Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Int J Mol Sci ; 25(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38928299

ABSTRACT

Bacterial nitroreductase enzymes capable of activating imaging probes and prodrugs are valuable tools for gene-directed enzyme prodrug therapies and targeted cell ablation models. We recently engineered a nitroreductase (E. coli NfsB F70A/F108Y) for the substantially enhanced reduction of the 5-nitroimidazole PET-capable probe, SN33623, which permits the theranostic imaging of vectors labeled with oxygen-insensitive bacterial nitroreductases. This mutant enzyme also shows improved activation of the DNA-alkylation prodrugs CB1954 and metronidazole. To elucidate the mechanism behind these enhancements, we resolved the crystal structure of the mutant enzyme to 1.98 Å and compared it to the wild-type enzyme. Structural analysis revealed an expanded substrate access channel and new hydrogen bonding interactions. Additionally, computational modeling of SN33623, CB1954, and metronidazole binding in the active sites of both the mutant and wild-type enzymes revealed key differences in substrate orientations and interactions, with improvements in activity being mirrored by reduced distances between the N5-H of isoalloxazine and the substrate nitro group oxygen in the mutant models. These findings deepen our understanding of nitroreductase substrate specificity and catalytic mechanisms and have potential implications for developing more effective theranostic imaging strategies in cancer treatment.


Subject(s)
Metronidazole , Nitroimidazoles , Nitroreductases , Nitroreductases/metabolism , Nitroreductases/chemistry , Nitroreductases/genetics , Nitroimidazoles/chemistry , Nitroimidazoles/metabolism , Metronidazole/chemistry , Metronidazole/metabolism , Metronidazole/pharmacology , Prodrugs/metabolism , Prodrugs/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Positron-Emission Tomography/methods , Escherichia coli/genetics , Escherichia coli/metabolism , Catalytic Domain , Protein Engineering , Models, Molecular , Aziridines/chemistry , Aziridines/metabolism
2.
Biotechnol Lett ; 43(1): 203-211, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32851465

ABSTRACT

OBJECTIVES: To use directed evolution to improve YfkO-mediated reduction of the 5-nitroimidazole PET-capable probe SN33623 without impairing conversion of the anti-cancer prodrug CB1954. RESULTS: Two iterations of error-prone PCR, purifying selection, and FACS sorting in a DNA damage quantifying GFP reporter strain were used to identify three YfkO variants able to sensitize E. coli host cells to at least 2.4-fold lower concentrations of SN33623 than the native enzyme. Two of these variants were able to be purified in a functional form, and in vitro assays revealed these were twofold and fourfold improved in kcat/KM with SN33623 over wild type YfkO. Serendipitously, the more-active variant was also nearly fourfold improved in kcat/KM versus wild type YfkO in converting CB1954 to a genotoxic drug. CONCLUSIONS: The enhanced activation of the PET imaging probe SN33623 and CB1954 prodrug exhibited by the lead evolved variant of YfkO offers prospects for improved enzyme-prodrug therapy.


Subject(s)
Bacillus subtilis , Bacterial Proteins/genetics , Directed Molecular Evolution/methods , Nitroimidazoles/metabolism , Nitroreductases/genetics , Antineoplastic Agents/metabolism , Aziridines/metabolism , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Bacterial Proteins/metabolism , Enzyme Therapy , Nitroreductases/metabolism
3.
Molecules ; 26(11)2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34206005

ABSTRACT

Phenanthroindolizidines, such as antofine and tylophorine, are a family of natural alkaloids isolated from different species of Asclepiadaceas. They are characterized by interesting biological activities, such as pronounced cytotoxicity against different human cancerous cell lines, including multidrug-resistant examples. Nonetheless, these derivatives are associated with severe neurotoxicity and loss of in vivo activity due to the highly lipophilic nature of the alkaloids. Here, we describe the development of highly polar prodrugs of antofine and tylophorine as hypoxia-targeted prodrugs. The developed quaternary ammonium salts of phenanthroindolizidines showed high chemical and metabolic stability and are predicted to have no penetration through the blood-brain barrier. The designed prodrugs displayed decreased cytotoxicity when tested under normoxic conditions. However, their cytotoxic activity considerably increased when tested under hypoxic conditions.


Subject(s)
Alkaloids/chemistry , Antineoplastic Agents/chemical synthesis , Indoles/chemistry , Indolizines/chemistry , Phenanthrenes/chemistry , Phenanthrolines/chemistry , Prodrugs/chemical synthesis , Quaternary Ammonium Compounds/chemical synthesis , Quaternary Ammonium Compounds/pharmacology , Tumor Hypoxia/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cricetulus , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , MCF-7 Cells , Molecular Structure , Prodrugs/chemistry , Prodrugs/pharmacology , Quaternary Ammonium Compounds/chemistry , Structure-Activity Relationship
4.
Bioorg Med Chem Lett ; 30(11): 127135, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32229061

ABSTRACT

Culicinin D (1), a 10 amino acid peptaibol originally isolated from Culicinomyces clavisporus, exhibits potent activity against a range of cancer cell lines. Building on our previous work exploring the structure-activity relationship (SAR) of the unusual (2S,4S,6R)-AHMOD residue, a series of analogues of culicinin D were prepared to further investigate the SAR of these peptaibols. Alanine scanning of a potent and readily accessible analogue 23 revealed the effect of each residue on antiproliferative activity, and a small panel of analogues were prepared to explore the SAR of the non-natural amino acid residue (2S,4R)-AMD. Results from the alanine scan were used to design an expanded library of culicinin D analogues, leading to the discovery of cyclohexylalanine analogue 52, which exhibited better antiproliferative activity than the natural product 1.


Subject(s)
Alanine/chemistry , Antineoplastic Agents/chemical synthesis , Oligopeptides/chemistry , Peptaibols/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Hypocreales/chemistry , Hypocreales/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Peptaibols/chemical synthesis , Peptaibols/pharmacology , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 30(16): 127331, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32631536

ABSTRACT

Culicinin D (1), a 10 amino acid peptaibol containing several unusual residues, has been shown to exhibit potent anticancer activity. Previous work in our group towards developing a structure-activity relationship (SAR) for this peptaibol has concentrated on replacement of the synthetically challenging AHMOD (3) and AMD (4) residues, resulting in the discovery of analogues with equivalent or better potency and simplified synthesis. The SAR of this peptaibol is extended in this work by investigating the effect of the N-terminal lipid tail and C-terminal amino alcohol, revealing the key contribution of each of these moieties on antiproliferative activity in a panel of breast and lung cancer cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Oligopeptides/pharmacology , Peptaibols/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Peptaibols/chemical synthesis , Peptaibols/chemistry , Structure-Activity Relationship
6.
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
7.
Angew Chem Int Ed Engl ; 59(33): 13764-13776, 2020 08 10.
Article in English | MEDLINE | ID: mdl-31889388

ABSTRACT

Drugs that function through allosteric inhibition of kinase signaling represent a promising approach for the targeted discovery of therapeutics. The majority of developed allosteric kinase inhibitors are characterized as type III and IV inhibitors that show good kinome selectivity but generally lack the subtype selectivity of same kinase family. Recently allosteric inhibitors have been developed that bind outside the catalytic kinase domain with high selectivity for specific kinase subtypes. Allosteric inhibitors that bind to the pseudokinase domain of pseudokinase or the extracellular domain of receptor tyrosine kinases are reviewed. We also review recent developments in the field of allosteric kinase inhibitors including examples of proteolysis targeting chimeras, and highlight the unique binding modes for each type of inhibitors and address future opportunities in this area.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Allosteric Regulation , Animals , Humans , Molecular Structure , Protein Kinase Inhibitors/chemistry , Proteolysis , Substrate Specificity
8.
Biochemistry ; 58(35): 3700-3710, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31403283

ABSTRACT

Gene-directed enzyme prodrug therapy (GDEPT) uses tumor-tropic vectors to deliver prodrug-converting enzymes such as nitroreductases specifically to the tumor environment. The nitroreductase NfsB from Escherichia coli (NfsB_Ec) has been a particular focal point for GDEPT and over the past 25 years has been the subject of several engineering studies seeking to improve catalysis of prodrug substrates. To facilitate clinical development, there is also a need to enable effective non-invasive imaging capabilities. SN33623, a 5-nitroimidazole analogue of 2-nitroimidazole hypoxia probe EF5, has potential for PET imaging exogenously delivered nitroreductases without generating confounding background due to tumor hypoxia. However, we show here that SN33623 is a poor substrate for NfsB_Ec. To address this, we used assay-guided sequence and structure analysis to identify two conserved residues that block SN33623 activation in NfsB_Ec and close homologues. Introduction of the rational substitutions F70A and F108Y into NfsB_Ec conferred high levels of SN33623 activity and enabled specific labeling of E. coli expressing the engineered enzyme. Serendipitously, the F70A and F108Y substitutions also substantially improved activity with the anticancer prodrug CB1954 and the 5-nitroimidazole antibiotic prodrug metronidazole, which is a potential biosafety agent for targeted ablation of nitroreductase-expressing vectors.


Subject(s)
Drug Monitoring/methods , Escherichia coli Proteins/metabolism , Etanidazole/analogs & derivatives , Hydrocarbons, Fluorinated/metabolism , Molecular Imaging/methods , Nitroimidazoles/therapeutic use , Nitroreductases/metabolism , Positron-Emission Tomography/methods , Prodrugs/therapeutic use , Antineoplastic Agents/therapeutic use , Biosensing Techniques/methods , Cell Hypoxia/physiology , Enzyme Activation , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Etanidazole/chemistry , Etanidazole/metabolism , Genetic Therapy/methods , HCT116 Cells , Humans , Hydrocarbons, Fluorinated/chemistry , Imidazoles/pharmacology , Imidazoles/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Nitroimidazoles/pharmacology , Nitroreductases/genetics , Prodrugs/metabolism , Protein Engineering
9.
Bioorg Med Chem Lett ; 29(10): 1215-1219, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30885680

ABSTRACT

Amide- and ester-linked kinase inhibitor-cytotoxin conjugates were rationally designed and synthesised as prototype hypoxia-activated anticancer mutual prodrugs. Chemical reduction of an aryl nitro trigger moiety was shown to initiate a spontaneous cyclisation/fragmentation reaction that simultaneously released the kinase inhibitor semaxanib (SU5416) and the amine- or alcohol-linked cytotoxin from the prodrugs. Preliminary cell testing and reduction potential measurements support optimisation of the compounds towards tumour-selective mutual prodrugs.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cytotoxins/chemistry , Prodrugs/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Alcohols/chemistry , Amines/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Cytotoxins/pharmacology , Drug Screening Assays, Antitumor/methods , Floxuridine/chemistry , Humans , Indoles/chemistry , Indoles/pharmacology , Molecular Structure , Prodrugs/pharmacology , Proof of Concept Study , Protein Kinase Inhibitors/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Structure-Activity Relationship , Tumor Hypoxia
10.
Med Res Rev ; 38(5): 1550-1581, 2018 09.
Article in English | MEDLINE | ID: mdl-29377179

ABSTRACT

Both the first-generation reversible epidermal growth factor receptor (EGFR) inhibitors gefitinib and erlotinib and the second-generation covalent epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) afatinib have significantly improved the survival of non-small-cell lung cancer (NSCLC) patients with activating EGFR mutations. However, a secondary EGFRT790M mutation leads to the clinically acquired resistance to the first- and second-generation EGFR-TKIs drugs. A number of the third-generation wild-type sparing EGFR inhibitors, for example, WZ4002, CO1686, AZD9291, HM61713, EGF816, ASP8173, and PF0674775, have been developed, among which AZD9291 has been approved by US FDA for the treatment of NSCLC patients with EGFRT790M . More recently, a tertiary EGFRC797S mutation was reported as the dominant resistance mechanism to the third-generation irreversible inhibitors. It is highly desirable to develop the fourth-generation EGFR inhibitors. This review summarizes the mechanisms of acquired resistance and the latest medicinal chemistry advances on the third- and fourth-generation EGFR inhibitors, with special attention being paid to the allosteric and reversible inhibitors combating the tertiary EGFRC797S mutation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Chemistry, Pharmaceutical , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/drug effects , Humans , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
11.
Biochem J ; 471(2): 131-53, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26431849

ABSTRACT

This review examines the vast catalytic and therapeutic potential offered by type I (i.e. oxygen-insensitive) nitroreductase enzymes in partnership with nitroaromatic prodrugs, with particular focus on gene-directed enzyme prodrug therapy (GDEPT; a form of cancer gene therapy). Important first indications of this potential were demonstrated over 20 years ago, for the enzyme-prodrug pairing of Escherichia coli NfsB and CB1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide]. However, it has become apparent that both the enzyme and the prodrug in this prototypical pairing have limitations that have impeded their clinical progression. Recently, substantial advances have been made in the biodiscovery and engineering of superior nitroreductase variants, in particular development of elegant high-throughput screening capabilities to enable optimization of desirable activities via directed evolution. These advances in enzymology have been paralleled by advances in medicinal chemistry, leading to the development of second- and third-generation nitroaromatic prodrugs that offer substantial advantages over CB1954 for nitroreductase GDEPT, including greater dose-potency and enhanced ability of the activated metabolite(s) to exhibit a local bystander effect. In addition to forging substantial progress towards future clinical trials, this research is supporting other fields, most notably the development and improvement of targeted cellular ablation capabilities in small animal models, such as zebrafish, to enable cell-specific physiology or regeneration studies.


Subject(s)
Aziridines/therapeutic use , Escherichia coli Proteins , Genetic Therapy/methods , Neoplasms, Experimental/therapy , Nitroreductases , Prodrugs/therapeutic use , Animals , Directed Molecular Evolution , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/genetics , Escherichia coli Proteins/therapeutic use , Humans , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Nitroreductases/biosynthesis , Nitroreductases/genetics , Nitroreductases/therapeutic use
12.
J Struct Biol ; 192(3): 539-544, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26522274

ABSTRACT

The discovery of genetic drivers of lung cancer in patient sub-groups has led to their use as predictive biomarkers and as targets for selective drug therapy. Some of the most important lung cancer drivers are mutations in the EGFR gene, for example, the exon 19 deletions and the L858R variant that confer sensitivity to the front line drugs erlotinib and gefitinib; the acquired T790M variants confer drug resistance and a poor prognosis. A challenge then in targeting EGFR is to produce drugs that inhibit both sensitising variants and resistance variants, leaving wild type protein in healthy cells unaffected. One such agent is AstraZeneca's "breakthrough" AZD9291 molecule that shows a 200-fold selectivity for T790M/L858R over wild type EGFR. Our X-ray crystal structure reveals the binding mode of AZD9291 to the kinase domain of wild type EGFR.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Lung Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Acrylamides/metabolism , Aniline Compounds/metabolism , Crystallography, X-Ray , Drug Resistance, Neoplasm/genetics , ErbB Receptors/ultrastructure , Erlotinib Hydrochloride/pharmacology , Gefitinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Protein Binding/physiology , Quinazolines/pharmacology
13.
J Biol Chem ; 288(52): 37138-53, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24196959

ABSTRACT

Hypoxia, a ubiquitous feature of tumors, can be exploited by hypoxia-activated prodrugs (HAP) that are substrates for one-electron reduction in the absence of oxygen. NADPH:cytochrome P450 oxidoreductase (POR) is considered one of the major enzymes responsible, based on studies using purified enzyme or forced overexpression in cell lines. To examine the role of POR in HAP activation at endogenous levels of expression, POR knock-outs were generated in HCT116 and SiHa cells by targeted mutation of exon 8 using zinc finger nucleases. Absolute quantitation by proteotypic peptide mass spectrometry of DNA sequence-confirmed multiallelic mutants demonstrated expression of proteins with residual one-electron reductase activity in some clones and identified two (Hko2 from HCT116 and S2ko1 from SiHa) that were functionally null by multiple criteria. Sensitivities of the clones to 11 HAP (six nitroaromatics, three benzotriazine N-oxides, and two quinones) were compared with wild-type and POR-overexpressing cells. All except the quinones were potentiated by POR overexpression. Knocking out POR had a marked effect on antiproliferative activity of the 5-nitroquinoline SN24349 in both genetic backgrounds after anoxic exposure but little or no effect on activity of most other HAP, including the clinical stage 2-nitroimidazole mustard TH-302, dinitrobenzamide mustard PR-104A, and benzotriazine N-oxide SN30000. Clonogenic cell killing and reductive metabolism of PR-104A and SN30000 under anoxia also showed little change in the POR knock-outs. Thus, although POR expression is a potential biomarker of sensitivity to some HAP, identification of other one-electron reductases responsible for HAP activation is needed for their rational clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , NADPH-Ferrihemoprotein Reductase/biosynthesis , Neoplasm Proteins/biosynthesis , Neoplasms/drug therapy , Prodrugs/pharmacology , Antineoplastic Agents/pharmacokinetics , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line, Tumor , Drug Screening Assays, Antitumor , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , NADPH-Ferrihemoprotein Reductase/genetics , Neoplasm Proteins/genetics , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Prodrugs/pharmacokinetics
14.
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
15.
J Med Chem ; 67(4): 2667-2689, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38348819

ABSTRACT

Fibroblast growth factor receptor 4 (FGFR4) has been considered as a potential anticancer target due to FGF19/FGFR4 mediated aberrant signaling in hepatocellular carcinoma (HCC). Several FGFR4 inhibitors have been reported, but none have gained approval. Herein, a series of 5-formyl-pyrrolo[3,2-b]pyridine-3-carboxamides and a series of 6-formylpyridyl ureas were characterized as selective reversible-covalent FGFR4 inhibitors. The representative 6-formylpyridyl urea 8z exhibited excellent potency against FGFR4WT, FGFR4V550L, and FGFR4V550M with IC50 values of 16.3, 12.6, and 57.3 nM, respectively. It also potently suppressed proliferation of Ba/F3 cells driven by FGFR4WT, FGFR4V550L, and FGFR4V550M, and FGFR4-dependent Hep3B and Huh7 HCC cells, with IC50 values of 1.2, 13.5, 64.5, 15.0, and 20.4 nM, respectively. Furthermore, 8z displayed desirable microsomal stability and significant in vivo efficacy in the Huh7 HCC cancer xenograft model in nude mice. The study provides a promising new lead for anticancer drug discovery directed toward overcoming FGFR4 gatekeeper mutation mediated resistance in HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Humans , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 4 , Urea/pharmacology , Urea/therapeutic use , Mice, Nude , Fibroblast Growth Factors/metabolism , Cell Line, Tumor
16.
Bioorg Med Chem ; 21(24): 7971-80, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24436995

ABSTRACT

We have synthesised a focused library of derivatives of natural products containing the pyranonaphthoquinone moiety including the first report of such a scaffold with an appended tetrazole functionality. Examples include kalafungin derivatives as well as analogues of nanaomycin and eleutherin. These compounds were assessed for cytotoxic activation by breast cancer cell lines engineered to express the prototypic human one- and two-electron quinone bioreductive enzymes, NADPH: cytochrome P450 oxidoreductase (POR) and NAD(P)H: quinoneoxidoreductase 1 (NQO1; DT-diaphorase), respectively. Several compounds were observed to be cytotoxic at sub-micromolar level and a pattern of increased aerobic potency was observed in cells over expressing POR. A subset of analogues was assessed under anoxic conditions, where cytotoxicity was reduced, implicating redox cycling as a major mechanism of toxicity. The substrate specificity for reductive enzymes is relevant to the future design of bioreductive prodrugs to treat cancer.


Subject(s)
Biological Products/chemical synthesis , Biological Products/toxicity , Naphthoquinones/chemistry , Naphthoquinones/toxicity , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Biological Products/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , NADPH-Ferrihemoprotein Reductase/metabolism , Naphthoquinones/chemical synthesis , Oxidation-Reduction/drug effects , Structure-Activity Relationship
17.
J Med Chem ; 66(17): 11656-11671, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37669428

ABSTRACT

Epidermal growth factor receptor (EGFR) targeted therapy is one of the most important and effective strategies to combat EGFR mutant nonsmall-cell lung cancer (NSCLC). However, a substantial number of patients bearing EGFR exon 20 insertion (Ex20ins) mutations respond poorly to common EGFR targeted therapies. This clinical need remained unmet until recently, when the EGFR Ex20ins mutation inhibitor mobocertinib was approved by the FDA. Despite this progress, the structural mechanisms of EGFR Ex20ins mutation resistance and characterization of inhibitor binding modes have not been systematically summarized. Herein, we analyze the structural mechanisms for ligand binding and resistance and summarize recent developments for the reported inhibitors of EGFR Ex20ins mutations. Furthermore, this Perspective aims to provide insights for the design of the next generation of EGFR Ex20ins inhibitors.


Subject(s)
ErbB Receptors , Lung Neoplasms , Humans , ErbB Receptors/genetics , Exons , Mutation
18.
ACS Med Chem Lett ; 14(11): 1517-1523, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37974941

ABSTRACT

Off-target aerobic activation of PR-104A by human aldo-keto reductase 1C3 (AKR1C3) has confounded the development of this dual hypoxia/gene therapy prodrug. Previous attempts to design prodrugs resistant to AKR1C3 activation have resulted in candidates that require further optimization. Herein we report the evaluation of a lipophilic series of PR-104A analogues in which a piperazine moiety has been introduced to improve drug-like properties. Octanol-water partition coefficients (LogD7.4) spanned >2 orders of magnitude. 2D antiproliferative and 3D multicellular clonogenic assays using isogenic HCT116 and H1299 cells confirmed that all examples were resistant to AKR1C3 metabolism while producing an E. coli NfsA nitroreductase-mediated bystander effect. Prodrugs 16, 17, and 20 demonstrated efficacy in H1299 xenografts where only a minority of tumor cells express NfsA. These prodrugs and their bromo/mesylate counterparts (25-27) were also evaluated for hypoxia-selective cell killing in vitro. These results in conjunction with stability assays recommended prodrug 26 (CP-506) for Phase I/II clinical trial.

19.
J Med Chem ; 66(11): 7405-7420, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37220641

ABSTRACT

Covalent kinase inhibitors (CKIs) hold great promise for drug development. However, examples of computationally guided design of CKIs are still scarce. Here, we present an integrated computational workflow (Kin-Cov) for rational design of CKIs. The design of the first covalent leucine-zipper and sterile-α motif kinase (ZAK) inhibitor was presented as an example to showcase the power of computational workflow for CKI design. The two representative compounds, 7 and 8, inhibited ZAK kinase with half-maximal inhibitory concentration (IC50) values of 9.1 and 11.5 nM, respectively. Compound 8 displayed an excellent ZAK target specificity in Kinome profiling against 378 wild-type kinases. Structural biology and cell-based Western blot washout assays validated the irreversible binding characteristics of the compounds. Our study presents a rational approach for the design of CKIs based on the reactivity and accessibility of nucleophilic amino acid residues in a kinase. The workflow is generalizable and can be applied to facilitate CKI-based drug design.


Subject(s)
Drug Design , Protein Kinases , Workflow , Protein Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry
20.
J Med Chem ; 65(1): 58-83, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34962782

ABSTRACT

Small molecule covalent kinase inhibitors (CKIs) have entered a new era in drug discovery, which have the advantage for sustained target inhibition and high selectivity. An increased understanding of binding kinetics of CKIs and discovery of additional irreversible and reversible-covalent cysteine-targeted warheads has inspired the development of this area. Herein, we summarize the major medicinal chemistry strategies employed in the discovery of these representative CKIs, which are categorized by the location of the target cysteine within seven main regions of the kinase: the front region, the glycine rich loop (P-loop), the hinge region, the DFG region, the activation loop (A-loop), the catalytic loop (C-loop), and the remote loop. The emphasis is placed on the design and optimization strategies of CKIs that are generated by addition of a warhead to a reversible lead/inhibitor scaffold. In addition, we address the challenges facing this area of drug discovery.


Subject(s)
Chemistry, Pharmaceutical , Cysteine/chemistry , Drug Design , Drug Discovery , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries/chemistry , Animals , High-Throughput Screening Assays , Humans , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL