Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Oncol Rep ; 52(1)2024 Jul.
Article in English | MEDLINE | ID: mdl-38757398

ABSTRACT

Following the publication of the above paper, it was drawn to the Editors' attention by a concerned reader that the data obtained from sphere­forming assay experiments shown in Figs. 4C­F and 8B and C, and western blotting data in Figs. 4A and 8A, were strikingly similar to data appearing in different form in other articles by different authors from different research institutes that had already been published, one of which has been retracted. Moreover, a pair of data panels comparing between Fig. 4E and 8C were partly overlapping, such that these data appear to have been derived from the same original source. Owing to the fact that the contentious data in the above article had already been published elsewhere prior to its submission to Oncology Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [Oncology Reports 35: 1204­1212, 2016; DOI: 10.3892/or.2015.4437].

2.
J Colloid Interface Sci ; 662: 857-869, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38382370

ABSTRACT

Copper-based nanomaterials show considerable potential in the chemodynamic therapy of cancers. However, their clinical application is restricted by low catalytic activity in tumor microenvironment and copper-induced tumor angiogenesis. Herein, a novel copper-doxorubicin-anlotinib (CDA) nanoconjugate was constructed by the combination of copper-hydrazide coordination, hydrazone linkage and Schiff base bond. The CDA nanoconjugate consists of a copper-3,3'-dithiobis(propionohydrazide)-doxorubicin core and an anlotinib-hyaluronan shell. Benefiting from hyaluronan camouflage and abundant disulfide bonds and Cu2+, the CDA nanoconjugate possessed excellent tumor-targeting and glutathione-depleting abilities and enhanced chemodynamic efficacy. Released doxorubicin significantly improved copper-mediated chemodynamic therapy by upregulating nicotinamide adenine dinucleotide phosphate oxidase 4 expression to increase intracellular H2O2 level. Furthermore, the nanoconjugate produced excessive •OH to induce lipid peroxidation and mitochondrial dysfunction, thus greatly elevating doxorubicin-mediated chemotherapy. Importantly, anlotinib effectively inhibited the angiogenic potential of copper ions. In a word, the CDA nanoconjugate is successfully constructed by combined coordination and pH-responsive linkages, and displays the great potential of copper-drug conjugate for targeted synergistic chemo/chemodynamic/antiangiogenic triple therapy against cancers.


Subject(s)
Carcinoma, Hepatocellular , Indoles , Liver Neoplasms , Nanoparticles , Neoplasms , Quinolines , Humans , Carcinoma, Hepatocellular/drug therapy , Copper , Hyaluronic Acid , Nanoconjugates , Hydrogen Peroxide , Liver Neoplasms/drug therapy , Doxorubicin/pharmacology , Glutathione , Cell Line, Tumor , Tumor Microenvironment
3.
ACS Appl Mater Interfaces ; 15(31): 37143-37156, 2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37498789

ABSTRACT

Ferroptosis holds great potential in cancer treatment, but its efficacy is severely limited by a low Fenton reaction efficacy. Meanwhile, the interactive relationship between Ferroptosis and the PD-1 blockade is still vague. Herein, a hydrazide/Cu/Fe/indocyanine green coordinated nanoplatform (TCFI) is constructed by a hydrazide-metal-sulfonate coordination process. The TCFI nanoplatform exhibits Fenton-/catalase-/glutathione oxidase-like triple activities and accordingly can trigger lipid peroxidation, relieve hypoxia, and downregulate the glutathione/glutathione peroxidase 4 axis, thus achieving positively and negatively dually enhanced Ferroptosis in B16F10 cancer cells. Under near-infrared laser irradiation, the TCFI nanoplatform induces robust immunogenic cancer cell death by elevating the intracellular reactive oxygen species level through synergistic photodynamic therapy/Ferroptosis, which significantly potentiates CD8+ T cell infiltration into tumors and interferon-γ secretion. Moreover, upregulated interferon-γ efficiently inhibits system xc- activity and sensitizes cancer cells to Ferroptosis. Interestingly, the PD-1 blockade may strengthen the reciprocal process. The combination of the TCFI nanoplatform and αPD-1 can eliminate primary tumors and inhibit distant tumor growth, lung metastasis, and tumor recurrence. This study presents a simple and novel coordination strategy to fabricate tumor microenvironment-responsive nanodrugs and highlights the enhancement effect of photodynamic therapy on reciprocal Ferroptosis and antitumor immunity.


Subject(s)
Ferroptosis , Melanoma , Neoplasms , Humans , Indocyanine Green , Interferon-gamma , Programmed Cell Death 1 Receptor , Hydrazines , Infrared Rays , Cell Line, Tumor , Tumor Microenvironment
4.
Adv Healthc Mater ; 12(13): e2202949, 2023 05.
Article in English | MEDLINE | ID: mdl-36716523

ABSTRACT

Cuproptosis is a recently discovered form of programmed cell death and shows great potential in cancer treatment. Herein, a copper-dithiocarbamate chelate-doped and artemisinin-loaded hollow nanoplatform (HNP) is developed via a chelation competition-induced hollowing strategy for cuproptosis-based combination therapy. The HNP exhibits tumor microenvironment-triggered catalytic activity, wherein liberated Cu2+ catalyzes artemisinin and endogenous H2 O2 to produce C-centered radicals and hydroxyl radicals, respectively. Meanwhile, the disulfide bonds-rich HNP can deplete intracellular glutathione, thus triply amplifying tumor oxidative stress. The augmented oxidative stress sensitizes cancer cells to the cuproptosis, causing prominent dihydrolipoamide S-acetyltransferase oligomerization and mitochondrial dysfunction. Moreover, the HNP can activate ferroptosis via inhibiting GPX4 activity and trigger apoptosis via dithiocarbamate-copper chelate-mediated ubiquitinated proteins accumulation, resulting in potent antitumor efficacy. Such a cuproptosis/ferroptosis/apoptosis synergetic strategy opens a new avenue for cancer therapy.


Subject(s)
Apoptosis , Artemisinins , Neoplasms , Cell Line, Tumor , Combined Modality Therapy , Copper/pharmacology , Neoplasms/drug therapy , Oxidative Stress , Tumor Microenvironment
5.
J Colloid Interface Sci ; 628(Pt B): 79-94, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-35985065

ABSTRACT

Hydrogel-based drug delivery holds great promise in topical tumor treatment. However, the simple construction of multifunctional therapeutic hydrogels under physiological conditions is still a huge challenge. Herein, for the first time, a multifunctional hyaluronan/MnO2 nanocomposite (HHM) hydrogel with injectable and self-healing capabilities was constructed under physiological conditions through innovative in situ mineralization-triggered Mn-hydrazide coordination crosslinking. The hydrogel formed from Mn2+ and hydrazided hyaluronan under optimized conditions exhibited a high elastic modulus >1 kPa, injectability, self-healing function, stimuli-responsiveness and catalase-like activity. In vitro and in vivo biological experiments demonstrated that our HHM hydrogel could not only efficiently relieve hypoxia by in situ catalytic decomposition of endogenous H2O2 into O2 but also achieve synergistic photodynamic/photothermal therapy of 4T1 breast cancer in a mouse tumor model. This study presented a novel mineralization-driven metal-hydrazide coordination crosslinking approach and developed a multifunctional therapeutic platform for O2-enhanced efficient topical dual-phototherapy of breast cancer.


Subject(s)
Hyaluronic Acid , Tumor Hypoxia , Mice , Animals , Nanogels , Catalase , Manganese Compounds/pharmacology , Hydrazines/pharmacology , Hydrogen Peroxide , Cell Line, Tumor , Oxides , Phototherapy , Hydrogels/pharmacology
6.
J Colloid Interface Sci ; 628(Pt B): 968-983, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36037718

ABSTRACT

Immune checkpoint blockade (ICB)-based immunotherapy is a revolutionary therapeutic strategy for hepatocellular carcinoma (HCC). However, tumor immune tolerance and escape severely restrict the therapeutic efficacy of ICB therapy. It is urgent to explore new strategies to potentiate ICB therapy in HCC. Herein, we developed manganese oxide-crosslinked bovine albumin/hyaluronic acid nanoparticles (BHM) by an innovative hydrazide-manganese coordination and desolvation process. Successive loading of doxorubicin (DOX) and indocyanine green (ICG) was achieved via hydrazone linkage and electrostatic interactions, respectively, obtaining DOX/ICG-coloaded BHM nanoplatform (abbreviated as BHMDI). The BHMDI nanoplatform exhibited a high drug content (>46%) and pH/reduction dual-responsive drug release behavior. The nanoplatform could efficiently alleviate tumor hypoxia by catalytic decomposition of intracellular H2O2 to O2 and significantly improve BHMDI-based photodynamic chemotherapy efficacy. The BHMDI nanoplatform downregulated the proportion of alternatively activated (M2) macrophages in tumors and simultaneously induced immunogenic death of HCC cells, thus promoting the maturation of dendritic cells and ensuing priming of CD4+ and CD8+ T cells. Importantly, programmed death-1 (PD-1) blockade in combination with BHMDI nanoplatform not only eradicated primary tumors but inhibited tumor recurrence, abscopal tumor growth and lung metastasis of HCC by triggering robust systemic antitumor immunity. This work proved the feasibility of BHMDI-based photodynamic chemotherapy for potentiating PD-1 blockade immunotherapy by reversing hypoxic and immunosuppressive tumor microenvironment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Cattle , Animals , Humans , Carcinoma, Hepatocellular/drug therapy , Manganese , Serum Albumin, Bovine , Hyaluronic Acid , Programmed Cell Death 1 Receptor/therapeutic use , Indocyanine Green/pharmacology , CD8-Positive T-Lymphocytes , Hydrazines/therapeutic use , Immune Checkpoint Inhibitors , Hydrogen Peroxide , Liver Neoplasms/drug therapy , Immunotherapy , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Hydrazones , Cell Line, Tumor
7.
Colloids Surf B Biointerfaces ; 209(Pt 2): 112159, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34687973

ABSTRACT

2D cell culture is widely utilized to develop anti-cancer drugs and to explore the mechanisms of cancer tumorigenesis and development. However, the findings obtained from 2D culture often fail to provide guidance for clinical tumor treatments since it cannot precisely replicate the features of real tumors. 3D tumor models capable of recapitulating native tumor microenvironments have been proved to be a promising alternative technique. Herein, we constructed a breast tumor model from novel hyaluronic acid (HA) hydrogel which was prepared through photocrosslinking of methacrylated HA. The hydrogel was used as a biomimetic extracellular matrix to incubate MCF-7 cells. It was found that methacrylation degree had great effects on hydrogel's microstructure, mechanical performances, and liquid-absorbing and degradation abilities. Optimized hydrogel exhibited highly porous morphology, high equilibrium swelling ratio, suitable mechanical properties, and hyaluronidase-responsive degradation behavior. The results demonstrated that the HA hydrogel facilitated MCF-7 cell proliferation and growth in an aggregation manner. Furthermore, 3D-cultured MCF-7 cells not only up-regulated the expression of VEGF, bFGF and interleukin-8 but exhibited greater invasion and tumorigenesis capabilities compared with 2D-cultured cells. Therefore, the HA hydrogel is a reliable substitute for tumor model construction.


Subject(s)
Breast Neoplasms , Hyaluronic Acid , Extracellular Matrix , Female , Humans , Hydrogels , MCF-7 Cells , Tumor Microenvironment
8.
Carbohydr Polym ; 276: 118810, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34823812

ABSTRACT

It is still a huge challenge for concurrent highly efficient loading of chemotherapeutic agent and photosensitizer into single nanocarrier via stimuli-responsive linkages due to their different physicochemical properties and pharmacokinetics. Herein, based on the discovery of unique cisplatin-hydrazide and cisplatin-indocyanine green (ICG) coordination reactions, a multifunctional coordination nanoprodrug, cisplatin/ICG co-loaded hydrazided hyaluronan/bovine serum albumin (HBCI) nanoparticles, was developed by a desolvation-dual coordination process. The nanoprodrug exhibited ultrahigh drug loading efficiency and glutathione/NIR light dual-responsive drug release behavior. In vitro cellular studies demonstrated efficient internalization and apoptosis-inducing ability of the nanoprodrug in HepG2 cells. In vivo results confirmed the efficacious tumor accumulation and biosafety of HBCI nanoprodrug and synergistic effect of HBCI-based combined photodynamic chemotherapy on inhibiting tumor growth. Overall, this work not only provides a novel dual coordination approach for highly efficient loading of cisplatin and ICG but also verifies the therapeutic potential of HBCI nanoprodrug in combating hepatocellular carcinoma.


Subject(s)
Cisplatin/pharmacology , Hyaluronic Acid/pharmacology , Indocyanine Green/pharmacology , Liver Neoplasms/drug therapy , Nanoparticles/chemistry , Photochemotherapy/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cisplatin/chemistry , Drug Delivery Systems/methods , Hep G2 Cells , Humans , Hyaluronic Acid/chemistry , Indocyanine Green/chemistry , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology
9.
Int J Biol Macromol ; 184: 768-775, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34174305

ABSTRACT

Polysaccharide hydrogels are promising candidate matrices for recapitulating the characteristics of extracellular matrix (ECM) in breast tumors in terms of their structure and composition. Herein, to obtain an ECM-mimetic matrix, hydroxyethyl chitosan (HECS) hydrogels were prepared through Schiff-base crosslinking reaction using dialdehyde hyaluronic acid as crosslinker. The obtained HECS hydrogels displayed a highly porous structure, a stiffness comparable to that of breast tissue, and a fast water-absorption speed. The amount of crosslinker had great effects on the swelling and rheological behaviors of the HECS hydrogels. Preliminary results from in vitro biological assessments confirmed that MCF-7 cells incubated within HECS hydrogels preferred to grow into three-dimensional spheroids. Importantly, the cells displayed enhanced migrative capability and upregulated expression levels of MMP-2, TGF-ß and VEGF in comparison to two-dimension cultured cells. Hence, the HECS hydrogels show great promise as a biomimetic ECM in constructing breast tumor models.


Subject(s)
Breast Neoplasms/metabolism , Chitosan/chemistry , Extracellular Matrix/metabolism , Hyaluronic Acid/chemistry , Hydrogels/chemical synthesis , Spheroids, Cellular/cytology , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , MCF-7 Cells , Matrix Metalloproteinase 2/metabolism , Porosity , Schiff Bases , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
10.
Pathol Res Pract ; 216(11): 153155, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32871536

ABSTRACT

BACKGROUND: The current recommendation is to reflex test HER2 immunohistochemistry (IHC) equivocal breast cancer cases with fluorescence in situ hybridization (FISH) analysis. Either IHC 3+ or FISH positive cancers are considered HER2 positive (HER2+) and treated with HER2 targeted therapy. This study examined the predictive value of HER IHC or FISH positivity in tumor response to HER2 targeted therapy. METHODS: Biopsies of 76 HER2+ breast cancer cases were evaluated. All patients were treated with neoadjuvant HER2 targeted therapy and chemotherapy. Tumor response was evaluated on the excisional specimens. Cancers with complete pathologic response (pCR) or MD Anderson residual cancer burden-I (RCB-I) were classified as responders and cancers with RCB-II/III as non-responders. Clinicopathologic parameters were correlated with response. RESULTS: In univariate analysis, small tumor size, low nuclear grade, high Ki67, HER2 IHC 3+, homogenous strong HER2 IHC staining, high HER2/CEP17 ratio, and high HER2 copy number were significantly associated with pCR/RCB-I. In multivariate analysis, homogenous strong HER2 IHC staining pattern was significantly associated with pCR/RCB-I. The receiver operating characteristics (ROC) model showed either high HER2/CEP17 ratio or HER2 copy number individually was predictive of tumor response. CONCLUSION: HER2 IHC staining pattern is significantly associated with tumor response to neoadjuvant chemotherapy, reiterating the importance of HER2 IHC evaluation. The ROC model shows either high HER2/CEP17 ratio or high HER2 copy number individually is predictive of tumor response to neoadjuvant HER2 targeted therapy in HER2+ breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Breast Neoplasms/drug therapy , Receptor, ErbB-2/analysis , Adult , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/metabolism , Chemotherapy, Adjuvant/methods , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Middle Aged , Molecular Targeted Therapy/methods , Neoadjuvant Therapy/methods , Trastuzumab/therapeutic use , Treatment Outcome
11.
Clin Breast Cancer ; 20(1): 19-24, 2020 02.
Article in English | MEDLINE | ID: mdl-31806448

ABSTRACT

BACKGROUND: HER2-targeted neoadjuvant therapy has high efficacy in treating HER2-positive breast cancer. Response to neoadjuvant therapy helps clinicians make treatment decisions and make estimates about prognosis. This study examined clinicopathologic features to determine which may be most predictive of response to neoadjuvant therapy in HER2+ breast cancer. PATIENTS AND METHODS: Patients with HER2+ breast cancer (n = 173) who had an initial biopsy performed between 2010 and 2016 were identified at our institution. Tumor response was evaluated on excisional specimens using the MD Anderson residual cancer burden (RCB) classification. Tumors with pathologic complete response (defined as no residual invasive carcinoma in the breast and lymph nodes) and RCB-I were classified as having response and tumors with RCB-II and -III as having no response. Patient age, tumor size, nuclear grade (1/2 vs. 3), mitosis, Nottingham grade, HER2 immunohistochemistry (1/2+ vs. 3+), HER2/CEP17 (chromosome enumeration probe 17) ratio, HER2 copy number, estrogen receptor, progesterone receptor, Ki-67, and tumor-infiltrating lymphocytes (TIL) were evaluated and correlated with response. TILs were evaluated for an average and also for the hot spot/total tumor stromal ratio. RESULTS: Small tumor size, low estrogen receptor and progesterone receptor expression, HER2 immunohistochemistry 3+, high Ki-67, high HER2/CEP17 ratio, and high HER2 copy number were significantly associated with response (all P < .05). TIL hot spot was associated with RCB in univariate (P < .05) but not multivariate analyses. CONCLUSION: Clinicopathologic features may help predict HER2+ breast cancer response to neoadjuvant therapy. Larger studies would be useful to confirm these associations, which may have relevance to clinical practice.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/analysis , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/therapy , Neoadjuvant Therapy/methods , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast/immunology , Breast/pathology , Breast/surgery , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/mortality , Chemotherapy, Adjuvant/methods , DNA Copy Number Variations , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Feasibility Studies , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mastectomy , Middle Aged , Predictive Value of Tests , Progression-Free Survival , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Estrogen/analysis , Receptors, Estrogen/metabolism , Receptors, Progesterone/analysis , Receptors, Progesterone/metabolism , Tumor Burden
12.
Carbohydr Polym ; 225: 115206, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31521263

ABSTRACT

Combination chemotherapy has attracted more and more attention in the field of anticancer treatment. Herein, a synergetic targeted combination chemotherapy of doxorubicin (DOX) and cisplatin in breast cancer was realized by HER2 antibody-decorated nanoparticles assembled from aldehyde hyaluronic acid (AHA) and hydroxyethyl chitosan (HECS). Cisplatin and DOX were successively conjugated onto AHA through chelation and Schiff's base reaction, respectively, forming DOX/cisplatin-loaded AHA inner core. The core was sequentially complexed with HECS and targeting HER2 antibody-conjugated AHA. The formed near-spherical nanoplatform had an average size of ∼160 nm and a zeta potential of -28 mV and displayed pH-responsive surface charge reversal and drug release behaviors. HER2 receptor-mediated active targeting significantly enhanced the cellular uptake of nanoplatform. Importantly, DOX and cisplatin exhibited a synergistic cell-killing effect in human breast cancer MCF-7 cells. These results clearly indicate that the novel nanoplatform is promising for synergistic combination chemotherapy of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Chitosan/therapeutic use , Drug Delivery Systems/methods , Hyaluronic Acid/therapeutic use , Nanoparticles/therapeutic use , Cisplatin/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/therapeutic use , Drug Combinations , Drug Liberation , Female , Humans , MCF-7 Cells
13.
Mater Sci Eng C Mater Biol Appl ; 103: 109854, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31349407

ABSTRACT

Combination of different therapy modalities with multiple tumoricidal mechanisms has emerged as a promising anticancer strategy. Herein, we reported an aldehyde/catechol-functionalized hyaluronic acid (DAHA) and hydroxyethyl chitosan (HECS) decorated gold nanorod (GNR) platform for combined chemo-photothermal therapy of breast cancer. The DAHA was synthesized by conjugating dopamine onto oxidized hyaluronic acid. The nanoplatform was prepared by successively modifying GNR with DAHA via Au-catechol bonds, conjugating DOX onto DAHA moiety through Schiff base linkage, and coating HECS interlayer for charge-reversal and hyaluronic acid corona for tumor cell targeting. The resulting nanoplatform GNR-HADOXCH exhibited acid-triggered surface charge-reversal and pH/NIR dual-responsive drug release behaviors. The nanoplatform could be efficiently internalized into MCF-7 breast cancer cells and displayed greater cancer cell killing than individual modalities. Therefore, polysaccharide decoration could ensure the co-delivery of GNR and DOX into cancer cells, and the developed GNR-HADOXCH holds great potential for breast cancer treatment.


Subject(s)
Breast Neoplasms/drug therapy , Chitosan/chemistry , Doxorubicin , Drug Delivery Systems , Gold , Hyaluronic Acid , Hyperthermia, Induced , Metal Nanoparticles , Nanotubes/chemistry , Phototherapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chitosan/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacology , Female , Gold/chemistry , Gold/pharmacology , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , MCF-7 Cells , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use
14.
Colloids Surf B Biointerfaces ; 181: 602-611, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31202131

ABSTRACT

The combination of chemotherapy and photothermal therapy is acknowledged as one of the most promising approaches in cancer treatment. Targeted delivery and controlled drug release are two important factors for combined chemo-photothermal therapy. In this study, a multifunctional nanoplatform based on gold nanorod (GNR) decorated with folate-conjugated poly(ethylene glycol)-b-poly(L-γ-glutamylhydrazine) (FEGGH) containing disulfide linker and dihydroxyphenyl groups was developed for targeted combined chemo-photothermal therapy of breast cancer. FEGGH was synthesized by ring-opening polymerization of γ-benzyl-l-glutamate-N-carboxyanhydride using folate/cystamine-heterobifunctionalized poly(ethylene glycol) as an initiator, following by hydrazinolysis and carbodiimide reactions. FEGGH was decorated onto GNR through Au-catechol bonds. Chemotherapeutic drug doxorubicin (DOX) was loaded onto the nanoplatform through pH-sensitive hydrazone linkage, obtaining final product FEGGHDOX-GNR. The DOX-loaded nanoplatform displayed excellent photostability and reduction/pH dual-responsive drug release behavior. Cytological studies demonstrated the effective internalization of FEGGHDOX-GNR into MCF-7 cells via folate-mediated endocytosis and additive therapeutic effect of combined photothermal-chemotherapy. These results indicate that our nanoplatform may be a promising strategy for targeted combined chemo-photothermal therapy of breast cancer.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/therapy , Doxorubicin/pharmacology , Gold/chemistry , Nanotubes/chemistry , Peptides/chemistry , Polyethylene Glycols/chemistry , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Drug Therapy, Combination , Female , Gold/pharmacology , Humans , MCF-7 Cells , Molecular Structure , Particle Size , Phototherapy , Structure-Activity Relationship , Surface Properties , Tumor Cells, Cultured
15.
Carbohydr Polym ; 211: 336-348, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30824098

ABSTRACT

In tumor biology, it is widely recognized that 3D rather than 2D cell culture can recapitulate key features of solid tumors, including cell-extracellular matrix (ECM) interactions. In this study, to mimick the ECM of breast cancer, hyaluronic acid (HA) hydrogels were synthesized from two polyvalent HA derivatives through a hydrazone and photo dual crosslinking process. HA hydrogels could be formed within 120 s. The hydrogels had similar topography and mechanical properties to breast tumor and displayed glutathione and hyaluronidase dual-responsive degradation behavior. Biological studies demonstrated that HA hydrogel could support the proliferation and clustering of breast cancer MCF-7 cells. The expression levels of VEGF, IL-8 and bFGF in hydrogel-cultured cells were significantly greater than those in 2D culture. Moreover, cells from hydrogel culture exhibited greater migration/invasion abilities and tumorigenicity than 2D-cultured cells. Therefore, the HA hydrogels are a promising ECM-mimicking matrix for in vitro construction of breast cancer.


Subject(s)
Cell Culture Techniques , Extracellular Matrix , Hyaluronic Acid , Hydrogels , Animals , Biomimetics , Cell Movement , Humans , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , MCF-7 Cells , Mice, Inbred BALB C , Tumor Burden
16.
Carbohydr Polym ; 212: 334-344, 2019 May 15.
Article in English | MEDLINE | ID: mdl-30832865

ABSTRACT

Chemo-photothermal combination therapy is a promising strategy for cancer treatment. In this study, to achieve the combined photothermal-chemotherapy of breast cancer, a pH-sensitive oxidized hyaluronic acid-decorated dihydroxyphenyl/hydrazide bifunctionalized hydroxyethyl chitosan (DHHC)-gold nanorod (GNR) conjugate was developed. DHHC was synthesized by successive dihydroxyphenylation and hydrazidation of hydroxyethyl chitosan through carbodiimide reaction and click chemistry, respectively. The conjugate was obtained by chemically bonding DHHC onto GNR via Au-catechol bonds. Doxorubicin (DOX) was loaded onto the conjugate via an acid-labile hydrazone linkage with a drug loading content of 5.1%. DOX-loaded conjugate displayed good stability in neutral aqueous solutions and exhibited pH-responsive drug release and surface charge reversal behaviors. In vitro biological studies indicated that the conjugate could be effectively internalized by breast cancer MCF-7 cells and synergistic therapeutic effects were demonstrated, suggesting its great potential in combined photothermal-chemotherapy of breast cancer.


Subject(s)
Breast Neoplasms/therapy , Doxorubicin/chemistry , Gold/chemistry , Nanotubes/chemistry , Phototherapy/methods , Polysaccharides/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Breast Neoplasms/pathology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Female , Gold/administration & dosage , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Polysaccharides/administration & dosage
17.
Carbohydr Polym ; 203: 203-213, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30318205

ABSTRACT

Combination of chemotherapy and photodynamic therapy has emerged as a promising anticancer strategy. Polysaccharide-based nanoparticles are being intensively explored as drug carriers for different forms of combination therapy. In this study, novel multifunctional polysaccharide-based nanocomplexes were prepared from aldehyde-functionalized hyaluronic acid and hydroxyethyl chitosan via sequential self-assembly method. Stable nanocomplexes were obtained through both Schiff's base bond and electrostatic interactions. Chemotherapeutics doxorubicin and pro-photosensitizer 5-aminolevulinic acid were chemically conjugated onto the nanocomplexes via Schiff base linkage. Anti-HER2 antibody as targeting moiety was decorated onto the surface of nanocomplexes. The obtained near-spherical shaped nanocomplexes had an average size of 140 nm and a zeta potential of -24.6 mV, and displayed pH-responsive surface charge reversal and drug release. Active targeting strategy significantly enhanced the cellular uptake of nanocomplexes and combined anticancer efficiency of chemo-photodynamic dual therapy in breast cancer MCF-7 cells. These results suggested that the nanocomplexes had great potential for targeted combination therapy of breast cancer.


Subject(s)
Aminolevulinic Acid/pharmacology , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Drug Carriers/chemistry , Nanoparticles/chemistry , Photosensitizing Agents/pharmacology , Aminolevulinic Acid/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Chitosan/analogs & derivatives , Doxorubicin/chemistry , Drug Liberation , Fluorescence , Humans , Hyaluronic Acid/analogs & derivatives , Light , MCF-7 Cells , Mice , Particle Size , Photosensitizing Agents/chemistry , Photosensitizing Agents/radiation effects , Protoporphyrins/biosynthesis , Protoporphyrins/chemistry , Protoporphyrins/radiation effects , Protoporphyrins/therapeutic use
18.
Acta Biomater ; 83: 400-413, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30465921

ABSTRACT

Multi-stimuli-responsive theranostic nanoplatform integrating functions of both imaging and multimodal therapeutics holds great promise for improving diagnosis and therapeutic efficacy. In this study, we reported a pH, glutathione (GSH) and hyaluronidase (HAase) triple-responsive nanoplatform for HER2 and CD44 dual-targeted and fluorescence imaging-guided PDT/PTT dual-therapy against HER2-overexpressed breast cancer. The nanoplatform was fabricated by functionalizing gold nanorods (GNRs) with hyaluronic acid (HA) bearing pendant hydrazide and thiol groups via Au-S bonds, and subsequently chemically conjugating 5-aminolevulinic acid (ALA), Cy7.5 and anti-HER2 antibody onto HA moiety for PDT, fluorescence imaging and active targeting, respectively. The resulting versatile nanoplatform GNR-HA-ALA/Cy7.5-HER2 had uniform sizes, favorable dispersibility, as well as pH, GSH and HAase triple-responsive drug release manner. In vitro studies demonstrated that HER2 and CD44 receptor-mediated dual-targeting strategy could significantly enhance the cellular uptake of GNR-HA-ALA/Cy7.5-HER2. Under near-infrared (NIR) irradiation, MCF-7 cells could efficiently generate reactive oxygen species (ROS) and heat, and be more efficiently killed by a combination of PDT and PTT as compared with individual therapy. Pharmacokinetic and biodistribution studies showed that the nanoplatform possessed a circulation half-life of 1.9 h and could be specifically delivered to tumor tissues with an accumulation ratio of 12.8%. Upon the fluorescence imaging-guided PDT/PTT treatments, the tumors were completely eliminated without obvious side effects. The results suggest that the GNR-HA-ALA/Cy7.5-HER2 holds great potential for breast cancer therapy. STATEMENT OF SIGNIFICANCE: A combination of photodynamic therapy (PDT) and photothermal therapy (PTT) is emerging as a promising cancer treatment strategy. However, its therapeutic efficacy is compromised by the nonspecific delivery and unintended release of photo-responsive agents. Herein, we developed a multifunctional theranostic nanoplatform GNR-HA-ALA/Cy7.5-HER2 with pH, glutathione and hyaluronidase triple-responsive drug release for HER2 and CD44 dual-targeted and fluorescence imaging-guided PDT/PTT therapy against breast cancer. We demonstrated that HER2 and CD44 receptors-mediated dual-targeting strategy significantly enhanced the cellular uptake of GNR-HA-ALA/Cy7.5-HER2. We also demonstrated that the combined PDT/PTT treatment had significantly superior antitumor effect than PDT or PTT alone both in vitro and in vivo. Therefore, GNR-HA-ALA/Cy7.5-HER2 could serve as a promising nanoplatform for HER2-positive breast cancer therapy.


Subject(s)
Breast Neoplasms , Gold , Hyaluronic Acid , Hyperthermia, Induced , Metal Nanoparticles , Nanotubes/chemistry , Photochemotherapy , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/pharmacokinetics , Aminolevulinic Acid/pharmacology , Animals , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Female , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacokinetics , Hyaluronic Acid/pharmacology , MCF-7 Cells , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
19.
Acta Biomater ; 82: 171-183, 2018 12.
Article in English | MEDLINE | ID: mdl-30336271

ABSTRACT

Integration of multimodal therapies into one nanoplatform holds great promise to overcome the drawbacks of conventional single-modal therapy and pursues enhanced anticancer efficacy. Herein, we developed a PEGylated gold nanorods (GNRs)-based nanoplatform (GNRs-MPH-ALA/DOX-PEG) with pH-responsive drug release property for triple-combined chemotherapy (CT), photodynamic therapy (PDT) and photothermal therapy (PTT) of breast cancer. GNRs were first decorated with mercaptopropionylhydrazide (MPH) and thiol-terminated monomethoxyl poly(ethylene glycol) (mPEG-SH) via Au-thiol linkage, and subsequently conjugated with chemotherapeutant doxorubicin (DOX) and pro-photosensitizer 5-aminolevulinic acid (ALA) through acid-liable hydrazone bonds between drugs and MPH molecules. The resulting nanoplatform GNRs-MPH-ALA/DOX-PEG exhibited excellent stability in physiological solutions and pH-responsive DOX and ALA release behaviors. In vitro studies showed that GNRs-MPH-ALA/DOX-PEG could efficiently enter human breast cancer MCF-7 cells and release DOX and ALA into cytoplasm. Furthermore, DOX could locate in the cell nucleus and ALA was productively metabolized into protoporphyrin IX (PpIX). Upon near-infrared (NIR) irradiation, PpIX produced enough reactive oxygen species for PDT and meanwhile GNRs could efficiently induce hyperthermia for PTT. Compared with single CT and dual-modal CT/PDT or CT/PTT treatment, the triple-combined CT/PDT/PTT treatment could more efficiently kill MCF-7 cells via a superadditive antitumor effect. Furthermore, the circulation half-life of GNRs-MPH-ALA/DOX-PEG in the blood was as long as approximately 52 min and it exhibited a tumor accumulation of 3.3%. The triple-combined CT/PDT/PTT treatment could completely suppress tumor growth without obvious systemic toxicity. Our study paves a new avenue for multimodal therapy of breast cancer. STATEMENT OF SIGNIFICANCE: The development of a simple but effective strategy to construct a versatile nanoplatform for multi-combined therapy still remains an enormous challenge. In this work, we developed a novel and simple nanoplatform GNRs-MPH-ALA/DOX-PEG with pH-responsive drug release for triple-combined chemotherapy (CT), photodynamic therapy (PDT) and photothermal therapy (PTT) of breast cancer. The nanoplatform could be efficiently internalized by MCF-7 cells. The intracellular GNRs-MPH-ALA/DOX-PEG could release DOX for CT, induce hyperthermia for PTT and generate high levels of ROS for PTT. Compared with single CT and dual-modal CT/PDT or CT/PTT treatments, the triple-combined CT/PDT/PTT treatment could more efficiently kill MCF-7 cells via a superadditive antitumor effect. Furthermore, upon triple-combined CT/PDT/PTT treatment, the tumor growth was completely suppressed without obvious systemic toxicity.


Subject(s)
Breast Neoplasms , Gold , Hyperthermia, Induced , Metal Nanoparticles , Nanotubes/chemistry , Photochemotherapy , Photosensitizing Agents , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Female , Gold/chemistry , Gold/pharmacology , Humans , MCF-7 Cells , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Xenograft Model Antitumor Assays
20.
ACS Appl Mater Interfaces ; 9(42): 36533-36547, 2017 Oct 25.
Article in English | MEDLINE | ID: mdl-28975790

ABSTRACT

Tumor-targeted delivery of photothermal agent and controlled release of concomitant chemotherapeutic drug are two key factors for combined photothermal chemotherapy. Herein, we developed a pH/near-infrared (NIR) dual-triggered drug release nanoplatform based on hyaluronic acid (HA)-functionalized gold nanorods (GNRs) for actively targeted synergetic photothermal chemotherapy of breast cancer. Targeting folate (FA), dopamine, and adipic acid dihydrazide triconjugated HA was first synthesized and used to decorate GNRs via Au-catechol bonds, and then an anticarcinogen doxorubicin (DOX) was conjugated onto HA moieties via an acid-labile hydrazone linkage, resulting in multifunctional nanoparticles GNRs-HA-FA-DOX. The nanoparticles exhibited excellent stability and had a pH and NIR dual-responsive drug release behavior. In vitro studies showed that the nanoparticles could be efficiently internalized into breast cancer MCF-7 cells and kill them under NIR irradiation in a synergistic fashion via inducing cell apoptosis. Pharmacokinetics and biodistribution studies in tumor-bearing mice indicated that the nanoparticles had a long blood circulation with a half-life of 2.4 h and exhibited a high accumulation of 11.3% in tumor site. The tumors of mice treated with combined chemotherapy and photothermal therapy were completely suppressed without obvious systemic toxicity after 20 d of treatment. These results demonstrated a great potential of GNRs-HA-FA-DOX nanoparticles for targeted synergistic therapy of breast cancer.


Subject(s)
Nanotubes , Animals , Breast Neoplasms , Doxorubicin , Drug Delivery Systems , Drug Liberation , Gold , Humans , Hyaluronic Acid , Hydrogen-Ion Concentration , Mice , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL