Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
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.
Biol Reprod ; 102(1): 248-260, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31435664

ABSTRACT

Ataxia-telangiectasia-mutated (ATM) protein recognizes and repairs DNA double strand breaks through activation of cell cycle checkpoints and DNA repair proteins. Atm gene mutations increase female reproductive cancer risk. Phosphoramide mustard (PM) induces ovarian DNA damage and destroys primordial follicles, and pharmacological ATM inhibition prevents PM-induced follicular depletion. Wild-type (WT) C57BL/6 or Atm+/- mice were dosed once intraperitoneally with sesame oil (95%) or PM (25 mg/kg) in the proestrus phase of the estrous cycle and ovaries harvested 3 days thereafter. Atm+/- mice spent ~25% more time in diestrus phase than WT. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) on ovarian protein was performed and bioinformatically analyzed. Relative to WT, Atm+/- mice had 64 and 243 proteins increased or decreased in abundance, respectively. In WT mice, PM increased 162 and decreased 20 proteins. In Atm+/- mice, 173 and 37 proteins were increased and decreased, respectively, by PM. Exportin-2 (XPO2) was localized to granulosa cells of all follicle stages and was 7.2-fold greater in Atm+/- than WT mice. Cytoplasmic FMR1-interacting protein 1 was 6.8-fold lower in Atm+/- mice and was located in the surface epithelium with apparent translocation to the ovarian medulla post-PM exposure. PM induced γH2AX, but fewer γH2AX-positive foci were identified in Atm+/- ovaries. Similarly, cleaved caspase-3 was lower in the Atm+/- PM-treated, relative to WT mice. These findings support ATM involvement in ovarian DNA repair and suggest that ATM functions to regulate ovarian atresia.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Repair/physiology , Ovary/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Female , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Mice , Mice, Knockout , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Ovary/drug effects , Phosphoramide Mustards/pharmacology
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.
Mol Pharmacol ; 95(6): 638-651, 2019 06.
Article in English | MEDLINE | ID: mdl-30979813

ABSTRACT

Evofosfamide (TH-302) is a hypoxia-activated DNA-crosslinking prodrug currently in clinical development for cancer therapy. Oxygen-sensitive activation of evofosfamide depends on one-electron reduction, yet the reductases that catalyze this process in tumors are unknown. We used RNA sequencing, whole-genome CRISPR knockout, and reductase-focused short hairpin RNA screens to interrogate modifiers of evofosfamide activation in cancer cell lines. Involvement of mitochondrial electron transport in the activation of evofosfamide and the related nitroaromatic compounds EF5 and FSL-61 was investigated using 143B ρ 0 (ρ zero) cells devoid of mitochondrial DNA and biochemical assays in UT-SCC-74B cells. The potency of evofosfamide in 30 genetically diverse cancer cell lines correlated with the expression of genes involved in mitochondrial electron transfer. A whole-genome CRISPR screen in KBM-7 cells identified the DNA damage-response factors SLX4IP, C10orf90 (FATS), and SLFN11, in addition to the key regulator of mitochondrial function, YME1L1, and several complex I constituents as modifiers of evofosfamide sensitivity. A reductase-focused shRNA screen in UT-SCC-74B cells similarly identified mitochondrial respiratory chain factors. Surprisingly, 143B ρ 0 cells showed enhanced evofosfamide activation and sensitivity but had global transcriptional changes, including increased expression of nonmitochondrial flavoreductases. In UT-SCC-74B cells, evofosfamide oxidized cytochromes a, b, and c and inhibited respiration at complexes I, II, and IV without quenching reactive oxygen species production. Our results suggest that the mitochondrial electron transport chain contributes to evofosfamide activation and that predicting evofosfamide sensitivity in patients by measuring the expression of canonical bioreductive enzymes such as cytochrome P450 oxidoreductase is likely to be futile.


Subject(s)
Electron Transport/drug effects , Mitochondria/genetics , Neoplasms/genetics , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Sequence Analysis, RNA/methods , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , HCT116 Cells , Humans , Mitochondria/drug effects , Neoplasms/drug therapy , Prodrugs , RNA, Small Interfering/pharmacology
6.
BMC Cancer ; 16: 644, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27535748

ABSTRACT

BACKGROUND: Neo-adjuvant chemoradiotherapy followed by surgery is the standard treatment with curative intent for oesophageal cancer patients, with 5-year overall survival rates up to 50 %. However, patients' quality of life is severely compromised by oesophagectomy, and eventually many patients die due to metastatic disease. Most solid tumours, including oesophageal cancer, contain hypoxic regions that are more resistant to chemoradiotherapy. The hypoxia-activated prodrug evofosfamide works as a DNA-alkylating agent under these hypoxic conditions, which directly kills hypoxic cancer cells and potentially minimizes resistance to conventional therapy. This drug has shown promising results in several clinical studies when combined with chemotherapy. Therefore, in this phase I study we investigate the safety of evofosfamide added to the chemoradiotherapy treatment of oesophageal cancer. METHODS/DESIGN: A phase I, non-randomized, single-centre, open-label, 3 + 3 trial with repeated hypoxia PET imaging, will test the safety of evofosfamide in combination with neo-adjuvant chemoradiotherapy in potentially resectable oesophageal adenocarcinoma patients. Investigated dose levels range from 120 mg/m2 to 340 mg/m2. Evofosfamide will be administered one week before the start of chemoradiotherapy (CROSS-regimen) and repeated weekly up to a total of six doses. PET/CT acquisitions with hypoxia tracer (18)F-HX4 will be made before and after the first administration of evofosfamide, allowing early assessment of changes in hypoxia, accompanied with blood sampling to measure hypoxia blood biomarkers. Oesophagectomy will be performed according to standard clinical practice. Higher grade and uncommon non-haematological, haematological, and post-operative toxicities are the primary endpoints according to the CTCAEv4.0 and Clavien-Dindo classifications. Secondary endpoints are reduction in hypoxic fraction based on (18)F-HX4 imaging, pathological complete response, histopathological negative circumferential resection margin (R0) rate, local and distant recurrence rate, and progression free and overall survival. DISCUSSION: This is the first clinical trial testing evofosfamide in combination with chemoradiotherapy. The primary objective is to determine the dose limiting toxicity of this combined treatment and herewith to define the maximum tolerated dose and recommended phase 2 dose for future clinical studies. The addition of non-invasive repeated hypoxia imaging ('window-of-opportunity') enables us to identify the biologically effective dose. We believe this approach could also be used for other hypoxia targeted drugs. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02598687 .


Subject(s)
Adenocarcinoma/diagnostic imaging , Adenocarcinoma/therapy , Chemoradiotherapy, Adjuvant/methods , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/therapy , Nitroimidazoles/administration & dosage , Phosphoramide Mustards/administration & dosage , Cell Hypoxia/drug effects , Dose-Response Relationship, Drug , Esophagectomy , Female , Humans , Male , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Positron-Emission Tomography/methods , Preoperative Care , Survival Analysis , Treatment Outcome
7.
Anticancer Drugs ; 27(8): 723-5, 2016 09.
Article in English | MEDLINE | ID: mdl-27232101

ABSTRACT

Evofosfamide, also formerly known as TH-302, is an investigational hypoxia-activated prodrug and is used to target cancerous cells under hypoxic conditions, which is a feature possessed by multiple solid tumors including pancreatic tumors. Gemcitabine, a cytotoxic agent, has for many years been the standard first-line treatment for metastatic pancreatic cancer in patients. In recent years, combination chemotherapeutic therapies have provided a new avenue for molecular targeting by increasing the probability of eliminating the cancer and minimizing the likelihood of resistance. We have evaluated multiple studies in an effort to shed light on an emerging prodrug, evofosfamide, which operates by selectively targeting the tumor hypoxic compartment. A web-based literature search was performed through PubMed and Google Scholar using the keywords 'evofosfamide', 'TH-302,' and 'pancreatic tumor.' Of the available results, 53 relevant studies were reviewed and summarized. Chemotherapeutic agents such as evofosfamide, which targets tumor hypoxia, are new agents against cancer cells. Current experience with these agents is limited as additional and longer prospective studies are needed to further evaluate the clinical efficacy and postmarketing safety profile.


Subject(s)
Antineoplastic Agents/pharmacology , Nitroimidazoles/pharmacology , Pancreatic Neoplasms/drug therapy , Phosphoramide Mustards/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Nitroimidazoles/therapeutic use , Pancreatic Neoplasms/pathology , Phosphoramide Mustards/therapeutic use , Prodrugs/pharmacology , Tumor Hypoxia
8.
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
9.
BMC Cancer ; 15: 422, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25994202

ABSTRACT

BACKGROUND: The hypoxia-activated prodrug TH-302 is reduced at its nitroimidazole group and selectively under hypoxic conditions releases the DNA cross-linker bromo-isophosphoramide mustard (Br-IPM). Here, we have explored the effect of Chk1 inhibition on TH-302-mediated pharmacological activities. METHODS: We employed in vitro cell viability, DNA damage, cellular signaling assays and the in vivo HT29 human tumor xenograft model to study the effect of Chk1inhibition on TH-302 antitumor activities. RESULTS: TH-302 cytotoxicity is greatly enhanced by Chk1 inhibition in p53-deficient but not in p53-proficient human cancer cell lines. Chk1 inhibitors reduced TH-302-induced cell cycle arrest via blocking TH-302-induced decrease of phosphorylation of histone H3 and increasing Cdc2-Y15 phosphorylation. Employing the single-cell gel electrophoresis (comet) assay, we observed a potentiation of the TH-302 dependent tail moment. TH-302 induced γH2AX and apoptosis were also increased upon the addition of Chk1 inhibitor. Potentiation of TH-302 cytotoxicity by Chk1 inhibitor was only observed in cell lines proficient in, but not deficient in homology-directed DNA repair. We also show that combination treatment led to lowering of Rad51 expression levels as compared to either agent alone. In vivo data demonstrate that Chk1 inhibitor enhances TH-302 anti-tumor activity in p53 mutant HT-29 human tumor xenografts, supporting the hypothesis that these in vitro results can translate to enhanced in vivo efficacy of the combination. CONCLUSIONS: TH-302-mediated in vitro and in vivo anti-tumor activities were greatly enhanced by the addition of Chk1 inhibitors. The preclinical data presented in this study support a new approach for the treatment of p53-deficient hypoxic cancers by combining Chk1 inhibitors with the hypoxia-activated prodrug TH-302.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Thiophenes/pharmacology , Urea/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , CDC2 Protein Kinase/metabolism , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Checkpoint Kinase 1 , DNA Damage/drug effects , Female , HT29 Cells , Histones/metabolism , Humans , Mice , Mice, Nude , Mutation , Nitroimidazoles/therapeutic use , Phosphoproteins/metabolism , Phosphoramide Mustards/therapeutic use , Phosphorylation , Protein Kinase Inhibitors/therapeutic use , Rad51 Recombinase/metabolism , Signal Transduction/drug effects , Thiophenes/therapeutic use , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Urea/pharmacology , Urea/therapeutic use
10.
Molecules ; 19(7): 8803-19, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24968335

ABSTRACT

A series of N-mustards, which was conjugated to mono- or bis-naphthalimides with a flexible amine link, were synthesized and evaluated for cytotoxicity against five cancer cell lines (HCT-116, PC-3, U87 MG, Hep G2 and SK-OV-3). Several compounds displayed better activities than the control compound amonafide. Further evaluations by fluorescence spectroscopy studies and DNA-interstrand cross-linking assays revealed that the derivatives showed both alkylating and intercalating properties. Among the derivatives, the bis-naphthalimide N-mustard derivative 11b was found to exhibit the highest cytotoxic activity and DNA cross-linking ability. Both 11b and 7b induce HCT-116 cell apoptosis by S phase arrest.


Subject(s)
Antineoplastic Agents, Alkylating/chemical synthesis , Naphthalimides/chemical synthesis , Phosphoramide Mustards/chemical synthesis , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Drug Screening Assays, Antitumor , HCT116 Cells , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Naphthalimides/pharmacology , Phosphoramide Mustards/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism
11.
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
12.
Free Radic Biol Med ; 222: 579-587, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38992394

ABSTRACT

Hypoxic tumor microenvironments pose a significant challenge in cancer treatment. Hypoxia-activated prodrugs like evofosfamide aim to specifically target and eliminate these resistant cells. However, their effectiveness is often limited by reoxygenation after cell death. We hypothesized that ascorbate's pro-oxidant properties could be harnessed to induce transient hypoxia, enhancing the efficacy of evofosfamide by overcoming reoxygenation. To test this hypothesis, we investigated the sensitivity of MIA Paca-2 and A549 cancer cells to ascorbate in vitro and in vivo. Ascorbate induced a cytotoxic effect at 5 mM that could be alleviated by endogenous administration of catalase, suggesting a role for hydrogen peroxide in its cytotoxic mechanism. In vitro, Seahorse experiments indicated that the generation of hydrogen peroxide consumes oxygen, which is offset at later time points by a reduction in oxygen consumption due to hydrogen peroxide's cytotoxic effect. In vivo, photoacoustic imaging showed pharmacologic ascorbate treatment at sublethal levels triggered a complex, multi-phasic response in tumor oxygenation across both cell lines. Initially, ascorbate generated transient hypoxia within minutes through hydrogen peroxide production, via reactions that consume oxygen. This initial hypoxic phase peaked at around 150 s and then gradually subsided. However, at longer time scales (approximately 300 s) a vasodilation effect triggered by ascorbate resulted in increased blood flow and subsequent reoxygenation. Combining sublethal levels of i. p. Ascorbate with evofosfamide significantly prolonged tumor doubling time in MIA Paca-2 and A549 xenografts compared to either treatment alone. This improvement, however, was only observed in a subpopulation of tumors, highlighting the complexity of the oxygenation response.


Subject(s)
Ascorbic Acid , Carcinoma, Pancreatic Ductal , Hydrogen Peroxide , Pancreatic Neoplasms , Prodrugs , Xenograft Model Antitumor Assays , Humans , Prodrugs/pharmacology , Ascorbic Acid/pharmacology , Animals , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Hydrogen Peroxide/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Phosphoramide Mustards/pharmacology , A549 Cells , Tumor Microenvironment/drug effects , Cell Line, Tumor , Cell Hypoxia/drug effects , Mice, Nude , Nitrogen Mustard Compounds
13.
Indian J Exp Biol ; 51(8): 615-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24228385

ABSTRACT

Oxazaphosphorines belong to a group of alkylating agents. Mafosfamide cyclohexylamine salt (D-17272), 4-hydro-peroxy-cyclophosphamide (D-18864) and glufosfamide (D-19575, beta-D-glucose-isophosphoramide mustard) are new generation oxazaphosphorines. The objective of the present study was to compare the cytotoxic action of these oxazaphosphorine compounds against human histiocytic lymphoma U937 cells. The chemical structures of the oxazaphosphorines were responsible for the different responses of U937 cells. The cytotoxic effects of D-17272, D-18864, and D-19575 on U937 cells depended on the agent tested, its dose, and the time intervals after the oxazaphosphorine application. Among the oxazaphosphorine agents, D-18864 appeared to be the most cytotoxic, and D-19575 was characterized by the lowest cytotoxicity. The in vitro cytotoxic activities of the oxazaphosphorines were strongly associated with their cell death inducing potential.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Cyclophosphamide/analogs & derivatives , Glucose/analogs & derivatives , Ifosfamide/analogs & derivatives , Lymphoma, Large B-Cell, Diffuse/pathology , Membrane Potential, Mitochondrial/drug effects , Phosphoramide Mustards/pharmacology , Cell Proliferation/drug effects , Cyclophosphamide/pharmacology , Flow Cytometry , Glucose/pharmacology , Humans , Ifosfamide/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Necrosis , Tumor Cells, Cultured
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.
Acta Pol Pharm ; 70(3): 481-7, 2013.
Article in English | MEDLINE | ID: mdl-23757939

ABSTRACT

A series of new prodrugs: [bis(2-chloroethylamino)phosphoryloxy]methyl acetate, [bis(2-chloroethylamino)phosphoryloxy]methyl pivalate and [bis(2-chloroethylamino)phosphoryloxy]methyl benzoate, was obtained in the reaction of isophosphoramide mustard (iPAM) with the corresponding acyloxymethyl halides. The cytotoxic activity of these new compounds is also shown. All compounds were highly active in the inhibition of cancer cell proliferation against the human lung (A594), prostate (PC-3) and breast (MCF-7) cancer cell lines.


Subject(s)
Antineoplastic Agents/chemical synthesis , Phosphoramide Mustards/chemical synthesis , Prodrugs/chemical synthesis , Antineoplastic Agents/pharmacology , Biotransformation , Cell Line, Tumor , Drug Stability , Humans , Phosphoramide Mustards/pharmacology , Prodrugs/pharmacology
16.
Blood ; 116(9): 1524-7, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20530289

ABSTRACT

Hypoxia is associated with increased metastatic potential and poor prognosis in solid tumors. In this study, we demonstrated in the murine 5T33MM model that multiple myeloma (MM) cells localize in an extensively hypoxic niche compared with the naive bone marrow. Next, we investigated whether hypoxia could be used as a treatment target for MM by evaluating the effects of a new hypoxia-activated prodrug TH-302 in vitro and in vivo. In severely hypoxic conditions, TH-302 induces G(0)/G(1) cell-cycle arrest by down-regulating cyclinD1/2/3, CDK4/6, p21(cip-1), p27(kip-1), and pRb expression, and triggers apoptosis in MM cells by up-regulating the cleaved proapoptotic caspase-3, -8, and -9 and poly ADP-ribose polymerase while having no significant effects under normoxic conditions. In vivo treatment of 5T33MM mice induces apoptosis of the MM cells within the bone marrow microenvironment and decreases paraprotein secretion. Our data support that hypoxia-activated treatment with TH-302 provides a potential new treatment option for MM.


Subject(s)
Apoptosis/drug effects , Hypoxia/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Nitroimidazoles/pharmacology , Oxygen/metabolism , Phosphoramide Mustards/pharmacology , Prodrugs/pharmacology , Animals , Blotting, Western , Bone Marrow/drug effects , Bone Marrow/metabolism , Caspases/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Multiple Myeloma/metabolism , Neovascularization, Pathologic , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
17.
Drug Dev Ind Pharm ; 38(9): 1047-53, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22185667

ABSTRACT

Cyclophosphamide, an alkylating agent widely used as anticancer agent, biotransformed in vivo to unstable phosphoramidic mustard and acrolein, where the latter metabolite has been found responsible for hemorrhagic cystitis and renal toxicity. Being one of the most popular strategies to avoid these deleterious effects, prodrug design has been attempted, which can, in addition, enable selective drug targeting. Our efforts to design, synthesize and evaluate the enzymatically activated prodrug phosphorodiamidic mustard as potential candidate for selective chemotherapy in antibody-directed enzyme prodrug therapy or prodrug monotherapy strategies are described. We propose an improved synthesis of prodrug 14, consisting of a galactose moiety, a spacer and a cytotoxic drug and its cytotoxicity has been investigated. The prodrug 14 has been found to be nontoxic (in vitro) which could be a valuable candidate for further development.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Breast Neoplasms/drug therapy , Drug Delivery Systems , Drug Design , Galactosides/pharmacology , Lysosomes/metabolism , Phosphoramide Mustards/pharmacology , Prodrugs/pharmacology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/metabolism , Cell Survival/drug effects , Cyclophosphamide/adverse effects , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/metabolism , Cyclophosphamide/pharmacology , Drug Delivery Systems/adverse effects , Drug Stability , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Female , Galactosides/adverse effects , Galactosides/chemical synthesis , Galactosides/metabolism , HeLa Cells , Humans , Hydrolysis , Inhibitory Concentration 50 , Kinetics , MCF-7 Cells , Phosphoramide Mustards/adverse effects , Phosphoramide Mustards/chemical synthesis , Phosphoramide Mustards/metabolism , Prodrugs/adverse effects , Prodrugs/chemical synthesis , Prodrugs/metabolism , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism
18.
ChemMedChem ; 17(17): e202200239, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35771689

ABSTRACT

A series of novel artemisinin-piperazine-phosphoramide mustard (PPM) hybrids were designed and synthesized by incorporating phosphoramide mustard (PM) into dihydroartemisinin (DHA) via an efficient, catalyst-free two-step sequential substitution. Artemisinin-PPM hybrids showed better cytotoxic potency against HepG2 cells than both the parent DHA and the reference, vincristine (VCR). Structure-activity relationship (SAR) studies showed that the cytotoxicity was significantly enhanced by the introduction of a thiazole moiety. Hybrid 7 h, the most potent compound with the highest selectivity index IC50 (HEK-293T)/IC50 (HepG2)=16, displayed 7.4-fold stronger potency than VCR against HepG2 cells. In addition, hybrid 7 h was substantially more cytotoxic on all human cancer cells tested than on the corresponding non-cancerous cells. Flow cytometric analysis showed that 7 h significantly blocked the cell cycle in the G0/G1 phase and induced apoptosis in a concentration-dependent manner.


Subject(s)
Antineoplastic Agents , Artemisinins , Antineoplastic Agents/pharmacology , Apoptosis , Artemisinins/pharmacology , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , Humans , Phosphoramide Mustards/pharmacology , Piperazine/pharmacology , Structure-Activity Relationship
19.
Anticancer Drugs ; 22(6): 488-93, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21427562

ABSTRACT

Glufosfamide (ß-D-glucose-isophosphoramide mustard, D-19575) belongs to the oxazaphosphorine class. Glufosfamide is a novel glucose conjugate of ifosfamide in which isophosphoramide mustard, the alkylating metabolite of ifosfamide, is glycosidically linked to the ß-D-glucose molecule. Glufosfamide represents an attractive new agent for cancer therapy. Its mode of action on normal and pathological cells is still under experimental and clinical investigations. An assessment of the anticancer potential of glufosfamide is of key importance in therapy. The researchers reviewed the current knowledge available on glufosfamide tested in the preclinical studies/clinical trials, based on a collection of the original papers and conference abstracts published and relevant articles searched in the SCOPUS and MEDLINE database and websites.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Glucose/analogs & derivatives , Ifosfamide/analogs & derivatives , Neoplasms/drug therapy , Phosphoramide Mustards/pharmacology , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Drug Design , Glucose/adverse effects , Glucose/pharmacokinetics , Glucose/pharmacology , Glucose/therapeutic use , Humans , Ifosfamide/adverse effects , Ifosfamide/pharmacokinetics , Ifosfamide/pharmacology , Ifosfamide/therapeutic use , Neoplasms/pathology , Phosphoramide Mustards/adverse effects , Phosphoramide Mustards/pharmacokinetics , Phosphoramide Mustards/therapeutic use
20.
Can J Physiol Pharmacol ; 89(5): 335-43, 2011 May.
Article in English | MEDLINE | ID: mdl-21609276

ABSTRACT

Ifosfamide-induced nephrotoxicity is a serious adverse effect in children undergoing chemotherapy. Our previous cell and rodent models have shown that the antioxidant N-acetylcysteine (NAC), used extensively as an antidote for acetaminophen poisoning, protects renal tubular cells from ifosfamide-induced nephrotoxicity at a clinically relevant concentration. For the use of NAC to be clinically relevant in preventing ifosfamide nephrotoxicity, we must ensure there is no effect of NAC on the antitumor activity of ifosfamide. Common pediatric tumors that are sensitive to ifosfamide, human neuroblastoma SK-N-BE(2) and rhabdomyosarcoma RD114-B cells, received either no pretreatment or pretreatment with 400 µmol/L of NAC, followed by concurrent treatment with NAC and either ifosfamide or the active agent ifosfamide mustard. Ifosfamide mustard significantly decreased the growth of both cancer cell lines in a dose-dependent manner (p < 0.001). The different combined treatments of NAC alone, sodium 2-mercaptoethanesulfonate alone, or NAC plus sodium 2-mercaptoethanesulfonate did not significantly interfere with the tumor cytotoxic effect of ifosfamide mustard. These observations suggest that NAC may improve the risk/benefit ratio of ifosfamide by decreasing ifosfamide-induced nephrotoxicity without interfering with its antitumor effect in cancer cells clinically treated with ifosfamide.


Subject(s)
Acetylcysteine/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Drug Interactions , Ifosfamide/pharmacology , Antineoplastic Agents, Alkylating/adverse effects , Cell Line, Tumor , Cell Survival/drug effects , Drug-Related Side Effects and Adverse Reactions/prevention & control , Humans , Ifosfamide/adverse effects , Mesna/pharmacology , Neuroblastoma , Phosphoramide Mustards/pharmacology , Rhabdomyosarcoma
SELECTION OF CITATIONS
SEARCH DETAIL