Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Chemistry ; : e202400870, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38736169

ABSTRACT

Alzheimer's disease (AD) is an age-related neurodegenerative disease with amyloid-ß (Aß) deposition as the main pathological feature. It's an important challenge to find new ways to clear Aß from the brain. The soluble amyloid precursor protein α (sAPPα) is a neuroprotective protein and can attenuate neuronal damage, including toxic Aß. However, the regulatory role of sAPPα in non-neuronal cells, such as microglia, is less reported and controversial. Here, we showed that sAPPα promoted the phagocytosis and degradation of Aß in both normal and damaged microglia. Moreover, the function of damaged microglia was improved by the sAPPα through normalizing mitochondrial function. Furthermore, the results of molecular docking simulation showed that sAPPα had a good affinity with Aß. We preliminarily reveal that sAPPα is similar to antibodies and can participate in the regulation of microglia phagocytosis and degradation of Aß after binding to Aß. sAPPα is expected to be a mild and safe peptide drug or drug carrier for AD.

2.
Proc Natl Acad Sci U S A ; 117(52): 32962-32969, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33318219

ABSTRACT

Clinical investigations have shown that a nonimmunogenic "cold" tumor is usually accompanied by few immunopositive cells and more immunosuppressive cells in the tumor microenvironment (TME), which is still the bottleneck of immune activation. Here, a fluorine assembly nanocluster was explored to break the shackles of immunosuppression, reawaken the immune system, and turn the cold tumor "hot." Once under laser irradiation, FS@PMPt produces sufficient reactive oxygen species (ROS) to fracture the ROS-sensitive linker, thus releasing the cisplatin conjugated PMPt to penetrate into the tumors and kill the regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). Meanwhile, ROS will induce potent immunogenic cell death (ICD) and further promote the accumulation of dendritic cells (DCs) and T cells, therefore not only increasing the infiltration of immunopositive cells from the outside but also reducing the immunosuppressive cells from the inside to break through the bottleneck of immune activation. The FS@PMPt nanocluster regulates the immune process in TME from negative to positive, from shallow to deep, to turn the cold tumor into a hot tumor and provoke a robust antitumor immune response.


Subject(s)
Antineoplastic Agents/chemical synthesis , Fluorine/chemistry , Immunologic Factors/chemical synthesis , Nanoconjugates/chemistry , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dendrimers/chemistry , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Immunologic Factors/pharmacology , Mice , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Platinum/chemistry , Reactive Oxygen Species/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology
3.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 52(3): 267-278, 2023 Jun 25.
Article in English, Zh | MEDLINE | ID: mdl-37476938

ABSTRACT

NK cell immunotherapy is a promising antitumor therapeutic modality after the development of T cell immunotherapy. Structural modification of NK cells with biomaterials may provide a precise, efficient, and low-cost strategy to enhance NK cell immunotherapy. The biomaterial modification of NK cells can be divided into two strategies: surface engineering with biomaterials and intracellular modification. The surface engineering strategies include hydrophobic interaction of lipids, receptor-ligand interaction between membrane proteins, covalent binding to amino acid residues, click reaction and electrostatic interaction. The intracellular modification strategies are based on manipulation by nanotechnology using membranous materials from various sources of NK cells (such as exosome, vesicle and cytomembranes). Finally, the biomaterials-based strategies regulate the recruitment, recognition and cytotoxicity of NK cells in the solid tumor site in situ to boost the activity of NK cells in the tumor. This article reviews the recent research progress in enhancing NK cell therapy based on biomaterial modification, to provide a reference for further researches on engineering NK cell therapy with biomaterials.


Subject(s)
Biocompatible Materials , Neoplasms , Humans , Biocompatible Materials/metabolism , Immunotherapy , Killer Cells, Natural/metabolism , Immunotherapy, Adoptive , Neoplasms/therapy
4.
Nano Lett ; 20(7): 4842-4849, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32578994

ABSTRACT

Development of T1/T2 dual-mode MRI contrast agents that can also treat cancer is an attractive prospect for personalized precision medicine. Unfortunately, conventional contrast agents can suffer from toxicity and lack any ability to treat cancer. An all-iron T1/T2 MR imaging agent with photothermal and drug delivery capability would overcome these issues. Here, an avocado-like Fe3+/Fe2O3 composed T1-T2 dual-mode contrast agent based on Fe-TA coordination network (CNMN) is developed. This material possesses suitable longitudinal and transverse relaxation coefficients. Moreover, the strong heat generation property of Fe-TA endows CNMN with the capability to act as a potent photothermal agent. Furthermore, CNMN can also act as an effective delivery platform for the chemotherapeutic drug doxorubicin (DOX) to achieve high effective chemo-photothermal combination therapy. The work demonstrates reliable T1-T2 MRI-guided chemo-photothermal therapy for safe and effective clinical application.


