Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
ACS Nano ; 18(10): 7346-7362, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38416031

ABSTRACT

The microenvironment after traumatic spinal cord injury (SCI) involves complex pathological processes, including elevated oxidative stress, accumulated reactive aldehydes from lipid peroxidation, excessive immune cell infiltration, etc. Unfortunately, most of current neuroprotection therapies cannot cope with the intricate pathophysiology of SCI, leading to scant treatment efficacies. Here, we developed a facile in situ reaction-induced self-assembly method to prepare aldehyde-scavenging polypeptides (PAH)-curcumin conjugate nanoassemblies (named as PFCN) for combined neuroprotection in SCI. The prepared PFCN could release PAH and curcumin in response to oxidative and acidic SCI microenvironment. Subsequently, PFCN exhibited an effectively neuroprotective effect through scavenging toxic aldehydes as well as reactive nitrogen and oxygen species in neurons, modulating microglial M1/M2 polarization, and down-regulating the expression of inflammation-related cytokines to inhibit neuroinflammation. The intravenous administration of PFCN could significantly ameliorate the malignant microenvironment of injured spinal cord, protect the neurons, and promote the motor function recovery in the contusive SCI rat model.


Subject(s)
Curcumin , Spinal Cord Injuries , Rats , Animals , Curcumin/pharmacology , Curcumin/therapeutic use , Aldehydes/metabolism , Aldehydes/pharmacology , Rats, Sprague-Dawley , Spinal Cord Injuries/drug therapy , Spinal Cord
2.
ACS Nano ; 18(6): 4957-4971, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38288709

ABSTRACT

Vaccine technology is effective in preventing and treating diseases, including cancers and viruses. The efficiency of vaccines can be improved by increasing the dosage and frequency of injections, but it would bring an extra burden to people. Therefore, it is necessary to develop vaccine-boosting techniques with negligible side effects. Herein, we reported a cupping-inspired noninvasive suction therapy that could enhance the efficacy of cancer/SARS-CoV-2 nanovaccines. Negative pressure caused mechanical immunogenic cell death and released endogenous adjuvants. This created a subcutaneous niche that would recruit and activate antigen-presenting cells. Based on this universal central mechanism, suction therapy was successfully applied in a variety of nanovaccine models, which include prophylactic/therapeutic tumor nanovaccine, photothermal therapy induced in situ tumor nanovaccine, and SARS-CoV-2 nanovaccine. As a well-established physical therapy method, suction therapy may usher in an era of noninvasive and high-safety auxiliary strategies when combined with vaccines.


Subject(s)
Cancer Vaccines , Nanoparticles , Neoplasms , Vaccines , Humans , Nanovaccines , Suction , Neoplasms/therapy , Physical Therapy Modalities , Immunotherapy
3.
Adv Mater ; 35(15): e2203291, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36326058

ABSTRACT

Biomineralization is a normal physiological process that includes nucleation, crystal growth, phase transformation, and orientation evolution. Notably, artificially induced biomineralization in the tumor tissue has emerged as an unconventional yet promising modality for malignancy therapy. However, the modest ion-chelating capabilities of carboxyl-containing biomineralization initiators lead to a deficient blockade, thus compromising antitumor efficacy. Herein, a biomineralization-inducing nanoparticle (BINP) is developed for blockade therapy of osteosarcoma. BINP is composed of dodecylamine-poly((γ-dodecyl-l-glutamate)-co-(l-histidine))-block-poly(l-glutamate-graft-alendronate) and combines a cytomembrane-insertion moiety, a tumor-microenvironment (TME)-responsive component, and an ion-chelating motif. After intravenous injection into osteosarcoma-bearing mice, BINP responds to the acidic TME to expose the dodecyl group on the surface of the expanded nanoparticles, facilitating their cytomembrane insertion. Subsequently, the protruding bisphosphonic acid group triggers continuous ion deposition to construct a mineralized barrier around the tumor, which blocks substance exchange between the tumor and surrounding normal tissues. The BINP-mediated blockade therapy displays tumor inhibition rates of 59.3% and 52.1% for subcutaneous and orthotopic osteosarcomas, respectively, compared with the Control group. In addition, the suppression of osteoclasts by the alendronate moiety alleviates bone dissolution and further inhibits pulmonary metastases. Hence, the BINP-initiated selective biomineralization provides a promising alternative for clinical osteosarcoma therapy.


Subject(s)
Bone Neoplasms , Nanoparticles , Osteosarcoma , Animals , Mice , Biomineralization , Alendronate , Glutamic Acid , Osteosarcoma/drug therapy , Peptides , Nanoparticles/chemistry , Bone Neoplasms/drug therapy , Cell Line, Tumor , Tumor Microenvironment
4.
Biomaterials ; 289: 121794, 2022 10.
Article in English | MEDLINE | ID: mdl-36113330

ABSTRACT

As a representative of tumor immunotherapy, tumor vaccine can inhibit tumor growth by activating tumor-specific immune response, which has the advantages of relatively low toxicity and high efficiency, and has attracted much attention in recent years. However, there are still difficulties in how to effectively deliver tumor vaccines in vivo and make them work efficiently. It is a relatively mature method to load tumor specific antigens with suitable carriers to produce tumor vaccines. Here, a generally minimalist construction method of tumor nanovaccine was developed. A high-efficiency tumor nanovaccine (NV) was prepared in one step by a biomineralization-like method, which contained ovalbumin (OVA, model antigen), unmethylated cytosine-phosphate-guanine (CpG, adjuvant) and Mn-NP (carrier and adjuvant). NV not only showed good tumor preventive effect, but also could successfully inhibited tumor development and metastasis when combined with anti-PD-L1, and induced long-term immune memory effect. However, the method of screening tumor specific antigen to construct nanovaccine is cumbersome and tumors are heterogeneous. Therefore, surgically resected tumor tissue is the best source of antigens for preparing tumor vaccines. Next, based on the strong loading ability of the carrier, we designed a personalized tumor nanovaccine (PNV) using the supernatant of tumor abrasive fluid (STAF) as antigen based on the generally minimalist tumor nanovaccine construction strategy. PNV combined with anti-PD-L1 could successfully inhibit post-surgical tumor recurrence and induce strong and durable immune memory effects. This study presents a novel, general, and minimalist strategy to construct high-efficiency personalized nanovaccine, which has a wide range of potential applications in the field of tumor treatment.


Subject(s)
Cancer Vaccines , Nanoparticles , Neoplasms , Animals , Antigens, Neoplasm , Cytosine , Guanine , Humans , Immune Checkpoint Inhibitors , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Ovalbumin , Phosphates
5.
J Mater Chem B ; 10(3): 456-467, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34982090

ABSTRACT

Bacterial infections and antibiotic resistance have become a global healthcare crisis. Herein, we designed and synthesized a series of cationic amphiphilic dendrons with cationic dendrons and hydrophobic alkyl chains for potential antibacterial applications. Our results showed that the antimicrobial activities of the cationic amphiphilic dendrons were highly dependent upon the length of the hydrophobic alkyl chain, whereas the number of cationic charges was less important. Among these cationic amphiphilic dendrons, a prime candidate was identified, which possessed excellent antimicrobial activity against various pathogens (minimum inhibitory concentrations of 9, 3, and 3 µg mL-1 for Escherichia coli, Staphylococcus aureus, and methicillin-resistant Staphylococcus aureus, respectively). Scanning electron microscopy and fluorescence microscopy analyses showed that it could disrupt the integrity of a pathogen's membrane, leading to cell lysis and death. In addition, in vitro bacteria-killing kinetics showed that it had rapid bactericidal efficiency. It also had excellent antimicrobial activities against MRSA in vivo and promoted wound healing. In general, the synthesized cationic amphiphilic dendrons, which exhibited rapid and broad-spectrum bactericidal activity, may have great potential in antimicrobial applications.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Dendrimers/therapeutic use , Staphylococcal Skin Infections/drug therapy , Surface-Active Agents/therapeutic use , Animals , Anti-Bacterial Agents/chemical synthesis , Cell Membrane/drug effects , Dendrimers/chemical synthesis , Drug Resistance, Bacterial/drug effects , Escherichia coli/drug effects , Hydrophobic and Hydrophilic Interactions , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Microbial Sensitivity Tests , Molecular Structure , Rats , Staphylococcal Skin Infections/pathology , Surface-Active Agents/chemical synthesis , Wound Healing/drug effects
6.
Macromol Biosci ; 21(6): e2100039, 2021 06.
Article in English | MEDLINE | ID: mdl-33818918

ABSTRACT

Despite the encouraging clinical responses of several human cancers to immunotherapy, the efficacy of this treatment remains limited by variable objective response rates and severe systemic immune-related adverse events. To overcome these issues, injectable hydrogels have been developed as local depots that permit the sustained release of single or multiple immunotherapy agents, including traditional immunomodulatory factors, immune checkpoint blocking antibodies, and exogenous immune cells. The antitumor efficacy of immunotherapy can also be enhanced by its combination with other therapeutic approaches, including chemotherapy, radiotherapy, and phototherapy. Despite local treatment strategies, potent systemic antitumor immune responses with low systemic toxicity can be obtained, leading to significant local and abscopal tumor-killing, reduced tumor metastasis, and the prevention of tumor recurrence. This review highlights recent progress in injectable hydrogel-based local depots for tumor immunotherapy and immune-based combination therapy. Moreover, the proposed mechanisms responsible for these antitumor effects are discussed.


Subject(s)
Combined Modality Therapy/methods , Drug Delivery Systems/methods , Hydrogels/administration & dosage , Neoplasm Recurrence, Local/prevention & control , Neoplasms/therapy , Animals , Antimetabolites, Antineoplastic/pharmacology , Cytokines/pharmacology , Disease Models, Animal , Humans , Hydrogels/chemistry , Immune Checkpoint Inhibitors/pharmacology , Immunologic Factors/pharmacology , Immunotherapy/methods , Induction Chemotherapy/methods , Molecular Targeted Therapy/methods , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasms/metabolism , Neoplasms/pathology , Phototherapy/methods
7.
Adv Mater ; 33(15): e2007426, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33675268

ABSTRACT

Subcellular organelle-targeted nanoformulations for cancer theranostics are receiving increasing attention owing to their benefits of precise drug delivery, maximized therapeutic index, and reduced off-target side effects. Herein, a multichannel calcium ion (Ca2+ ) nanomodulator (CaNMCUR+CDDP ), i.e., a cisplatin (CDDP) and curcumin (CUR) co-incorporating calcium carbonate (CaCO3 ) nanoparticle, is prepared by a facile one-pot strategy in a sealed container with in situ synthesized polydopamine (PDA) as a template to enhance Ca2+ -overload-induced mitochondrial dysfunction in cancer therapy. After systemic administration, the PEGylated CaNMCUR+CDDP (PEG CaNMCUR+CDDP ) selectively accumulates in tumor tissues, enters tumor cells, and induces multilevel destruction of mitochondria by the combined effects of burst Ca2+ release, Ca2+ efflux inhibition by CUR, and chemotherapeutic CDDP, thereby observably boosting mitochondria-targeted tumor inhibition. Fluorescence imaging of CUR combined with photoacoustic imaging of PDA facilitates the visualization of the nanomodulator. The facile and practical design of this multichannel Ca2+ nanomodulator will contribute to the development of multimodal bioimaging-guided organelle-targeted cancer therapy in the future.


Subject(s)
Antineoplastic Agents/chemistry , Calcium Channel Agonists/chemistry , Cisplatin/chemistry , Curcumin/chemistry , Mitochondria/drug effects , Nanocapsules/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Calcium Carbonate/chemistry , Calcium Channel Agonists/pharmacokinetics , Cell Line, Tumor , Cell Membrane Permeability , Cisplatin/pharmacokinetics , Curcumin/pharmacokinetics , Drug Liberation , Drug Therapy, Combination , Humans , Indoles/chemistry , Mice, Nude , Polymers/chemistry , Signal Transduction
8.
ACS Appl Mater Interfaces ; 12(37): 41127-41137, 2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32808767

ABSTRACT

Weak T cell responses and immune checkpoints within tumors could be two key factors for limiting antitumor efficacy in the field of cancer immunotherapy. Thus, the combined strategy of tumor vaccines and immune checkpoint blockade has been widely studied and expected to boost antitumor immune responses. Herein, we first developed a two-barreled strategy to combine the nanovaccine with a gene-mediated PD-L1 blockade. On the one hand, polyethyleneimine (PEI) worked as a vaccine carrier to codeliver the antigen ovalbumin (OVA) and the adjuvant unmethylated cytosine-phosphate-guanine (CpG) to formulate the PEI/OVA/CpG nanovaccine through electrostatic binding, which realized both dendritic cell activation and antigen cross-presentation enhancement. On the other hand, the PD-L1 silence gene was loaded by PEI to form PEI/pshPD-L1 complexes, which were further in situ shielded by aldehyde-modified polyethylene glycol (OHC-PEG-CHO) via pH-responsive Schiff base bonds. The formed pshPD-L1@NPs could decrease PD-L1 expression on the tumor cells. However, such a combined two-barreled strategy improved feebly for tumor inhibition in comparison with monotherapy, exhibiting the antagonistic effect, which might be due to the limited T cell response enhancement in the tumor microenvironment. To solve this problem, we have further developed a three-barreled strategy to combine oral administration of l-arginine, which worked as an amplifier to induce robust T cell response enhancement, without causing the upregulation of other negative immune regulators. Superior antitumor behavior and tumor rechallenge protection were realized by the three-barreled strategy in B16F10-OVA (B16-OVA)-bearing mice. The unique three-barreled strategy we developed might offer a novel clinical therapeutic treatment.


Subject(s)
Arginine/immunology , B7-H1 Antigen/antagonists & inhibitors , Cancer Vaccines/immunology , Immunotherapy , Nanoparticles/chemistry , Animals , Arginine/chemistry , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cancer Vaccines/chemistry , Cytosine/chemistry , Cytosine/immunology , Guanine/chemistry , Guanine/immunology , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Ovalbumin/chemistry , Ovalbumin/immunology , Particle Size , Phosphates/chemistry , Phosphates/immunology , Polyethyleneimine/chemistry , Surface Properties
9.
Nanoscale ; 12(3): 1349-1355, 2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31913380

ABSTRACT

Nowadays, Fenton reaction-based chemodynamic therapy (CDT) strategies have drawn extensive attention as tumor-specific nanomedicine-based therapy. Nevertheless, current existing CDTs normally suffer from therapeutic bottlenecks such as the scavenging of hydroxyl radical (˙OH) by intracellular antioxidants and unideal therapeutic outcome of single treatment modality. Herein, we constructed novel all-in-one AFP nanoparticles (NPs) as CDT agents through a one-pot process for multifunctional nanotheranostics. The as-constructed AFP NPs could simultaneously produce ˙OH through the Fenton reaction and scavenge intracellular glutathione, functioning as self-reinforced CDT agents to achieve tumor-triggered enhanced CDT (ECDT). In addition, the AFP NPs possessed the capability of H2O2 and acid-boosted photoacoustic imaging and photothermal therapy, enabling a precise and effective tumor therapeutic outcome with minimal nonspecific damage in combination with ECDT. Our novel nanoplatform would open new perspectives on multi-functional CDT agents for accurate and non-invasive tumor theranostics.


Subject(s)
Glutathione/metabolism , Hyperthermia, Induced , Nanoparticles , Neoplasms, Experimental , Photoacoustic Techniques , Phototherapy , Animals , Cell Line, Tumor , Humans , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , Theranostic Nanomedicine
10.
ACS Appl Mater Interfaces ; 11(43): 39503-39512, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31580067

ABSTRACT

Covalent organic frameworks (COFs) have received considerable interest because of their advanced applications. However, their low dispersibility and aqueous stability are intractable issues limiting their biomedical application. To address the issue, water-dispersible nanocomposites (COF@IR783) produced through the assembly of cyanines and COFs are proposed and prepared. Therefore, a strategy of "killing three birds with one stone" is developed. First, the nanocomposites exhibit superior dispersibility and aqueous stability compared to COFs. The nanocomposites have a nanosized morphology and negative charges, which are in favor of improving the blood circulation and enhanced permeability and retention-mediated tumor-targeting delivery therapy for in vivo application. Second, the nanocomposites have enhanced photothermal therapy (PTT) ability in the near-infrared region compared to cyanines. The nanocomposites also have a photoacoustic imaging ability, which can guide the antitumor therapy in vivo. Lastly, the nanocomposites can be further used as drug-delivery carriers for loading the anticancer cis-aconityl-doxorubicin (CAD) prodrug. In comparison with individual PTT or chemotherapy, the combination of PTT and chemotherapy achieved with COF@IR783@CAD synergistically induced the death of cancer cells in vitro, and an intravenous injection of COF@IR783@CAD in mice resulted in significant tumor ablation. This work indicates that the dispersibility and aqueous stability of COFs can be appropriately overcome through a rational design and can further expand the biomedical applications of COFs.


Subject(s)
Doxorubicin , Hyperthermia, Induced , Nanocomposites , Neoplasms, Experimental/therapy , Phototherapy , Prodrugs , Animals , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacology , Humans , Mice , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Prodrugs/chemistry , Prodrugs/pharmacology , Xenograft Model Antitumor Assays
11.
Biomaterials ; 223: 119470, 2019 12.
Article in English | MEDLINE | ID: mdl-31526950

ABSTRACT

Nowadays, two-dimensional (2D) nanomaterials with many fascinating physicochemical properties have drawn extensive attention as drug delivery platforms for cancer theranostics. Nevertheless, current existing 2D nanomaterial-based drug delivery systems normally undergo the bottlenecks of hash preparation process, low drug loading content and unsatisfactory therapeutic outcome. Herein, we developed a novel nanoparticles-induced assemble strategy to construct 2D nanosheets with ultra-high curcumin loading content of 59.6 % and excellent stability in water. Furthermore, a distinct photothermal effect and multimodal imaging property after polydopamine coating could be obtained, thereby leading to precise and efficient ablation of tumor in combination of curcumin-induced chemotherapy. More importantly, the design principle of our work offers novel facile strategy to assemble metal-binding drugs into 2D nanomedicine with high drug content and well-defined shapes.


Subject(s)
Curcumin/chemistry , Nanoparticles/chemistry , Neoplasms/therapy , Phototherapy/methods , Animals , Drug Delivery Systems , HeLa Cells , Humans , Hyperthermia, Induced , Indoles/chemistry , MCF-7 Cells , Metals/chemistry , Mice , Microscopy, Confocal , Multimodal Imaging , Nanomedicine , Neoplasm Transplantation , Polymers/chemistry , Protein Binding , Theranostic Nanomedicine , Treatment Outcome , Water/chemistry
12.
Biomaterials ; 223: 119459, 2019 12.
Article in English | MEDLINE | ID: mdl-31499253

ABSTRACT

Combination of photodynamic therapy (PDT) and photothermal therapy (PTT) generally requires different components to build a composite irradiated with different excitation lights. One component photoactive agent for enhanced combination of PDT and PTT under the excitation of a single wavelength light source is more urgent in tumor phototherapy via adjusting spatial arrangement of photoactive units. Herein, porphyrin-based covalent organic framework nanoparticles (COF-366 NPs) were synthesized to control the orderly spatial arrangement of the photoactive building units and firstly used for antitumor therapy in vivo. COF-366 NPs provide the simultaneous therapy of PDT and PTT under a single wavelength light source with the monitoring of photoacoustic (PA) imaging, which makes the operation simpler and more convenient. COF-366 NPs had achieved good phototherapy effect even in the face of large tumors. The prepared multifunctional COF-366 NPs open up a new avenue to phototherapeutic materials and expand the application range of covalent organic framework.


Subject(s)
Nanoparticles/chemistry , Neoplasms/therapy , Photoacoustic Techniques , Phototherapy , Porphyrins/chemistry , Animals , Benzofurans/chemistry , Cell Line, Tumor , Combined Modality Therapy , Female , Hemolysis , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/therapy , Organic Chemicals , Rabbits , Reactive Oxygen Species/chemistry
13.
Adv Mater ; 31(40): e1902885, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31423690

ABSTRACT

Activatable theranostic agents that can be activated by tumor microenvironment possess higher specificity and sensitivity. Here, activatable nanozyme-mediated 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) loaded ABTS@MIL-100/poly(vinylpyrrolidine) (AMP) nanoreactors (NRs) are developed for imaging-guided combined tumor therapy. The as-constructed AMP NRs can be specifically activated by the tumor microenvironment through a nanozyme-mediated "two-step rocket-launching-like" process to turn on its photoacoustic imaging signal and photothermal therapy (PTT) function. In addition, simultaneously producing hydroxyl radicals in response to the high H2 O2 level of the tumor microenvironment and disrupting intracellular glutathione (GSH) endows the AMP NRs with the ability of enhanced chemodynamic therapy (ECDT), thereby leading to more efficient therapeutic outcome in combination with tumor-triggered PTT. More importantly, the H2 O2 -activated and acid-enhanced properties enable the AMP NRs to be specific to tumors, leaving the normal tissues unharmed. These remarkable features of AMP NRs may open a new avenue to explore nanozyme-involved nanoreactors for intelligent, accurate, and noninvasive cancer theranostics.


Subject(s)
Biomimetic Materials/chemistry , Peroxidases/metabolism , Photoacoustic Techniques/methods , Phototherapy/methods , Theranostic Nanomedicine/methods , Tumor Microenvironment , Animals , Benzothiazoles/chemistry , Combined Modality Therapy , Mice , Organometallic Compounds/chemistry , Povidone/chemistry , Sulfonic Acids/chemistry
14.
Acta Biomater ; 92: 229-240, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31100462

ABSTRACT

Effective systemic therapy is highly desired for the treatment of hepatocellular carcinoma (HCC). In this study, a combination of nanoparticles of poly(L-glutamic acid)-graft-methoxy poly(ethylene glycol)/combretastatin A4 sodium salt (CA4-NPs) plus sorafenib is developed for the cooperative systemic treatment of HCC. The CA4-NPs leads to the disruption of established tumor blood vessels and extensive tumor necrosis, however, inducing increased expression of VEGF-A and angiogenesis. Sorafenib reduces the VEGF-A induced angiogenesis and further inhibits tumor proliferation, cooperating with the CA4-NPs. A significant decrease in tumor volume and prolonged survival time are observed in the combination group of CA4-NPs plus sorafenib compared with CA4-NPs or sorafenib monotherapy in subcutaneous and orthotopic H22 hepatic tumor models. Seventy-one percent of the mice are alive without residual tumor at 96 days post tumor inoculation for the subcutaneous models treated with CA4-NPs 30 or 35 mg·kg-1 plus sorafenib 30 mg·kg-1. Our findings suggest that co-administration of sorafenib and CA4-NPs possesses significant antitumor efficacy for HCC treatment. STATEMENT OF SIGNIFICANCE: Effective systemic therapy is highly desired for the treatment of hepatocellular carcinoma (HCC). Herein, we demonstrate that a combination of nanoparticles of poly(L-glutamic acid)-graft-methoxy poly(ethylene glycol)/combretastatin A4 sodium salt (CA4-NPs) plus sorafenib is a promising synergistic approach for systemic treatment of HCC. The CA4-NPs leads to the disruption of established tumor blood vessels and extensive tumor necrosis, however, inducing increased expression of VEGF-A and angiogenesis. Sorafenib reduces the VEGF-A induced angiogenesis and further inhibits tumor proliferation, cooperating with the CA4-NPs.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Nanoparticles/chemistry , Sorafenib/administration & dosage , Sorafenib/therapeutic use , Stilbenes/administration & dosage , Stilbenes/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/pathology , Cell Death/drug effects , Cell Survival/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ki-67 Antigen/metabolism , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Stilbenes/chemical synthesis , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
15.
Theranostics ; 9(5): 1426-1452, 2019.
Article in English | MEDLINE | ID: mdl-30867842

ABSTRACT

Hepatoma is one of the most severe malignancies usually with poor prognosis, and many patients are insensitive to the existing therapeutic agents, including the drugs for chemotherapy and molecular targeted therapy. Currently, researchers are committed to developing the advanced formulations with controlled drug delivery to improve the efficacy of hepatoma therapy. Numerous inoculated, induced, and genetically engineered hepatoma rodent models are now available for formulation screening. However, animal models of hepatoma cannot accurately represent human hepatoma in terms of histological characteristics, metastatic pathways, and post-treatment responses. Therefore, advanced animal hepatoma models with comparable pathogenesis and pathological features are in urgent need in the further studies. Moreover, the development of nanomedicines has renewed hope for chemotherapy and molecular targeted therapy of advanced hepatoma. As one kind of advanced formulations, the polymer-based nanoformulated drugs have many advantages over the traditional ones, such as improved tumor selectivity and treatment efficacy, and reduced systemic side effects. In this article, the construction of rodent hepatoma model and much information about the current development of polymer nanomedicines were reviewed in order to provide a basis for the development of advanced formulations with clinical therapeutic potential for hepatoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/therapy , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Nanostructures/therapeutic use , Polymers/therapeutic use , Animals , Drug Therapy/methods , Molecular Targeted Therapy/methods , Rodentia
16.
Biomaterials ; 197: 284-293, 2019 03.
Article in English | MEDLINE | ID: mdl-30677557

ABSTRACT

Despite multifunctional nanoparticles using for photothermal therapy can efficiently kill cancer cells, their further application is still hindered by the intrinsic high uptake in the reticuloendothelial system (RES) organs, causing the slow elimination from the body and potential toxicity to the body. Therefore, it is ideal to develop multifunctional nanoparticles which process the ability to effectively accumulate in tumors, while the nanoparticles can be rapidly excreted from the body via renal clearance after effective treatment. Herein, we report the multifunctional nanoparticles (FeTNPs) based on the coordination interaction of phenolic group and metal iron, which are composed of ferric iron, tannic acid (TA) and poly (glutamic acid)-graft-methoxypoly (ethylene glycol) (PLG-g-mPEG). FeTNPs exhibit the following highlighted features: (1) The effective accumulation in the tumor tissue is achieved based on EPR effect. (2) The dual photoacoustic (PA)/magnetic resonance (MR) imaging capacity can provide guidance for the photothermal therapy (PTT). (3) FeTNPs can be dynamically disassembled by deferoxamine mesylate (DFO) to accelerate elimination of the nanoparticles, thus reducing the potential toxicity for the body. The DFO triggered dynamic disassembling strategy may open a new avenue to overcome the dilemma between EPR effect and renal clearance.


Subject(s)
Breast Neoplasms/therapy , Multifunctional Nanoparticles/therapeutic use , Animals , Breast Neoplasms/diagnostic imaging , Female , Humans , Hyperthermia, Induced , MCF-7 Cells , Magnetic Resonance Imaging , Mice, Inbred BALB C , Mice, Nude , Multifunctional Nanoparticles/chemistry , Photoacoustic Techniques , Rats, Sprague-Dawley , Theranostic Nanomedicine
17.
ACS Appl Mater Interfaces ; 10(48): 41035-41045, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30403471

ABSTRACT

Imaging-guided therapy has considerable potential in tumor treatment. Different treatments have been integrated to realize combinational tumor therapy with improved therapeutic efficiency. Herein, the conventional metal-organic framework (MOF) MIL-100 is utilized to load curcumin with excellent encapsulation capacity. Polydopamine-modified hyaluronic acid (HA-PDA) is coated on the MIL-100 surface to construct engineering MOF nanoparticles (MCH NPs). The HA-PDA coating not only improves the dispersibility and stability of NPs but also introduces a tumor-targeting ability to this nanosystem. A two-stage augmented photothermal conversion capability is introduced to this nanosystem by encapsulating curcumin in MIL-100 pores and then coating HA-PDA on the surface, which confer the MCH NPs with strong photothermal conversional efficiency. After being intravenously injected into xenograft HeLa tumor-bearing mice, MCH NPs prefer to accumulate at the tumor site and achieve photoacoustic imaging-guided chemo-/photothermal combinational tumor therapy, generating nearly complete tumor ablation. Engineering MOFs is an efficient platform for imaging-guided combinational tumor therapy, as confirmed by in vitro and in vivo evaluations.


Subject(s)
Doxorubicin , Hyperthermia, Induced , Metal-Organic Frameworks , Nanoparticles , Neoplasms, Experimental , Photoacoustic Techniques , Phototherapy , A549 Cells , Animals , CHO Cells , Cricetulus , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , HeLa Cells , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacokinetics , Hyaluronic Acid/pharmacology , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacokinetics , Metal-Organic Frameworks/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Polymers/chemistry , Polymers/pharmacokinetics , Polymers/pharmacology , Xenograft Model Antitumor Assays
18.
Macromol Biosci ; 18(12): e1800240, 2018 12.
Article in English | MEDLINE | ID: mdl-30303620

ABSTRACT

Different strategies of chemotherapeutics-based combination cancer therapy have presented enhanced antitumor efficiency and are widely used in clinical cancer treatments. However, several drawbacks of the systems for systemic administration, including low drug accumulation at tumor sites and significant systemic side effects limit their efficacy and application in the clinic. Local drug co-delivery systems based on injectable hydrogels may have considerable advantages, such as a facile drug-delivery procedure, targeted delivery of antitumor agents to tumor sites in a sustained manner, and markedly reduced systemic toxicities. Thus, in recent years, these systems have received increasing attention and consequently various injectable hydrogels have been tested as platforms for local chemotherapeutics-based combination antitumor therapy. In this review, the focus is on recent advances in injectable hydrogel-based drug co-delivery systems for local combination antitumor therapy, including multiple chemotherapeutics combination therapy, chemo-immunotherapy, chemo-radiotherapy, and hyperthermia-chemotherapy. Moreover, the rationale and preparation of local co-delivery systems are summarized and discussed.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Delivery Systems/methods , Hydrogels/chemistry , Neoplasms/therapy , Animals , Antineoplastic Agents/pharmacokinetics , Combined Modality Therapy , Cross-Linking Reagents/chemistry , Gamma Rays/therapeutic use , Humans , Hyperthermia, Induced/methods , Immunotherapy/methods , Injections , Mice , Neoplasms/metabolism , Neoplasms/pathology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Xenograft Model Antitumor Assays
19.
Acta Biomater ; 73: 388-399, 2018 06.
Article in English | MEDLINE | ID: mdl-29694920

ABSTRACT

Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown significant activity against P-glycoprotein (P-gp) mediated multi drug resistant cancer cells. However, because of the poor aqueous solubility and high toxicity, PPT cannot be used in clinical cancer therapy. In order to enhance the efficiency and reduce side effect of PPT, a polypeptide based PPT conjugate PLG-g-mPEG-PPT was developed and used for the treatment of multi drug resistant breast cancer. The PLG-g-mPEG-PPT was prepared by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol) (PLG-g-mPEG) via ester bonds. The PPT conjugates self-assembled into nanoparticles with average sizes about 100 nm in aqueous solution. Western blotting assay showed that the PLG-g-mPEG-PPT could effectively inhibit the expression of P-gp in the multiple drug resistant MCF-7/ADR cells. In vitro cytotoxicity assay indicated that the resistance index (RI) values of PLG-g-mPEG-PPT on different drug-resistant cancer cell lines exhibited 57-270 folds reduction than of traditional microtubule inhibitor chemotherapeutic drug PTX or DTX. Hemolysis assay demonstrated that the conjugation greatly decreased the hemolytic activity of free PPT. Maximum tolerated dose (MTD) of PLG-g-mPEG-PPT increased greatly (13.3 folds) as compared to that of free PPT. In vivo study showed that the PLG-g-mPEG-PPT conjugate remarkably enhanced the antitumor efficacy against MCF-7/ADR xenograft tumors with a tumor suppression rate (TSR) of 82.5%, displayed significantly improved anticancer efficacy as compared to free PPT (TSR = 37.1%) with minimal toxicity when both of the two formulations were used in MTD. STATEMENT OF SIGNIFICANCE: The development of multiple drug resistance (MDR) of cancer cells is the main cause of chemotherapy failure. The over-expression of P-glycoprotein (P-gp) has been recognized to be the most important cause of MDR in cancer. Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown strong activity against P-gp mediated multidrug resistant cancer cells by simultaneously inhibiting the over-expression of P-gp and the growth of cancer cells. However, PPT can not be used in clinical cancer treatment due to its poor aqueous solubility and high toxicity. Herein, we developed a polypeptide based PPT conjugate PLG-g-mPEG-PPT by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol). The PLG-g-mPEG-PPT shows significantly decreased hemolytic activity, greatly improved maximum tolerated dose and remarkably enhanced antitumor efficacy against MCF-7/ADR xenograft tumors as compared to free PPT.


Subject(s)
Breast Neoplasms/drug therapy , Drug Delivery Systems , Drug Resistance, Neoplasm , Peptides/chemistry , Podophyllotoxin/chemistry , A549 Cells , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , Cell Line, Tumor , Drug Evaluation, Preclinical , Drug Resistance, Multiple/drug effects , Female , Hemolysis , Humans , MCF-7 Cells , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Neoplasm Transplantation , Polyethylene Glycols/chemistry , Polyglutamic Acid/chemistry
20.
Acta Biomater ; 47: 124-134, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27721008

ABSTRACT

Near infrared light responsive nanoparticles can transfer the absorbed NIR optical energy into heat, offering a desirable platform for photoacoustic (PA) imaging guided photothermal therapy (PTT) of tumor. However, a key issue in exploiting this platform is to achieve optimal combination of PA imaging and PTT therapy in single nanoparticle. Here, we demonstrate that the biodegradable polydopamine nanoparticles (PDAs) are excellent PA imaging agent and highly efficient for PTT therapy, thus enabling the optimal combination of PA imaging and PTT therapy in single nanoparticle. Upon modification with arginine-glycine-aspartic-cysteine acid (RGDC) peptide, PDA-RGDC can successfully target tumor site. Moreover, PDA-RGDC can load a chemotherapy drug, doxorubicin (DOX), whose release can be triggered by near-infrared (NIR) light and pH dual-stimuli. The in vitro and in vivo experiments show that this platform can deliver anti-cancer drugs to target cells, release them intracellular upon NIR irradiation, and effectively eliminate tumors through chemo-photothermal synergistic therapeutic effect. Our results offer a way to harness PDA-based theranostic agents to achieve PA imaging-guided cancer therapy. STATEMENT OF SIGNIFICANCE: NIR-light adsorbed nanoparticles combing the advantage of PAI and PTT (TNP-PAI/PTT) are expected to play a significant role in the dawning era of personalized medicine. However, the reported Au-, Ag-, Cu-, Co-, and other metal based, carbon-based TNP-PAI/PTT suffer from complex multicomponent system and poor biocompatibility and biodegradability. To overcome this limitation, biocompatible polydopamine nanoparticles (PDAs), structurally similar to naturally occurring melanin, were designed as both PA imaging contrast agent and a chemo-thermotherapy therapy agent for tumor. RGDC peptide modified PDAs can improve the PA imaging and PTT efficiency and specific targeted deliver doxorubicin (DOX) to perinuclear region of tumor cells. Our finding may help the development of PDA-based nanoplatform for PA imaging-directed synergistic therapy of tumor in clinic.


Subject(s)
Hyperthermia, Induced , Indoles/chemistry , Nanoparticles/chemistry , Neoplasms/therapy , Photoacoustic Techniques/methods , Phototherapy , Polymers/chemistry , Animals , Biocompatible Materials/pharmacology , Combined Modality Therapy , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Liberation , HeLa Cells , Humans , Indoles/chemical synthesis , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Polymers/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL