Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Acta Biomater ; 181: 308-316, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38570107

ABSTRACT

Acute lung injury (ALI) represents a critical respiratory condition typified by rapid-onset lung inflammation, contributing to elevated morbidity and mortality rates. Central to ALI pathogenesis lies macrophage dysfunction, characterized by an overabundance of pro-inflammatory cytokines and a shift in metabolic activity towards glycolysis. This study emphasizes the crucial function of glucose metabolism in immune cell function under inflammatory conditions and identifies hexokinase 2 (HK2) as a key regulator of macrophage metabolism and inflammation. Given the limitations of HK2 inhibitors, we propose the CRISPR/Cas9 system for precise HK2 downregulation. We developed an aerosolized core-shell liposomal nanoplatform (CSNs) complexed with CaP for efficient drug loading, targeting lung macrophages. Various CSNs were synthesized to encapsulate an mRNA based CRISPR/Cas9 system (mCas9/gHK2), and their gene editing efficiency and HK2 knockout were examined at both gene and protein levels in vitro and in vivo. The CSN-mCas9/gHK2 treatment demonstrated a significant reduction in glycolysis and inflammation in macrophages. In an LPS-induced ALI mouse model, inhaled CSN-mCas9/gHK2 mitigated the proinflammatory tumor microenvironment and reprogrammed glucose metabolism in the lung, suggesting a promising strategy for ALI prevention and treatment. This study highlights the potential of combining CRISPR/Cas9 gene editing with inhalation delivery systems for effective, localized pulmonary disease treatment, underscoring the importance of targeted gene modulation and metabolic reprogramming in managing ALI. STATEMENT OF SIGNIFICANCE: This study investigates an inhalable CRISPR/Cas9 gene editing system targeting pulmonary macrophages, with the aim of modulating glucose metabolism to alleviate Acute Lung Injury (ALI). The research highlights the role of immune cell metabolism in inflammation, as evidenced by changes in macrophage glucose metabolism and a notable reduction in pulmonary edema and inflammation. Additionally, observed alterations in macrophage polarization and cytokine levels in bronchoalveolar lavage fluid suggest potential therapeutic implications. These findings not only offer insights into possible ALI treatments but also contribute to the understanding of immune cell metabolism in inflammatory diseases, which could be relevant for various inflammatory and metabolic disorders.


Subject(s)
Acute Lung Injury , CRISPR-Cas Systems , Hexokinase , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Animals , Mice , Hexokinase/genetics , Hexokinase/metabolism , Mice, Inbred C57BL , Macrophages/metabolism , Macrophages/drug effects , Administration, Inhalation , Liposomes/chemistry , RAW 264.7 Cells , Male , Cellular Reprogramming/drug effects , Gene Editing , Glycolysis/drug effects
2.
Int J Pharm ; 625: 122076, 2022 Sep 25.
Article in English | MEDLINE | ID: mdl-35931394

ABSTRACT

Small cell lung cancer (SCLC), considered a mortal recalcitrant cancer, is a severe healthcare issue because of its poor prognosis, early metastasis, drug resistance and limited clinical treatment options. In our previous study, we established a MRP1-targeted antibody-IR700 system (Mab-IR700) for near infrared photoimmunotherapy (NIR-PIT) which exhibited a promising therapeutic effect on drug resistant H69AR cells both in vitro and in vivo, though the tumor growth suppression effect did not last long with a single round of PIT treatment. To achieve a better anticancer effect, we have combined Mab-IR700-mediated NIR-PIT with liposomal doxorubicin (Doxil®) and investigated the in vitro and in vivo cytotoxicity by using a H69AR/3T3 cell co-culture model in which 3T3 cells were used to mimic stromal cells. Cytotoxicity experiments demonstrated the specificity of Mab-IR700 to H69AR cells, while cytotoxicity and flow cytometry experiments confirmed that H69AR cells were doxorubicin-resistant. Compared with Mab-IR700-mediated PIT or Doxil-mediated chemotherapy, the combination therapy exhibited the best cell killing effect in vitro and superior tumor growth inhibition and survival prolongation effect in vivo. Super enhanced permeability and retention (SUPR) effect was observed in both co-culture spheroids and tumor-bearing mice. Owing to an approximately 9-fold greater accumulation of Doxil within the tumors, NIR-PIT combined with Doxil resulted in enhanced antitumor effects compared to NIR-PIT alone. This photoimmunochemotherapy is a practical strategy for the treatment of chemoresistant SCLC and should be further investigated for clinical translation.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Animals , Cell Line, Tumor , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Immunotherapy/methods , Lung Neoplasms/drug therapy , Mice , Multidrug Resistance-Associated Proteins , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Phototherapy/methods , Polyethylene Glycols , Small Cell Lung Carcinoma/drug therapy , Xenograft Model Antitumor Assays
3.
Sci Transl Med ; 14(648): eabh1261, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35675434

