Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Adv Mater ; 36(40): e2407927, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39185788

ABSTRACT

The combination of nanoparticles and tumor-targeting bacteria for cancer immunotherapy can overcome the shortcomings of poor nanoparticle accumulation, limited penetration, and restricted distribution. However, it remains a great challenge for the hybrid system to improve therapeutic efficacy through the simultaneous and controllable regulation of immune cells and tumor cells. Herein, a hybrid therapeutic platform is rationally designed to achieve immune cascade-augmented cancer immunotherapy. To construct the hybrids, photothermal nanoparticles responsive to light in the second near-infrared (NIR-II) region are conjugated onto the surface of engineered bacteria through pH-responsive Schiff base bonds. Taking advantage of the hypoxia targeting and deep penetration characteristics of the bacteria, the hybrids can accumulate at tumor sites. Then nanoparticles detach from the bacteria to realize genetic engineering of tumor cells, which induces tumor cell apoptosis and down-regulate the expression of programmed cell death ligand 1 to alleviate immunosuppressive tumor microenvironment. The mild photothermal heating can not only induce tumor-associated antigen release, but also trigger sustainable expression of cytokine interleukin-2. Notably, a synergistic antitumor effect is achieved between the process of p53 transfection and NIR-II light-activated genetic engineering of bacteria. This work proposes a facile strategy for the construction of hybrid system to achieve cascade-augmented cancer immunotherapy.


Subject(s)
Genetic Engineering , Immunotherapy , Infrared Rays , Nanoparticles , Animals , Cell Line, Tumor , Nanoparticles/chemistry , Mice , Humans , Neoplasms/therapy , Apoptosis , Tumor Microenvironment
2.
Nano Lett ; 23(17): 8081-8090, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37615340

ABSTRACT

Since central cells are more malignant and aggressive in solid tumors, improving penetration of therapeutic agents and activating immunity in tumor centers exhibit great potential in cancer therapies. Here, polydopamine-coated Escherichia coli Nissle 1917 (EcN) bearing CRISPR-Cas9 plasmid-loaded liposomes (Lipo-P) are applied for enhanced immunotherapy in deep tumors through activation of innate and adaptive immunity simultaneously. After accumulation in the tumor center through hypoxia targeting, Lipo-P could be detached under the reduction of reactive oxygen species (ROS)-responsive linkers, lowering the thermal resistance of cancer cells via Hsp90α depletion. Owing to that, heating induced by polydopamine upon near-infrared irradiation could achieve effective tumor ablation. Furthermore, mild photothermal therapy induces immunogenic cell death, as bacterial infections in tumor tissues trigger innate immunity. This bacteria-assisted approach provides a promising photothermal-sensitized immunotherapy in deep tumors.


Subject(s)
Neoplasms , Probiotics , Humans , CRISPR-Cas Systems/genetics , Immunotherapy , Neoplasms/therapy , Escherichia coli/genetics , Liposomes
3.
ACS Nano ; 17(14): 13158-13175, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37436002

ABSTRACT

Tumour hypoxia plays an important role in modulating tumorigenesis, angiogenesis, invasion, immunosuppression, resistance to treatment, and even maintenance of the stemness of cancer stem cells (CSCs). Moreover, the targeting and treatment of hypoxic cancer cells and CSCs to reduce the influence of tumor hypoxia on cancer therapy remains an imperative clinical problem that needs to be addressed. Since cancer cells upregulate the expression of glucose transporter 1 (GLUT1) through the Warburg effect, we considered the possibility of GLUT1-mediated transcytosis in cancer cells and developed a tumor hypoxia-targeting nanomedicine. Our experimental results indicate that glucosamine-labeled liposomal ceramide can be efficiently transported between cancer cells by GLUT1 transporters and substantially accumulated in the hypoxic area in in vitro CSC spheroids and in vivo tumor xenografts. We also verified the effects of exogenous ceramide on tumor hypoxia, including important bioactivities such as upregulation of p53 and retinoblastoma protein (RB), downregulation of hypoxia-inducible factor-1 alpha (HIF-1α) expression, disruption of the OCT4-SOX2 network of stemness, and inhibition of CD47 and PD-L1 expression. To achieve an ideal therapeutic outcome, we combined treatment of glucosamine-labeled liposomal ceramide with paclitaxel and carboplatin, and we found an excellent synergistic effect, with tumor clearance being noted in three-fourths of the mice. Overall, our findings provide a potential therapeutic strategy for the treatment of cancer.


Subject(s)
Hypoxia , Neoplasms , Humans , Mice , Animals , Glucose Transporter Type 1/metabolism , Hypoxia/metabolism , Cell Hypoxia , Liposomes/pharmacology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Transcytosis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Line, Tumor , Neoplasms/pathology
4.
Colloids Surf B Biointerfaces ; 225: 113277, 2023 May.
Article in English | MEDLINE | ID: mdl-36996630

ABSTRACT

Poor drug penetration in hypoxia area of solid tumor is a big challenge for intestinal tumor therapy and thus it is crucial to develop an effective strategy to overcome this challenge. Compared with other bacteria used for construction of hypoxia targeted bacteria micro-robot, the Escherichia coli Nissle 1917 (EcN) bacteria are nonpathogenic Gram-negative probiotic and can especially target and identify the signal molecules in the hypoxic region of tumor, and thus, in this study, we choose EcN to construct a bacteria propelled micro-robot for targeting intestinal tumor therapy. Firstly, the MSNs@DOX with average diameter of 200 nm were synthesized and conjugated with EcN bacteria using EDC/NHS chemical crosslinking method to construct a EcN propelled micro-robot. The motility of micro-robot was then evaluated and the motion velocity of EcN-pMSNs@DOX was 3.78 µm/s. Compared with pMSNs@DOX without EcN driven, EcN bacteria propelled micro-robot transported much more pMSNs@DOX into the inner of HCT-116 3D multicellular tumor spheroids. However, the EcN bacteria are non-intracelluar bacteria which lead to the micro-robot can not directly enter into tumor cells. Therefore, we utilized acid-labile linkers of cis-aconitic amido bone to link EcN with MSNs@DOX nanoparticles to achieve the pH sensitive separation of EcN with MSNs@DOX from the micro-robot. At 4 h of incubation, the isolated MSNs@DOX began to enter into the tumor cells through CLSM observation. In vitro live/dead staining results show that EcN-pMSNs@DOX induced much more cell death than pMSNs@DOX at 24 and 48 h of incubation with HCT-116 tumor cells in acid culture media (pH 5.3). For the validation of the therapeutic efficacy of the micro-robot for intestinal tumor, we established the HCT-116 subcutaneous transplantation tumor model. After 28 days of treatment, EcN-pMSNs@DOX dramatically inhibit tumor growth with tumor volume was around 689 mm3, induce much more tumor tissues necrosis and apoptosis. Finally, the toxicity of this micro-robot was investigated by pathological analysis the liver and heart tissues. We expect that the pH sensitive EcN propelled micro-robot here we constructed may be a safe and feasible strategy for intestinal tumor therapy.


Subject(s)
Intestinal Neoplasms , Probiotics , Robotics , Humans , Escherichia coli , Hypoxia , Hydrogen-Ion Concentration
5.
Asian J Pharm Sci ; 17(6): 892-907, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36600894

ABSTRACT

Colorectal cancer is often accompanied by multiple organ metastasis. Anaerobic Bifidobacterium Infantis (BI) bacterial can selectively grow in hypoxic colorectal tumor microenvironment (TME), to own the natural advantage of preferentially colorectal tumor targeting. Herein, a self-guidance biological hybrid drug delivery system (BI-ES-FeAlg/DOX) based on BI was constructed to inhibit the proliferation and metastasis of colon cancer. Results demonstrated that BI-ES-FeAlg/DOX could overcome physical barriers to target and accumulate in colon tumor tissues. Then DOX was released to kill tumor cells along with the phase transition (solid to liquid) of FeAlg hydrogel, due to Fe3+ was reduced to Fe2+by intracellular GSH. Meanwhile, BI-ES selectively colonized into tumors and expressed endostatin (ES) protein to down-regulate VEGF and bFGF expression, exerting anti-angiogenic effect. Moreover, FeAlg catalyzed H2O2 in the local tumor to generate cytotoxic ·OH, further enhancing the antitumor effect. The pharmacodynamic result in AOM/DSS model proved that BI-ES-FeAlg/DOX had the best therapeutic effect, with the final V/V0 of 2.19 ±â€¯0.57, which was significantly lower than the other groups. Meanwhile, on CT-26 tumor-bearing model, it also showed an outstanding anti-tumor effect with inhibition rate of 82.12% ± 3.08%. In addition, lung metastases decreased significantly in tumor metastasis model after BI-ES-FeAlg/DOX treatment.

6.
Int J Pharm ; 605: 120817, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34166726

ABSTRACT

Bacteria-driven drug-delivery systems have drawn considerable interests for their highly selective hypoxia-targeting and efficacy in tumor inhibition. For the first time, a supramolecular biohybrid bacterium (SA@HU) is constructed by coating attenuated Salmonella typhimurium (S. typhimurium ΔppGpp/Lux) with nanoassemblies. In addition, the host-guest inclusion complexes based on hydroxypropyl-ß-cyclodextrin (HPCD) and amantadine (AMA) was developed to encapsulate the natural antineoplastic product, ursolic acid (UA). It is found that the drug-carried coating layer has no significant impact on the antitumor activity or tumor-targeting capacity of bacteria. Significant restraint of tumor progression is achieved by SA@HU due to the synergy of cellular immune activation and apoptosis enhancement. Most importantly, intravenous delivery of UA by this biohybrid vector can cause tumor lysis, as the bacteria-attracting nutrients beneficial for preferential accumulation of bacteria in tumor. The mutual promotion of bacteria and UA may also contribute to a superior anticancer effect. Hence, the SA@HU-based biotic/abiotic supramolecular therapeutic system represents a novel strategy for combined chemo-bacterial therapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Apoptosis , Bacteria , Humans , Hypoxia , Neoplasms/drug therapy
7.
ACS Nano ; 15(1): 1100-1110, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33236885

ABSTRACT

Hypoxia can increase the resistance of tumor cells to radiotherapy and chemotherapy. However, the dense extracellular matrix, high interstitial fluid pressure, and irregular blood supply often serve as physical barriers to inhibit penetration of drugs or nanodrugs across tumor blood microvessels into hypoxic regions. Therefore, it is of great significance and highly desirable to improve the efficiency of hypoxia-targeted therapy. In this work, living photosynthetic bacteria (PSB) are utilized as hypoxia-targeted carriers for hypoxic tumor therapy due to their near-infrared (NIR) chemotaxis and their physiological characteristics as facultative aerobes. More interestingly, we discovered that PSB can serve as a kind of photothermal agent to generate heat through nonradiative relaxation pathways due to their strong photoabsorption in the NIR region. Therefore, PSB integrate the properties of hypoxia targeting and photothermal therapeutic agents in an "all-in-one" manner, and no postmodification is needed to achieve hypoxia-targeted cancer therapy. Moreover, as natural bacteria, noncytotoxic PSB were found to enhance immune response that induced the infiltration of cytotoxicity T lymphocyte. Our results indicate PSB specifically accumulate in hypoxic tumor regions, and they show a high efficiency in the elimination of cancer cells. This proof of concept may provide a smart therapeutic system in the field of hypoxia-targeted photothermal therapeutic platforms.


Subject(s)
Hyperthermia, Induced , Neoplasms , Drug Delivery Systems , Humans , Hypoxia , Neoplasms/drug therapy , Phototherapy
8.
ACS Appl Bio Mater ; 3(1): 327-334, 2020 Jan 21.
Article in English | MEDLINE | ID: mdl-35019449

ABSTRACT

The need for in situ accurate identification of tumor assisted real-time image-guided surgical resection calls for new near-infrared fluorescence agents with high tumor-sensitivity and excellent biocompatibility. Here, an albumin-conjugate nanoparticle system HSA-Er-RI-Cl was designed, synthesized, and applied in cancer imaging, which simultaneously achieved the EPR effect, hypoxia-targeting, and EGFR-targeting property. Our novel nanoprobe is composed of human serum albumin (HSA) and double-targeting small molecule conjugate (Er-RI-Cl): a hypoxia-targeting heptamethine carbocyanine dye (RI-Cl) conjugated with a clinic anti-EGFR antagonist (Erlotinib) by covalent bonding. This conjugate could bind to albumin proteins, forming albumin-conjugate complexes, and those complexes self-assemble into particles with diameters of approximately 100 nm in the aqueous solution. The tumor hypoxia and EGFR targeting specificity of HSA-Er-RI-Cl was, respectively, evaluated in vitro and in vivo. Using murine xenograft subcutaneous and brain metastatic tumor models, we demonstrated that HSA-Er-RI-Cl is a highly potent tumor-targeting NIR agent for noninvasive imaging with remarkable tumor localization and excellent pharmacokinetic properties.

9.
Adv Exp Med Biol ; 1136: 141-157, 2019.
Article in English | MEDLINE | ID: mdl-31201722

ABSTRACT

Patients with the low levels of O2 (hypoxia) in their primary tumors have a higher risk for metastasis and death, indicating a need to therapeutically inhibit the effectors of hypoxia. Many strategies have been developed and investigated to block the hypoxic response. For example, inhibitors of HIF-1 and HIF-2 function by altering the transcription, translation, dimerization, nuclear translocation, DNA-binding, or ubiquitination of the HIF proteins. Hypoxia-activated prodrugs inhibit the hypoxic response through hypoxia-mediated reduction of an inactive, or minimally active, chemical to a cytotoxic agent. Most hypoxia-activated prodrugs function by inducing DNA damage, but others with more novel functions, including prodrugs that release EGFR/HER2 inhibitors also exist. Despite the existence of many therapeutics to combat the hypoxic response, there has been very little success in late phase clinical trials, potentially due to a lack of biomarkers that can be used to stratify patients who would benefit from a hypoxia-targeted therapy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Hypoxia-Inducible Factor 1 , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Hypoxia , Cell Hypoxia , Humans
10.
Biomaterials ; 214: 119226, 2019 09.
Article in English | MEDLINE | ID: mdl-31174068

ABSTRACT

Bacteria-driven drug-delivery systems have attracted great attention for their enhanced therapeutic specificity and efficacy in cancer treatment. YB1, a particularly attractive genetically modified safe Salmonella Typhimurium strain, is known to penetrate hypoxic tumor cores with its self-driven properties while remarkably avoiding damage to normal tissues. Herein, nanophotosensitizers (indocyanine green (ICG)-loaded nanoparticles, INPs) were covalently attached to the surface of YB1 with amide bonds to develop a biotic/abiotic cross-linked system (YB1-INPs) for tumor precision therapy. YB1 microswimmer retained its viability after efficiently linking with INPs. This YB1-INPs treatment strategy demonstrated specific hypoxia targeting to solid tumors, perfect photothermal conversion, and efficient fluorescence (FL) imaging properties. Benefited from the combined contribution of tumor tissue destruction and the bacteria-attracting nutrients generation after photothermal treatment, the bioaccumulation of YB1-INPs was significantly improved 14-fold compared to no photothermal intervention. Furthermore, YB1-INPs pervaded throughout the large solid tumor (≥500 mm3). Under near-infrared (NIR) laser irradiation, YB1-INPs exhibited a dependable and highly efficient photothermal killing ability for eradicating the large solid tumor without relapse. This strategy of bacteria-driven hypoxia-targeting delivery has a great value for large solid tumors therapy with low toxicity and high efficiency.


Subject(s)
Salmonella/physiology , Urinary Bladder Neoplasms/therapy , Animals , Bioaccumulation , Female , Fluorescent Antibody Technique , Hyperthermia, Induced/methods , Immunohistochemistry , Indocyanine Green/chemistry , Mice , Mice, Inbred C57BL , Microbial Viability , Nanoparticles/chemistry , Phototherapy/methods , Salmonella/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/physiology , Temperature , Urinary Bladder Neoplasms/microbiology
11.
Biomaterials ; 213: 119195, 2019 08.
Article in English | MEDLINE | ID: mdl-31132643

ABSTRACT

Hypoxia is one of the critical features in solid tumors, and hypoxia-targeting contrast agents (CAs) could greatly enhance the magnetic resonance imaging (MRI) of tumors by increasing its specificity and providing more diagnosis information. In this article, an internally hydroxy dendrimer of high molecular weight was facilely synthesized by "epoxy-amine" and "thiol-ene" reactions with a ß-cyclodextrin (ß-CD) as the core, and a hypoxia-targeting dendritic contrast agent (DCA) was synthesized through conjugating Gd chelates onto the internal hydroxyl groups and grafting the hypoxia-targeting groups sulfonamides and zwitterionic groups onto the exterior groups of the dendrimer. The zwitterionic surface without disturbance from internally conjugated CAs could reduce unspecific cellular uptake by normal cells, while the hypoxia-targeting group mediate the cellular uptake by hypoxic tumor cells. The in vitro and in vivo study showed that the hypoxia-targeting DCA could be selectively uptake by hypoxic cancer cells and greatly enhance the MRI of orthotopic breast tumor in a mouse model.


Subject(s)
Contrast Media/pharmacology , Dendrimers/chemistry , Hypoxia , Magnetic Resonance Imaging , Animals , Cell Line, Tumor , Chelating Agents/pharmacology , Drug Design , Female , Gadolinium/chemistry , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Mammary Neoplasms, Experimental , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Neoplasm Transplantation , Rhodamines/chemistry , beta-Cyclodextrins/pharmacology
12.
Biomaterials ; 183: 280-294, 2018 11.
Article in English | MEDLINE | ID: mdl-30179778

ABSTRACT

Drug resistance is one of the significant clinical burden in renal cell carcinoma (RCC). The development of drug resistance is attributed to many factors, including impairment of apoptosis, elevation of carbonic anhydrase IX (CA IX, a marker of tumor hypoxia), and infiltration of tumorigenic immune cells. To alleviate the drug resistance, we have used Sorafenib (Sor) in combination with tumor hypoxia directed nanoparticle (NP) loaded with a new class of apoptosis inducer, CFM 4.16 (C4.16), namely CA IX-C4.16. The NP is designed to selectively deliver the payload to the hypoxic tumor (core), provoke superior cell death in parental (WT) and Everolimus-resistant (Evr-res) RCC and selectively downmodulate tumorigenic M2-macrophage. Copper-free 'click' chemistry was utilized for conjugating SMA-TPGS with Acetazolamide (ATZ, a CA IX-specific targeting ligand). The NP was further tagged with a clinically approved NIR dye (S0456) for evaluating hypoxic tumor core penetration and organ distribution. Imaging of tumor spheroid treated with NIR dye-labeled CA IX-SMA-TPGS revealed remarkable tumor core penetration that was modulated by CA IX-mediated targeting in hypoxic-A498 RCC cells. The significant cell killing effect with synergistic combination index (CI) of CA IX-C4.16 and Sor treatment suggests efficient reversal of Evr-resistance in A498 cells. The CA IX directed nanoplatform in combination with Sor has shown multiple benefits in overcoming drug resistance through (i) inhibition of p-AKT, (ii) upregulation of tumoricidal M1 macrophages resulting in induction of caspase 3/7 mediated apoptosis of Evr-res A498 cells in macrophage-RCC co-culturing condition, (iii) significant in vitro and in vivo Evr-res A498 tumor growth inhibition as compared to individual therapy, and (iv) untraceable liver and kidney toxicity in mice. Near-infrared (NIR) imaging of CA IX-SMA-TPGS-S0456 in Evr-res A498 RCC model exhibited significant accumulation of CA IX-oligomer in tumor core with >3-fold higher tumor uptake as compared to control. In conclusion, this proof-of-concept study demonstrates versatile tumor hypoxia directed nanoplatform that can work in synergy with existing drugs for reversing drug-resistance in RCC accompanied with re-education of tumor-associated macrophages, that could be applied universally for several hypoxic tumors.


Subject(s)
Acetazolamide/chemistry , Carcinoma, Renal Cell/therapy , Drug Carriers/chemistry , Drug Resistance, Neoplasm , Kidney Neoplasms/therapy , Nanoparticles/chemistry , Tumor Hypoxia/physiology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase IX/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Cellular Reprogramming , Combined Modality Therapy , Everolimus/pharmacology , Female , Humans , Macrophages/metabolism , Mice , Mice, Nude , Nanoparticles/metabolism , Permeability , Proof of Concept Study , Sorafenib/pharmacology , Theranostic Nanomedicine
13.
ACS Nano ; 12(6): 5995-6005, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29786420

ABSTRACT

The facultative anaerobe Salmonella strain VNP20009 selectively colonizes into tumors following systemic injection due to its preference for the hypoxia in the tumor cores. However, the phase 1 clinical trial of VNP20009 has been terminated mainly due to its weak antitumor effects and exhibition of dose-dependent toxicity. Here, we leveraged the advantages of VNP20009 biotherapy together with polydopamine-mediated photothermal therapy in order to enhance the antitumor efficacy toward malignant melanoma. VNP20009 was coated with polydopamine via oxidation and self-polymerization, which was then injected into tumor-bearing mice via the tail vein. Polydopamine-coated VNP20009 targeted hypoxic areas of the solid tumors, and near-infrared laser irradiation of the tumors induced heating due to polydopamine. This combined approach eliminated the tumors without relapse or metastasis with only one injection and laser irradiation. More importantly, we found both VNP and pDA potentiate the therapeutic ability of each other, resulting in a superior anticancer effect.


Subject(s)
Antineoplastic Agents/pharmacology , Hypoxia/metabolism , Indoles/pharmacology , Melanoma, Experimental/therapy , Phototherapy , Polymers/pharmacology , Salmonella/metabolism , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/metabolism , Cell Survival/drug effects , Indoles/adverse effects , Indoles/metabolism , Lasers , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Polymers/adverse effects , Polymers/metabolism , Salmonella/growth & development , Tissue Distribution , Tumor Cells, Cultured
14.
Int J Mol Sci ; 19(3)2018 Mar 13.
Article in English | MEDLINE | ID: mdl-29534020

ABSTRACT

Triple negative breast cancer (TNBC) is a difficult to treat disease due to the absence of the three unique receptors estrogen, progesterone and herceptin-2 (HER-2). To improve the current therapy and overcome the resistance of TNBC, there is unmet need to develop an effective targeted therapy. In this regard, one of the logical and economical approaches is to develop a tumor hypoxia-targeting drug formulation platform for selective delivery of payload to the drug-resistant and invasive cell population of TNBC tumors. Toward this, we developed a Carbonic Anhydrase IX (CA IX) receptor targeting human serum albumin (HSA) carriers to deliver the potent anticancer drug, Paclitaxel (PTX). We used Acetazolamide (ATZ), a small molecule ligand of CA IX to selectively deliver HSA-PTX in TNBC cells. A novel method of synthesis involving copper free 'click' chemistry (Dibenzocyclooctyl, DBCO) moiety with an azide-labeled reaction partner, known as Strain-Promoted Alkyne Azide Cycloaddition (SPAAC) along with a desolvation method for PTX loading were used in the present study to arrive at the CA IX selective nano-carriers, HSA-PTX-ATZ. The anticancer effect of HSA-PTX-ATZ is higher compared to HSA, PTX and non-targeted HSA-PTX in MDA-MB-231 and MDA-MB-468 cells. The cell killing effect is associated with induction of early and late phases of apoptosis. Overall, our proof-of-concept study shows a promising avenue for hypoxia-targeted drug delivery that can be adapted to several types of cancers.


Subject(s)
Acetazolamide/chemistry , Antineoplastic Agents/administration & dosage , Carbonic Anhydrase Inhibitors/chemistry , Click Chemistry/methods , Nanoparticles/chemistry , Paclitaxel/administration & dosage , Acetazolamide/pharmacology , Albumins/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase Inhibitors/pharmacology , Cell Hypoxia , Cell Line, Tumor , Copper/chemistry , Drug Liberation , Humans , Paclitaxel/pharmacology
15.
ACS Appl Mater Interfaces ; 9(16): 13976-13987, 2017 Apr 26.
Article in English | MEDLINE | ID: mdl-28383881

ABSTRACT

Hypoxia occurs in most solid tumors, and it has been shown to be an independent prognostic indicator of a poor clinical outcome for patients with various cancers. Therefore, constructing a nanosystem specifically targeting cancer cells under hypoxia conditions is a promising approach for cancer therapy. Herein, we develop a porous silicon (PSi)-based nanosystem for targeted cancer therapy. VD11-4-2, a novel inhibitor for carbonic anhydrase IX (CA IX), is anchored on PSi particles (VD-PSi). As CA IX is mainly expressed on the cancer cell membrane under hypoxia condition, this nanocomplex inherits a strong affinity toward hypoxic human breast adenocarcinoma (MCF-7) cells; thus, a better killing efficiency for the hypoxia-induced drug resistance cancer cell is observed. Furthermore, the release of doxorubicin (DOX) from VD-PSi showed pH dependence, which is possibly due to the hydrogen-bonding interaction between DOX and VD11-4-2. The fluorescence resonance energy transfer effect between DOX and VD11-4-2 is observed and applied for monitoring the DOX release intracellularly. Protein inhibition and binding assays showed that VD-PSi binds and inhibits CA IX. Overall, we developed a novel nanosystem inheriting several advantageous properties, which has great potential for targeted treatment of cancer cells under hypoxic conditions.


Subject(s)
Carbonic Anhydrase IX/chemistry , Antigens, Neoplasm , Cell Hypoxia , Cell Line, Tumor , Doxorubicin , Humans , Silicon
16.
Oncotarget ; 7(34): 54795-54810, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27458162

ABSTRACT

Adoptively transferred mesenchymal stem cells (MSCs) home to solid tumors. Biologic features within the tumor environment can be used to selectively activate transgenes in engineered MSCs after tumor invasion. One of the characteristic features of solid tumors is hypoxia. We evaluated a hypoxia-based imaging and therapy strategy to target expression of the sodium iodide symporter (NIS) gene to experimental hepatocellular carcinoma (HCC) delivered by MSCs.MSCs engineered to express transgenes driven by a hypoxia-responsive promoter showed robust transgene induction under hypoxia as demonstrated by mCherry expression in tumor cell spheroid models, or radioiodide uptake using NIS. Subcutaneous and orthotopic HCC xenograft mouse models revealed significant levels of perchlorate-sensitive NIS-mediated tumoral radioiodide accumulation by tumor-recruited MSCs using 123I-scintigraphy or 124I-positron emission tomography. Functional NIS expression was further confirmed by ex vivo 123I-biodistribution analysis. Administration of a therapeutic dose of 131I in mice treated with NIS-transfected MSCs resulted in delayed tumor growth and reduced tumor perfusion, as shown by contrast-enhanced sonography, and significantly prolonged survival of mice bearing orthotopic HCC tumors. Interestingly, radioiodide uptake into subcutaneous tumors was not sufficient to induce therapeutic effects. Our results demonstrate the potential of using tumor hypoxia-based approaches to drive radioiodide therapy in non-thyroidal tumors.


Subject(s)
Iodine Radioisotopes/therapeutic use , Liver Neoplasms, Experimental/radiotherapy , Liver Neoplasms/radiotherapy , Mesenchymal Stem Cells/metabolism , Symporters/metabolism , Animals , Cell Line, Tumor , Combined Modality Therapy , Female , Humans , Hypoxia , Iodine Radioisotopes/pharmacokinetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mice, Nude , Symporters/genetics , Transfection , Xenograft Model Antitumor Assays
17.
Biochem Pharmacol ; 89(2): 224-35, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24632291

ABSTRACT

The nitro-chloromethylbenzindoline prodrug SN29428 has been rationally designed to target tumour hypoxia. SN29428 is metabolised to a DNA minor groove alkylator via oxygen-sensitive reductive activation initiated by unknown one-electron reductases. The present study sought to identify reductases capable of activating SN29428 in tumours. Expression of candidate reductases in cell lines was modulated using forced expression and, for P450 (cytochrome) oxidoreductase (POR), by zinc finger nuclease-mediated gene knockout. Affymetrix microarray mRNA expression of flavoreductases was correlated with SN29428 activation in a panel of 23 cancer cell lines. Reductive activation and cytotoxicity of prodrugs were measured using mass spectrometry and antiproliferative assays, respectively. SN29428 activation under hypoxia was strongly attenuated by the pan-flavoprotein inhibitor diphenyliodonium, but less so by knockout of POR suggesting other flavoreductases contribute. Forced expression of 5-methyltetrahydrofolate-homocysteine methyltransferase reductase (MTRR), as well as POR, increased activation of SN29428 in hypoxic HCT 116 cells. SN29428 activation strongly correlated with expression of POR and also FAD-dependent oxidoreductase domain containing 2 (FOXRED2), in cancer cell lines. This association persisted after removing the effect of POR enzyme activity using first-order partial correlation. Forced expression of FOXRED2 increased SN29428 activation and cytotoxicity in hypoxic HEK293 cells and also increased activation of hypoxia-targeted prodrugs PR-104A, tirapazamine and SN30000, and increased cytotoxicity of the clinical-stage prodrug TH-302. Thus this study has identified three flavoreductases capable of enzymatically activating SN29428, one of which (FOXRED2) has not previously been implicated in xenobiotic metabolism. These results will inform future development of biomarkers predictive of SN29428 sensitivity.


Subject(s)
Flavoproteins/biosynthesis , Oxidoreductases/metabolism , Prodrugs/chemistry , Prodrugs/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/pharmacology , HCT116 Cells , HEK293 Cells , Hep G2 Cells , Humans , Indoles/administration & dosage , Indoles/chemistry , Indoles/metabolism , Oxidation-Reduction , Oxidoreductases/biosynthesis , Prodrugs/administration & dosage , Triazines/chemistry , Triazines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL