Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Nanobiotechnology ; 20(1): 42, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35062959

ABSTRACT

Despite considerable progress has been achieved in hypoxia-associated anti-tumor therapy, the efficacy of utilizing hypoxia-activated prodrugs alone is not satisfied owing to the inadequate hypoxia within the tumor regions. In this work, a mitochondrial targeted nanoplatform integrating photodynamic therapy, photothermal therapy and hypoxia-activated chemotherapy has been developed to synergistically treat cancer and maximize the therapeutic window. Polydopamine coated hollow copper sulfide nanoparticles were used as the photothermal nanoagents and thermosensitive drug carriers for loading the hypoxia-activated prodrug, TH302, in our study. Chlorin e6 (Ce6) and triphenyl phosphonium (TPP) were conjugated onto the surface of the nanoplatform. Under the action of TPP, the obtained nanoplatform preferentially accumulated in mitochondria to restore the drug activity and avoid drug resistance. Using 660 nm laser to excite Ce6 can generate ROS and simultaneously exacerbate the cellular hypoxia. While under the irradiation of 808 nm laser, the nanoplatform produced local heat which can increase the release of TH302 in tumor cells, ablate cancer cells as well as intensify the tumor hypoxia levels. The aggravated tumor hypoxia then significantly boosted the anti-tumor efficiency of TH302. Both in vitro and in vivo studies demonstrated the greatly improved anti-cancer activity compared to conventional hypoxia-associated chemotherapy. This work highlights the potential of using a combination of hypoxia-activated prodrugs plus phototherapy for synergistic cancer treatment.


Subject(s)
Cell Hypoxia/drug effects , Drug Delivery Systems/methods , Mitochondria/metabolism , Nanoparticles/chemistry , Photochemotherapy/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Female , Mice , Mice, Inbred C57BL , Nitroimidazoles/chemistry , Nitroimidazoles/pharmacokinetics , Nitroimidazoles/pharmacology , Phosphoramide Mustards/chemistry , Phosphoramide Mustards/pharmacokinetics , Phosphoramide Mustards/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Tissue Distribution
2.
J Nanobiotechnology ; 19(1): 147, 2021 May 19.
Article in English | MEDLINE | ID: mdl-34011362

ABSTRACT

BACKGROUND: Triple negative breast cancer (TNBC) is an aggressive tumor with extremely high mortality that results from its lack of effective therapeutic targets. As an adhesion molecule related to tumorigenesis and tumor metastasis, cluster of differentiation-44 (also known as CD44) is overexpressed in TNBC. Moreover, CD44 can be effectively targeted by a specific hyaluronic acid analog, namely, chitosan oligosaccharide (CO). In this study, a CO-coated liposome was designed, with Photochlor (HPPH) as the 660 nm light mediated photosensitizer and evofosfamide (also known as TH302) as the hypoxia-activated prodrug. The obtained liposomes can help diagnose TNBC by fluorescence imaging and produce antitumor therapy by synergetic photodynamic therapy (PDT) and chemotherapy. RESULTS: Compared with the nontargeted liposomes, the targeted liposomes exhibited good biocompatibility and targeting capability in vitro; in vivo, the targeted liposomes exhibited much better fluorescence imaging capability. Additionally, liposomes loaded with HPPH and TH302 showed significantly better antitumor effects than the other monotherapy groups both in vitro and in vivo. CONCLUSION: The impressive synergistic antitumor effects, together with the superior fluorescence imaging capability, good biocompatibility and minor side effects confers the liposomes with potential for future translational research in the diagnosis and CD44-overexpressing cancer therapy, especially TNBC.


Subject(s)
Chitosan/pharmacology , Liposomes/chemistry , Nitroimidazoles/pharmacology , Oligosaccharides/pharmacology , Phosphoramide Mustards/pharmacology , Photochemotherapy/methods , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Chitosan/chemistry , Female , Humans , Hyaluronan Receptors , Hyaluronic Acid , Mice , Mice, Inbred BALB C , Mice, Nude , Nanomedicine , Nitroimidazoles/chemistry , Oligosaccharides/chemistry , Optical Imaging , Phosphoramide Mustards/chemistry , Photosensitizing Agents/chemistry , Prodrugs/chemistry , Triple Negative Breast Neoplasms/pathology
3.
Antioxid Redox Signal ; 35(11): 904-915, 2021 10 10.
Article in English | MEDLINE | ID: mdl-32787454

ABSTRACT

Aims: In hypoxic tumor microenvironments, the strongly reducing redox environment reduces evofosfamide (TH-302) to release a cytotoxic bromo-isophosphoramide (Br-IPM) moiety. This drug therefore preferentially attacks hypoxic regions in tumors where other standard anticancer treatments such as chemotherapy and radiation therapy are often ineffective. Various combination therapies with evofosfamide have been proposed and tested in preclinical and clinical settings. However, the treatment effect of evofosfamide monotherapy on tumor hypoxia has not been fully understood, partly due to the lack of quantitative methods to assess tumor pO2in vivo. Here, we use quantitative pO2 imaging by electron paramagnetic resonance (EPR) to evaluate the change in tumor hypoxia in response to evofosfamide treatment using two pancreatic ductal adenocarcinoma xenograft models: MIA Paca-2 tumors responding to evofosfamide and Su.86.86 tumors that do not respond. Results: EPR imaging showed that oxygenation improved globally after evofosfamide treatment in hypoxic MIA Paca-2 tumors, in agreement with the ex vivo results obtained from hypoxia staining by pimonidazole and in apparent contrast to the decrease in Ktrans observed in dynamic contrast-enhanced magnetic resonance imaging (DCE MRI). Innovations: The observation that evofosfamide not only kills the hypoxic region of the tumor but also improves oxygenation in the residual tumor regions provides a rationale for combination therapies using radiation and antiproliferatives post evofosfamide for improved outcomes. Conclusion: This study suggests that reoxygenation after evofosfamide treatment is due to decreased oxygen demand rather than improved perfusion. Following the change in pO2 after treatment may therefore yield a way of monitoring treatment response. Antioxid. Redox Signal. 35, 904-915.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/therapy , Cell Hypoxia/drug effects , Nitroimidazoles/pharmacology , Pancreatic Neoplasms/therapy , Phosphoramide Mustards/pharmacology , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemistry , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Nitroimidazoles/chemistry , Oxidation-Reduction , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphoramide Mustards/chemistry , Prodrugs/chemistry
4.
Bioorg Chem ; 98: 103747, 2020 05.
Article in English | MEDLINE | ID: mdl-32208207

ABSTRACT

The tumor microenvironment in chondrosarcoma (CHS), a chemo- and radio-resistant cancer provides unique hallmarks for developing a chondrosarcoma targeted drug-delivery system. Tumor targeting could be achieved using a quaternary ammonium function (QA) as a ligand for aggrecan, the main high negative charged proteoglycan of the extracellular matrix of CHS, and a 2-nitroimidazole as trigger that enables hypoxia-responsive drug release. In a previous work, ICF05016 was identified as efficient proteoglycan-targeting hypoxia-activated prodrug in a human extraskeletal myxoid chondrosarcoma model in mice and a first study of the structure-activity relationship of the QA function and the alkyl linker length was conducted. Here, we report the second part of the study, namely the modification of the nitro-aromatic trigger and the position of the proteoglycan-targeting ligand at the aromatic ring as well as the nature of the alkylating mustard. Synthetic approaches have been established to functionalize the 2-nitroimidazole ring at the N-1 and C-4 positions with a terminal tertiary alkyl amine, and to perform the phosphorylation step namely through the use of an amine borane complex, leading to phosphoramide and isophosphoramide mustards and also to a phosphoramide mustard bearing four 2-chloroethyl chains. In a preliminary study using a reductive chemical activation, QA-conjugates, except the 4-nitrobenzyl one, were showed to undergo efficient cleavage with release of the corresponding mustard. However N,N,N-trimethylpropylaminium tethered to the N-1 or C-4 positions of the imidazole seemed to hamper the enzymatic reduction of the prodrugs and all tested compounds featured moderate selectivity toward hypoxic cells, likely not sufficient for application as hypoxia-activated prodrugs.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Chondrosarcoma/drug therapy , Drug Design , Neoplasms, Connective and Soft Tissue/drug therapy , Phosphoramide Mustards/pharmacology , Prodrugs/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chondrosarcoma/pathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Neoplasms, Connective and Soft Tissue/pathology , Phosphoramide Mustards/chemical synthesis , Phosphoramide Mustards/chemistry , Prodrugs/chemical synthesis , Prodrugs/chemistry , Structure-Activity Relationship
5.
Anticancer Drugs ; 31(6): 617-622, 2020 07.
Article in English | MEDLINE | ID: mdl-32044797

ABSTRACT

Cyclophosphamide is an inert prodrug converted into 4-hydroxycyclophosphamide (OHCP) by hepatic hydroxylation. OHCP is in equilibrium with its tautomeric aldophosphamide (ALDO). From ALDO, the cytotoxic active metabolites are formed enzymatically by phosphodiesterases; these are the alkylating metabolite phosphoramide mustard (PAM) and the proapoptotic aldehyde 3-hydroxypropanal (HPA). PAM damages the DNA by alkylation; HPA amplifies the thereby induced apoptosis. The generally accepted view that acrolein, which is believed to be formed in the formation of PAM by ß-elimination from ALDO would be mainly responsible for the toxicity of cyclophosphamide, has to be revised because no acrolein is formed in the systemic circulation of patients after cyclophosphamide administration. It is shown that not acrolein, but OHCP itself is the true toxic metabolite of cyclophosphamide. Toxicity tests with OHCP and PAM were carried out, which demonstrated that OHCP unfolds its toxicity, not as a carrier of PAM but is toxic itself by reacting with nucleophilic groups of macromolecules, for example, thiol groups of membrane proteins. Further experiments demonstrate that the toxicity of oxazaphosphorine cytostatics may be drastically reduced if the formation of the pharmacologically active metabolite ALDO bypasses the formation of OHCP. Toxicity experiments in mice with S-ethanol-cyclophosphamide (SECP) that hydrolyzes to OHCP show that SECP is as toxic as OHCP, whereas the thiazolidine of ALDO, which hydrolyzes to ALDO bypassing OHCP is 7-9 times less toxic without loss of antitumor activity.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Cell Proliferation/drug effects , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/toxicity , Leukemia P388/pathology , Phosphoramide Mustards/toxicity , Animals , Antineoplastic Agents, Alkylating/chemistry , Cyclophosphamide/chemistry , Female , Leukemia P388/drug therapy , Male , Mice , Phosphoramide Mustards/chemistry , Toxicity Tests
6.
Biochem Pharmacol ; 156: 265-280, 2018 10.
Article in English | MEDLINE | ID: mdl-30134191

ABSTRACT

Evofosfamide (TH-302) is a clinical-stage hypoxia-activated prodrug with proven efficacy against hypoxic cells in preclinical tumour models. TH-302 is designed to release the DNA crosslinking agent bromo-isophosphoramide mustard (Br-IPM) when reduced in hypoxic tissue. Br-IPM is considered to diffuse locally from hypoxic regions, eliciting additional tumour cell killing, but the latter 'bystander effect' has not been demonstrated directly. Previous studies with multicellular co-cultures that included cells expressing the E. coli nitroreductase NfsA as a model TH-302 reductase have provided clear evidence of a bystander effect (which we confirm in the present study). However, NfsA is an oxygen-insensitive two-electron reductase that is not expected to generate the nitro radical intermediate that has been demonstrated to fragment to release Br-IPM. Here, we use mass spectrometry methods to characterise TH-302 metabolites generated by one-electron reduction (steady-state radiolysis by ionising radiation and cellular metabolism under hypoxia, including HCT116 cells that overexpress P450 oxidoreductase, POR) or by NfsA expressed in HCT116 cells under oxic conditions, and investigate the stability and cytotoxicity of these products. Br-IPM is shown to have very low cytotoxic potency when added to extracellular culture medium and to be rapidly converted to other hydrophilic products including dichloro-isophosphoramide mustard (IPM). Only traces of Br-IPM or IPM were detected in the extracellular medium when generated by cellular metabolism of TH-302. We identify, in NfsA-expressing cells, the hydroxylamine metabolite of TH-302, and downstream products resulting from rearrangement or hydration of the imidazole ring, and demonstrate that formation of these candidate bystander effect mediators is suppressed by hypoxia. This characterisation of the cellular pharmacology of TH-302 implies that bystander effects from hypoxic activation of TH-302 are unlikely to contribute to its anticancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , Bystander Effect , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Escherichia coli Proteins , HCT116 Cells , Humans , Molecular Structure , NADPH-Ferrihemoprotein Reductase , Nitroimidazoles/chemistry , Nitroreductases , Phosphoramide Mustards/chemistry , Prodrugs/pharmacology
7.
Molecules ; 23(8)2018 Aug 06.
Article in English | MEDLINE | ID: mdl-30082625

ABSTRACT

Novel mustard functionalized sophoridine derivatives were synthesized and evaluated for their cytotoxicity against of a panel of various cancer cell lines. They were shown to be more sensitive to S180 and H22 tumor cells with IC50 values ranging from 1.01⁻3.65 µM, and distinctly were more cytotoxic to cancer cells than normal cell L929. In addition, compounds 7a, 7c, and 7e displayed moderate tumor suppression without apparent organ toxicity in vivo against mice bearing H22 liver tumors. Furthermore, they arrested tumor cells in the G1 phase and induced cellular apoptosis. Their potential binding modes with DNA-Top I complex have also been investigated.


Subject(s)
Antineoplastic Agents/therapeutic use , Liver Neoplasms/drug therapy , Phosphoramide Mustards/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Mice , Structure-Activity Relationship
8.
Eur J Med Chem ; 127: 442-458, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28092860

ABSTRACT

A series of novel compounds with phosphoramide mustard functionality incorporated into the quinazoline scaffold of EGFR/HER2 inhibitors were designed and synthesized as multi-target-directed ligands against tumor cells. In vitro assays showed that tumor cell lines with high HER2 level were more sensitive to the compounds than tumor cells with low HER2 level. Compound 10d (EMB-3) was one of the most potent inhibitors with IC50 of 7.4 nM and 82 nM against EGFR and HER2, respectively. The mechanism studies were also supported by the effect of 10d-induced DNA damage in MDA-MB-468 cells. In vivo efficacy study showed that 10d could significantly inhibit H522 tumor xenograft model with a TGI of 68% at dose of 100 mg/kg (QDx28, p.o.) and no significant body weight loss was observed. MTD study indicated that compound 10d had no acute toxicity to mice at doses up to 900 mg/kg (single dose).


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Drug Design , Phosphoramide Mustards/chemical synthesis , Phosphoramide Mustards/pharmacology , Quinazolines/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chemistry Techniques, Synthetic , Dose-Response Relationship, Drug , Humans , Male , Mice , Phosphoramide Mustards/chemistry , Phosphoramide Mustards/pharmacokinetics , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
Bioorg Med Chem ; 24(12): 2697-706, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27156193

ABSTRACT

A series of Glutaryl-Hyp-Ala-Ser-Chg-Gln-4-aminobenzyl phosphoramide mustard conjugates (1a-e) was designed and synthesized as potential prodrugs for site-specific activation by PSA in prostate cancer cells. All conjugates were found to be substrates of PSA with cleavage occurring between Gln and the para-aminobenzyl (PAB) linker. Structure-activity relationship studies on these conjugates indicated that introduction of electron-withdrawing fluorine(s) on the phenyl ring in the PAB linker uniformly improved the chemical stability of the conjugates while the position of substitution affected differently the self-immolative process of conjugates upon proteolysis. Introduction of a fluorine at ortho position to benzylic phosphoramide as in 1b results in better stability of the conjugate prior to activation while maintaining its antiproliferative activity upon activation by PSA. The conjugate 1b with 2-fluoro substitution was identified as a promising lead for further evaluation and optimization in the development of prostate cancer-targeted prodrugs.


Subject(s)
Antineoplastic Agents/chemistry , Drug Design , Peptides/chemistry , Phosphoramide Mustards/chemistry , Prodrugs/chemistry , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Peptides/chemical synthesis , Peptides/metabolism , Peptides/pharmacology , Phosphoramide Mustards/chemical synthesis , Phosphoramide Mustards/metabolism , Phosphoramide Mustards/pharmacology , Prodrugs/chemical synthesis , Prodrugs/metabolism , Prodrugs/pharmacology , Prostatic Neoplasms/metabolism , Structure-Activity Relationship
10.
Chem Res Toxicol ; 29(2): 190-202, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26692166

ABSTRACT

N,N-Bis-(2-chloroethyl)-phosphorodiamidic acid (phosphoramide mustard, PM) and N,N-bis-(2-chloroethyl)-amine (nornitrogen mustard, NOR) are the two biologically active metabolites of cyclophosphamide, a DNA alkylating drug commonly used to treat lymphomas, breast cancer, certain brain cancers, and autoimmune diseases. PM and NOR are reactive bis-electrophiles capable of cross-linking cellular biomolecules to form covalent DNA-DNA and DNA-protein cross-links (DPCs). In the present work, a mass spectrometry-based proteomics approach was employed to characterize PM- and NOR-mediated DNA-protein cross-linking in human cells. Following treatment of human fibrosarcoma cells (HT1080) with cytotoxic concentrations of PM, over 130 proteins were found to be covalently trapped to DNA, including those involved in transcriptional regulation, RNA splicing/processing, chromatin organization, and protein transport. HPLC-ESI(+)-MS/MS analysis of proteolytic digests of DPC-containing DNA from NOR-treated cells revealed a concentration-dependent formation of N-[2-[cysteinyl]ethyl]-N-[2-(guan-7-yl)ethyl]amine (Cys-NOR-N7G) conjugates, confirming that it cross-links cysteine thiols of proteins to the N7 position of guanines in DNA. Cys-NOR-N7G adduct numbers were higher in NER-deficient xeroderma pigmentosum cells (XPA) as compared with repair proficient cells. Furthermore, both XPA and FANCD2 deficient cells were sensitized to PM treatment as compared to that of wild type cells, suggesting that Fanconi anemia and nucleotide excision repair pathways are involved in the removal of cyclophosphamide-induced DNA damage.


Subject(s)
Alkylating Agents/chemistry , DNA/chemistry , Nitrogen Mustard Compounds/chemistry , Phosphoramide Mustards/chemistry , Proteins/chemistry , Cell Line, Tumor , Chromatography, High Pressure Liquid , DNA/metabolism , DNA Adducts/analysis , Humans , Peptides/analysis , Proteins/metabolism , Proteomics , Spectrometry, Mass, Electrospray Ionization
11.
Bioorg Med Chem ; 22(21): 5747-56, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25440502

ABSTRACT

A series of bis(sulfonylethyl) and mono(sulfonylethyl) phenyl phosphotriesters of zidovudine (3'-azido-3'-deoxythymidine, AZT) were synthesized as potential anticancer prodrugs that liberate AZT monophosphate via nonenzymatic ß-elimination mechanism. Stability studies demonstrated that all the synthesized prodrugs spontaneously liberate AZT monophosphate with half-lives in the range of 0.07-278.8h under model physiological conditions in 0.1M phosphate buffer at pH 7.4 and 37 °C. Analogous to aldophosphamide, the elimination rates were accelerated in the presence of reconstituted human plasma under the same conditions. Among the compounds, 3, 4, 8, and 10 were comparable or superior to AZT against five established human cancerous cell lines in vitro. Moreover, the selected compounds were equally sensitive to both the wild-type osteosarcoma 143 B and the thymidine kinase-deficient 143 B/TK(-) cell lines. The findings are consistent with that these compounds deliver AZT monophosphate intracellularly.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Sulfones/chemistry , Zidovudine/chemistry , Zidovudine/pharmacology , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Esters , Half-Life , Humans , MCF-7 Cells , Phosphoramide Mustards/chemistry , Prodrugs/pharmacokinetics , Zidovudine/pharmacokinetics
12.
Chin J Cancer ; 33(2): 80-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23845143

ABSTRACT

Hypoxia, a state of low oxygen, is a common feature of solid tumors and is associated with disease progression as well as resistance to radiotherapy and certain chemotherapeutic drugs. Hypoxic regions in tumors, therefore, represent attractive targets for cancer therapy. To date, five distinct classes of bioreactive prodrugs have been developed to target hypoxic cells in solid tumors. These hypoxia-activated prodrugs, including nitro compounds, N-oxides, quinones, and metal complexes, generally share a common mechanism of activation whereby they are reduced by intracellular oxidoreductases in an oxygen-sensitive manner to form cytotoxins. Several examples including PR-104, TH-302, and EO9 are currently undergoing phase II and phase III clinical evaluation. In this review, we discuss the nature of tumor hypoxia as a therapeutic target, focusing on the development of bioreductive prodrugs. We also describe the current knowledge of how each prodrug class is activated and detail the clinical progress of leading examples.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Hypoxia/drug effects , Neoplasms , Prodrugs/pharmacology , Anthraquinones/chemistry , Anthraquinones/pharmacology , Antineoplastic Agents/chemistry , Aziridines/chemistry , Aziridines/pharmacology , Humans , Indolequinones/chemistry , Indolequinones/pharmacology , Molecular Structure , NAD(P)H Dehydrogenase (Quinone)/chemistry , NAD(P)H Dehydrogenase (Quinone)/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Nitrogen Mustard Compounds/chemistry , Nitrogen Mustard Compounds/pharmacology , Nitroimidazoles/chemistry , Nitroimidazoles/pharmacology , Phosphoramide Mustards/chemistry , Phosphoramide Mustards/pharmacology , Prodrugs/chemistry , Tirapazamine , Triazines/chemistry , Triazines/pharmacology
13.
Bioorg Med Chem ; 21(23): 7507-14, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24139844

ABSTRACT

In our continued effort to develop prodrugs of phosphoramide mustard, conjugates of 4-aminocyclophosphamide (4-NH2-CPA) with three PSA-specific peptides were synthesized and evaluated as substrates of PSA. These include conjugates of cis-(2R,4R)-4-NH2-CPA with a tetrapeptide Succinyl-Ser-Lys-Leu-Gln-OH, a hexapeptide Succinyl-His-Ser-Ser-Lys-Leu-Gln-OH, and a pentapeptide Glutaryl-Hyp-Ala-Ser-Chg-Gln-OH. These conjugates were cleaved by PSA efficiently and exclusively after the expected glutamine residue to release 4-NH2-CPA, the activated prodrug form of phosphoramide mustard. The cleavage was most efficient for the pentapeptide conjugate 3 (Glutaryl-Hyp-Ala-Ser-Chg-Gln-NH-CPA), which showed a half-life of 55 min with PSA, followed by the hexapeptide conjugate 2 (Succinyl-His-Ser-Ser-Lys-Leu-Gln-NH-CPA) and the tertrapeptide conjugate 1 (Succinyl-Ser-Lys-Leu-Gln-NH-CPA) with half-lives of 6.5 and 12h, respectively. These results indicate a potential of the conjugate 3 as an anticancer prodrug of phosphoramide mustard for selective PSA activation.


Subject(s)
Antineoplastic Agents/chemistry , Oligopeptides/chemistry , Phosphoramide Mustards/chemistry , Prodrugs/chemistry , Prostate-Specific Antigen/metabolism , Antineoplastic Agents/metabolism , Humans , Oligopeptides/metabolism , Phosphoramide Mustards/metabolism , Prodrugs/metabolism , Prostate-Specific Antigen/chemistry
14.
Folia Biol (Krakow) ; 61(1-2): 31-40, 2013.
Article in English | MEDLINE | ID: mdl-23767290

ABSTRACT

Mafosfamide cyclohexylamine salt (D-17272), 4-hydro-peroxy-cyclophosphamide (D-18864) and glufosfamide (D-19575, beta-D-glucose-isophosphoramide mustard) are new generation oxazaphosphorine agents. The present investigation was undertaken to determine the activity of these three oxazaphosphorines in human promyelocytic leukemia HL-60 cells. The research was conducted using the spectrophotometric MTT assay and the electronic Beckman Coulter and microscopy methods. Functional and morphological changes were observed after exposure of HL-60 cells to the oxazaphosphorine agents. The various patterns of temporary alterations in cell viability, size and count, and also in the frequency of leukemic cells undergoing mitotic catastrophe, apoptosis and necrosis, were shown. Different leukemic cell responses to the action of the three oxazaphosphorines were evaluated. These are the first data comparing the in vitro activity of D-17272, D-18864 and D-19575 against human promyelocytic leukemia cells.


Subject(s)
Antineoplastic Agents/pharmacology , Phosphoramide Mustards/pharmacology , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Molecular Structure , Phosphoramide Mustards/chemistry
15.
Mol Cancer Ther ; 11(3): 740-51, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22147748

ABSTRACT

TH-302 is a 2-nitroimidazole triggered hypoxia-activated prodrug (HAP) of bromo-isophosphoramide mustard currently undergoing clinical evaluation. Here, we describe broad-spectrum activity, hypoxia-selective activation, and mechanism of action of TH-302. The concentration and time dependence of TH-302 activation was examined as a function of oxygen concentration, with reference to the prototypic HAP tirapazamine, and showed superior oxygen inhibition of cytotoxicity and much improved dose potency relative to tirapazamine. Enhanced TH-302 cytotoxicity under hypoxia was observed across 32 human cancer cell lines. One-electron reductive enzyme dependence was confirmed using cells overexpressing human NADPH:cytochrome P450 oxidoreductase and radiolytic reduction established the single-electron stoichiometry of TH-302 fragmentation (activation). Examining downstream effects of TH-302 activity, we observed hypoxia-dependent induction of γH2AX phosphorylation, DNA cross-linking, and cell-cycle arrest. We used Chinese hamster ovary cell-based DNA repair mutant cell lines and established that lines deficient in homology-dependent repair, but not lines deficient in base excision, nucleotide excision, or nonhomologous end-joining repair, exhibited marked sensitivity to TH-302 under hypoxia. Consistent with this finding, enhanced sensitivity to TH-302 was also observed in lines deficient in BRCA1, BRCA2, and FANCA. Finally, we characterized TH-302 activity in the three-dimensional tumor spheroid and multicellular layer models. TH-302 showed much enhanced potency in H460 spheroids compared with H460 monolayer cells under normoxia. Multicellular layers composed of mixtures of parental HCT116 cells and HCT116 cells engineered to express an oxygen-insensitive bacterial nitroreductase showed that TH-302 exhibits a significant bystander effect.


Subject(s)
Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Prodrugs/pharmacology , Spheroids, Cellular/drug effects , Animals , CHO Cells , Cell Hypoxia , Cell Line, Tumor , Cell Survival/drug effects , Comet Assay , Cricetinae , Cricetulus , DNA Damage , Dose-Response Relationship, Drug , HCT116 Cells , HT29 Cells , Histones/metabolism , Humans , Inhibitory Concentration 50 , Molecular Structure , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nitroimidazoles/chemistry , Oxidation-Reduction/radiation effects , Oxygen/pharmacology , Phosphoramide Mustards/chemistry , Phosphorylation/drug effects , Prodrugs/chemistry , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Stem Cell Assay
16.
J Chromatogr A ; 1218(32): 5393-400, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21737081

ABSTRACT

A new derivatization and extraction technique termed as dispersive derivatization liquid-liquid extraction (DDLLE) speeds up the analysis process by removing the requirement for drying of the sample. The derivatization process takes place at the interface between the analyte containing aqueous phase and derivatization agent laden organic phase. The organic phase is highly dispersed using disperser solvent so that the total surface area is large. The derivatizing agent used is 1-(heptafluorobutyryl)imidazole and the resulting heptafluorobutyryl (HFB) derivatized analytes are partitioned into the organic phase. In addition to reduced sample preparation time, for some of the analytes, the HFB derivatives provide better spectral differentiation between isomers than conventional trimethylsilyl (TMS) derivatives. Method parameters for the DDLLE, such as extraction, and disperser solvent and their volume, type and amount of base, amount of heptafluorobutyrylimidazole and extraction time were optimized on diisopropylaminoethanol (DiPAE), ethyldiethanolamine (EDEA), triethanolamine (TEA) and thiodiglycol (TDG). The DDLLE was also used on various real world samples, which also includes few OPCW organized proficiency test and a spiked urine sample. The observed limit of detection (LOD) with 1mL of sample for DDLLE in full scan with AMDIS was 10ng/mL and with methane chemical ionization, multiple reaction monitoring (MRM) was 100pg/mL, i.e., 100fg on-column.


Subject(s)
Chemical Fractionation/methods , Chemical Warfare Agents/isolation & purification , Ethanolamines/isolation & purification , Phosphoramide Mustards/chemistry , Water Pollutants, Chemical/isolation & purification , Chemical Fractionation/instrumentation , Chemical Warfare Agents/analysis , Ethanolamines/analysis , Limit of Detection , Mustard Plant , Phosphoramide Mustards/isolation & purification , Water Pollutants, Chemical/analysis
17.
Biol Trace Elem Res ; 136(3): 294-301, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19838642

ABSTRACT

Selenium-containing thioredoxin reductase (TrxR) is an important target of cancer therapy. Many useful anticancer agents including bis-alkylating agents, cisplatin, and arsenic trioxide are known to interact with the selenocysteine dipeptide in the carboxy terminal region of thioredoxin reductase and inactivate its ability to reduce thioredoxin. Some investigators have postulated that the inactivation of TrxR may add to the cytotoxic potential of these anticancer agents. TH-302 is a newly developed antineoplastic drug which represents a potential new class of tumor selective hypoxia-activated prodrugs (HAPs). TH-302 is an inactive prodrug created by the covalent conjugation of 2-nitroimidazole as an oxygen sensor to bromo-isophosphoramide (Br-IPM). In the presence of severe hypoxia and near anoxia, the two imidazole sensor moiety undergoes reduction and the Br-IPM is released in situ. Bromo-IPM is a more potential analog of Chloro-IPM, the active alkylating moiety that is derived by activation of ifosfamide (IFO). We previously demonstrated that IFO could inhibit tumor TrxR activity and chloro-IPM is known to bind covalently to the seleno-cysteine dipeptide in thioredoxin reductase. The present study assessed the ability of TH-302 to activate in the tumors of mice-bearing hepatoma 22 (H22) and inactivate the tumor TrxR. In mice-bearing hepatoma 22 (H22) solid tumors, intraperitoneal (i.p.) injection with TH-302 at the dose of 200 mg/kg administered twice, a regimen which was well tolerated by the mice, significantly inhibited tumor growth. Also in this mice model, i.p. TH-302 at the dose of 300 mg/kg, which would be the maximum single i.p. administration dose tolerated by mice, and which induced only 2% body weight loss, significantly inhibited both TrxR and glutathione reductase (GR) activities by 46% (P < 0.001) and 60% (P < 0.001) as compared with the controls, respectively, at 3 h after the injection. Since TrxR is a key player in thioredoxin system and GR is the major reductase for the reduction of oxidized glutathione in glutathione system, the present results imply the anticancer effect of TH-302 is associated concurrently with modulation of TrxR and GR. These findings suggest that the anticancer activity of TH-302 in this model system may associate with both DNA alkylation and the modulation of TrxR and GR. In addition, they suggest that, by inhibition of these two critical reductases, with less glutathione available to intercept the reactive intermediates involved in DNA alkylation, the antitumor effects of the chemotherapy would be enhanced.


Subject(s)
Glutathione Reductase/antagonists & inhibitors , Liver Neoplasms, Experimental/prevention & control , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enzyme Assays , Glutathione Reductase/metabolism , Injections, Intraperitoneal , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/pathology , Male , Mice , Molecular Structure , Nitroimidazoles/administration & dosage , Nitroimidazoles/chemistry , Phosphoramide Mustards/administration & dosage , Phosphoramide Mustards/chemistry , Thioredoxin-Disulfide Reductase/metabolism , Time Factors , Treatment Outcome , Tumor Burden/drug effects
18.
Antimicrob Agents Chemother ; 54(3): 1193-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20028822

ABSTRACT

Nitroheterocyclic prodrugs have been used to treat trypanosomal diseases for more than 40 years. Recently, the key step involved in the activation of these compounds has been elucidated and shown to be catalyzed by a type I nitroreductase (NTR). This class of enzyme is normally associated with bacteria and is absent from most eukaryotes, with trypanosomes being a major exception. Here we exploit this difference by evaluating the trypanocidal activity of a library of nitrobenzylphosphoramide mustards against bloodstream-form Trypanosoma brucei parasites. Biochemical screening against the purified enzyme revealed that a subset of halogenated nitroaromatic compounds were effective substrates for T. brucei NTR (TbNTR), having apparent K(cat)/K(m) values approximately 100 times greater than nifurtimox. When tested against T. brucei, cytotoxicity mirrored enzyme activity, with 50% inhibitory concentrations of the most potent substrates being less than 10 nM. T. brucei NTR plays a key role in parasite killing: heterozygous lines displayed resistance to the compounds, while parasites overexpressing the enzyme showed hypersensitivity. We also evaluated the cytotoxicities of substrates with the highest trypanocidal activities by using mammalian THP-1 cells. The relative toxicities of these newly identified compounds were much lower than that of nifurtimox. We conclude that halogenated nitrobenzylphosphoramide mustards represent a novel class of antitrypanosomal agents, and their efficacy validates the strategy of specifically targeting NTR activity to develop new therapeutics.


Subject(s)
Nitroreductases/metabolism , Phosphoramide Mustards , Trypanocidal Agents , Trypanosoma brucei brucei/drug effects , Animals , Cell Line , Enzyme Activation , Humans , Oxidation-Reduction , Parasitic Sensitivity Tests , Phosphoramide Mustards/chemistry , Phosphoramide Mustards/metabolism , Phosphoramide Mustards/pharmacology , Phosphoramide Mustards/toxicity , Structure-Activity Relationship , Substrate Specificity , Trypanocidal Agents/chemistry , Trypanocidal Agents/metabolism , Trypanocidal Agents/pharmacology , Trypanocidal Agents/toxicity , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/growth & development
19.
Bioorg Med Chem Lett ; 18(14): 4059-63, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18556201

ABSTRACT

N-(2,2-Dimethyl-2-(2-nitrophenyl)acetyl)-4-aminocyclophosphamide isomers (DMNA-NH-CPA, 4) were synthesized stereospecifically from Boc-L-Hse(OBn)-OH and the degradation of the corresponding reduced amine 5a was investigated by UV/vis spectroscopy and LC/MS. The rate of cyclization of 5a was found to increase with decreasing pH, with half-lives ranging from 3.2 to 54 min at pH 4-7.4, suggesting that the cyclization is catalyzed by the hydronium ions. LC/MS analysis of the degradation products of 5a indicates that 4-aminocyclophosphamide is rapidly released from 4 upon reductive activation under acidic conditions and further decomposes into the cytotoxic phosphoramide mustard. These results validated 4-aminocyclophosphamide as a prodrug form of phosphoramide mustard and suggest that compound 4 can potentially be used as a prodrug of phosphoramide mustard for bioreductive activation.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/pharmacology , Phosphoramide Mustards/pharmacology , Catalysis , Chromatography, Liquid/methods , Drug Design , Hydrogen-Ion Concentration , Ions , Kinetics , Mass Spectrometry/methods , Models, Chemical , Onium Compounds/chemistry , Phosphoramide Mustards/chemistry , Protein Isoforms , Spectrophotometry, Ultraviolet/methods , Time Factors
20.
Curr Pharm Des ; 13(9): 963-78, 2007.
Article in English | MEDLINE | ID: mdl-17430192

ABSTRACT

The oxazaphosphorines including cyclophosphamide (CPA, Cytoxan, or Neosar), ifosfamide (IFO, Ifex) and trofosfamide (Ixoten) represent an important group of therapeutic agents due to their substantial antitumor and immunomodulating activity. However, several intrinsic limitations have been uncounted during the clinical use of these oxazaphosphorines, including substantial pharmacokinetic variability, resistance and severe host toxicity. To circumvent these problems, new oxazaphosphorines derivatives have been designed and evaluated with an attempt to improve the selectivity and response with reduced host toxicity. These include mafosfamide (NSC 345842), glufosfamide (D19575, beta-D-glucosylisophosphoramide mustard), S-(-)-bromofosfamide (CBM-11), NSC 612567 (aldophosphamide perhydrothiazine) and NSC 613060 (aldophosphamide thiazolidine). Mafosfamide is an oxazaphosphorine analog that is a chemically stable 4-thioethane sulfonic acid salt of 4-hydroxy-CPA. Glufosfamide is IFO derivative in which the isophosphoramide mustard, the alkylating metabolite of IFO, is glycosidically linked to a beta-D-glucose molecule. Phase II studies of glufosfamide in the treatment of pancreatic cancer, non-small cell lung cancer (NCSLC), and recurrent glioblastoma multiform (GBM) have recently completed and Phase III trials are ongoing, while Phase I studies of intrathecal mafosfamide have recently completed for the treatment of meningeal malignancy secondary to leukemia, lymphoma, or solid tumors. S-(-)-bromofosfamide is a bromine-substituted IFO analog being evaluated in a few Phase I clinical trials. The synthesis and development of novel oxazaphosphorine analogs with favourable pharmacokinetic and pharmacodynamic properties still constitutes a great challenge for medicinal chemists and cancer pharmacologists.


Subject(s)
Antineoplastic Agents , Drug Design , Neoplasms/drug therapy , Phosphoramide Mustards , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Clinical Trials as Topic , Humans , Lethal Dose 50 , Molecular Structure , Phosphoramide Mustards/adverse effects , Phosphoramide Mustards/chemistry , Phosphoramide Mustards/therapeutic use , Phosphoramide Mustards/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...