Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Nano ; 18(13): 9713-9735, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38507590

ABSTRACT

Cancer-associated fibroblasts (CAFs) assist in breast cancer (BRCA) invasion and immune resistance by overproduction of extracellular matrix (ECM). Herein, we develop FPC@S, a photodynamic immunomodulator that targets the ECM, to improve the photodynamic immunotherapy for fibrotic BRCA. FPC@S combines a tumor ECM-targeting peptide, a photosensitizer (protoporphyrin IX) and an antifibrotic drug (SIS3). After anchoring to the ECM, FPC@S causes ECM remodeling and BRCA cell death by generating reactive oxygen species (ROS) in situ. Interestingly, the ROS-mediated ECM remodeling can normalize the tumor blood vessel to improve hypoxia and in turn facilitate more ROS production. Besides, upon the acidic tumor microenvironment, FPC@S will release SIS3 for reprograming CAFs to reduce their activity but not kill them, thus inhibiting fibrosis while preventing BRCA metastasis. The natural physical barrier formed by the dense ECM is consequently eliminated in fibrotic BRCA, allowing the drugs and immune cells to penetrate deep into tumors and have better efficacy. Furthermore, FPC@S can stimulate the immune system and effectively suppress primary, distant and metastatic tumors by combining with immune checkpoint blockade therapy. This study provides different insights for the development of fibrotic tumor targeted delivery systems and exploration of synergistic immunotherapeutic mechanisms against aggressive BRCA.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Reactive Oxygen Species/metabolism , Extracellular Matrix/metabolism , Immunotherapy , Fibrosis , Tumor Microenvironment
2.
Biomaterials ; 303: 122392, 2023 12.
Article in English | MEDLINE | ID: mdl-37984245

ABSTRACT

Tumor cells overexpress programmed cell death ligand 1 (PD-L1) to impede immune responses and escape immune elimination. Development of effective combination regimens to sensitize immunotherapy is promising but always challenging. Herein, a self-reinforced photodynamic immunostimulator (designated as PCS) is constructed for metastatic breast cancer treatment through simultaneous downregulation and blockade of PD-L1. Specifically, PCS is prepared by encapsulating signal transducer and activator of transcription 3 (STAT3) inhibitor (Stattic) into photosensitizer (protoporphyrin IX) modified PD-L1 blockade peptide (CVRARTR) through drug self-assembly. PCS can facilitate the targeted drug accumulation in PD-L1 overexpressed breast cancer cells to block PD-L1 and inhibit the phosphorylation of STAT3 to downregulate PD-L1. Moreover, PCS increases intracellular oxidative stress to show a robust anti-proliferation effect through photodynamic therapy (PDT), which also triggers an immunogenic cell death (ICD) to expose the immunostimulatory signals. Consequently, the efficient PD-L1 inhibition and robust PDT of PCS synergistically suppress the malignant growth of breast cancer, and concurrently activate the systemic anti-tumor immunity for metastatic inhibition with no obvious side effects. Such a photodynamic immunostimulator may provide an effective combination regimen for therapies activated immunotherapy against metastatic breast cancer.


Subject(s)
Breast Neoplasms , Photochemotherapy , Humans , Female , Breast Neoplasms/drug therapy , B7-H1 Antigen/metabolism , Immunotherapy , Photosensitizing Agents/therapeutic use , Cell Line, Tumor
3.
Theranostics ; 13(7): 2176-2191, 2023.
Article in English | MEDLINE | ID: mdl-37153731

ABSTRACT

Background: Currently, the prognosis and survival rate for patients bearing non-small cell lung cancer (NSCLC) is still quite poor, mainly due to lack of efficient theranostic paradigms to exert in time diagnostics and therapeutics. Methods: Herein, for NSCLC treatment, we offer a customized theranostic paradigm, termed NIR-IIb fluorescence diagnosis and synergistic surgery/starvation/chemodynamic therapeutics, with a newly designed theranostic nanoplatform PEG/MnCuDCNPs@GOx. The nanoplatform is composed of brightly NIR-II emissive downconversion nanoparticles (DCNPs)-core and Mn/Cu-silica shell loaded with glucose oxidase (GOx) to achieve synergistic starvation and chemodynamic therapy (CDT). Results: It is found that 10% Ce3+ doped in the core and 100% Yb3+ doped in the middle shell greatly improves the NIR-IIb emission up to even 20.3 times as compared to the core-shell DCNPs without Ce3+ doping and middle shell. The bright NIR-IIb emission of the nanoplatform contributes to sensitive margin delineation of early-stage NSCLC (diameter < 1 mm) with a signal-to-background ratio (SBR) of 2.18, and further assists in visualizing drug distribution and guiding surgery/starvation/chemodynamic therapy. Notably, the starvation therapy mediated by GOx-driven oxidation reaction efficiently depletes intratumoral glucose, and supplies H2O2 to boost the CDT mediated by the Mn2+ and Cu2+, which consequently realized a highly effective synergistic treatment for NSCLC. Conclusion: This research demonstrates an efficient treatment paradigm for NSCLC with NIR-IIb fluorescence diganosis and image-guided synergistic surgery/starvation/chemodynamic therapeutics.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Nanoparticles , Neoplasms , Small Cell Lung Carcinoma , Starvation , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Fluorescence , Hydrogen Peroxide , Lung Neoplasms/drug therapy , Glucose Oxidase , Cell Line, Tumor , Tumor Microenvironment
4.
Adv Healthc Mater ; 12(4): e2202307, 2023 02.
Article in English | MEDLINE | ID: mdl-36349844

ABSTRACT

Safe and effective strategies are urgently needed to fight against the life-threatening diseases of various cancers. However, traditional therapeutic modalities, such as radiotherapy, chemotherapy and surgery, exhibit suboptimal efficacy for malignant tumors owing to the serious side effects, drug resistance and even relapse. Phototherapies, including photodynamic therapy (PDT) and photothermal therapy (PTT), are emerging therapeutic strategies for localized tumor inhibition, which can produce a large amount of reactive oxygen species (ROS) or elevate the temperature to initiate cell death by non-invasive irradiation. In consideration of the poor bioavailability of phototherapy agents (PTAs), lots of drug delivery systems have been developed to enhance the tumor targeted delivery. Nevertheless, the carriers of drug delivery systems inevitably bring biosafety concerns on account of their metabolism, degradation, and accumulation. Of note, carrier-free nanomedicine attracts great attention for clinical translation with synergistic antitumor effect, which is characterized by high drug loading, simplified synthetic method and good biocompatibility. In this review, the latest advances of phototherapy with various carrier-free nanomedicines are summarized, which may provide a new paradigm for the future development of nanomedicine and tumor precision therapy.


Subject(s)
Neoplasms , Photochemotherapy , Humans , Nanomedicine , Phototherapy , Neoplasms/drug therapy , Drug Delivery Systems , Cell Line, Tumor , Theranostic Nanomedicine
5.
Mater Sci Eng C Mater Biol Appl ; 131: 112524, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34857303

ABSTRACT

Currently, the combining photodynamic therapy (PDT) with photothermal therapy (PTT) modalities based on a single near infrared (NIR) laser irradiation and highly selective internalization still remain a challenge. Herein, a hierarchical dual-responsive cleavable nanosystem for synergetic NIR triggered PDT/PTT is reported. The engineered nanoplatform (Au NRs/Cur/UCNPs@PBE) is designed by loading curcumin (Cur, photosensitizer) on gold nanarods (Au NRs) to build PDT/PTT therapy system, which was encapsulated outside with upconversion nanoparticles (UCNPs) and then modified with phenylboronic double ester (PBE). The pH and ROS-responsive feature made Au NRs/Cur/UCNPs@PBE provide a fundamental structural evolution and improve the specificity and intracellular accumulation to tumors. Au NRs/Cur/UCNPs@PBE exhibited significant PDT and PTT efficiency against two type melanoma cells due to upconversion nanoparticles and Au NRs induced by an 808 nm laser. Notably, the platform can mainly activate apoptosis and partial ferroptosis to achieve the synergistic PDT/PTT, furthermore, the integrated PDT with PTT using Au NRs/Cur/UCNPs@PBE showcased a great antitumor efficacy in vivo superior to the other alone treatment. Our findings highlight that this intelligent nanoagents for synergistic phototherapy facilitate enhanced fighting melanoma and provide a promising strategy for melanoma theranostics.


Subject(s)
Melanoma , Nanoparticles , Photochemotherapy , Cell Line, Tumor , Gold , Humans , Melanoma/drug therapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Phototherapy , Photothermal Therapy
6.
ACS Appl Bio Mater ; 4(3): 2639-2653, 2021 03 15.
Article in English | MEDLINE | ID: mdl-35014380

ABSTRACT

Combinatory photodynamic and chemotherapy have demonstrated superior performance in cancer ablation over singular therapeutics. However, photodynamic therapy (PDT) often exhibits suboptimal efficacy for deep-seated tumors, owing to the limited penetration depth of illumination light, while chemotherapy is generally accompanied by severe side effects. Therefore, it is imperative to develop a functional nanoplatform for combinatory PDT and chemotherapy, which could, for PDT, achieve enhanced light penetration and, for chemotherapy, realize reduced therapeutic threshold dosage and a controllable drug release profile (e.g., minimized release in blood circulation but bursting release in the tumor microenvironment). Herein, we demonstrate a therapeutic nanoplatform composed of poly(acrylic acid) (PAA)-modified silica-coated Nd3+-doped upconversion nanoparticles decorated with methylene blue (MB) and doxorubicin (DOX) in silica and a PAA layer, respectively. Notably, 808 nm light is used to excite upconversion nanoparticles and further trigger the photosensitization behavior of MB in PDT, while the quick acid response of the PAA layer in the tumor acid environment introduces DOX bursting for optimized chemotherapy with significantly decreased therapeutic threshold dosage and minimized side effects. Importantly, the anticancer efficiency of the nanoplatform in vitro and in vivo shows an IC50 and a tumor inhibition rate of 12.55 µg mL-1 and 89.81%, respectively. This study provides a strategy for combinatory cancer therapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Biocompatible Materials/pharmacology , Doxorubicin/pharmacology , Lasers , Photochemotherapy , Photosensitizing Agents/pharmacology , Antibiotics, Antineoplastic/chemistry , Biocompatible Materials/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/chemistry , Drug Liberation , Drug Screening Assays, Antitumor , Humans , Materials Testing , Molecular Structure , Particle Size , Photosensitizing Agents/chemistry , Tumor Cells, Cultured
7.
J Nanobiotechnology ; 17(1): 44, 2019 Mar 27.
Article in English | MEDLINE | ID: mdl-30917812

ABSTRACT

BACKGROUND: The combination of multiple chemotherapeutics has been used in the clinic for enhanced cancer chemotherapy, however, frequent relapse, chemo-resistance and side effects remains therapeutic hurdles. Thus, the development of co-delivery system with enhanced targeting and synergistic different modal treatments has been proposed as promising strategies for intensive improvement of the therapeutic outcomes. RESULTS: We fabricated a nanocarrier based on gold nanorods (Au NRs), cRGD peptide-modified and multi-stimuli-responsive paclitaxel (PTX) and curcumin (CUR) release for synergistic anticancer effect and chemo-photothermal therapy (PTX/CUR/Au NRs@cRGD). The specific banding of cRGD to αvß3 integrin receptor on the tumor cell surfaces facilitated the endocytosis of PTX/CUR/Au NRs@cRGD, and the near-infrared ray (NIR) further enhanced the drug release and chemotherapeutical efficiency. Compared to single drug, single model treatment or undecorated-PTX/CUR/Au NRs, the PTX/CUR/Au NRs@cRGD with a mild NIR showed significantly enhanced apoptosis and S phase arrest in three cancer cell lines in vitro, and improved drug accumulation in tumor sites as well as tumor growth inhibition in vivo. CONCLUSIONS: The tumor targeted chemo-photothermal therapy with the synergistic effect of dual drugs provided a versatile strategy for precise cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Curcumin/administration & dosage , Drug Carriers/chemistry , Drug Liberation , Gold/chemistry , Nanotubes/chemistry , Paclitaxel/administration & dosage , A549 Cells , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Survival/drug effects , Curcumin/chemistry , Hep G2 Cells , Humans , Infrared Rays , Integrin alphaVbeta3/metabolism , KB Cells , Mice, Inbred BALB C , Paclitaxel/chemistry , Particle Size , Peptides, Cyclic/metabolism , Phototherapy/methods , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...