP) and photocaged chemistry to prepare hyperbranched and linear polypeptides. The photocaged Nε-(o-nitrobenzyloxycarbonyl)-l-lysine-N-carboxyanhydride possesses intrinsic photosensitivity and will be transformed into an activated AB* inimer-type α-amino acid N-carboxyanhydride (NCA) containing a primary ε-amine, which further triggers ROP to produce linear and/or hyperbranched polypeptides in one pot and at room temperature. The microstructure and topology of the resulting polypeptide were clarified by means of mass spectroscopy and various NMR techniques including 1H NMR, 1H, 1H-COSY, and quantitative 13C NMR. By tuning the UV irradiation time or intensity, this methodology can produce a linear polypeptide with a high Mw,GPC of 109 kDa and/or (hyper)branched counterparts with tunable Mw,GPC's of 1.4-73.5 kDa and degree of branching of 0.09-0.60.

SELECTION OF CITATIONS
SEARCH DETAIL
...