ABSTRACT

Tumor evasion of immune destruction is associated with the production of immunosuppressive adenosine in the tumor microenvironment (TME). Anticancer therapies can trigger adenosine triphosphate (ATP) release from tumor cells, causing rapid formation of adenosine by the ectonucleotidases CD39 and CD73, thereafter exacerbating immunosuppression in the TME. The goal of this study was to develop an approach to facilitate cancer therapy-induced immunogenic cell death including ATP release and to limit ATP degradation into adenosine, in order to achieve durable antitumor immune response. Our approach was to construct reactive oxygen species (ROS)-producing nanoparticles that carry an ectonucleotidase inhibitor ARL67156 by electronic interaction and phenylboronic ester. Upon near-infrared irradiation, nanoparticle-produced ROS induced ATP release from MOC1 cancer cells in vitro and triggered the cleavage of phenylboronic ester, facilitating the release of ARL67156 from the nanoparticles. ARL67156 prevented conversion of ATP to adenosine and enhanced anticancer immunity in an MOC1-based coculture model. We tested this approach in mouse tumor models. Nanoparticle-based ROS-responsive drug delivery reprogramed the immunogenic landscape in tumors, eliciting tumor-specific T cell responses and tumor regression, conferring long-term survival in mouse models. We demonstrated that TME reprograming sets the stage for response to anti-programmed cell death protein 1 (PD1) immunotherapy, and the combination resulted in tumor regression in a 4T1 breast cancer mouse model that was resistant to PD1 blockade. Furthermore, our approach also induced immunological effects in patient-derived organotypic tumor spheroid model, suggesting potential translation of our nanoparticle approach for treating human cancers.


Subject(s)
Nanoparticles , Neoplasms , Adenosine/pharmacology , Adenosine/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Cell Line, Tumor , Esters , Humans , Immunosuppression Therapy , Mice , Neoplasms/drug therapy , Reactive Oxygen Species , Tumor Microenvironment
4.
Nat Biomed Eng ; 5(11): 1306-1319, 2021 11.
Article in English | MEDLINE | ID: mdl-34725506

ABSTRACT

Antigen release resulting from the death of tumour cells induced by chemotherapies and targeted therapies can augment the antitumour responses induced by immune checkpoint blockade (ICB). However, tumours responding to ICB therapies often become resistant to them. Here we show that the specific targeting of tumour cells promotes the growth of tumour-cell variants that are resistant to ICB, and that the acquired resistance can be overcome via the concurrent depletion of tumour cells and of major types of immunosuppressive cell via a monoclonal antibody binding the enzyme CD73, which we identified as highly expressed on tumour cells and on regulatory T cells, myeloid-derived suppressor cells and tumour-associated macrophages, but not on cytolytic T lymphocytes, natural killer cells and dendritic cells. In mice with murine tumours, the systemic administration of anti-PD1 antibodies and anti-CD73 antibodies conjugated to a near-infrared dye prevented near-infrared-irradiated tumours from acquiring resistance to ICB and resulted in the eradication of advanced tumours. The elimination of immunosuppressive cells may overcome acquired resistance to ICB across a range of tumour types and combination therapies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Drug Resistance, Neoplasm , Neoplasms , Programmed Cell Death 1 Receptor , 5'-Nucleotidase/antagonists & inhibitors , Animals , Killer Cells, Natural , Mice , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Regulatory
5.
Sheng Wu Gong Cheng Xue Bao ; 37(9): 3088-3100, 2021 Sep 25.
Article in Chinese | MEDLINE | ID: mdl-34622619

ABSTRACT

Photoimmunotherapy (PIT) is an emerging tumor-targeted phototherapy that combines the tumor specificity of monoclonal antibodies with the phototoxicity of light absorbers to rapidly and selectively induce the immunogenic death of target tumor cells. PIT has minimal side effects due to its high specificity. The immunogenic cell death induced by PIT results in rapid maturation of immature dendritic cells proximal to dying tumor cells. Subsequently, the mature dendritic cells present the tumor antigens to CD8+ T cells and induce their activation and proliferation, thus enhancing the antitumor immune response of the host. PIT can also improve the anticancer efficacy by enhancing the penetration of nanomedicines into tumor tissues. In view of the excellent application prospects of PIT, this review summarizes the advances in the immune activation mechanism of PIT, the superenhanced permeability and retention effect, and the new strategies for combinatory therapy, providing references for future research and clinical translation.


Subject(s)
Neoplasms , Photosensitizing Agents , Antibodies, Monoclonal/therapeutic use , Humans , Immunotherapy , Neoplasms/therapy , Phototherapy
6.
Int J Pharm ; 604: 120760, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34077781

ABSTRACT

Small cell lung cancer (SCLC), one of the most aggressive cancers, has a high mortality rate and poor prognosis, and the clinical therapeutic outcomes of multidrug resistant SCLC are even worse. Multidrug resistance protein 1 (MRP1), one of the ATP-binding cassette (ABC) transporter proteins that cause decreased drug accumulation in cancer cells, is overexpressed in drug resistant SCLC cells and could be a promising target for treating the patients suffering from this illness. Near infrared photoimmunotherapy (NIR-PIT) is a newly developed approach for targeted cancer treatment which uses a conjugate of a monoclonal antibody and photoabosorber IR700 followed by NIR light irradiation to induce rapid cancer cell death. In the present study, an anti-MRP1 antibody (Mab) -IR700 conjugate (Mab-IR700) was synthesized, purified and used to treat chemoresistant SCLC H69AR cells that overexpressed MRP1, while non-MRP1-expressing H69 cells were used as a control. Then, the photokilling and tumor suppression effect were separately evaluated in H69AR cells both in vitro and in vivo. Higher cellular delivery of Mab-IR700 was detected in H69AR cells, whereas there was little uptake of IgG-IR700 in both H69 and H69AR cells. Due to the targeting activity of Mab, stronger photokilling effect was found both in H69AR cells and spheroids treated with Mab-IR700, while superior tumor suppression effect was also observed in the mice treated with Mab-IR700 and light illumination. Photoacoustic imaging results proved that oxygen was involved in NIR-PIT treatment, and TUNEL staining images showed the occurrence of cell apoptosis, which was also testified by HE staining. This research provides MRP1 as a novel target for PIT and presents a prospective way for treating drug resistant SCLC and, thus, should be further studied.


Subject(s)
Lung Neoplasms , Pharmaceutical Preparations , Small Cell Lung Carcinoma , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Animals , Cell Line, Tumor , Humans , Immunotherapy , Infrared Rays , Lung Neoplasms/drug therapy , Mice , Photosensitizing Agents , Phototherapy , Prospective Studies , Small Cell Lung Carcinoma/drug therapy , Xenograft Model Antitumor Assays
7.
Biomater Sci ; 6(11): 3063-3074, 2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30298866

ABSTRACT

P-glycoprotein (Pgp) has been considered as a major cause of cancer multidrug resistance; however, clinical solutions to overcome this drug resistance do not exist despite the tremendous endeavors. The lack of cancer specificity is a main reason for clinical failure of conventional approaches. Targeted photodynamic therapy (PDT) is highly cancer specific by combining antibody targeting and locoregional light irradiation. We aimed to develop Pgp-targeted PDT using antibody-photosensitizer conjugates made of a recombinant Fab fragment. We prepared the photosensitizer conjugates by expressing a recombinant Fab fragment and specifically linking IR700-maleimide at the C-terminal of the Fab heavy chain. In vitro studies showed that the Fab conjugates specifically bind to Pgp. Their phototoxicity was comparable to full antibody conjugates when assayed with conventional 2-D cell culture, but they outperformed the full antibody conjugates in a 3-D tumor spheroid model. In a mouse xenograft model of chemoresistant tumors, Fab conjugates showed Pgp specific delivery to chemoresistant tumors. Upon irradiation with near-infrared light, they caused rapid tumor shrinkage and significantly prolonged the survival of tumor-bearing mice. Compared to the full antibody conjugates, Fab conjugates took a shorter time to reach peak tumor levels and achieved a more homogeneous tumor distribution. This allows light irradiation to be initiated at a shorter time interval after the conjugate injection, and thus may facilitate clinical translation. We conclude that our targeted PDT approach provides a highly cancer-specific approach to combat chemoresistant tumors, and that the conjugates made of recombinant antibody fragments are superior to full antibody conjugates for targeted PDT.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibodies/chemistry , Antineoplastic Agents/chemistry , Drug Resistance, Neoplasm , Immunoglobulin Fab Fragments/chemistry , Photochemotherapy/methods , 3T3 Cells , Animals , Antibodies/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Combined Modality Therapy/methods , Drug Resistance, Multiple , Female , Heterografts , Humans , Immunoglobulin Fab Fragments/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/diagnostic imaging , Neoplasms/pathology , Neoplasms/therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tissue Distribution
8.
ACS Appl Mater Interfaces ; 10(39): 33464-33473, 2018 Oct 03.
Article in English | MEDLINE | ID: mdl-30188117

ABSTRACT

P-Glycoprotein (Pgp)-medicated multidrug resistance (MDR) remains a formidable challenge to cancer therapy. As conventional approaches using small-molecule inhibitors failed in clinical development because of the lack of cancer specificity, we develop Pgp-targeted carbon nanotubes to achieve highly cancer-specific therapy through combining antibody-based cancer targeting and locoregional tumor ablation with photothermal therapy. Through a dense coating with phospholipid-poly(ethylene glycol), we have engineered multiwalled carbon nanotubes (MWCNTs) which show minimum nonspecific cell interactions and maximum intercellular diffusion. After chemically modifying with an anti-Pgp antibody, these MWCNTs showed highly Pgp-specific cellular uptake. Treatment of the targeted MWCNTs caused dramatic cytotoxicity in MDR cancer cells upon photoirradiation, whereas they did not cause any toxicity in the dark or phototoxicity toward normal cells that do not express Pgp. Because of excellent intratumor diffusion and Pgp-specific cellular uptake, the targeted MWCNTs produced strong phototoxicity in tumor spheroids of MDR cancer cells, a 3-D tumor model for studying tumor penetration and therapy. In conclusion, we have developed highly Pgp-specific MWCNTs that may provide an effective therapy for MDR cancers where other approaches have failed.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Nanotubes, Carbon/chemistry , Photochemotherapy/methods , Animals , Cell Line , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Flow Cytometry , Mice , Phospholipids/metabolism , Polyethylene Glycols/chemistry , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology
9.
J Control Release ; 286: 289-300, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30081143

ABSTRACT

Drug resistance remains a formidable challenge to cancer therapy. P-glycoprotein (Pgp) contributes to multidrug resistance in numerous cancers by preventing accumulation of anticancer drugs in cancer cells. Strategies to overcome this resistance have been vigorously sought for over 3 decades, yet clinical solutions do not exist. The main reason for the failure is lack of cancer specificity of small-molecule Pgp inhibitors, thus causing severe toxicity in normal tissues. In this study, Pgp-targeted photodynamic therapy (PDT) was developed to achieve superior cancer specificity through antibody targeting plus locoregional light activation. Thus, a Pgp monoclonal antibody was chemically modified with IR700, a porphyrin photosensitizer. In vitro studies showed that the antibody-photosensitizer conjugates specifically bind to Pgp-expressing drug resistant cancer cells, and caused dramatic cytotoxicity upon irradiation with a near infrared light. We then tested our Pgp-targeted approach in mouse xenograft models of chemoresistant ovarian cancer and head and neck cancer. In both models, targeted PDT produced rapid tumor shrinkage, and significantly prolonged survival of tumor-bearing mice. We conclude that our targeted PDT approach produces molecularly targeted and spatially selective ablation of chemoresistant tumors, and thereby provides an effective approach to overcome Pgp-mediated multidrug resistance in cancer, where conventional approaches have failed.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Drug Delivery Systems/methods , Immunoconjugates/administration & dosage , Neoplasms/drug therapy , Photosensitizing Agents/administration & dosage , Animals , Cell Line, Tumor , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Infrared Rays , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/metabolism , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use , Xenograft Model Antitumor Assays
10.
Mol Pharm ; 15(7): 2559-2569, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29764120

ABSTRACT

Photodynamic therapy, a procedure that uses a photosensitizer to enable light therapy selectively at diseased sites, remains underutilized in oncological clinic. To further improve its cancer selectivity, we developed a polymeric nanosystem by conjugating a photosensitizer IRDye 700DX (IR700) and cancer targeting RGD peptide to 8-arm polyethylene glycol (PEG). The resulting nanoconjugates (RGD-8PEG-IR700) exhibited a hydrodynamic size of 6.6 nm with narrow distribution of size. The targeted nanoconjugates showed significantly higher intracellular uptake of IR700 in integrin αvß3-expressing A375 and SKOV3 cells when compared with nontargeted control 8PEG-IR700, and an excess amount of RGD peptides could abolish this enhancement, indicating a receptor-mediated uptake mechanism for the targeted polymer conjugates. Phototoxicity studies indicated that RGD-8PEG-IR700 produced massive cell killing in A375 cells after photoirradiation with an IC50 value of 57.8 nM for IR700. In contrast, free IR700 and the control 8PEG-IR700 conjugates did not produce any phototoxicity at the concentrations up to 1 µM IR700. Upon photoirradiation, the RGD-8PEG-IR700 could produce sufficient singlet oxygen in the cells and induced cell apoptosis. The studies with three-dimensional tumor spheroids showed that they penetrated tumor spheroids deeply and produced strong phototoxicity. Thus, we conclude that the polymer nanoconjugates may provide a promising delivery system for targeted photodynamic therapy of cancers due to their small size, cancer cell specificity, and minimal side effects.


Subject(s)
Nanoconjugates/chemistry , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , 3T3 Cells , Animals , Apoptosis/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Mice , Neoplasms/pathology , Oligopeptides/administration & dosage , Polyethylene Glycols/chemistry , Spheroids, Cellular
11.
Nucleic Acids Res ; 46(4): 1601-1613, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29361039

ABSTRACT

The pharmacological effects of antisense and siRNA oligonucleotides are hindered by the tendency of these molecules to become entrapped in endomembrane compartments thus failing to reach their targets in the cytosol or nucleus. We have previously used high throughput screening to identify small molecules that enhance the escape of oligonucleotides from intracellular membrane compartments and have termed such molecules OECs (oligonucleotide enhancing compounds). Here, we report on the structure-activity relationships of a family of OECs that are analogs of a hit that emerged from our original screen. These studies demonstrate key roles for the lipophilic aromatic groups, the tertiary nitrogen, and the carbamate moiety of the parent compound. We have also investigated the intracellular site of action of the OECs and have shown that activity is due to the release of oligonucleotides from intermediate endosomal compartments rather than from early endosomes or from highly acidic downstream compartments. At high concentrations of OECs toxicity occurs in a manner that is independent of caspases or of lysosomal cathepsins but instead involves increased plasma membrane permeability. Thus, in addition to describing specific characteristics of this family of OECs, the current study provides insights into basic mechanisms of oligonucleotide trafficking and their implications for oligonucleotide delivery.


Subject(s)
Oligonucleotides/metabolism , Pyrazines/pharmacology , Pyridines/pharmacology , HeLa Cells , Humans , Intracellular Membranes/drug effects , Oligonucleotides/analysis , Pyrazines/chemistry , Pyridines/chemistry , Structure-Activity Relationship
12.
Theranostics ; 8(22): 6274-6290, 2018.
Article in English | MEDLINE | ID: mdl-30613297

ABSTRACT

Cancer nanomedicines only modestly improve the overall survival of patients because their anticancer activity is limited by biological barriers posed by the tumor microenvironment. Currently, all the drugs in FDA-approved cancer nanomedicines are substrates for P-glycoprotein (Pgp), and thus, Pgp-mediated multidrug resistance (MDR) remains a hurdle for cancer nanomedicines. Methods: In this study, Pgp-targeted photodynamic therapy (PDT) was developed to enhance the anticancer efficacy of nanomedicines by depleting MDR cancer cells as well as enhancing tumor penetration of nanomedicines. We first examined the Pgp specificity of our targeted PDT approach, and then tested combination therapy of PDT with Doxil in mixed tumor models of MDR cancer cells and stromal cells, mimicking human heterogeneous tumors. Results:In vitro studies showed that the antibody-photosensitizer conjugates produced Pgp-specific cytotoxicity towards MDR cancer cells upon irradiation with a near-infrared light. The studies with a co-culture model of MDR cancer cells and stromal cells revealed synergistic effects in the combination therapy of PDT with Doxil. Using a mouse model of mixed tumors containing MDR cancer cells and stroma cells, we observed markedly enhanced tumor delivery of Doxil after PDT in vivo. We further examined the effects of the two modalities on individual cell populations and their synergism using an in vivo dual substrate bioluminescence assay. The results indicated that Pgp-targeted PDT specifically depleted MDR cancer cells and further enhanced Doxil's actions on both MDR cancer cells and stromal cells. Conclusion: We conclude that our targeted PDT approach markedly enhances anticancer actions of nanomedicines by depleting MDR cancer cells and increasing their tumor penetration, and thereby, may provide an effective approach to facilitate translation of cancer nanomedicines.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/administration & dosage , Tumor Microenvironment/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Cell Line, Tumor , Combined Modality Therapy , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanomedicine , Polyethylene Glycols/administration & dosage
13.
Mol Pharm ; 14(10): 3391-3398, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28813596

ABSTRACT

Overexpression of P-glycoprotein (Pgp) has been considered a primary cause for multidrug resistance in a variety of cancers for three decades. However, clinical translation of Pgp targeted therapeutics has been hindered by lack of patient preselection based on the Pgp presence in tumors. We aim to develop a molecularly targeted probe for imaging tumoral Pgp in vivo with positron emission tomography (PET) and fluorescence, and to provide a tool for preselecting the patients with tumoral Pgp expression. Thus, a Pgp monoclonal antibody 15D3 was chemically modified with IRDye800 (IR800) and DOTA chelator. The specificity of the antibody conjugates DOTA-Pab-IR800 was verified in Pgp-expressing 3T3-MDR1 and control 3T3 cells. After radiolabeling with 64Cu, the probe was applied in small animal PET imaging of Pgp in a mouse xenograft model of NCI/ADR-Res cells, which are chemoresistant through overexpression of Pgp. Quantification analysis of the PET images demonstrated that the tumor uptake of the radioactive probe was 9.9 ± 1.4, 12.1 ± 1.2, and 10.5 ± 1.0%ID/g at 4, 24, and 48 h post injection. The tumor-to-muscle ratio was 20.9 at 48 h post injection based on biodistribution studies. Fluorescence imaging was performed following PET experiments, and it demonstrated excellent tumor accumulation of this dual-modality probe in the NCI/ADR-Res tumors. Further, an image-guided surgery was successfully performed using the fluorescence modality of the probe, demonstrating potential utility of this probe in image-guided surgical removal of Pgp-positive drug resistant tumors in the patients. In conclusion, this study clearly demonstrated that the Pgp-targeted antibody probe, 64Cu-DOTA-Pab-IR800, could provide a promising diagnosis tool for detection of Pgp-expressing tumors in vivo.


Subject(s)
Antibodies, Monoclonal/chemistry , Molecular Imaging/methods , Molecular Probes/chemistry , Neoplasm Proteins/metabolism , Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemistry , ATP Binding Cassette Transporter, Subfamily B/immunology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Chelating Agents/chemistry , Copper Radioisotopes/chemistry , Drug Resistance, Neoplasm , Female , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Hybridomas , Indoles/chemistry , Mice , Mice, Nude , Molecular Probes/immunology , Molecular Probes/pharmacology , Neoplasm Proteins/immunology , Neoplasms/immunology , Neoplasms/pathology , Optical Imaging/methods , Organometallic Compounds/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays
14.
Mol Pharm ; 14(8): 2793-2804, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28700237

ABSTRACT

Advances in photodynamic therapy of cancer have been restrained by lack of cancer specificity and side effects to normal tissues. Molecularly targeted photodynamic therapy can achieve higher cancer specificity by combination of active cancer targeting and localized laser activation. We aimed to use albumin as a carrier to prepare targeted nanoconjugates that are selective to cancer cells and smaller than conventional nanoparticles for superior tumor penetration. IRDye 700DX (IR700), a porphyrin photosensitizer, was covalently conjugated to human serum albumin that was also linked with tumor-targeting RGD peptides. With multiple IR700 and RGD molecules in a single albumin molecule, the resultant nanoconjugates demonstrated monodispersed and uniform size distribution with a diameter of 10.9 nm. These targeted nanoconjugates showed 121-fold increase in cellular delivery of IR700 into TOV21G ovarian cancer cells compared to control nanoconjugates. Mechanistic studies revealed that the integrin specific cellular delivery was achieved through dynamin-mediated caveolae-dependent endocytosis pathways. They produced massive cell killing in TOV21G cells at low nanomolar concentrations upon light irradiation, while NIH/3T3 cells that do not express integrin αvß3 were not affected. Because of their small size, targeted albumin nanoconjugates could penetrate tumor spheroids of SKOV-3 ovarian cancer cells and produced strong phototoxicity in this 3-D model. Owing to their cancer-specific delivery and small size, these targeted nanoconjugates may become an effective drug delivery system for enabling molecularly targeted photodynamic therapy of cancer.


Subject(s)
Albumins/chemistry , Nanoconjugates/chemistry , Nanoparticles/chemistry , Photosensitizing Agents/chemistry , Photochemotherapy
15.
Angew Chem Int Ed Engl ; 56(5): 1319-1323, 2017 01 24.
Article in English | MEDLINE | ID: mdl-27981718

ABSTRACT

Efficient delivery of biomacromolecules (e.g., proteins, nucleic acids) into cell cytosol remains a critical challenge for the development of macromolecular therapeutics or diagnostics. To date, most common approaches to assess cytosolic delivery rely on fluorescent labeling of macromolecules with an "always on" reporter and subcellular imaging of endolysosomal escape by confocal microscopy. This strategy is limited by poor signal-to-noise ratio and only offers low throughput, qualitative information. Herein we describe a quantitative redox-activatable sensor (qRAS) for the real-time monitoring of cytosolic delivery of macromolecules. qRAS-labeled macromolecules are silent (off) inside the intact endocytic organelles, but can be turned on by redox activation after endolysosomal disruption and delivery into the cytosol, thereby greatly improving the detection accuracy. In addition to confocal microscopy, this quantitative sensing technology allowed for a high-throughput screening of a panel of polymer carriers toward efficient cytosolic delivery of model proteins on a plate reader. The simple and versatile qRAS design offers a useful tool for the investigation of new strategies for endolysosomal escape of biomacromolecules to facilitate the development of macromolecular therapeutics for a variety of disease indications.


Subject(s)
Cytosol/metabolism , Drug Delivery Systems/methods , Macromolecular Substances/metabolism , A549 Cells , Endosomes/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Glutathione/chemistry , Glutathione/metabolism , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Macromolecular Substances/chemistry , Microscopy, Confocal , Oxidation-Reduction , Phosphines/chemistry
16.
Biomed Res Int ; 2016: 7478650, 2016.
Article in English | MEDLINE | ID: mdl-27579319

ABSTRACT

Liver fibrosis is a major cause of morbidity and mortality worldwide due to chronic viral hepatitis and, more recently, from fatty liver diseases. Activation and proliferation of hepatic stellate cells (HSCs) represent a key aspect of fibrogenesis and are associated with progressive reduction of HSC apoptosis. Bcl-x, an antiapoptotic member of Bcl-2 gene family, plays a role in apoptosis regulation in mammalian cells. Through alternative splicing, the Bcl-x gene yields two major protein isoforms with opposing functions, antiapoptotic Bcl-xL and proapoptotic Bcl-xS. This study aimed to investigate the role of Bcl-x and its alternate splicing in HSC apoptosis. The results indicated that the expression of Bcl-xL was dramatically higher than Bcl-2 in activated human HSCs. The relative expression of Bcl-xL over Bcl-xS increased gradually when HSCs were activated in cell culture, which was consistent with the increase in apoptosis resistance of activated HSCs. Redirection of Bcl-x splicing by an antisense oligonucleotide from the antiapoptotic isoform to the proapoptotic isoform induced death of HSCs without other apoptosis stimuli. We conclude that Bcl-x plays a role in regulation of HSC apoptosis and modulation of Bcl-x alternative splicing may become a novel molecular therapy for liver fibrosis.


Subject(s)
Alternative Splicing/genetics , Apoptosis/genetics , Gene Expression Regulation/genetics , Hepatic Stellate Cells/physiology , bcl-X Protein/genetics , Cells, Cultured , Humans
17.
J Control Release ; 192: 114-21, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25016158

ABSTRACT

There is no effective clinical therapy yet for triple-negative breast cancer (TNBC) without particular human epidermal growth factor receptor-2, estrogen and progesterone receptor expression. In this study, we report a molecularly targeted and synthetic lethality-based siRNA therapy for TNBC treatment, using cationic lipid assisted poly(ethylene glycol)-b-poly(d,l-lactide) (PEG-PLA) nanoparticles as the siRNA carrier. It is demonstrated that only in c-Myc overexpressed TNBC cells, while not in normal mammary epithelial cells, delivery of siRNA targeting cyclin-dependent kinase 1 (CDK1) with the nanoparticle carrier (NPsiCDK1) induces cell viability decreasing and cell apoptosis through RNAi-mediated CDK1 expression inhibition, indicating the synthetic lethality between c-Myc with CDK1 in TNBC cells. Moreover, systemic delivery of NPsiCDK1 is able to suppress tumor growth in mice bearing SUM149 and BT549 xenograft and cause no systemic toxicity or activate the innate immune response, suggesting the therapeutic promise with such nanoparticles carrying siCDK1 for c-Myc overexpressed triple negative breast cancer.


Subject(s)
CDC2 Protein Kinase/genetics , Drug Carriers/chemistry , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , RNA Interference , RNA, Small Interfering/therapeutic use , Triple Negative Breast Neoplasms/therapy , Animals , Breast/metabolism , Breast/pathology , Cell Line, Tumor , Female , Genetic Therapy , Humans , Mice , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
18.
Biomaterials ; 35(26): 7622-34, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24929619

ABSTRACT

Systemic delivery of small interfering RNA (siRNA) into cancer cells remains the major obstacle to siRNA drug development. An ideal siRNA delivery vehicle for systemic administration should have long circulation time in blood, accumulate at tumor site, and sufficiently internalize into cancer cells for high-efficiency of gene silence. Herein, we report a core-shell Micelleplex delivery system that made from block copolymer bearing poly(ethylene glycol) (PEG), matrix metalloproteinase 2 (MMP-2)-degradable peptide PLG*LAG, cationic cell penetrating peptide polyarginine r9 and poly(ε-caprolactone) (PCL) for siRNA delivery. We show clear evidences in vitro and in vivo to prove that the micelle carrying siRNA can circulate enough time in blood, enrich accumulation at tumor sites, shed the PEG layer when triggered by tumor overexpressing MMP-2, and then the exposing cell penetrating peptide r9 enhanced cellular uptake of siRNA. Accordingly, this design strategy enhances the inhibition of breast tumor growth following systemic injection of this system carrying siRNA against Polo-like kinase 1, which demonstrating this Micelleplex can be a potential delivery system for systemic siRNA delivery in cancer therapy.


Subject(s)
Drug Carriers/metabolism , Matrix Metalloproteinase 2/metabolism , Peptides/metabolism , RNA, Small Interfering/administration & dosage , Transfection , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Cycle Proteins/genetics , Cell Line, Tumor , Drug Carriers/chemistry , Female , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Micelles , Molecular Sequence Data , Peptides/chemistry , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Polo-Like Kinase 1
19.
Mol Ther ; 22(5): 964-73, 2014 May.
Article in English | MEDLINE | ID: mdl-24496383

ABSTRACT

The KRAS mutation is present in ~20% of lung cancers and has not yet been effectively targeted for therapy. This mutation is associated with a poor prognosis in non-small-cell lung carcinomas (NSCLCs) and confers resistance to standard anticancer treatment drugs, including epidermal growth factor receptor tyrosine kinase inhibitors. In this study, we exploited a new therapeutic strategy based on the synthetic lethal interaction between cyclin-dependent kinase 4 (CDK4) downregulation and the KRAS mutation to deliver micellar nanoparticles (MNPs) containing small interfering RNA targeting CDK4 (MNPsiCDK4) for treatment in NSCLCs harboring the oncogenic KRAS mutation. Following MNPsiCDK4 administration, CDK4 expression was decreased, accompanied by inhibited cell proliferation, specifically in KRAS mutant NSCLCs. However, this intervention was harmless to normal KRAS wild-type cells, confirming the proposed mechanism of synthetic lethality. Moreover, systemic delivery of MNPsiCDK4 significantly inhibited tumor growth in an A549 NSCLC xenograft murine model, with depressed expression of CDK4 and mutational KRAS status, suggesting the therapeutic promise of MNPsiCDK4 delivery in KRAS mutant NSCLCs via a synthetic lethal interaction between KRAS and CDK4.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cyclin-Dependent Kinase 4/genetics , Genetic Therapy , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , ras Proteins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Humans , Mice , Nanoparticles/therapeutic use , Proto-Oncogene Proteins p21(ras) , RNA, Small Interfering/therapeutic use , Xenograft Model Antitumor Assays
20.
Biotechnol Adv ; 32(4): 789-803, 2014.
Article in English | MEDLINE | ID: mdl-23933109

ABSTRACT

pH-responsive nanoparticles (NPs) are currently under intense development as drug delivery systems for cancer therapy. Among various pH-responsiveness, NPs that are designed to target slightly acidic extracellular pH environment (pHe) of solid tumors provide a new paradigm of tumor targeted drug delivery. Compared to conventional specific surface targeting approaches, the pHe-targeting strategy is considered to be more general due to the common occurrence of acidic microenvironment in solid tumors. This review mainly focuses on the design and applications of pHe-activated NPs, with special emphasis on pHe-activated surface charge reversal NPs, for drug and siRNA delivery to tumors. The novel development of NPs described here offers great potential for achieving better therapeutic effects in cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Nanomedicine/methods , Nanoparticles/administration & dosage , Nanotechnology/methods , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Endocytosis , Female , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Mice , Nanoparticles/chemistry , Neoplasm Transplantation , RNA, Small Interfering/metabolism , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...