Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 43
1.
Chem Biol Drug Des ; 103(5): e14531, 2024 May.
Article En | MEDLINE | ID: mdl-38726798

Inhibition of prolylhydroxylase-2 (PHD-2) in both normoxic and hypoxic cells is a critical component of solid tumours. The present study aimed to identify small molecules with PHD-2 activation potential. Virtually screening 4342 chemical compounds for structural similarity to R59949 and docking with PHD-2. To find the best drug candidate, hits were assessed for drug likeliness, antihypoxic and antineoplastic potential. The selected drug candidate's PHD-2 activation, cytotoxic and apoptotic potentials were assessed using 2-oxoglutarate, MTT, AO/EtBr and JC-1 staining. The drug candidate was also tested for its in-vivo chemopreventive efficacy against DMBA-induced mammary gland cancer alone and in combination with Tirapazamine (TPZ). Virtual screening and 2-oxoglutarate assay showed BBAP-6 as lead compound. BBAP-6 exhibited cytotoxic and apoptotic activity against ER+ MCF-7. In carmine staining and histology, BBAP-6 alone or in combination with TPZ restored normal surface morphology of the mammary gland after DMBA produced malignant alterations. Immunoblotting revealed that BBAP-6 reduced NF-κB expression, activated PHD-2 and induced intrinsic apoptotic pathway. Serum metabolomics conducted with 1H NMR confirmed that BBAP-6 prevented HIF-1α and NF-κB-induced metabolic changes in DMBA mammary gland cancer model. In a nutshell, it can be concluded that BBAP-6 activates PHD-2 and exhibits anticancer potential.


Apoptosis , Breast Neoplasms , Hypoxia-Inducible Factor-Proline Dioxygenases , Humans , Female , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/prevention & control , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Apoptosis/drug effects , Mice , Cell Hypoxia/drug effects , Molecular Docking Simulation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , MCF-7 Cells , Cell Line, Tumor , NF-kappa B/metabolism , Tirapazamine/pharmacology , Tirapazamine/chemistry , Tirapazamine/metabolism
2.
J Nanobiotechnology ; 22(1): 205, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658965

The elevated level of hydrogen sulfide (H2S) in colon cancer hinders complete cure with a single therapy. However, excessive H2S also offers a treatment target. A multifunctional cascade bioreactor based on the H2S-responsive mesoporous Cu2Cl(OH)3-loaded hypoxic prodrug tirapazamine (TPZ), in which the outer layer was coated with hyaluronic acid (HA) to form TPZ@Cu2Cl(OH)3-HA (TCuH) nanoparticles (NPs), demonstrated a synergistic antitumor effect through combining the H2S-driven cuproptosis and mild photothermal therapy. The HA coating endowed the NPs with targeting delivery to enhance drug accumulation in the tumor tissue. The presence of both the high level of H2S and the near-infrared II (NIR II) irradiation achieved the in situ generation of photothermic agent copper sulfide (Cu9S8) from the TCuH, followed with the release of TPZ. The depletion of H2S stimulated consumption of oxygen, resulting in hypoxic state and mitochondrial reprogramming. The hypoxic state activated prodrug TPZ to activated TPZ (TPZ-ed) for chemotherapy in turn. Furthermore, the exacerbated hypoxia inhibited the synthesis of adenosine triphosphate, decreasing expression of heat shock proteins and subsequently improving the photothermal therapy. The enriched Cu2+ induced not only cuproptosis by promoting lipoacylated dihydrolipoamide S-acetyltransferase (DLAT) heteromerization but also performed chemodynamic therapy though catalyzing H2O2 to produce highly toxic hydroxyl radicals ·OH. Therefore, the nanoparticles TCuH offer a versatile platform to exert copper-related synergistic antitumor therapy.


Copper , Hyaluronic Acid , Hydrogen Sulfide , Mitochondria , Nanoparticles , Photothermal Therapy , Prodrugs , Tirapazamine , Photothermal Therapy/methods , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Animals , Copper/chemistry , Copper/pharmacology , Mice , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Prodrugs/pharmacology , Prodrugs/chemistry , Tirapazamine/pharmacology , Tirapazamine/chemistry , Nanoparticles/chemistry , Hyaluronic Acid/chemistry , Cell Line, Tumor , Colonic Neoplasms/therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/drug therapy , Mice, Inbred BALB C , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Nude
3.
Adv Mater ; 36(3): e2307929, 2024 Jan.
Article En | MEDLINE | ID: mdl-37856705

Combination therapy has emerged as a promising approach for effective tumor treatment. However, the combination of sonodynamic therapy (SDT) and hypoxia-activated prodrugs (HAPs) has not been explored due to the contradictory requirement of oxygen (O2 ) for reactive oxygen species (ROS) generation and the necessity to avoid O2 for the activation of HAPs. In this study, this challenge is addressed by developing BiOCl-Au-Ag2 S Z-scheme heterostructure nanoparticles loaded with tirapazamine (TPZ) to achieve O2 -independent therapy. These nanoparticles demonstrate efficient electron-hole separation under ultrasound irradiation while maintaining a high redox potential. The generated holes react with water to efficiently produce hydroxyl radicals, while the electrons autonomously activate TPZ, negating the need for O2 . In vitro and in vivo assessments validate the effective tumor elimination by these Z-scheme nanoparticles without disrupting the hypoxic environment. This innovative design overcomes the limitations associated with O2 requirement in SDT and introduces a novel strategy for HAP activation and synergistic therapy between ROS and HAPs-based therapy.


Nanoparticles , Neoplasms , Prodrugs , Humans , Oxygen , Reactive Oxygen Species , Prodrugs/chemistry , Tirapazamine/chemistry , Hypoxia , Neoplasms/drug therapy , Cell Line, Tumor
4.
J Photochem Photobiol B ; 248: 112798, 2023 Nov.
Article En | MEDLINE | ID: mdl-37820499

BACKGROUND: Photodynamic therapy (PDT) has a promising application prospect in Echinococcus granulosus (Egs), however, the hypoxic environment of Egs and the hypoxia associated with PDT will greatly limit its effects. As a hypoxic-activated pre-chemotherapeutic drug, tirapazamine (TPZ) can be only activated and produce cytotoxicity under hypoxia environment. Albendazole sulfoxide (ABZSO) is the first choice for the treatment of Egs. This study aimed to explore the effects of ABZSO nanoparticles (ABZSO NPs), TPZ combined with PDT on the activity of Egs in vitro and in vivo. METHODS: The Egs were divided into control, ABZSO NPs, ABZSO NPs + PDT, and ABZSO NPs + TPZ + PDT groups, and the viability of Egs was determined using methylene blue staining. Then, the ROS, LDH and ATP levels were measured using their corresponding assay kit, and H2AX and TopoI protein expression was detected by western blot. The morphology of Egs with different treatments was observed using hematoxylin eosin (HE) staining and scanning electron microscopy (SEM). After that, the in vivo efficacy of ABZSO NPs, TPZ and PDT on Egs was determined in a Egs infected mouse model. RESULTS: In vitro experiments showed that the combined treatment of TPZ, ABZSO NPs and PDT significantly inhibited Egs viability; and significantly increased ROS levels and LDH contents, while decreased ATP contents in Egs; as well as up-regulated H2AX and down-regulated TopoI protein expression. HE staining and SEM results showed that breaking-then-curing treatment seriously damaged the Egs wall. Additionally, in vivo experiments found that the combination of ABZSO NPs, PDT and TPZ had more serious calcification and damage of the wall structure of cysts. CONCLUSIONS: ABZSO NPs combined with TPZ and PDT has a better inhibitory effect on the growth of Egs in vitro and in vivo based on the strategy of "breaking-then-curing".


Echinococcosis , Echinococcus granulosus , Nanoparticles , Photochemotherapy , Animals , Mice , Tirapazamine/pharmacology , Tirapazamine/chemistry , Tirapazamine/therapeutic use , Echinococcus granulosus/metabolism , Reactive Oxygen Species/metabolism , Hypoxia , Photochemotherapy/methods , Echinococcosis/drug therapy , Nanoparticles/chemistry , Adenosine Triphosphate
5.
Biomater Sci ; 11(16): 5674-5679, 2023 Aug 08.
Article En | MEDLINE | ID: mdl-37439102

Moderate oxygen (O2) supply and uneven distribution of oxygen at the tumor site usually hinder the therapeutic efficacy of hypoxia-activated prodrugs. In this report, we designed a ferrocene-containing supramolecular nanomedicine (PFC/GOD-TPZ) with the PEG corona and disulfide-bond cross-linked core to co-encapsulate 4-di-N-oxide tirapazamine (TPZ) and glucose oxidase (GOD). The PEG corona of PFC/GOD-TPZ could be weakly acidic tumor pH-responsively detached for an enhanced cellular internalization, while the disulfide-bond cross-linked core could be cleavaged by intracellular glutathione (GSH) to present a GSH-triggered drug-release behavior. Subsequently, the cascade reactions, including catalytic reactions among the released GOD, glucose, and O2 to generate H2O2 and the subsequent Fenton reaction between ferrocene and H2O2, occurred. With the depletion of O2, the non-toxic TPZ was activated and converted into the cytotoxic therapeutic agent benzotriazinyl (BTZ) radical under the exacerbated hypoxic microenvironment. Collectively, the PFC/GOD-TPZ provides a promising strategy for effective combination therapy of GOD-mediated starvation therapy, chemodynamic therapy (CDT), and hypoxia-activated chemotherapy (CT).