Subject(s)
Nanoparticles , Neoplasms , Persea , Doxorubicin/therapeutic use , Iron , Magnetic Resonance Imaging , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Phototherapy , Precision Medicine
5.
Nanomedicine ; 23: 102071, 2020 01.
Article in English | MEDLINE | ID: mdl-31442581

ABSTRACT

Developing various kinds of nanoplatforms with integrated diagnostic and therapeutic functions would be significant for imaging-guided precision treatment of cancer. However, it is still a challenge to organically integrate therapeutic and imaging components into a single nano-system rather than simply mixing. Herein, an iron-gallic acid network-based nanoparticle (Fe-GA@PEG-PLGA) was designed for magnetic resonance imaging (MRI)-guided chemo-photothermal synergistic therapy of tumors. The tumor spatial location and size information can be accurately achieved due to T1 MRI based on Fe3+ coordination with GA in Fe-GA network. Furthermore, the nanoparticle exhibited extraordinary photostability and photothermal therapy capacity exceeded 42 °C within 100 s under 808 nm laser irradiation. Meanwhile, the Fe-GA polymeric network can be disassembled in tumor acidic environment and the released drug GA can induce apoptosis. This study demonstrated that the Fe-GA network-based nanoparticle is a promising diagnostic and therapeutic agent for theranostic application and further clinic translation.


Subject(s)
Magnetic Resonance Imaging , Nanostructures , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/therapy , Phototherapy , Animals , Gallic Acid/chemistry , Gallic Acid/pharmacology , Humans , Hydrogen-Ion Concentration , Hyperthermia, Induced , Iron/chemistry , Iron/pharmacology , Mice , Nanostructures/chemistry , Nanostructures/therapeutic use
6.
Nano Lett ; 19(7): 4334-4342, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31179709

ABSTRACT

Nanozymes as artificial enzymes that mimicked natural enzyme-like activities have received great attention in cancer diagnosis and therapy. Biomimetic nanozymes require more consideration regarding complicated tumor microenvironments to mimic biological enzymes, thus achieving superior nanozyme activity in vivo. Here we report a biomimetic hybrid nanozyme (named rMGB) which integrates natural enzyme glucose oxidase (GOx) with nanozyme manganese dioxide (MnO2) by mutual promotion for maximizing the enzymatic activity of MnO2 and GOx. Under hypoxia environment, we observed that MnO2 could react with endogenous H2O2 to produce O2 for enhancing the catalytic efficiency of GOx for starvation therapy. Meanwhile, we confirmed that glucose oxidation generated gluconic acid and further improved the catalytic efficiency of MnO2 subsequently. The biochemical reaction cycle, consisting of MnO2, O2, GOx, and H+, was triggered by the tumor microenvironment and accelerated each other so as to achieve self-supplied H+ and accelerate O2 generation, enhancing the starvation therapy, alleviating tumor hypoxia and accelerating the reactive oxygen species generation in photodynamic therapy. This biomimetic hybrid nanozyme would further facilitate the development of biological nanozymes for cancer treatment.


Subject(s)
Biomimetic Materials , Glucose Oxidase , Manganese Compounds , Nanostructures , Neoplasms, Experimental , Oxides , Oxygen/metabolism , Photochemotherapy , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , Glucose Oxidase/chemistry , Glucose Oxidase/pharmacology , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Mice , Nanostructures/chemistry , Nanostructures/therapeutic use , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oxides/chemistry , Oxides/pharmacology , Reactive Oxygen Species/metabolism
7.
Small ; 15(17): e1900212, 2019 04.
Article in English | MEDLINE | ID: mdl-30941900

ABSTRACT

A multimodal cancer therapeutic nanoplatform is reported. It demonstrates a promising approach to synergistically regulating the tumor microenvironment. The combination of intracellular reactive oxygen species (ROS) generated by irradiation of photosensitizer and endoplasmic reticulum (ER) stress induced by 2-deoxy-glucose (2-DG) has a profound effect on necrotic or apoptotic cell death. Especially, targeting metabolic pathway by 2-DG is a promising strategy to promote the effect of photodynamic therapy and chemotherapy. The nanoplatform can readily release its cargoes inside cancer cells and combines the advantages of ROS-sensitive releasing chemotherapeutic drugs, upregulating apoptosis pathways under ER stress, light-induced generation of cytotoxic ROS, achieving tumor accumulation, and in vivo fluorescence imaging capability. This work highlights the importance of considering multiple intracellular stresses as design parameters for nanoscale functional materials in cell biology, immune response, as well as medical treatments of cancer, Alzheimer's disease, etc.


Subject(s)
Antineoplastic Agents/pharmacology , Deoxyglucose/pharmacology , Endoplasmic Reticulum Stress , Light , Tumor Microenvironment/drug effects , Apoptosis , Combined Modality Therapy , Humans , Kinetics , MCF-7 Cells , Nanomedicine , Necrosis , Phagocytosis , Photochemotherapy , Photosensitizing Agents/pharmacology , Reactive Oxygen Species
8.
Angew Chem Int Ed Engl ; 58(36): 12661-12666, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31243832

ABSTRACT

A photosensitizing monofunctional Pt complex, Pt-BDPA, was prepared with a BODIPY chromophore. Apart from its DNA binding ability, this complex displays emission at ca. 578 nm and a singlet oxygen quantum yield of 0.133. Confocal imaging revealed that this complex was sequestered in lysosomes via endocytosis in the dark, preventing its access to the nucleus. Profiting from its photoinduced ROS generation ability, this complex undergoes lysosomal escape to access the nucleus upon photoirradiation. The photoinduced ROS still cause a drop in intracellular GSH, favoring the stability of Pt-BDPA and contributing to its nuclear DNA accessibility. This complex displayed distinct cytotoxicity to all tested tumor cell lines upon photoirradiation, and the IC50 values were ca. 3-6 µm, which are distinctly lower than those found with only dark incubation (IC50 >50 µm). These results are consistent with photoactivated lysosomal escape of this photosensitizing Pt complex to access the nucleus.

9.
Nanomedicine ; 14(7): 2283-2294, 2018 10.
Article in English | MEDLINE | ID: mdl-29981460

ABSTRACT

Tumor angiogenesis is a key step in the process of tumor development, and antitumor angiogenesis has a profound influence on tumor growth. Herein we report a dual-function drug delivery system comprising a Near-infrared (NIR) dye and an anti-angiogenic drug within liposomes (Lip-IR780-Sunitinib) for enhanced antitumor therapy. The hydrophobic NIR dye IR780 was loaded into the liposome phospholipid bilayer, and the bilayer would be disrupted by laser irradiation so that anti-angiogenic drug sunitinib release would be activated remotely at the tumor site. The released hydrophilic sunitinib could potentially target multiple VEGF receptors on the tumor endothelial cell surface to inhibit angiogenesis. Meanwhile, IR780-loaded liposomes kill the cancer cells by photothermal therapy. Lip-IR780-Sunitinib exhibited enhanced anti-tumor and anti-angiogenic effects in vitro and in vivo. This system facilitates easy and controlled release of cargos to achieve anti-tumor angiogenesis and photothermal therapy.


Subject(s)
Breast Neoplasms/therapy , Hyperthermia, Induced , Indoles/chemistry , Liposomes/administration & dosage , Neovascularization, Pathologic/therapy , Phototherapy , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis , Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Combined Modality Therapy , Drug Carriers/chemistry , Drug Delivery Systems , Female , Humans , Indoles/administration & dosage , Lasers , Liposomes/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/pathology , Spectroscopy, Near-Infrared , Sunitinib/chemistry , Sunitinib/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Nano Lett ; 17(2): 733-739, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28079384

ABSTRACT

A glucose-responsive closed-loop insulin delivery system mimicking pancreas activity without long-term side effect has the potential to improve diabetic patients' health and quality of life. Here, we developed a novel glucose-responsive insulin delivery device using a painless microneedle-array patch containing insulin-loaded vesicles. Formed by self-assembly of hypoxia and H2O2 dual-sensitive diblock copolymer, the glucose-responsive polymersome-based vesicles (d-GRPs) can disassociate and subsequently release insulin triggered by H2O2 and hypoxia generated during glucose oxidation catalyzed by glucose specific enzyme. Moreover, the d-GRPs were able to eliminate the excess H2O2, which may lead to free radical-induced damage to skin tissue during the long-term usage and reduce the activity of GOx. In vivo experiments indicated that this smart insulin patch could efficiently regulate the blood glucose in the chemically induced type 1 diabetic mice for 10 h.


Subject(s)
Drug Carriers/chemistry , Glucose/metabolism , Hydrogen Peroxide/metabolism , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Animals , Blood Glucose/metabolism , Cell Hypoxia , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Drug Liberation , Glucose Oxidase/metabolism , HeLa Cells , Humans , Hypoglycemic Agents/chemistry , Insulin/chemistry , Male , Mice, Inbred C57BL , Nitroimidazoles/chemistry , Oxidation-Reduction , Peptides/chemistry , Polyethylene Glycols/chemistry
11.
Acta Pharmacol Sin ; 38(6): 764-781, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28552910

ABSTRACT

Conjugated polymer nanomaterials (CPNs), as optically and electronically active materials, hold promise for biomedical imaging and drug delivery applications. This review highlights the recent advances in the utilization of CPNs in theranostics. Specifically, CPN-based in vivo imaging techniques, including near-infrared (NIR) imaging, two-photon (TP) imaging, photoacoustic (PA) imaging, and multimodal (MM) imaging, are introduced. Then, CPN-based photodynamic therapy (PDT) and photothermal therapy (PTT) are surveyed. A variety of stimuli-responsive CPN systems for drug delivery are also summarized, and the promising trends and translational challenges are discussed.


Subject(s)
Drug Delivery Systems , Nanostructures/chemistry , Polymers/chemistry , Theranostic Nanomedicine , Photochemotherapy
12.
Angew Chem Int Ed Engl ; 56(10): 2588-2593, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28140504

ABSTRACT

Anaerobic bacteria, such as Clostridium and Salmonella, can selectively invade and colonize in tumor hypoxic regions (THRs) and deliver therapeutic products to destroy cancer cells. Herein, we present an anaerobe nanovesicle mimic that can not only be activated in THRs but also induce hypoxia in tumors by themselves. Moreover, inspired by the oxygen metabolism of anaerobes, we construct a light-induced hypoxia-responsive modality to promote dissociation of vehicles and activation of bioreductive prodrugs simultaneously. In vitro and in vivo experiments indicate that this anaerobe-inspired nanovesicle can efficiently induce apoptotic cell death and significantly inhibit tumor growth. Our work provides a new strategy for engineering stimuli-responsive drug delivery systems in a bioinspired and synergistic fashion.


Subject(s)
Antineoplastic Agents/pharmacology , Clostridium/chemistry , Hypoxia/metabolism , Nanoparticles/chemistry , Prodrugs/pharmacology , Salmonella/chemistry , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , Clostridium/metabolism , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Delivery Systems , Drug Screening Assays, Antitumor , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Prodrugs/chemistry , Salmonella/metabolism , Tirapazamine/chemistry , Tirapazamine/pharmacology
13.
Natl Sci Rev ; 8(9): nwaa286, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34691728

ABSTRACT

Synthetic lethality was proposed nearly a century ago by geneticists and recently applied to develop precision anti-cancer therapies. To exploit the synthetic lethality concept in the design of chemical anti-cancer agents, we developed a bio-orthogonally catalyzed lethality (BCL) strategy to generate targeting anti-tumor metallodrugs both in vitro and in vivo. Metallodrug Ru-rhein was generated from two non-toxic species Ru-N3 and rhein-alkyne via exclusive endogenous copper-catalyzed azide alkyne cycloaddition (CuAAC) reaction without the need of an external copper catalyst. The non-toxic species Ru-arene complex Ru-N3 and rhein-alkyne were designed to perform this strategy, and the mitochondrial targeting product Ru-rhein was generated in high yield (>83%) and showed high anti-tumor efficacy in vitro. This BCL strategy achieved a remarkable tumor suppression effect on the tumor-bearing mice models. It is interesting that the combination of metal-arene complexes with rhein via CuAAC reaction could transform two non-toxic species into a targeting anti-cancer metallodrug both in vitro and in vivo, while the product Ru-rhein was non-toxic towards normal cells. This is the first example that exclusive endogenous copper was used to generate metal-based anti-cancer drugs for cancer treatment. The anti-cancer mechanism of Ru-rhein was studied and autophagy was induced by increased reactive oxygen species and mitochondrial damage. The generality of this BCL strategy was also studied and it could be extended to other metal complexes such as Os-arene and Ir-arene complexes. Compared with the traditional methods for cancer treatment, this work presented a new approach to generating targeting metallodrugs in vivo via the BCL strategy from non-toxic species in metal-based chemotherapy.

14.
Sci Adv ; 6(20): eaaz9240, 2020 05.
Article in English | MEDLINE | ID: mdl-32440550

ABSTRACT

Anti-programmed cell death 1 ligand 1 (PD-L1) therapy is extraordinarily effective in select patients with cancer. However, insufficient lymphocytic infiltration, weak T cell-induced inflammation, and immunosuppressive cell accumulation in the tumor microenvironment (TME) may greatly diminish the efficacy. Here, we report development of the FX@HP nanocomplex composed of fluorinated polymerized CXCR4 antagonism (FX) and paclitaxel-loaded human serum albumin (HP) for pulmonary delivery of anti-PD-L1 small interfering RNA (siPD-L1) to treat orthotopic lung tumors. FX@HP induced T cell infiltration, increased expression of calreticulin on tumor cells, and reduced the myeloid-derived suppressor cells/regulatory T cells in the TME, thereby acting synergistically with siPD-L1 for effective immunotherapy. Our work suggests that the CXCR4-inhibiting nanocomplex decreases tumor fibrosis, facilitates T cell infiltration and relieves immunosuppression to modulate the immune process to improve the objective response rate of anti-PD-L1 immunotherapy.


Subject(s)
B7-H1 Antigen , Lung Neoplasms , Humans , Immunotherapy , Lung Neoplasms/drug therapy , Receptors, CXCR4 , Signal Transduction , Tumor Microenvironment
15.
Sci Adv ; 6(36)2020 09.
Article in English | MEDLINE | ID: mdl-32917602

ABSTRACT

The high redox level of tumor microenvironment inhibits the oxidation treatment and the immune response. Here, we innovatively develop maleimide liposome (ML) adjuvants to promote immunogenic cell death (ICD) induction and dendritic cells (DCs) maturation by glutathione (GSH) depletion for augmenting the photothermal immunotherapy of breast cancer. The ML effectively depletes the intracellular GSH and up-regulates reactive oxygen species (ROS) in both tumor cells and DCs. In tumor cells, the ROS boosted the ABTS·+ production to activate photothermal-induced ICD. In DCs, it relieved the immunosuppression, promoting DC maturation (57%) and antigen presenting. As a result of the ML assistant, the therapeutic systems improved the infiltration of CD8+ T cells to 53% in tumor tissues, eliciting strong abscopal effect and antimetastasis effect. The MLs were believed to be a superior candidate of adjuvants for enhancing immune response and cancer therapeutic efficacy.


Subject(s)
Breast Neoplasms , Liposomes , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Female , Glutathione , Humans , Immunotherapy , Reactive Oxygen Species , Tumor Microenvironment
16.
ACS Appl Bio Mater ; 3(11): 8000-8011, 2020 Nov 16.
Article in English | MEDLINE | ID: mdl-35019539

ABSTRACT

Chemotherapy plays an important role in cancer treatment, yet its clinical application is inhibited by side effects. Chemotherapeutic agents accumulate at nonspecific sites and induce oxidative stress damage in noncancer tissues. A selective approach would be ideal, which would not only enhance anticancer efficacy in the tumor sites but also reduce chemotherapy-induced adverse effects on normal tissues. Therefore, we reported an adenosine-5'-triphosphate (ATP)-responsive oxidative stress nanoregulator (DePQu-DOX) to achieve the tissue-specific therapy. The DePQu-DOX NPs coloading doxorubicin (DOX) and quercetin (Qu) enhanced oxidative stress in murine breast cancer cells and scavenged DOX-induced oxygen free radicals in normal cardiac myocytes and podocytes. The released Qu could accelerate free radical scavenging more efficiently in oxygen-rich myocardium than in hypoxic tumors. Additionally, the ATP-specific responsiveness of nanocarriers enable cargos to selectively accumulate at tumor sites and decline the accumulation amount at normal tissues, resulting in lower system toxicity and improved anticancer effects. In vitro and in vivo experiments showed that this oxidative stress nanoregulator could efficiently protect normal tissues and significantly inhibit tumor growth. This study suggests that nanomedicine-mediated oxidative stress regulation could provide selective tumor therapeutics and reduce anthracycline-induced system toxicity.

17.
J Control Release ; 326: 615-627, 2020 10 10.
Article in English | MEDLINE | ID: mdl-32735880

ABSTRACT

Invasion and metastasis of tumor cells is one of the major obstacles in cancer therapy. The process of tumor metastasis and diffusion is coordinated by multiple pathways associated with chemokine signals and migration microenvironment. In our previous work, chemokine CXC receptor 4 (CXCR4) antagonists showed significant anti-metastatic effects by blocking the CXCR4/stromal cell-derived factor-1(SDF-1) axis in pancreatic cancer and breast cancer. Here, we proposed to achieve migration chain-treatment for metastatic tumors by introducing a cell adhesion molecules CD44 inhibitor (Star miR-34a) to deprive of cell migration capability on the basis of CXCR4 antagonism (cyclam monomer, CM). Dextrin modified 1.8 k PEI with CM-end was prepared to deliver therapeutic miR-34a (named DPC/miR-34a) for efficient anti-metastasis by downregulating adhesion protein CD44 and targeting the CXCR4/SDF-1 axis. Additionally, reduced expression of the anti-apoptotic protein Bcl2 caused by miR-34a could enhance the anti-tumor efficacy of DPC/miR-34a nanoplex administration. Compared with inhibition of the CXCR4/SDF-1 axis or CD44 expression, the multidimensional therapy (DPC/miR-34a) exhibited considerable suppression of cancer cell invasion as assessed by an in vitro cell invasion assay and in vivo anti-metastasis model. Moreover, DPC/miR-34a demonstrated a superior antitumor and anti-metastatic efficacy both in lung metastatic model and orthotopic MDA-MB-231 tumor models, thus providing an efficient approach for combating metastatic tumors.


Subject(s)
Breast Neoplasms , MicroRNAs/therapeutic use , Receptors, CXCR4/antagonists & inhibitors , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement , Chemokine CXCL12 , Female , Humans , Tumor Microenvironment
18.
iScience ; 23(2): 100872, 2020 Feb 21.
Article in English | MEDLINE | ID: mdl-32059177

ABSTRACT

Protein drugs own a large share in the market and hold great prospects for the treatment of many diseases. However, the available protein drugs are limited to the extracellular target, owing to the inefficient transduction and activity modulation of proteins targeting intracellular environment. In this study, we constructed ATP-charged platforms to overcome the above-mentioned barriers for cancer theranostics. The phenylboronic acid-modified polycations (PCD) were synthesized to assemble with enzymes and shield its activity in the blood circulation. When the PCD nanoclusters reached tumor site, they effectively transported the enzymes into the cells, followed by recovering its catalytic activity after being charged with ATP. Importantly, the cascaded enzyme systems (GOx&HRPA) selectively induced starvation therapy as well as photoacoustic imaging of tumor. Our results revealed that the intelligent nanoclusters were broadly applicable for protein transduction and enzyme activity modulation, which could accelerate the clinical translation of protein drugs toward intracellular target.

19.
Biomater Sci ; 7(12): 5359-5368, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31621699

ABSTRACT

Photodynamic therapy (PDT) is a clinically approved cancer treatment approach that relies on the generation of excess reactive oxygen species (ROS) to eradicate tumor cells by inducing oxidative stress. Unfortunately, if the tumor's endogenous glutathione (GSH) is overexpressed, it will eliminate the ROS and restrict the therapeutic efficacy of PDT. Herein, we report a H2O2-activated oxidative stress amplifier (OSA) for enhancing the ROS generation for PDT via GSH scavenging. Cinnamaldehyde (Cin) and chlorin e6 (Ce6) were applied as the GSH scavenger and photosensitizer, respectively, which were assembled with the ROS-responsive amphipathic polymer (DPL) to form DPL@CC micelles as the OSA. In the circulation of blood, the OSA can effectively protect the Cin from albumin binding to retain its GSH depletion ability. Once the OSA reached the tumor site, the high level of H2O2 triggered the degradation of DPL and led to the release of Cin and Ce6. Subsequently, the released Cin reacted with the intracellular GSH by Michael Addition and downregulated the GSH level to about 18.9%, versus untreated cells, to weaken the anti-oxidation ability of tumor cells. Thus, it provided a suitable environment for PDT to obtain an amplifying effect on oxidative stress and superior anti-cancer efficacy of 94% growth inhibition. The preparation of the H2O2-activated oxidative stress amplifier is a convincing strategy for promoting intracellular ROS generation and enhancing the tumor PDT efficacy, which could also augment the clinical application of PDT.


Subject(s)
Acrolein/analogs & derivatives , Breast Neoplasms/drug therapy , Glutathione/metabolism , Hydrogen Peroxide/administration & dosage , Porphyrins/administration & dosage , Acrolein/administration & dosage , Acrolein/pharmacology , Administration, Intravenous , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chlorophyllides , Combined Modality Therapy , Female , Humans , Hydrogen Peroxide/pharmacology , Mice , Oxidative Stress , Photochemotherapy , Porphyrins/pharmacology , Xenograft Model Antitumor Assays
20.
Acta Biomater ; 94: 435-446, 2019 08.
Article in English | MEDLINE | ID: mdl-31216493

ABSTRACT

Metallic materials are widely emerging as photothermal agents owing to their superior photothermal transduction efficiency and satisfactory photostability. In this study, an iron-based coordination polymer (Fe-CNP) loaded with doxorubicin (DOX) was assessed as a dual-function agent for photothermal therapy (PTT) and tumor-targeted chemotherapy. Fe-CNPs were synthesized by a one-step coordination reaction between Fe3+, hydrocaffeic acid, and dopamine-modified hyaluronic acid. A drug-loading method was developed to entrap DOX within Fe-CNPs through the formation of coordination bonds by Fe3+ and DOX (Scheme 1). DOX release was rapidly triggered in the cellular acidic environment and further enhanced by hyperpyrexia in the part of tumor, which will kill the remaining tumor cells after PTT. Animal experiments demonstrated complete inhibition of tumor growth without recurrence in 21 days after injection of DOX@Fe-CNPs with NIR laser irradiation. These results confirmed the enhanced anti-tumor efficiency of the chemo-photothermal nanosystem. Our work may reveal a photothermal coordination polymer as a drug-loading framework and highlight the development of metal-organic materials in combined chemo-photothermal therapy. STATEMENT OF SIGNIFICANCE: Photothermal therapy (PTT), which could directly act on tumors, has been considered as a promising treatment method for cancer. The combination of PTT with chemotherapy is attracting tremendous attention because such advanced application can achieve personalized precise medicine. Unfortunately, most PTT materials have photobleaching property, which results in reduced photothermal efficiency. Furthermore, their clinical applications also suffer from low loading capacity of chemotherapeutic drugs or nonbiodegradability in the biological system. In this study, we hypothesized that iron-based coordination polymers (Fe-CNPs) could function dually as agents to deliver both PTT and tumor-targeted chemotherapy by coordination loading of the chemotherapeutic drug doxorubicin (DOX). Our work may open up new avenues to rationally design versatile platforms for photothermal-chemotherapy to obtain synergistically enhanced therapeutic efficacy.


Subject(s)
Coordination Complexes , Doxorubicin , Drug Carriers , Hyperthermia, Induced , Neoplasms, Experimental , Phototherapy , Animals , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , Coordination Complexes/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy
SELECTION OF CITATIONS
SEARCH DETAIL