Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 501
Filter
Add more filters

Publication year range
1.
J Nanobiotechnology ; 22(1): 358, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907270

ABSTRACT

BACKGROUND: Hypoxia-activated prodrug (HAP) is a promising candidate for highly tumor-specific chemotherapy. However, the oxygenation heterogeneity and dense extracellular matrix (ECM) of tumor, as well as the potential resistance to chemotherapy, have severely impeded the resulting overall efficacy of HAP. RESULTS: A HAP potentiating strategy is proposed based on ultrasound responsive nanodroplets (PTP@PLGA), which is composed of protoporphyrin (PpIX), perfluoropropane (PFP) and a typical HAP, tirapazamine (TPZ). The intense vaporization of PFP upon ultrasound irradiation can magnify the sonomechanical effect, which loosens the ECM to promote the penetration of TPZ into the deep hypoxic region. Meanwhile, the PpIX enabled sonodynamic effect can further reduce the oxygen level, thus activating the TPZ in the relatively normoxic region as well. Surprisingly, abovementioned ultrasound effect also results in the downregulation of the stemness of cancer cells, which is highly associated with drug-refractoriness. CONCLUSIONS: This work manifests an ideal example of ultrasound-based nanotechnology for potentiating HAP and also reveals the potential acoustic effect of intervening cancer stem-like cells.


Subject(s)
Fluorocarbons , Nanoparticles , Prodrugs , Protoporphyrins , Tirapazamine , Humans , Tirapazamine/pharmacology , Tirapazamine/chemistry , Protoporphyrins/pharmacology , Protoporphyrins/chemistry , Fluorocarbons/chemistry , Fluorocarbons/pharmacology , Prodrugs/pharmacology , Prodrugs/chemistry , Cell Line, Tumor , Nanoparticles/chemistry , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Ultrasonic Waves , Animals , Extracellular Matrix/metabolism , Mice , Neoplasms/drug therapy
2.
J Nanobiotechnology ; 22(1): 205, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658965

ABSTRACT

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.


Subject(s)
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.
Int J Mol Sci ; 24(7)2023 Apr 06.
Article in English | MEDLINE | ID: mdl-37047836

ABSTRACT

3-Amino-1,2,4-benzotriazine-1,4-dioxide (tirapazamine, TPZ) and other heteroaromatic N-oxides (ArN→O) exhibit tumoricidal, antibacterial, and antiprotozoal activities. Their action is attributed to the enzymatic single-electron reduction to free radicals that initiate the prooxidant processes. In order to clarify the mechanisms of aerobic mammalian cytotoxicity of ArN→O, we derived a TPZ-resistant subline of murine hepatoma MH22a cells (resistance index, 5.64). The quantitative proteomic of wild-type and TPZ-resistant cells revealed 5818 proteins, of which 237 were up- and 184 down-regulated. The expression of the antioxidant enzymes aldehyde- and alcohol dehydrogenases, carbonyl reductases, catalase, and glutathione reductase was increased 1.6-5.2 times, whereas the changes in the expression of glutathione peroxidase, superoxide dismutase, thioredoxin reductase, and peroxiredoxins were less pronounced. The expression of xenobiotics conjugating glutathione-S-transferases was increased by 1.6-2.6 times. On the other hand, the expression of NADPH:cytochrome P450 reductase was responsible for the single-electron reduction in TPZ and for the 2.1-fold decrease. These data support the fact that the main mechanism of action of TPZ under aerobic conditions is oxidative stress. The unchanged expression of intranuclear antioxidant proteins peroxiredoxin, glutaredoxin, and glutathione peroxidase, and a modest increase in the expression of DNA damage repair proteins, tend to support non-site-specific but not intranuclear oxidative stress as a main factor of TPZ aerobic cytotoxicity.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Tirapazamine/pharmacology , Triazines/pharmacology , Antineoplastic Agents/pharmacology , Antioxidants , Proteomics , Oxidation-Reduction , Glutathione Peroxidase , Mammals
4.
Mol Pharm ; 19(6): 1766-1777, 2022 06 06.
Article in English | MEDLINE | ID: mdl-34473523

ABSTRACT

Tirapazamine (TPZ) and its derivatives (TPZD) have shown their great potential for efficiently killing hypoxic cancer cells. However, unsatisfactory clinical outcomes resulting from the low bioavailability of the low-molecular TPZ and TPZD limited their further applications. Precise delivery and release of these prodrugs via functional nanocarriers can significantly improve the therapeutic effects due to the targeted drug delivery and enhanced permeability and retention (EPR) effect. Herein, zwitterionic block copolymer (BCP) micelles with aldehyde functional groups are prepared from the self-assembly of poly(2-methacryloyloxyethyl phosphorylcholine-b-poly(di(ethylene glycol) methyl ether methacrylate-co-4-formylphenyl methacrylate) [PMPC-b-P(DEGMA-co-FPMA)]. TPZD is then grafted onto PMPC-b-P(DEGMA-co-FPMA) to obtain a polymer-drug conjugate, PMPC-b-P(DEGMA-co-FPMA-g-TPZD) (BCP-TPZ), through the formation of a pH-responsive imine bond, exhibiting a pH-dependent drug release profile owing to the cleavage of the imine bond under acidic conditions. Outstandingly, BCP-TPZ shows around 13.7-fold higher cytotoxicity to hypoxic cancer cells in comparison to normoxic cancer cells evaluated through an in vitro cytotoxicity assay. The pH-responsiveness and hypoxia-specific cytotoxicity confer BCP-TPZ micelles a great potential to achieve precise delivery of TPZD and thus enhance the therapeutic effect toward tumor-hypoxia.


Subject(s)
Micelles , Prodrugs , Doxorubicin/chemistry , Humans , Hydrogen-Ion Concentration , Hypoxia , Imines , Methacrylates/chemistry , Polymers/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Tirapazamine
5.
J Vasc Interv Radiol ; 33(8): 926-933.e1, 2022 08.
Article in English | MEDLINE | ID: mdl-35504436

ABSTRACT

PURPOSE: To investigate the safety of replacing doxorubicin with tirapazamine in conventional transarterial chemoembolization (TACE) in an Asian population with hepatocellular carcinoma (HCC), and to determine the optimal tirapazamine dose for phase II studies. MATERIALS AND METHODS: This was a phase I, 3 + 3 dose-escalation study for patients with unresectable early- and intermediate-stage HCC who received 5, 10, or 20 mg/m2 of intra-arterial (IA) tirapazamine followed by ethiodized oil/gelatin sponge-based embolization. Key eligibilities included HCCs no more than 10 cm in diameter, prior embolization allowed, Eastern Cooperative Oncology Group performance status of 0 or 1, Child-Pugh score of 5-7, and platelet count of ≥60,000 µL. Dose-limiting toxicity (DLT) was defined as any grade 3 nonhematological or grade 4 hematological toxicity, with the exception of transient elevation of aminotransferase levels after the procedure. RESULTS: Seventeen patients were enrolled, 59% of whom had progression from a prior HCC therapy and 35% of whom had progression or recurrence after TACE. All patients tolerated the tirapazamine TACE well without any DLT or serious adverse event. Using the modified Response Evaluation Criteria in Solid Tumors, the complete response (CR) rate was 47%, and the CR + partial response rate was 65%. The median duration of response was not reached. The median time to progression was 12.6 months (95% confidence interval, 5.1-not reached). The median overall survival was 29.3 months. The selected phase II dose was set at a fixed dose of 35 mg of IA tirapazamine. CONCLUSIONS: IA tirapazamine with transarterial embolization was well tolerated and showed promising efficacy signals in intermediate-stage HCC, justifying pursuit of a phase II study.


Subject(s)
Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Liver Neoplasms , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic/adverse effects , Chemoembolization, Therapeutic/methods , Ethiodized Oil , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/therapy , Tirapazamine/adverse effects , Treatment Outcome
6.
J Nanobiotechnology ; 20(1): 43, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35062953

ABSTRACT

BACKGROUND: Chemodynamic therapy is a promising cancer treatment with specific therapeutic effect at tumor sites, as toxic hydroxyl radical (·OH) could only be generated by Fenton or Fenton-like reaction in the tumor microenvironment (TME) with low pH and high level of endogenous hydrogen peroxide. However, the low concentration of catalytic metal ions, excessive glutathione (GSH) and aggressive hypoxia at tumor site seriously restrict the curative outcomes of conventional chemodynamic therapy. RESULTS: In this study, polyethylene glycol-phenylboronic acid (PEG-PBA)-modified generation 5 (G5) poly(amidoamine) (PAMAM) dendrimers were synthesized as a targeted nanocarrier to chelate Cu(II) and then encapsulate hypoxia-sensitive drug tirapazamine (TPZ) by the formation of hydrophobic Cu(II)/TPZ complex for hypoxia-enhanced chemo/chemodynamic therapy. The formed G5.NHAc-PEG-PBA@Cu(II)/TPZ (GPPCT) nanoplatform has good stability and hemocompatibility, and could release Cu(II) ions and TPZ quickly in weakly acidic tumor sites via pH-sensitive dissociation of Cu(II)/TPZ. In vitro experiments showed that the GPPCT nanoplatforms can efficiently target murine breast cancer cells (4T1) cells overexpressing sialic acid residues, and show a significantly enhanced inhibitory effect on hypoxic cells by the activation of TPZ. The excessive GSH in tumors could be depleted by the reduction of Cu(II) to Cu(I), and abundant of toxic ·OH would be generated in tumor cells by Fenton reaction for chemodynamic therapy. In vivo experiments demonstrated that the GPPCT nanoplatform could specifically accumulate at tumors, effectively inhibit the growth and metastasis of tumors by the combination of CDT and chemotherapy, and be metabolized with no systemic toxicity. CONCLUSIONS: The targeted GPPCT nanoplatform may represent an effective model for the synergistic inhibition of different tumor types by hypoxia-enhanced chemo/chemodynamic therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Hypoxia/drug effects , Dendrimers , Nanostructures/chemistry , Tumor Microenvironment/drug effects , Animals , Dendrimers/chemistry , Dendrimers/pharmacology , Mice , Tirapazamine/pharmacology
7.
Biochem Biophys Res Commun ; 567: 118-124, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34147710

ABSTRACT

Osteosarcoma is the most common primary orthopedic malignant bone tumor in adolescents. However, the traditional neoadjuvant chemotherapy regimen has reached the bottleneck. TPZ is a hypoxic prodrug that has a powerful anti-tumor effect in the hypoxic microenvironment of tumors. And ferroptosis is a newly discovered cell death in 2012, and ferroptosis inducers have been used in anti-tumor therapy research in recent decades. Though, the role of TPZ and ferroptosis in osteosarcoma remains unclear. The aim of this study was to investigate the role of TPZ in osteosarcoma and the specific mechanism. MTT assay showed the extraordinary inhibition of TPZ on three osteosarcoma cells under hypoxia. And fluorescence of Fe2+ staining was enhanced by TPZ. Western blotting showed decreased expression of SLC7A11 and GPX4. Lipid peroxidation was confirmed by MDA assay and C11 BODIPY 581/591 staining. SLC7A11 overexpression could restored the proliferation and migration abilities inhibited by TPZ. Thus, we for the first time demonstrated that TPZ could inhibit the proliferation and migration of osteosarcoma cells, and induce ferroptosis in part through inhibiting SLC7A11.


Subject(s)
Amino Acid Transport System y+/metabolism , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Ferroptosis/drug effects , Osteosarcoma/drug therapy , Tirapazamine/pharmacology , Bone Neoplasms/metabolism , Cell Line, Tumor , Humans , Osteosarcoma/metabolism
8.
Invest New Drugs ; 39(3): 747-755, 2021 06.
Article in English | MEDLINE | ID: mdl-33428079

ABSTRACT

Background Tirapazamine's (TPZ) tolerability after an intra-arterial (IA) injection remains unclear. We investigated TPZ's safety and tolerability in rats by first injecting into the left hepatic artery and then performing a hepatic artery ligation, which recapitulates the transarterial embolization used clinically. Research design and methods: Forty-six rats in five groups were respectively injected with 0, 0.25, 0.50, 1.0, or more than 1.5 mL IA of TPZ (0.7 mg/mL) into the left hepatic artery and then subjected to hepatic artery ligation under laparotomy. Blood samples were collected four times daily up to day 15 after which the rats were euthanized and necropsied. The toxicity profile of IA injection of TPZ followed by hepatic artery ligation was then assessed. Results No significant changes to the rats' body weight and serum total bilirubin were observed. Serum alanine aminotransferase (ALT) levels increased slightly but remained below 100 U/L one day after treatment for most rats. Three rats in Groups 3 and 4 exhibited an over two-fold transient elevation of ALT. All ALT recovered to the baseline at day 14. Liver tissues were collected on day 15 using H&E staining. One rat in Group 3 showed ischemic coagulative necrosis in its liver tissue. Other sporadic pathological changes not related to TPZ doses were observed in Groups 2, 3, 4, and 5. Conclusion TPZ by IA injection followed by embolization is tolerated up to 7 mg/kg. This finding supports the strategy of administering an IA injection of TPZ followed by trans-arterial embolization to the liver.


Subject(s)
Antineoplastic Agents/toxicity , Tirapazamine/toxicity , Alanine Transaminase/blood , Animals , Bilirubin/blood , Female , Hepatic Artery/surgery , Injections, Intra-Arterial , Ligation , Liver/drug effects , Liver/pathology , Male , Rats , Tumor Hypoxia
9.
Nanotechnology ; 32(46)2021 Aug 23.
Article in English | MEDLINE | ID: mdl-34325415

ABSTRACT

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.


Subject(s)
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
10.
J Nanobiotechnology ; 19(1): 298, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34592996

ABSTRACT

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.


Subject(s)
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
11.
J Nanobiotechnology ; 19(1): 89, 2021 Mar 29.
Article in English | MEDLINE | ID: mdl-33781277

ABSTRACT

BACKGROUND: Areas of hypoxia are often found in triple-negative breast cancer (TNBC), it is thus more difficult to treat than other types of breast cancer, and may require combination therapies. A new strategy that combined bioreductive therapy with photodynamic therapy (PDT) was developed herein to improve the efficacy of cancer treatment. Our design utilized the characteristics of protoporphyrin IX (PpIX) molecules that reacted and consumed O2 at the tumor site, which led to the production of cytotoxic reactive oxygen species (ROS). The low microenvironmental oxygen levels enabled activation of a bioreductive prodrug, tirapazamine (TPZ), to become a toxic radical. The TPZ radical not only eradicated hypoxic tumor cells, but it also promoted therapeutic efficacy of PDT. RESULTS: To achieve the co-delivery of PpIX and TPZ for advanced breast cancer therapy, thin-shell hollow mesoporous Ia3d silica nanoparticles, designated as MMT-2, was employed herein. This nanocarrier designed to target the human breast cancer cell MDA-MB-231 was functionalized with PpIX and DNA aptamer (LXL-1), and loaded with TPZ, resulting in the formation of TPZ@LXL-1-PpIX-MMT-2 nanoVector. A series of studies confirmed that our nanoVectors (TPZ@LXL-1-PpIX-MMT-2) facilitated in vitro and in vivo targeting, and significantly reduced tumor volume in a xenograft mouse model. Histological analysis also revealed that this nanoVector killed tumor cells in hypoxic regions efficiently. CONCLUSIONS: Taken together, the synergism and efficacy of this new therapeutic design was confirmed. Therefore, we concluded that this new therapeutic strategy, which exploited a complementary combination of PpIX and TPZ, functioned well in both normoxia and hypoxia, and is a promising medical procedure for effective treatment of TNBC.


Subject(s)
Antineoplastic Agents/pharmacology , Nanoparticles/therapeutic use , Photochemotherapy/methods , Triple Negative Breast Neoplasms/drug therapy , Animals , Aptamers, Nucleotide , Cell Line, Tumor , Combined Modality Therapy , Female , Humans , Mice , Oxygen , Prodrugs , Reactive Oxygen Species , Silicon Dioxide , Tirapazamine , Tumor Burden , Tumor Hypoxia/drug effects , Xenograft Model Antitumor Assays
12.
Int J Mol Sci ; 22(6)2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33808887

ABSTRACT

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.


Subject(s)
Electrons , Tirapazamine/chemistry , Algorithms , Anions/chemistry , Models, Chemical , Radiation-Sensitizing Agents/chemistry
13.
J Pharm Pharm Sci ; 23: 231-242, 2020.
Article in English | MEDLINE | ID: mdl-32574141

ABSTRACT

The reduction potentials of bioreductively-activated drugs represent an important design parameter to be accommodated in the course of creating lead compounds and improving the efficacy of older generation drugs.  Reduction potentials are traditionally reported as single-electron reduction potentials, E(1), measured against reference electrodes under strictly defined experimental conditions.  More recently, computational chemists have described redox properties in terms of a molecule's highest occupied molecular orbital (HOMO) and lowest unoccupied molecular orbital (LUMO), in electron volts (eV).  The relative accessibility of HOMO/LUMO data through calculation using today's computer infrastructure and simplified algorithms make the calculated value (LUMO) attractive in comparison to the accepted but rigorous experimental determination of E(1).  This paper describes the correlations of eV (LUMO) to E(1) for three series of bioreductively-activated benzotriazine di-N-oxides (BTDOs), ring-substituted BTDOs, ring-added BTDOs and a selection of aromatic nitro compounds. The current computational approach is a closed-shell calculation with a single optimization.  Gas phase geometry optimization was followed by a single-point DFT (Density Functional Theory) energy calculation in the gas phase or in the presence of polar solvent.  The resulting DFT-derived LUMO energies (eV) calculated for BTDO analogues in gas phase and in presence of polar solvent (water) exhibited very strong linear correlations with high computational efficiency (r2 = 0.9925) and a very high predictive ability (MAD = 7 mV and RMSD = 9 mV) when compared to reported experimentally determined single-electron reduction potentials.


Subject(s)
Density Functional Theory , Electrons , Hypoxia , Tirapazamine/chemistry , Oxidation-Reduction , Tirapazamine/analogs & derivatives
14.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33228195

ABSTRACT

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.


Subject(s)
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
15.
Molecules ; 25(21)2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33105798

ABSTRACT

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.


Subject(s)
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
16.
Acc Chem Res ; 51(10): 2502-2511, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30234960

ABSTRACT

The common existence of hypoxia in solid tumors has been heavily researched because it renders tumors more resistant to most standard therapeutic methods, such as radiotherapy (RT), chemotherapy, and photodynamic therapy (PDT), and is associated with a more malignant phenotype and poor survival in patients with tumors. The development of hypoxia modulation methods for advanced therapeutic activity is therefore of great interest but remains a considerable challenge. Since the significant development of nanotechnology and nanomedicine, functionalized nanomaterials can be exploited as adjuvant "drugs" for these oxygen-dependent standard therapies or as hypoxia initiators for advanced new therapies to solid tumors. In this Account, we summarize our recent studies on the design and synthesis of nanomaterials with a set of desired chemistry benefits achievable by modulating hypoxia, suggesting a valid therapeutic option for tumors. The investigated strategies can be categorized into three groups: The first strategy is based on countering hypoxia. Considering that O2 deficiency is the major obstacle for the oxygen-dependent therapies, we initially developed methods to supply O2 by taking advantage of the hypoxia-responsive properties of nano-MnO2 or nanomaterials' photothermal effects for increased intratumoral blood flow. The second approach is to disregard hypoxia. Possible benefits of nanoagents include reducing/eliminating reliance on O2 or making O2 replacements as adjuvants to standard therapies. To this end, we investigated a nano-upconversion/scintillator with the capacity toup-/down-convert near-infrared light (NIR)/X-ray to luminescence in the ultraviolet/visible region fortype-I PDT with minimized oxygen-tension dependency or developed Fe-based nanomaterials for chemodynamic therapy (CDT) without external energy and oxygen participation for efficient free radical killing of deep tumors. The third strategy involves exploiting hypoxia. The unique biological characteristics of hypoxia are exploited to activate nanoagents for new therapies. To address the discrepancy between the nanoagents' demand and supply within the hypoxia region, a smart "molecule-nano" medicine that stays small-molecule-like in the bloodstream and turns into self-assembled nanovesicles after entry into the hypoxia region was constructed for hypoxia-adaptive photothermal therapy (PTT). In addition to traditional anti-angiogenesis therapy, we prepared Mg2Si nanoparticles by a special self-propagating high-temperature synthesis approach. These nanoparticles can directly remove the intratumoral oxygen via the oxidation reactions of Mg2Si and later efficiently block the rapid reoxygenation via tumor blood vessels by the resultant SiO2 microsheets for cancer starvation therapy. Taken together, these findings indicate that nanomaterials will assume a valuable role for anticancer exploration based on either their properties to make up oxygen deficiency or the use of hypoxia for therapeutic applications.


Subject(s)
Cell Hypoxia , Nanostructures/chemistry , Animals , HeLa Cells , Humans , Infrared Rays , Magnesium Silicates/chemistry , Mice , Nanostructures/therapeutic use , Neoplasms/drug therapy , Oxygen/chemistry , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Silicon Dioxide/chemistry , Tirapazamine/chemistry , Tirapazamine/therapeutic use , Transplantation, Heterologous
17.
Mol Pharm ; 16(5): 2172-2183, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30978027

ABSTRACT

Although photodynamic therapy (PDT) has been an attractive strategy for several cancer treatments in the clinical setting, PDT efficacy is attenuated by consumption of oxygen. To address this photodynamic issue, we adopted a phototherapy-chemotherapy combination strategy based on targeted delivery of the near-infrared photosensitizer indocyanine green (ICG), photothermal conversion agent polydopamine (PDA), and tirapazamine (TPZ), a hypoxia-activated prodrug. Under laser irradiation, ICG consumption of oxygen and aggravated hypoxia in tumor sites can activate TPZ to damage DNA. In parallel, ICG produces reactive oxygen species which work in synergy with PDA to enhance phototherapeutic efficiency. Herein, hybrid CaCO3/TPGS nanoparticles delivering ICG, PDA, and TPZ (ICG-PDA-TPZ NPs) were designed for effective and safe cancer therapy. ICG-PDA-TPZ NPs showed significantly improved cellular uptake and accumulation in tumors. Furthermore, we demonstrated that ICG-PDA-TPZ NPs showed intensive photodynamic and photothermal effects in vitro and in vivo, which synergized with TPZ in subcutaneous U87 malignant glioma growth and orthotopic B16F10 tumor inhibition, with negligible side effects. Thus, ICG-PDA-TPZ NPs could be an effective strategy for improvement of PDT.


Subject(s)
Hyperthermia, Induced , Indocyanine Green , Indoles , Nanoparticles , Neoplasms , Photochemotherapy , Prodrugs , Radiation-Sensitizing Agents , Tirapazamine , Animals , Humans , Mice , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems , Hyperthermia, Induced/methods , Indocyanine Green/metabolism , Indocyanine Green/therapeutic use , Indoles/metabolism , Indoles/therapeutic use , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasms/drug therapy , Photochemotherapy/adverse effects , Photochemotherapy/methods , Polymers/metabolism , Polymers/therapeutic use , Prodrugs/metabolism , Prodrugs/therapeutic use , Radiation-Sensitizing Agents/metabolism , Radiation-Sensitizing Agents/therapeutic use , Reactive Oxygen Species/radiation effects , Tirapazamine/metabolism , Tirapazamine/therapeutic use , Tissue Distribution , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
18.
Biomacromolecules ; 20(7): 2649-2656, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31125209

ABSTRACT

Stimuli-responsive drug delivery has rendered promising utilities in cancer treatment. Nevertheless, cancer selectivity as well as sensitivity still remains critical challenges that would undermine the therapeutic efficacy of chemodrugs and cause undesired systemic toxicity. Herein, a dual hypoxia-responsive drug delivery system was developed to enable photodynamic therapy (PDT)-induced drug release and drug activation intermediated via PDT-induced hypoxia. Particularly, tumor-targeting and hypoxia-dissociable nanoparticles (NPs) were self-assembled from the amphiphilic polyethylenimine-alkyl nitroimidazole [PEI-ANI, (PA)] and hyaluronic acid-chlorin e6 (HA-Ce6) to encapsulate bioreductive chemodrug, tirapazamine (TPZ). After systemic administration, the obtained PA/HA-Ce6@TPZ NPs enabled effective tumor accumulation due to HA-mediated cancer targeting. Upon receptor-mediated endocytosis, light irradiation (660 nm, 10 mW/cm2) produced high levels of reactive oxygen species to mediate PDT and generated a severe local hypoxic environment to dissociate the NPs and selectively release TPZ, as a consequence of hypoxia-triggered hydrophobic-to-hydrophilic transformation of ANI. In the meantime, TPZ was activated under hypoxia, finally contributing to a synergistic anticancer treatment between PDT and hypoxia-strengthened bioreductive chemotherapy. This study, therefore, demonstrates a suitable strategy for cancer-selective drug delivery as well as programmed combination therapy.


Subject(s)
Antineoplastic Agents , Drug Delivery Systems , Nanoparticles , Neoplasms, Experimental , Photochemotherapy , Photosensitizing Agents , Tirapazamine , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Nanomedicine , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Tirapazamine/chemistry , Tirapazamine/pharmacology
19.
Proc Natl Acad Sci U S A ; 113(42): 11937-11942, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27702890

ABSTRACT

Transarterial chemoembolization (TACE) is the main treatment for intermediate stage hepatocellular carcinoma (HCC) with Barcelona Clinic Liver Cancer classification because of its exclusive arterial blood supply. Although TACE achieves substantial necrosis of the tumor, complete tumor necrosis is uncommon, and the residual tumor generally rapidly recurs. We combined tirapazamine (TPZ), a hypoxia-activated cytotoxic agent, with hepatic artery ligation (HAL), which recapitulates transarterial embolization in mouse models, to enhance the efficacy of TACE. The effectiveness of this combination treatment was examined in HCC that spontaneously developed in hepatitis B virus X protein (HBx) transgenic mice. We proved that the tumor blood flow in this model was exclusively supplied by the hepatic artery, in contrast to conventional orthotopic HCC xenografts that receive both arterial and venous blood supplies. At levels below the threshold oxygen levels created by HAL, TPZ was activated and killed the hypoxic cells, but spared the normoxic cells. This combination treatment clearly limited the toxicity of TPZ to HCC, which caused the rapid and near-complete necrosis of HCC. In conclusion, the combination of TPZ and HAL showed a synergistic tumor killing activity that was specific for HCC in HBx transgenic mice. This preclinical study forms the basis for the ongoing clinical program for the TPZ-TACE regimen in HCC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Trans-Activators/genetics , Triazines/pharmacology , Animals , Biomarkers , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Hepatic Artery/surgery , Humans , Immunohistochemistry , Ligation , Liver Neoplasms/therapy , Magnetic Resonance Imaging , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Necrosis , Recurrence , Tirapazamine , Tumor Burden/drug effects , Viral Regulatory and Accessory Proteins , Xenograft Model Antitumor Assays
20.
Int J Mol Sci ; 20(18)2019 Sep 17.
Article in English | MEDLINE | ID: mdl-31533349

ABSTRACT

Derivatives of tirapazamine and other heteroaromatic N-oxides (ArN→O) exhibit promising antibacterial, antiprotozoal, and tumoricidal activities. Their action is typically attributed to bioreductive activation and free radical generation. In this work, we aimed to clarify the mechanism(s) of aerobic mammalian cell cytotoxicity of ArN→O performing the parallel studies of their reactions with NADPH:cytochrome P-450 reductase (P-450R), adrenodoxin reductase/adrenodoxin (ADR/ADX), and NAD(P)H:quinone oxidoreductase (NQO1); we found that in P-450R and ADR/ADX-catalyzed single-electron reduction, the reactivity of ArN→O (n = 9) increased with their single-electron reduction midpoint potential (E17), and correlated with the reactivity of quinones. NQO1 reduced ArN→O at low rates with concomitant superoxide production. The cytotoxicity of ArN→O in murine hepatoma MH22a and human colon adenocarcinoma HCT-116 cells increased with their E17, being systematically higher than that of quinones. The cytotoxicity of both groups of compounds was prooxidant. Inhibitor of NQO1, dicoumarol, and inhibitors of cytochromes P-450 α-naphthoflavone, isoniazid and miconazole statistically significantly (p < 0.02) decreased the toxicity of ArN→O, and potentiated the cytotoxicity of quinones. One may conclude that in spite of similar enzymatic redox cycling rates, the cytotoxicity of ArN→O is higher than that of quinones. This is partly attributed to ArN→O activation by NQO1 and cytochromes P-450. A possible additional factor in the aerobic cytotoxicity of ArN→O is their reductive activation in oxygen-poor cell compartments, leading to the formation of DNA-damaging species similar to those forming under hypoxia.


Subject(s)
Antineoplastic Agents/pharmacology , Oxidants/pharmacology , Tirapazamine/pharmacology , Antineoplastic Agents/chemistry , Biomarkers , Humans , Molecular Structure , NAD(P)H Dehydrogenase (Quinone)/metabolism , NADP/metabolism , Oxidants/chemistry , Oxidation-Reduction/drug effects , Reactive Oxygen Species , Tirapazamine/analogs & derivatives , Tirapazamine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL