Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
ACS Chem Biol ; 14(4): 751-757, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30840432

ABSTRACT

Colloidal drug aggregates have been a nuisance in drug screening, yet, because they inherently comprise drug-rich particles, they may be useful in vivo if issues of stability can be addressed. As the first step toward answering this question, we optimized colloidal drug aggregate formulations using a fluorescence-based assay to study fulvestrant colloidal formation and stability in high (90%) serum conditions in vitro. We show, for the first time, that the critical aggregation concentration of fulvestrant depends on media composition and increases with serum concentration. Excipients, such as polysorbate 80, stabilize fulvestrant colloids in 90% serum in vitro for over 48 h. Using fulvestrant and an investigational pro-drug, pentyloxycarbonyl-( p-aminobenzyl) doxazolidinylcarbamate (PPD), as proof-of-concept colloidal formulations, we demonstrate that the in vivo plasma half-life for stabilized colloids is greater than their respective monomeric forms. These studies demonstrate the potential of turning the nuisance of colloidal drug aggregation into an opportunity for drug-rich formulations.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Carbamates/chemistry , Carbamates/pharmacokinetics , Doxorubicin/analogs & derivatives , Oxazoles/chemistry , Oxazoles/pharmacokinetics , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Animals , Antineoplastic Agents/blood , Carbamates/blood , Colloids , Doxorubicin/blood , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Stability , Excipients , Female , Fulvestrant/chemistry , Humans , MCF-7 Cells , Mice , Neoplasm Transplantation , Oxazoles/blood , Polysorbates/chemistry , Proof of Concept Study , Serum
2.
J Med Chem ; 62(4): 1959-1970, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30703330

ABSTRACT

Natural lipid nanocarriers, exosomes, carry cell-signaling materials such as DNA and RNA for intercellular communications. Exosomes derived from cancer cells contribute to the progression and metastasis of cancer cells by transferring oncogenic signaling molecules to neighboring and remote premetastatic sites. Therefore, applying the unique properties of exosomes for cancer therapy has been expected in science, medicine, and drug discovery fields. Herein, we report that an exosome-targeting prodrug system, designated MARCKS-ED-photodoxaz, could spatiotemporally control the activation of an exquisitely cytotoxic agent, doxazolidine (doxaz), with UV light. The MARCKS-ED peptide enters a cell by forming a complex with the exosomes in situ at its plasma membrane and in the media. MARCKS-ED-photodoxaz releases doxaz under near-UV irradiation to inhibit cell growth with low nanomolar IC50 values. The MARCKS-ED-photodoxaz system targeting exosomes and utilizing photochemistry will potentially provide a new approach for the treatment of cancer, especially for highly progressive and invasive metastatic cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Doxorubicin/analogs & derivatives , Exosomes/drug effects , Nitrobenzenes/pharmacology , Oxazoles/pharmacology , Prodrugs/pharmacology , Amino Acid Sequence , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Cell Line, Tumor , Cell Membrane/metabolism , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/pharmacology , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/pharmacology , Cross-Linking Reagents/radiation effects , Doxorubicin/chemical synthesis , Doxorubicin/pharmacology , Doxorubicin/radiation effects , Humans , Nitrobenzenes/chemical synthesis , Nitrobenzenes/radiation effects , Oxazoles/chemical synthesis , Oxazoles/radiation effects , Photolysis , Prodrugs/chemical synthesis , Prodrugs/radiation effects , Ultraviolet Rays
3.
Mol Pharm ; 14(6): 1852-1860, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28502177

ABSTRACT

While limited drug loading continues to be problematic for chemotherapeutics formulated in nanoparticles, we found that we could take advantage of colloidal drug aggregation to achieve high loading when combined with polymeric excipients. We demonstrate this approach with two drugs, fulvestrant and pentyl-PABC doxazolidine (PPD; a prodrug of doxazolidine, which is a DNA cross-linking anthracycline), and two polymers, polysorbate 80 (UP80) and poly(d,l-lactide-co-2-methyl-2-carboxytrimethylene carbonate)-graft-poly(ethylene glycol) (PLAC-PEG; a custom-synthesized, self-assembling amphiphilic polymer). In both systems, drug-loaded nanoparticles had diameters < 200 nm and were stable for up to two days in buffered saline solution and for up to 24 h in serum-containing media at 37 °C. While colloidal drug aggregates alone are typically unstable in saline and serum-containing media, we attribute the colloid stability observed herein to the polymeric excipients and consequent decreased protein adsorption. We expect this strategy of polymer-stabilized colloidal drug aggregates to be broadly applicable in delivery formulations.


Subject(s)
Colloids/chemistry , Drug Delivery Systems/methods , Nanoparticles/chemistry , Doxorubicin/analogs & derivatives , Doxorubicin/chemistry , Drug Carriers/chemistry , Micelles , Oxazoles/chemistry , Polymers/chemistry
4.
J Med Chem ; 59(5): 2205-21, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26881291

ABSTRACT

Anthracyclines are a class of antitumor compounds that are successful and widely used but suffer from cardiotoxicity and acquired tumor resistance. Formaldehyde interacts with anthracyclines to enhance antitumor efficacy, bypass resistance mechanisms, improve the therapeutic profile, and change the mechanism of action from a topoisomerase II poison to a DNA cross-linker. Contrary to current dogma, we show that both efficient DNA cross-linking and potent synergy in combination with formaldehyde correlate with the anthracycline's ability to form cyclic formaldehyde conjugates as oxazolidine moieties and that the cyclic conjugates are better cross-linking agents and cytotoxins than acyclic conjugates. We also provide evidence that suggests that the oxazolidine forms in situ, since cotreatment with doxorubicin and formaldehyde is highly cytotoxic to dox-resistant tumor cell lines, and that this benefit is absent in combinations of formaldehyde and epirubicin, which cannot form stable oxazolidines. These results have potential clinical implications in the active field of anthracycline prodrug design and development.


Subject(s)
Antineoplastic Agents/pharmacology , Cross-Linking Reagents/pharmacology , DNA, Neoplasm/chemistry , Doxorubicin/pharmacology , Formaldehyde/pharmacology , Oxazoles/pharmacology , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/chemistry , DNA, Neoplasm/drug effects , Dose-Response Relationship, Drug , Doxorubicin/analogs & derivatives , Doxorubicin/biosynthesis , Doxorubicin/chemistry , Drug Screening Assays, Antitumor , Esterases/metabolism , Formaldehyde/chemistry , Humans , Liver/enzymology , Molecular Structure , Oxazoles/chemical synthesis , Oxazoles/chemistry , Prodrugs/chemical synthesis , Prodrugs/chemistry , Structure-Activity Relationship , Swine , Tumor Cells, Cultured
5.
Biochem Pharmacol ; 85(3): 439-47, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23228697

ABSTRACT

Orlistat has been the most used anti-obesity drug and the mechanism of its action is to reduce lipid absorption by inhibiting gastrointestinal lipases. These enzymes, like carboxylesterases (CESs), structurally belong to the α/ß hydrolase fold superfamily. Lipases and CESs are functionally related as well. Some CESs (e.g., human CES1) have been shown to hydrolyze lipids. This study was designed to test the hypothesis that orlistat inhibits CESs with higher potency toward CES1 than CES2, a carboxylesterase with little lipase activity. Liver microsomes and recombinant CESs were tested for the inhibition of the hydrolysis of standard substrates and the anticancer prodrugs pentyl carbamate of p-aminobenzyl carbamate of doxazolidine (PPD) and irinotecan. Contrary to the hypothesis, orlistat at 1 nM inhibited CES2 activity by 75% but no inhibition on CES1, placing CES2 one of the most sensitive targets of orlistat. The inhibition varied among some CES2 polymorphic variants. Pretreatment with orlistat reduced the cell killing activity of PPD. Certain mouse but not rat CESs were also highly sensitive. CES2 is responsible for the hydrolysis of many common drugs and abundantly expressed in the gastrointestinal track and liver. Inhibition of this carboxylesterase probably presents a major source for altered therapeutic activity of these medicines if co-administered with orlistat. In addition, orlistat has been linked to various types of organ toxicities, and this study provides an alternative target potentially involved in these toxicological responses.


Subject(s)
Anti-Obesity Agents/pharmacology , Antineoplastic Agents/pharmacology , Carboxylic Ester Hydrolases/antagonists & inhibitors , Lactones/pharmacology , Prodrugs/pharmacology , Animals , Anti-Obesity Agents/metabolism , Antineoplastic Agents/metabolism , Camptothecin/analogs & derivatives , Camptothecin/metabolism , Camptothecin/pharmacology , Carboxylic Ester Hydrolases/classification , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Cell Line , Cell Survival/drug effects , Doxorubicin/analogs & derivatives , Doxorubicin/chemistry , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Interactions , Female , Humans , Irinotecan , Liver/drug effects , Liver/enzymology , Male , Mice , Orlistat , Oxazoles/chemistry , Oxazoles/metabolism , Oxazoles/pharmacology , Prodrugs/metabolism , Rats , Species Specificity
6.
J Med Chem ; 55(14): 6595-607, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22742660

ABSTRACT

Doxazolidine (doxaz) is a new anthracycline anticancer agent. While structurally similar to doxorubicin (dox), doxaz acts via a distinct mechanism to selectively enhance anticancer activity over cardiotoxicity, the most significant clinical impediment to successful anthracycline treatment. Here, we describe the synthesis and characterization of a prodrug platform designed for doxaz release mediated by secreted proteolytic activity, a common association with invasiveness and poor prognosis in cancer patients. GaFK-Doxaz is hydrolyzable by the proteases plasmin and cathepsin B, both strongly linked with cancer progression, as well as trypsin. We demonstrate that activation of GaFK-Doxaz releases highly potent doxaz that powerfully inhibits the growth of a wide variety of cancer cells (average IC(50) of 8 nM). GaFK-Doxaz is stable in human plasma and is poorly membrane permeable, thereby limiting activation to locally secreted proteolytic activity and reducing the likelihood of severe side effects.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Doxorubicin/analogs & derivatives , Oxazoles/metabolism , Oxazoles/pharmacology , Peptide Hydrolases/metabolism , Antineoplastic Agents/blood , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Doxorubicin/blood , Doxorubicin/chemical synthesis , Doxorubicin/chemistry , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Stability , Humans , Kinetics , Oxazoles/blood , Oxazoles/chemical synthesis , Oxazoles/chemistry , Phosphates/chemistry , Proteolysis
7.
PLoS One ; 5(12): e15004, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21165148

ABSTRACT

BACKGROUND: The interrogation of proteomes ("proteomics") in a highly multiplexed and efficient manner remains a coveted and challenging goal in biology and medicine. METHODOLOGY/PRINCIPAL FINDINGS: We present a new aptamer-based proteomic technology for biomarker discovery capable of simultaneously measuring thousands of proteins from small sample volumes (15 µL of serum or plasma). Our current assay measures 813 proteins with low limits of detection (1 pM median), 7 logs of overall dynamic range (~100 fM-1 µM), and 5% median coefficient of variation. This technology is enabled by a new generation of aptamers that contain chemically modified nucleotides, which greatly expand the physicochemical diversity of the large randomized nucleic acid libraries from which the aptamers are selected. Proteins in complex matrices such as plasma are measured with a process that transforms a signature of protein concentrations into a corresponding signature of DNA aptamer concentrations, which is quantified on a DNA microarray. Our assay takes advantage of the dual nature of aptamers as both folded protein-binding entities with defined shapes and unique nucleotide sequences recognizable by specific hybridization probes. To demonstrate the utility of our proteomics biomarker discovery technology, we applied it to a clinical study of chronic kidney disease (CKD). We identified two well known CKD biomarkers as well as an additional 58 potential CKD biomarkers. These results demonstrate the potential utility of our technology to rapidly discover unique protein signatures characteristic of various disease states. CONCLUSIONS/SIGNIFICANCE: We describe a versatile and powerful tool that allows large-scale comparison of proteome profiles among discrete populations. This unbiased and highly multiplexed search engine will enable the discovery of novel biomarkers in a manner that is unencumbered by our incomplete knowledge of biology, thereby helping to advance the next generation of evidence-based medicine.


Subject(s)
Aptamers, Nucleotide , Biomarkers/metabolism , Proteomics/methods , Aged , Evidence-Based Medicine , Female , Gene Library , Genetic Techniques , Glomerular Filtration Rate , Humans , Kidney Failure, Chronic/metabolism , Kinetics , Male , Mass Spectrometry/methods , Middle Aged , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Proteome , Reproducibility of Results
8.
J Med Chem ; 52(23): 7678-88, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19634903

ABSTRACT

Doxazolidine (Doxaz) is a functionally distinct formaldehyde conjugate of doxorubicin (Dox) that induces cancer cell death in Dox-sensitive and resistant cells. Pentyl PABC-Doxaz (PPD) is a prodrug of Doxaz that is activated by carboxylesterase 2 (CES2), which is expressed by liver, non-small-cell lung, colon, pancreatic, renal, and thyroid cancer cells. Here, we demonstrate that in two murine models, PPD was effective at slowing tumor growth and demonstrated markedly reduced cardiotoxic and nephrotoxic effects, as well as better tolerance, relative to Dox. Hepatotoxicity, consistent with liver expression of the murine CES2 homologue, was induced by PPD. Unlike irinotecan, a clinical CES2-activated prodrug, PPD produced no visible gastrointestinal damage. Finally, we demonstrate that cellular response to PPD may be predicted with good accuracy using CES2 expression and Doxaz sensitivity, suggesting that these metrics may be useful as clinical biomarkers for sensitivity of a specific tumor to PPD treatment.


Subject(s)
Carbamates/metabolism , Carbamates/pharmacology , Carboxylesterase/metabolism , Doxorubicin/analogs & derivatives , Oxazoles/metabolism , Prodrugs/metabolism , Prodrugs/pharmacology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Carbamates/chemistry , Carbamates/toxicity , Carboxylesterase/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/chemistry , Doxorubicin/metabolism , Doxorubicin/pharmacology , Doxorubicin/toxicity , Drug Evaluation, Preclinical , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mice , Myocytes, Cardiac/drug effects , Neoplasms/enzymology , Neoplasms/pathology , Oxazoles/pharmacology , Prodrugs/chemistry , Prodrugs/toxicity , Rats , Regression Analysis
9.
J Med Chem ; 51(2): 298-304, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18173233

ABSTRACT

Doxazolidine (Doxaz), a formaldehyde-doxorubicin (Dox) conjugate, exhibits markedly increased tumor toxicity with respect to Dox without a concurrent increase in toxicity to cardiomyocytes. Pentyl PABC-Doxaz (PPD) is a Doxaz carbamate prodrug that is hydrolyzed by carboxylesterases. Here, we identify human intestinal carboxylesterase (hiCE) as the agent of activation for PPD. Upon prodrug treatment, cells that express higher levels of hiCE responded with lower IC50 values for growth inhibition. Exposing MCF-7 human breast cancer cells, which respond poorly and express little hiCE, to PPD together with hiCE resulted in a dramatic decrease in the IC50, a decrease that was absent when human carboxylesterase 1 was added to prodrug treatment. Finally, U373MG glioblastoma cells overexpressing hiCE displayed approximately 100-fold reduction in the IC50 for PPD compared to cells lacking the carboxylesterase. Overall, our studies indicate that PPD is selectively hydrolyzed to the active metabolite by hiCE.


Subject(s)
Antineoplastic Agents/metabolism , Carbamates/metabolism , Carboxylic Ester Hydrolases/metabolism , Doxorubicin/analogs & derivatives , Intestines/enzymology , Prodrugs/metabolism , Antineoplastic Agents/pharmacology , Carbamates/pharmacology , Cell Line, Tumor , Doxorubicin/metabolism , Doxorubicin/pharmacology , Humans , Prodrugs/pharmacology , Recombinant Proteins/metabolism
10.
Top Curr Chem ; 283: 141-70, 2008.
Article in English | MEDLINE | ID: mdl-23605631

ABSTRACT

The sequence of research leading to a proposal for anthracycline cross-linking of DNA is presented.The clinical anthracycline antitumor drugs are anthraquinones, and as such are redox active. Their redoxchemistry leads to induction of oxidative stress and drug metabolites. An intermediate in reductive glycosidiccleavage is a quinone methide, once proposed as an alkylating agent of DNA. Subsequent research nowimplicates formaldehyde as a mediator of anthracycline-DNA cross-linking. The cross-link at 5'-GC-3'sites consists of a covalent linkage from the amino group of the anthracycline to the 2-amino groupof the G-base through a methylene from formaldehyde, hydrogen bonding from the 9-OH to the G-base onthe opposing strand, and hydrophobic interactions through intercalation of the anthraquinone. The combinationof these interactions has been described as a virtual cross-linkof DNA. The origin of the formaldehyde in vivo remains a mystery. In vitro, doxorubicin reacts withformaldehyde to give firstly a monomeric oxazolidine, doxazolidine, and secondly a dimeric oxazolidine,doxoform. Doxorubicin reacts with formaldehyde in the presence of salicylamide to give the N-Mannich baseconjugate, doxsaliform. Doxsaliform is several fold more active in tumor cell growth inhibition than doxorubicin,but doxazolidine and doxoform are orders of magnitude more active than doxorubicin. Exploratory researchon the potential for doxsaliform and doxazolidine as targeted cytotoxins is presented. A promisinglead design is pentyl PABC-Doxaz, targeted to a carboxylesterase enzyme overexpressed in liver cancercells and/or colon cancer cells.

11.
Mutat Res ; 638(1-2): 110-21, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17961607

ABSTRACT

Doxorubicin, a widely used anthracycline anticancer agent, acts as a topoisomerase II poison but can also form formaldehyde-mediated DNA adducts. This has led to the development of doxorubicin derivatives such as doxoform, which can readily form adducts with DNA. This work aimed to determine which DNA repair pathways are involved in the recognition and possible repair of anthracycline-DNA adducts. Cell lines lacking functional proteins involved in each of the five main repair pathways, mismatch repair (MMR), base excision repair (BER), nucleotide excision repair (NER), homologous recombination (HR) and non-homologous end-joining (NHEJ) were examined for sensitivity to various anthracycline adduct-forming treatments. The treatments used were doxorubicin, barminomycin (a model adduct-forming anthracycline) and doxoform (a doxorubicin-formaldehyde conjugate). Cells with deficiencies in MMR, BER and NHEJ were equally sensitive to adduct-forming treatments compared to wild type cells and therefore these pathways are unlikely to play a role in the repair of these adducts. Some cells with deficiencies in the NER pathway (specifically, those lacking functional XPB, XPD and XPG), displayed tolerance to adducts induced by both barminomycin and doxoform and also exhibited a decreased level of apoptosis in response to adduct-forming treatments. Conversely, two HR deficient cell lines were shown to be more sensitive to barminomycin and doxoform than HR proficient cells, indicating that this pathway is also involved in the repair response to anthracycline-DNA adducts. These results suggest an unusual damage response pathway to anthracycline adducts involving both NER and HR that could be used to optimise cancer therapy for tumours with either high levels of NER or defective HR. Tumours with either of these characteristics would be predicted to respond particularly well to anthracycline-DNA adduct-forming treatments.


Subject(s)
Anthracyclines/metabolism , Colonic Neoplasms/genetics , DNA Adducts/metabolism , DNA Repair , Recombination, Genetic , Cell Line, Tumor , Cell Proliferation , DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA-Binding Proteins , Endonucleases , Humans , Nuclear Proteins , Transcription Factors
12.
J Med Chem ; 50(18): 4493-500, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17696516

ABSTRACT

The mechanism of doxorubicin is compared with that of doxazolidine, a doxorubicin-formaldehyde conjugate. The IC(50) for growth inhibition of 67 human cancer cell lines, but not cardiomyocytes, is 32-fold lower with doxazolidine than with doxorubicin. Growth inhibition by doxazolidine correlates better with growth inhibition by DNA cross-linking agents than with growth inhibition by doxorubicin. Doxorubicin induces G2/M arrest in HCT-116 colon cancer cells and HL-60 leukemia cells through a well-documented topoisomerase II dependent mechanism. Doxazolidine fails to induce a G2/M arrest in HCT-116 cells but induces apoptosis 4-fold better than doxorubicin. The IC(50) for doxazolidine growth inhibition of HL-60/MX2 cells, a topoisomerase II deficient derivative of HL-60 cells, is 1420-fold lower than the IC(50) for doxorubicin, and doxazolidine induces apoptosis 15-fold better. Further, doxazolidine has little effect in a topoisomerase II activity assay. These data indicate that doxorubicin and doxazolidine induce apoptosis via different mechanisms and doxazolidine cytotoxicity is topoisomerase II independent.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , DNA Topoisomerases, Type II/physiology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Formaldehyde/analogs & derivatives , Formaldehyde/pharmacology , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type II/genetics , Humans , Models, Molecular , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Topoisomerase II Inhibitors
13.
J Med Chem ; 49(24): 7002-12, 2006 Nov 30.
Article in English | MEDLINE | ID: mdl-17125253

ABSTRACT

The synthesis and tumor cell growth inhibition by doxazolidine carbamate prodrugs are reported. The carbamates were designed for selective hydrolysis by one or more human carboxylesterases to release doxazolidine (Doxaz), the formaldehyde-oxazolidine of doxorubicin that cross-links DNA to trigger cell death. Simple butyl and pentyl, but not ethyl, carbamate prodrugs inhibited the growth of cancer cells that overexpress carboxylesterase CES1 (hCE1) and CES2 (hiCE). Relative CES1 and CES2 expression levels were determined by reverse transcription of the respective mRNAs, followed by polymerase chain reaction amplification. More complex structures with a p-aminobenzyl alcohol (PABA) self-eliminating spacer showed better growth inhibition (IC50=50 nM for Hep G2 liver cancer cells) while exhibiting reduced toxicity toward rat cardiomyocytes, relative to the parent drug doxorubicin. Pentyl 4-(N-doxazolidinylcarbonyloxymethyl)phenylcarbamate, the lead compound for further investigation, appears to be activated in Hep G2 cells that express both CES1 and CES2.


Subject(s)
Antineoplastic Agents/chemical synthesis , Carbamates/chemical synthesis , Carboxylic Ester Hydrolases/metabolism , Doxorubicin/analogs & derivatives , Oxazoles/chemical synthesis , Prodrugs/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbamates/chemistry , Carbamates/pharmacology , Carboxylic Ester Hydrolases/biosynthesis , Cell Line , Cell Line, Tumor , Doxorubicin/chemical synthesis , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Design , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Conformation , Myocytes, Cardiac/drug effects , Oxazoles/chemistry , Oxazoles/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , RNA, Messenger/biosynthesis , Rats , Structure-Activity Relationship
14.
J Med Chem ; 48(24): 7648-57, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16302805

ABSTRACT

A crystal structure establishes doxoform as a dimeric formaldehyde conjugate of the oxazolidine of doxorubicin. Doxoform is a prodrug of doxazolidine, a monomeric doxorubicin formaldehyde-oxazolidine. Both doxoform and doxazolidine inhibit the growth of cancer cells at 1-4 orders of magnitude lower concentration than doxorubicin. They also inhibit the growth of cancer cells better than doxsaliform, a prodrug for an acyclic doxorubicin-formaldehyde conjugate. Doxoform rapidly hydrolyzes to doxazolidine, which then hydrolyzes to doxorubicin with a half-life of 3 min in human serum at 37 degrees C. Both doxoform and doxazolidine are taken up by multidrug-resistant MCF-7/Adr cells 3- to 4-fold better than doxorubicin. A molecular model suggests that doxazolidine can cross-link DNA by direct reaction with a G-base in a tautomeric form with synchronous ring opening of the oxazolidine. These results point to doxoform being a prodrug for doxazolidine that is the reactive species that directly cross-links DNA.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Cross-Linking Reagents/chemistry , DNA/chemistry , Doxorubicin/analogs & derivatives , Doxorubicin/chemistry , Oxazoles/chemistry , Prodrugs/chemistry , Antibiotics, Antineoplastic/chemical synthesis , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/pharmacology , Crystallography, X-Ray , Doxorubicin/chemical synthesis , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Drug Stability , Humans , Hydrolysis , Models, Molecular , Oxazoles/chemical synthesis , Oxazoles/metabolism , Oxazoles/pharmacology , Prodrugs/chemical synthesis , Prodrugs/pharmacology
15.
J Med Chem ; 47(26): 6509-18, 2004 Dec 16.
Article in English | MEDLINE | ID: mdl-15588086

ABSTRACT

The anthracycline antitumor drug, doxorubicin (DOX), has long been used as a broad spectrum chemotherapeutic. The literature now documents the role of formaldehyde in the cytotoxic mechanism, and anthracycline-formaldehyde conjugates possess substantially enhanced activity in vitro and in vivo. We have recently reported the design, synthesis, and preliminary evaluation of a doxorubicin-formaldehyde conjugate targeted, via 4-hydroxytamoxifen, to the estrogen receptor (ER) and antiestrogen binding site (AEBS), which are commonly present in breast cancer cells. The lead targeted doxorubicin-formaldehyde conjugate, called DOX-TEG-TAM, was found to possess superior cell growth inhibition characteristics relative to clinical doxorubicin and an untargeted control conjugate, especially in ER-negative, multidrug resistant MCF-7/Adr cells. The enhanced activity in the absence of estrogen receptor raised the possibility that targeting was also mediated via AEBS. Fluorescence microscopy of an ER-negative, AEBS-positive cell line as a function of time showed initial DOX-TEG-TAM localization in cytosol, in contrast to initial DOX and untargeted doxorubicin-formaldehyde conjugate localization in the nucleus. DOX-TEG-TAM was taken up by four AEBS-positive cell lines to a greater extent than doxorubicin and an untargeted doxorubicin-formaldehyde conjugate. Of the four cell lines, three were ER negative. DOX-TEG-TAM uptake was inhibited in a dose-dependent manner by the presence of a competing AEBS ligand. DOX-TEG-TAM retains 60% of the affinity of 4-hydroxytamoxifen for AEBS. DOX-TEG-TAM was also taken up by the AEBS-negative, ER-positive cancer cell line Rtx-6; with these cells uptake was inhibited in a dose-dependent manner by the ER ligand, estradiol. The data support the hypothesis that uptake of 4-hydroxytamoxifen targeted doxorubicin-formaldehyde conjugate is mediated by both the antiestrogen binding site and estrogen receptor.


Subject(s)
Antineoplastic Agents/chemical synthesis , Doxorubicin/analogs & derivatives , Doxorubicin/chemical synthesis , Formaldehyde/analogs & derivatives , Formaldehyde/chemical synthesis , Receptors, Drug/metabolism , Receptors, Estrogen/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Binding Sites , Breast Neoplasms , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/metabolism , Doxorubicin/pharmacokinetics , Female , Formaldehyde/metabolism , Formaldehyde/pharmacokinetics , Humans , Microscopy, Fluorescence , Structure-Activity Relationship , Tamoxifen/metabolism , Tamoxifen/pharmacokinetics
16.
J Med Chem ; 47(23): 5690-9, 2004 Nov 04.
Article in English | MEDLINE | ID: mdl-15509168

ABSTRACT

The synthesis of a doxorubicin-formaldehyde conjugate bound to the nonsteroidal anti-androgen cyanonilutamide, via a cleavable tether, and binding of the construct to cell free androgen receptor (AR) as a function of tether design were previously reported. Cyanonilutamide bearing a linear alkyne tether bound to the AR better than other designs. Fluorescence microscopy studies of binding of the lead targeted drug, as well as various tethered cyanonilutamides, to the AR and subsequent trafficking of the resulting AR complex in live PC3 prostate cancer cells transfected with AR-green fluorescent protein (GFP) chimera are now described. Cyanonilutamide and cyanonilutamide bonded to a linear alkyne tether caused translocation of AR-GFP to the nucleus. In general, the ability of tethered cyanonilutamides to cause translocation paralleled their binding affinity for the AR. However, a noncleavable form of the lead cyanonilutamide-doxorubicin-formaldehyde conjugate bound to AR-GFP but the resulting complex did not translocate to the nucleus. Binding was apparent from the drugs inhibition of Mibolerone-induced translocation. Direct observation of anthraquinone fluorescence of targeted drug in PC3 cells showed initial cytosolic localization, independent of AR expression, with predominant nuclear localization after sufficient time for release of drug from the targeting moiety. The results indicate that doxorubicin-formaldehyde conjugate bonded to cyanonilutamide via a cleavable linear tether enters PC3 cells, resides in cytosol, binds to the AR if present, and ultimately releases doxorubicin or a doxorubicin derivative to the nucleus.


Subject(s)
Androgen Antagonists/pharmacokinetics , Doxorubicin/analogs & derivatives , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Formaldehyde/chemistry , Green Fluorescent Proteins/genetics , Imidazoles/pharmacokinetics , Mannich Bases/pharmacokinetics , Receptors, Androgen/metabolism , Recombinant Fusion Proteins/metabolism , Androgen Antagonists/chemistry , Humans , Imidazoles/chemistry , Ligands , Male , Mannich Bases/chemistry , Microscopy, Fluorescence , Prostatic Neoplasms , Receptors, Androgen/genetics , Recombinant Fusion Proteins/genetics , Structure-Activity Relationship , Tumor Cells, Cultured
17.
J Mol Biol ; 336(5): 1159-73, 2004 Mar 05.
Article in English | MEDLINE | ID: mdl-15037076

ABSTRACT

ssDNA oligonucleotides containing bromodeoxyuridine, BrdU-photoaptamers, are rapidly emerging as specific protein capture reagents in protein microarray technologies. A mathematical model for the kinetic analysis of photoaptamer-protein photocross-linking reactions is presented. The model is based on specific aptamer/protein binding followed by laser excitation that can lead to either covalent cross-linking of the photoaptamer and protein in the complex or irreversible photodamage to the aptamer. Two distinct kinetic regimes, (1) frozen and (2) rapid equilibrium, are developed analytically to model binding kinetics between laser pulses. The models are used to characterize the photocross-linking between three photoaptamers and their cognate protein targets; photoaptamers 0650 and 0615 cross-link human basic fibroblast growth factor and 0518 cross-links HIV MN envelope glycoprotein. Data for cross-linking reaction yields as a function of both laser energy dose and target protein concentration are analyzed for affinity constants and cross-link reaction rates. The binding dissociation constants derived from the cross-linking data are in good accord with independent measurements; the rapid equilibrium model appears to produce results more consistent with the experimental observations, although there is significant overlap between the two models for most conditions explored here. The rate of photodamage for 0615 and 0518 is 3.5 and 2.5 times that of the specific cross-link, giving low maximum reaction yields of approximately 20% and approximately 30%, whereas 0650 cross-links with a rate over five times higher than its photodamage rate and has a maximum reaction yield exceeding 80%. Quantum yields for the three systems are estimated from the data; photoaptamer 0650 has a reasonably high quantum yield of approximately 0.2 for protein cross-linking, while 0518 and 0615 have quantum yields of 0.07 and 0.02. The work presented here provides a useful set of metrics that allow for refinement of photoaptamer properties.


Subject(s)
Cross-Linking Reagents/chemistry , DNA, Single-Stranded/chemistry , Oligodeoxyribonucleotides/chemistry , Photochemistry/methods , Proteins/chemistry , Binding Sites , Bromodeoxyuridine/chemistry , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/metabolism , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/metabolism , Humans , Kinetics , Models, Theoretical , Protein Array Analysis/methods , Protein Binding , Proteins/metabolism
18.
J Med Chem ; 47(5): 1193-206, 2004 Feb 26.
Article in English | MEDLINE | ID: mdl-14971899

ABSTRACT

The anthracycline antitumor drug doxorubicin (DOX) has been utilized for decades as a broad-spectrum chemotherapeutic. Recent literature evidence documents the role of formaldehyde in the cytotoxic mechanism, and anthracycline-formaldehyde conjugates possess substantially enhanced activity in vitro and in vivo. Targeting a doxorubicin-formaldehyde conjugate specifically to cancer cells may provide a more efficacious chemotherapeutic. The design and 11-step synthesis of doxorubicin-formaldehyde conjugates targeted to the estrogen receptor, which is commonly overexpressed in breast cancer cells, are reported. The formaldehyde is incorporated in a masked form as an N-Mannich linkage between doxorubicin and salicylamide. The salicylamide triggering molecule, previously developed to release the doxorubicin-formaldehyde active metabolite, is tethered via derivatized ethylene glycols to an E and Z mixture of 4-hydroxytamoxifen. The targeting group, E/Z-4-hydroxytamoxifen, was selected for its ability to tightly bind the estrogen receptor and antiestrogen binding sites. The targeted doxorubicin-formaldehyde conjugates' estrogen receptor binding and in vitro growth inhibition were evaluated as a function of tether length. The lead compound, DOX-TEG-TAM, bearing a triethylene glycol tether, binds the estrogen receptor with a binding affinity of 2.5% relative to E/Z-4-hydroxytamoxifen and inhibits the growth of four breast cancer cell lines with 4-fold up to 140-fold enhanced activity relative to doxorubicin.


Subject(s)
Antineoplastic Agents/chemical synthesis , Doxorubicin/chemistry , Estrogen Receptor Modulators/chemical synthesis , Formaldehyde/chemistry , Tamoxifen/analogs & derivatives , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding, Competitive , Breast Neoplasms , Cell Division/drug effects , Cell Line , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Drug Stability , Estrogen Receptor Modulators/chemistry , Estrogen Receptor Modulators/pharmacology , Female , Humans , Hydrolysis , Models, Molecular , Polyethylene Glycols/chemistry , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Stereoisomerism , Structure-Activity Relationship , Tamoxifen/chemistry
19.
Mol Cancer Ther ; 3(12): 1593-604, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15634653

ABSTRACT

We have reported the synthesis and biological evaluation of a prodrug to a doxorubicin active metabolite. Under physiologic conditions, release of the active metabolite, a conjugate of doxorubicin with formaldehyde, occurs with a half-life of 1 hour. To direct this prodrug to tumor, we designed two conjugates of the prodrug, doxsaliform, with the alphavbeta3-targeting peptides, CDCRGDCFC (RGD-4C) and cyclic-(N-Me-VRGDf) (Cilengitide). We now report the synthesis of these doxsaliform-peptide conjugates and their evaluation using MDA-MB-435 cancer cells. A hydroxylamine ether tether was used to attach 5''-formyldoxsaliform to RGD-4C in its acyclic form via an oxime functional group. The construct acyclic-RGD-4C-doxsaliform showed good binding affinity for alphavbeta3 in the vitronection cell adhesion assay (IC50 = 10 nmol/L) and good growth inhibition of MDA-MB-435 breast cancer cells (IC50 = 50 nmol/L). In its bicyclic forms, RGD-4C showed less affinity for alphavbeta3 and significantly less water solubility. Cyclic-(N-Me-VRGDf) was modified by substitution of D-4-aminophenylalanine for D-phenylalanine to provide a novel attachment point for doxsaliform. The conjugate, cyclic-(N-Me-VRGDf-NH)-doxsaliform, maintained a high affinity for alphavbeta3 (IC50 = 5 nmol/L) in the vitronectin cell adhesion assay relative to the peptide bearing only the tether (0.5 nmol/L). The IC50 for growth inhibition of MDA-MB-435 cells was 90 nmol/L. Flow cytometry and growth inhibition experiments suggest that the complete drug construct does not penetrate through the plasma membrane, but the active metabolite does on release from the targeting group. These drug conjugates could have significantly reduced side effects and are promising candidates for in vivo evaluation in tumor-bearing mice.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Integrin alphaVbeta3/metabolism , Prodrugs/therapeutic use , Breast Neoplasms/metabolism , Cell Adhesion , Cell Membrane/metabolism , Cell Proliferation/drug effects , Doxorubicin/administration & dosage , Drug Design , Female , Formaldehyde/administration & dosage , Humans , Inhibitory Concentration 50 , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Receptors, Vitronectin/metabolism , Tumor Cells, Cultured , Vitronectin/metabolism
20.
J Med Chem ; 46(24): 5258-70, 2003 Nov 20.
Article in English | MEDLINE | ID: mdl-14613328

ABSTRACT

The synthesis and preliminary evaluation of a doxorubicin-formaldehyde conjugate tethered to the nonsteroidal antiandrogen, cyanonilutamide (RU 56279), for the treatment of prostate cancer are reported. The relative ability of the targeting group to bind to the human androgen receptor was studied as a function of tether. The tether served to attach the antiandrogen to the doxorubicin-formaldehyde conjugate via an N-Mannich base of a salicylamide derivative. The salicylamide was selected to serve as a trigger release mechanism to separate the doxorubicin-formaldehyde conjugate from the targeting group after it has bound to the androgen receptor. The remaining part of the tether consisted of a linear group that spanned from the 5-position of the salicylamide to the 3'-position of cyanonilutamide. The structures explored for the linear region of the tether were derivatives of di(ethylene glycol), tri(ethylene glycol), N,N'-disubstituted-piperazine, and 2-butyne-1,4-diol. Relative binding affinity of the tethers bound to the targeting group for human androgen receptor were measured using a (3)H-Mibolerone competition assay and varied from 18% of nilutamide binding for the butynediol-based linear region to less than 1% for one of the piperazine derivatives. The complete targeted drug with the butynediol-based linear region has a relative binding affinity of 10%. This relative binding affinity is encouraging in light of the cocrystal structure of human androgen receptor ligand binding domain bound to the steroid Metribolone which predicts very limited space for a tether connecting the antiandrogen on the inside to the cytotoxin on the outside.


Subject(s)
Androgen Antagonists/chemical synthesis , Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Formaldehyde/chemistry , Prodrugs/chemical synthesis , Receptors, Androgen/drug effects , Androgen Antagonists/chemistry , Antineoplastic Agents/chemistry , Binding, Competitive , Cell Line , Doxorubicin/chemistry , Drug Delivery Systems , Drug Design , Humans , Ligands , Microscopy, Fluorescence , Models, Molecular , Prodrugs/chemistry , Prodrugs/pharmacology , Receptors, Androgen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...