Antineoplastic Agents , Neoplasms , Humans , Nanomedicine , Metallocenes/pharmacology , Metallocenes/therapeutic use , Hydrogen Peroxide/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Tirapazamine/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Oxygen , Hypoxia/drug therapy , Glutathione , Disulfides/pharmacology , Hydrogen-Ion Concentration , Cell Line, Tumor , Tumor Microenvironment
6.
J Colloid Interface Sci ; 634: 495-508, 2023 Mar 15.
Article En | MEDLINE | ID: mdl-36542978

Chemodynamic therapy (CDT), an emerging oncology treatment, has received considerable attention owing to its high selectivity, less aggressiveness, and endogenous stimulation. However, the complex intra-tumor environment limits the therapeutic effect. In this study, Cu+ was directly doped into the structure of the UiO-66 matrix using an in situ one-pot oil bath method. The as-formed UiO-66/Cu possessed a large surface area, making it feasible to modify folic acid (FA) and carry more chemotherapeutic agents like tirapazamine (TPZ), thus forming UiO-66/Cu-FA-TPZ nanoplatforms. For CDT, the nanoplatform catalyzed the cyclic generation of the highly oxidizing hydroxyl radical (·OH) from H2O2. Particularly, low-frequency ultrasound enhanced the curative effect. Notably, in a tumor, a severe hypoxic environment and ultrasound can activate more TPZ for safe and efficient chemotherapy, achieving synergistic and hypoxia-activated tumor treatment with a low risk of side effects. Moreover, the nanoplatform exhibits computed tomography imaging functions for combined diagnosis and treatment. Our designed nanoplatform overcomes the dilemma of insufficient efficacy from conventional therapy attributed to a hypoxic environment, expecting to guide the design of future treatment regimens for hypoxic tumors.


Antineoplastic Agents , Neoplasms , Humans , Tirapazamine/pharmacology , Tirapazamine/chemistry , Antineoplastic Agents/chemistry , Hydrogen Peroxide , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Hypoxia/drug therapy , Cell Line, Tumor
7.
J Photochem Photobiol B ; 234: 112535, 2022 Sep.
Article En | MEDLINE | ID: mdl-35930948

The treatment efficacy of anticancer drugs in complex physiological environments is still restricted by multi-drug resistance. To overcome this issue, a nanodrug system of HA-SS@CuS@ZIF-8@TPZ&TBMACN (HSCZTT) that breaks through the detoxification barrier for tirapazamine (TPZ) delivery was developed in this manuscript. In addition to the photothermal effect aroused by CuS in HSCZTT, which can damage tumour cells, TBMACN with photostable fluorescence in the aggregate state can also generate sufficient reactive oxygen species (ROS) to destroy tumour cells. The continuous consumption of oxygen in PDT aggravates the hypoxic environment of tumours, which further activates the TPZ released in the acidic microenvironment of the tumour to achieve apoptosis of the tumour cells. The HSCZTT can not only target the CD44 receptor overexpressed on the surface of the cancer cell, but can also effectively consume a large amount of glutathione (GSH) through the disulphide bond-modified hyaluronic acid, which serves as a targeted disulphide bond, interfering with the detoxification barrier. Our finding presents a rational strategy to overcome multidrug resistance for the improved efficacy of anticancer drugs by the targeting of Hyaluronic acid (HA), release of the drug by the acid response of ZIF-8, breakthrough of the detoxification barrier, precise positioning of the drug release and combined treatment with phototherapy and hypoxia-activated chemotherapy.


Antineoplastic Agents , Nanoparticles , Neoplasms , Photochemotherapy , Antineoplastic Agents/chemistry , Cell Line, Tumor , Disulfides , Humans , Hyaluronic Acid/chemistry , Hypoxia , Nanoparticles/chemistry , Neoplasms/pathology , Photosensitizing Agents/chemistry , Tirapazamine/chemistry , Tirapazamine/metabolism , Tirapazamine/pharmacology , Tumor Microenvironment
8.
J Mater Chem B ; 10(20): 3849-3860, 2022 05 25.
Article En | MEDLINE | ID: mdl-35470367

With the advantages of deep tissue penetration and controllability, external X-ray-induced photodynamic therapy (X-PDT) is highly promising for combined cancer therapy. In addition to the low efficiency of photosensitizer (PS) delivery to tumor sites, however, the radiation- and drug-resistance of hypoxic cells inside the tumor after X-PDT also limit its benefits. Herein, we develop a combined therapeutic modality based on an intelligent nanosized platform (DATAT-NPVT) with tumor acidity-activated TAT presenting and redox-boosted release of tirapazamine (TPZ) for more precise and synchronous X-PDT and selective hypoxia-motivated chemotherapy. After DATAT-NPVT has accumulated in tumor tissues via decreased blood clearance by masking of the TAT ligand, its targeting ability is reactivated by tumor pH (∼6.8), which enhances tumoral cellular uptake. Upon low-dose X-ray irradiation, the encapsulated verteporfin (VP) generates reactive oxygen species (ROS) to carry out X-PDT against MDA-MB-231 breast tumors. As a result of the abundant GSH-triggered degradation of ditelluride bridged bonds, the cascaded TPZ release and activation in the hypoxic environment following X-PDT would produce highly cytotoxic radicals to serve as antitumor agents to kill the remaining hypoxic tumor cells. This concept provides new avenues for the design of hierarchical-responsive drug delivery systems and represents a proof-of-concept combinatorial tumor treatment.


Antineoplastic Agents , Nanoparticles , Photochemotherapy , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Hypoxia , Nanoparticles/chemistry , Oxidation-Reduction , Tirapazamine/chemistry , X-Rays
9.
ACS Biomater Sci Eng ; 8(4): 1604-1612, 2022 04 11.
Article En | MEDLINE | ID: mdl-35348331

The low sensitivity of hypoxic regions in solid tumors to radiotherapy and chemotherapy remains a major obstacle to cancer treatment. By taking advantage of hypoxic-activated prodrugs, tirapazamine (TPZ), generating cytotoxic reductive products and the glucose oxidase (GOx)-based glucose oxidation reaction, we designed a nanodrug-loading system that combined TPZ-induced chemotherapy with GOx-mediated cancer-orchestrated starvation therapy and cancer oxidation therapy. In this work, we first prepared mesoporous silica (MSN) loaded with TPZ. Then, in order to prevent the leakage of TPZ in advance, the surface was coated with a layer of carMOF formed by Fe3+ and carbenicillin (car), and GOx was adsorbed on the outermost layer to form the final nanosystem MSN-TPZ@carMOF-GOx (MT@c-G). GOx could effectively consume oxygen and catalyzed glucose into gluconic acid and hydrogen peroxide. First, the generated gluconic acid lowered the pH of tumor tissues, promoted the decomposition of carMOF, and released TPZ. Second, oxygen consumption could improve the degree of hypoxia in tumor tissues, so that enhanced the activity of TPZ. Furthermore, GOx could generate cancer-orchestrated starvation/oxidation therapy. Therefore, our study provided a new strategy that TPZ combined with GOx achieved starvation/oxidation/chemotherapy for enhancing anticancer effects in hypoxic regions.


Prodrugs , Cell Line, Tumor , Glucose , Humans , Hypoxia , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/therapeutic use , Tirapazamine/chemistry , Tirapazamine/pharmacology
10.
J Mater Chem B ; 9(44): 9142-9152, 2021 11 17.
Article En | MEDLINE | ID: mdl-34693960

Multimodal synergistic therapy has gained increasing attention in cancer treatment to overcome the limitations of monotherapy and achieve high anticancer efficacy. In this study, a synergistic phototherapy and hypoxia-activated chemotherapy nanoplatform based on natural melanin nanoparticles (MPs) loaded with the bioreduction prodrug tirapazamine (TPZ) and decorated with hyaluronic acid (HA) was developed. A self-reporting aggregation-induced emission (AIE)-active photosensitizer (PS) (BATTMN) was linked to the prepared nanoparticles by boronate ester bonds. The MPs and BATTMN-HA played roles as quenchers for PS and cancer targeting/photodynamic moieties, respectively. As a pH sensitive bond, the borate ester bonds between HA and BATTMN are hydrolysed in the acidic cancer environment, thereby separating BATTMN from the nanoparticles and leading to the induction of fluorescence for imaging-guided synergistic phototherapy/hypoxia-activated chemotherapy under dual irradiation. TPZ can be released upon activation by pH, near-infrared (NIR) and hyaluronidase (Hyal). Particularly, the hypoxia-dependent cytotoxicity of TPZ was amplified by oxygen consumption in the tumor intracellular environment induced by the AIE-active PS in photodynamic therapy (PDT). The nanoparticles developed in our research showed favorable photothermal conversion efficiency (η = 37%), desired cytocompatibility, and excellent synergistic therapeutic efficacy. The proposed nanoplatform not only extends the application scope of melanin materials with AIE-active PSs, but also offers useful insights into developing multistimulus as well as multimodal synergistic tumor treatment.


Antineoplastic Agents/therapeutic use , Drug Carriers/chemistry , Melanins/therapeutic use , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Animals , Antineoplastic Agents/chemistry , Boronic Acids/chemistry , Boronic Acids/radiation effects , Boronic Acids/therapeutic use , Combined Modality Therapy , Drug Therapy , Female , Humans , MCF-7 Cells , Melanins/chemistry , Melanins/radiation effects , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/radiation effects , Photosensitizing Agents/chemistry , Photosensitizing Agents/radiation effects , Photothermal Therapy , Prodrugs/chemistry , Prodrugs/therapeutic use , Tirapazamine/chemistry , Tirapazamine/therapeutic use , Tumor Hypoxia/physiology , Xenograft Model Antitumor Assays
11.
J Nanobiotechnology ; 19(1): 298, 2021 Sep 30.
Article En | MEDLINE | ID: mdl-34592996

BACKGROUND: Hypoxia is a characteristic of solid tumors that can lead to tumor angiogenesis and early metastasis, and addressing hypoxia presents tremendous challenges. In this work, a nanomedicine based on oxygen-absorbing perfluorotributylamine (PFA) and the bioreductive prodrug tirapazamine (TPZ) was prepared by using a polydopamine (PDA)-coated UiO-66 metal organic framework (MOF) as the drug carrier. RESULTS: The results showed that TPZ/PFA@UiO-66@PDA nanoparticles significantly enhanced hypoxia, induced cell apoptosis in vitro through the oxygen-dependent HIF-1α pathway and decreased oxygen levels in vivo after intratumoral injection. In addition, our study demonstrated that TPZ/PFA@UiO-66@PDA nanoparticles can accumulate in the tumor region after tail vein injection and effectively inhibit tumor growth when combined with photothermal therapy (PTT). TPZ/PFA@UiO-66@PDA nanoparticles increased HIF-1α expression while did not promote the expression of CD31 in vivo during the experiment. CONCLUSIONS: By using TPZ and PFA and the enhanced permeability and retention effect of nanoparticles, TPZ/PFA@UiO-66@PDA can target tumor tissues, enhance hypoxia in the tumor microenvironment, and activate TPZ. Combined with PTT, the growth of osteosarcoma xenografts can be effectively inhibited.


Fluorocarbons , Metal-Organic Frameworks , Osteosarcoma/metabolism , Phthalic Acids , Tirapazamine , Tumor Hypoxia , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Fluorocarbons/chemistry , Fluorocarbons/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Male , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Mice , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/toxicity , Phthalic Acids/chemistry , Phthalic Acids/pharmacology , Polymers/chemistry , Polymers/pharmacology , Tirapazamine/chemistry , Tirapazamine/pharmacology
12.
Adv Mater ; 33(41): e2104504, 2021 Oct.
Article En | MEDLINE | ID: mdl-34436814

One of the main challenges for tumor vascular infarction in combating cancer lies in failing to produce sustained complete thrombosis. Inspired by the capability of vascular infarction in blocking the delivery of oxygen to aggravate tumor hypoxia, the performance of selective tumor thrombus inducing hypoxia activation therapy to improve the therapeutic index of coagulation-based tumor therapy is presented. By encapsulating coagulation-inducing protease thrombin and a hypoxia-activated prodrug (HAP) tirapazamine into metal-organic framework nanoparticles with a tumor-homing ligand, the obtained nanoplatform selectively activates platelet aggregation at the tumor to induce thrombosis and vascular obstruction therapy by the exposed thrombin. Meanwhile, the thrombus can cut off the blood oxygen supply and potentiate the hypoxia levels to enhance the HAP therapy. This strategy not only addresses the dissatisfaction of vascular therapy, but also conquers the dilemma of inadequate hypoxia in HAP treatment. Since clinical operations such as surgery can be used to induce coagulation, coagulation-based synergistic therapy is promising for translation into a clinical combination regimen.


Prodrugs/chemistry , Thrombin/chemistry , Tumor Hypoxia , Animals , Cell Survival/drug effects , Hep G2 Cells , Humans , Metal-Organic Frameworks/chemistry , Mice , Mice, Nude , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Platelet Aggregation/drug effects , Prodrugs/pharmacology , Prodrugs/therapeutic use , Thrombosis/pathology , Tirapazamine/chemistry , Transplantation, Heterologous , Tumor Hypoxia/drug effects
13.
J Mater Chem B ; 9(26): 5318-5328, 2021 07 07.
Article En | MEDLINE | ID: mdl-34231629

For cancer treatment, the traditional monotherapy has the problems of low drug utilization rate, poor efficacy and easy recurrence of the cancer. Herein, nanoparticles (NPs) based on a novel semiconducting molecule (ITTC) are developed with excellent photostability, high photothermal conversion efficiency and good 1O2 generation ability. The chemotherapy of the hypoxia-activated prodrug tirapazamine (TPZ) was improved accordingly after oxygen consumption by the photodynamic therapy of ITTC NPs. Additionally, the metabolic process of ITTC NPs in vivo could be monitored in real time for fluorescence imaging guided phototherapy, which presented great passive targeting ability to the tumor site. Remarkably, both in vitro and in vivo experiments demonstrated that the combination of ITTC NPs and TPZ presented excellent synergistic tumor ablation through photothermal therapy, photodynamic therapy and hypoxia-activated chemotherapy with great potential for clinical applications in the future.


Antineoplastic Agents/pharmacology , Hypoxia/diagnostic imaging , Hypoxia/drug therapy , Nanoparticles/chemistry , Optical Imaging , Photosensitizing Agents/pharmacology , Tirapazamine/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Nanoparticles/administration & dosage , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Semiconductors , Tirapazamine/administration & dosage , Tirapazamine/chemistry
14.
Nanotechnology ; 32(46)2021 Aug 23.
Article En | MEDLINE | ID: mdl-34325415

The therapeutic effect of oxygen-concentration-dependent photodynamic therapy (PDT) can be diminished in the hypoxic environment of solid tumours, the effective solution to this problem is utilising hypoxic-activated bioreduction therapy (BRT). In this research, a biocompatible HA-C60/TPENH2nanogel which can specifically bind to CD44 receptor was developed for highly efficient PDT-BRT synergistic therapy. The nanogel was degradable in acidic microenvironments of tumours and facilitated the release of biological reduction prodrug tirapazamine (TPZ). Importantly, HA-C60/TPENH2nanogel produced reactive oxygen species and consumed oxygen content in the cell to activate TPZ, leading to higher cytotoxicity than the free TPZ did. The intracellular observation of nanogel indicated that the HA-C60/TPENH2nanogel was self-fluorescence for cell imaging. This study applied PDT-BRT to design smart HA-based nanogel with targeted delivery, pH response, and AIEgen feature for efficient cancer therapy.


Fullerenes/chemistry , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Hypoxia/drug therapy , Nanogels/chemistry , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Tirapazamine/administration & dosage , Tirapazamine/chemistry , Antineoplastic Agents/administration & dosage , Cell Line , Cell Line, Tumor , HEK293 Cells , Hep G2 Cells , Humans , Hypoxia/metabolism , Nanoparticles/chemistry , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Prodrugs/administration & dosage , Reactive Oxygen Species/metabolism , Tumor Microenvironment/drug effects
15.
J Mol Model ; 27(6): 177, 2021 May 22.
Article En | MEDLINE | ID: mdl-34021836

Tirapazamine (TP) has been shown to enhance the cytotoxic effects of ionizing radiation in hypoxic cells, thus making it a candidate for a radiosensitizer. This selective behavior is often directly linked to the abundance of O2. In this paper, we study the electronic properties of TP in vacuum, micro-hydrated from one up to three molecules of water and embedded in a continuum of water. We discuss electron affinities, charge distribution, and bond dissociation energies of TP, and find that these properties do not change significantly upon hydration. In agreement with its large electron affinity, and bond breaking triggered by electron attachment requires energies higher than 2.5 eV, ruling out the direct formation of bioactive TP radicals. Our results suggest, therefore, that the selective behavior of TP cannot be explained by a one-electron reduction from a neighboring O2 molecule. Alternatively, we propose that TP's hypoxic selectivity could be a consequence of O2 scavenging hydrogen radicals.


Models, Chemical , Models, Molecular , Radiation-Sensitizing Agents/chemistry , Tirapazamine/chemistry
16.
J Med Chem ; 64(9): 6161-6178, 2021 05 13.
Article En | MEDLINE | ID: mdl-33857374

The ubiquitin-like protein NEDD8 is a critical signaling molecule implicated in the functional maintenance and homeostasis of cells. Dysregulation of this process is involved in a variety of human diseases, including cancer. Therefore, NEDD8-activating enzyme E1 (NAE), the only activation enzyme of the neddylation pathway, has been an emergent anticancer target. In view of the single-agent modest response of the clinical NAE inhibitor, pevonedistat (compound 1, MLN4924), efforts on development of new inhibitors with both high potency and better safety profiles are urgently needed. Here, we report a structural hopping strategy by optimizing the central deazapurine framework and the solvent interaction region of compound 1, leading to compound 26 bearing a pyrimidotriazole scaffold. Compound 26 not only has compatible potency in the biochemical and cell assays but also possesses improved pharmacokinetic (PK) properties than compound 1. In vivo, compound 26 showed significant antitumor efficacy and good safety in xenograft models.


Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Tirapazamine/chemistry , Tirapazamine/pharmacology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Cell Line, Tumor , Cisplatin , Enzyme Inhibitors/pharmacokinetics , Humans , Ifosfamide , Mitomycin , Tirapazamine/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays
17.
Int J Mol Sci ; 22(6)2021 Mar 19.
Article En | MEDLINE | ID: mdl-33808887

We investigate dissociative electron attachment to tirapazamine through a crossed electron-molecule beam experiment and quantum chemical calculations. After the electron is attached and the resulting anion reaches the first excited state, D1, we suggest a fast transition into the ground electronic state through a conical intersection with a distorted triazine ring that almost coincides with the minimum in the D1 state. Through analysis of all observed dissociative pathways producing heavier ions (90-161 u), we consider the predissociation of an OH radical with possible roaming mechanism to be the common first step. This destabilizes the triazine ring and leads to dissociation of highly stable nitrogen-containing species. The benzene ring is not altered during the process. Dissociation of small anionic fragments (NO2-, CN2-, CN-, NH2-, O-) cannot be conclusively linked to the OH predissociation mechanism; however, they again do not require dissociation of the benzene ring.


Electrons , Tirapazamine/chemistry , Algorithms , Anions/chemistry , Models, Chemical , Radiation-Sensitizing Agents/chemistry
18.
J Mater Chem B ; 9(11): 2613-2622, 2021 03 21.
Article En | MEDLINE | ID: mdl-33683252

Photothermal therapy (PTT) based on semiconducting polymer nanoparticles (SPNs) is a promising strategy to treat solid tumors, but its ability to combine with chemotherapy for immune remodeling to efficiently suppress metastatic cancers has rarely been studied. Here, we demonstrate that PTT combined with chemotherapy can efficiently elicit immunity to suppress metastatic tumor growth. Specifically, we rationally designed a new SPN (PDPSe NPs) as a photothermal agent for PTT with a large mass extinction coefficient in the near-infrared region (e.g., 44.9 L g-1 cm-1 at 808 nm), high photothermal conversion efficiency (62.5%) and excellent biocompatibility. A hypoxia-activated anti-tumor drug, tirapazamine (TPZ), was selected for chemotherapy. Strikingly, the combination therapy not only induced tumor cell death in the primary tumor, but also effectively suppressed the growth of distant tumors (mimicking metastatic tumors) without PTT. Importantly, the combined therapies exhibit synergistic effects on immune remodeling. Immunofluorescence data suggest that the inhibition of metastatic tumor growth is attributed to the immune remodeling triggered by PTT and chemotherapy. This work demonstrates a new paradigm of utilizing PTT together with hypoxia-activated drugs to effectively retard metastatic tumor growth.


Antineoplastic Agents/pharmacology , Liver Neoplasms/drug therapy , Nanoparticles/chemistry , Photothermal Therapy , Polymers/pharmacology , Splenic Neoplasms/drug therapy , Tirapazamine/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Female , Injections, Subcutaneous , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Mice , Polymers/chemical synthesis , Polymers/chemistry , Semiconductors , Splenic Neoplasms/immunology , Splenic Neoplasms/secondary , Tirapazamine/administration & dosage , Tirapazamine/chemistry
19.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article En | MEDLINE | ID: mdl-33228195

Derivatives of tirapazamine and other heteroaromatic N-oxides (ArN→O) exhibit tumoricidal, antibacterial, and antiprotozoal activities, which are typically attributed to bioreductive activation and free radical generation. In this work, we aimed to clarify the role of NAD(P)H:quinone oxidoreductase (NQO1) in ArN→O aerobic cytotoxicity. We synthesized 9 representatives of ArN→O with uncharacterized redox properties and examined their single-electron reduction by rat NADPH:cytochrome P-450 reductase (P-450R) and Plasmodium falciparum ferredoxin:NADP+ oxidoreductase (PfFNR), and by rat NQO1. NQO1 catalyzed both redox cycling and the formation of stable reduction products of ArN→O. The reactivity of ArN→O in NQO1-catalyzed reactions did not correlate with the geometric average of their activity towards P-450R- and PfFNR, which was taken for the parameter of their redox cycling efficacy. The cytotoxicity of compounds in murine hepatoma MH22a cells was decreased by antioxidants and the inhibitor of NQO1, dicoumarol. The multiparameter regression analysis of the data of this and a previous study (DOI: 10.3390/ijms20184602) shows that the cytotoxicity of ArN→O (n = 18) in MH22a and human colon carcinoma HCT-116 cells increases with the geometric average of their reactivity towards P-450R and PfFNR, and with their reactivity towards NQO1. These data demonstrate that NQO1 is a potentially important target of action of heteroaromatic N-oxides.


Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Antiprotozoal Agents/pharmacology , Cyclic N-Oxides/pharmacology , Ferredoxin-NADP Reductase/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NADPH-Ferrihemoprotein Reductase/antagonists & inhibitors , Aerobiosis , Animals , Anti-Bacterial Agents/chemical synthesis , Antioxidants/chemical synthesis , Antiprotozoal Agents/chemical synthesis , Cell Line, Tumor , Cell Survival/drug effects , Cyclic N-Oxides/chemical synthesis , Dicumarol/pharmacology , Enzyme Assays , Enzyme Inhibitors/pharmacology , Ferredoxin-NADP Reductase/chemistry , Ferredoxin-NADP Reductase/metabolism , HCT116 Cells , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Kinetics , Mice , NAD(P)H Dehydrogenase (Quinone)/chemistry , NAD(P)H Dehydrogenase (Quinone)/metabolism , NADPH-Ferrihemoprotein Reductase/chemistry , NADPH-Ferrihemoprotein Reductase/metabolism , Oxidation-Reduction , Plasmodium falciparum/chemistry , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Rats , Tirapazamine/chemistry , Tirapazamine/pharmacology
20.
Molecules ; 25(21)2020 Oct 22.
Article En | MEDLINE | ID: mdl-33105798

Hypoxia is an adverse prognostic feature of solid cancers that may be overcome with hypoxia-activated prodrugs (HAPs). Tirapazamine (TPZ) is a HAP which has undergone extensive clinical evaluation in this context and stimulated development of optimized analogues. However the subcellular localization of the oxidoreductases responsible for mediating TPZ-dependent DNA damage remains unclear. Some studies conclude only nuclear-localized oxidoreductases can give rise to radical-mediated DNA damage and thus cytotoxicity, whereas others identify a broader role for endoplasmic reticulum and cytosolic oxidoreductases, indicating the subcellular location of TPZ radical formation is not a critical requirement for DNA damage. To explore this question in intact cells we engineered MDA-231 breast cancer cells to express the TPZ reductase human NADPH: cytochrome P450 oxidoreductase (POR) harboring various subcellular localization sequences to guide this flavoenzyme to the nucleus, endoplasmic reticulum, cytosol or inner surface of the plasma membrane. We show that all POR variants are functional, with differences in rates of metabolism reflecting enzyme expression levels rather than intracellular TPZ concentration gradients. Under anoxic conditions, POR expression in all subcellular compartments increased the sensitivity of the cells to TPZ, but with a fall in cytotoxicity per unit of metabolism (termed 'metabolic efficiency') when POR is expressed further from the nucleus. However, under aerobic conditions a much larger increase in cytotoxicity was observed when POR was directed to the nucleus, indicating very high metabolic efficiency. Consequently, nuclear metabolism results in collapse of hypoxic selectivity of TPZ, which was further magnified to the point of reversing O2 dependence (oxic > hypoxic sensitivity) by employing a DNA-affinic TPZ analogue. This aerobic hypersensitivity phenotype was partially rescued by cellular copper depletion, suggesting the possible involvement of Fenton-like chemistry in generating short-range effects mediated by the hydroxyl radical. In addition, the data suggest that under aerobic conditions reoxidation strictly limits the TPZ radical diffusion range resulting in site-specific cytotoxicity. Collectively these novel findings challenge the purported role of intra-nuclear reductases in orchestrating the hypoxia selectivity of TPZ.


Antineoplastic Agents/chemistry , Hypoxia/drug therapy , NADPH-Ferrihemoprotein Reductase/genetics , Prodrugs/chemistry , Tirapazamine/chemistry , Antineoplastic Agents/pharmacology , Cell Engineering , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Nucleus/drug effects , Cell Survival/drug effects , Copper/metabolism , DNA Damage/drug effects , DNA Damage/genetics , Humans , Models, Biological , NADPH-Ferrihemoprotein Reductase/metabolism , NADPH-Ferrihemoprotein Reductase/ultrastructure , Oxygen/metabolism , Prodrugs/metabolism , Tirapazamine/metabolism
...