Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Oncotarget ; 14: 1-13, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36634212

ABSTRACT

Overexpression of CD74, a type II transmembrane glycoprotein involved in MHC class II antigen presentation, has been reported in many B-cell non-Hodgkin lymphomas (NHLs) and in multiple myeloma (MM). STRO-001 is a site-specific, predominantly single-species antibody-drug conjugate (ADC) that targets CD74 and has demonstrated efficacy in xenograft models of MM and tolerability in non-human primates. Here we report results of preclinical studies designed to elucidate the potential role of STRO-001 in B-cell NHL. STRO-001 displayed nanomolar and sub-nanomolar cytotoxicity in 88% (15/17) of cancer cell lines tested. STRO-001 showed potent cytotoxicity on proliferating B cells while limited cytotoxicity was observed on naïve human B cells. A linear dose-response relationship was demonstrated in vivo for DLBCL models SU-DHL-6 and U2932. Tumor regression was induced at doses less than 5 mg/kg, while maximal activity with complete cures were observed starting at 10 mg/kg. In MCL Mino and Jeko-1 xenografts, STRO-001 starting at 3 mg/kg significantly prolonged survival or induced tumor regression, respectively, leading to tumor eradication in both models. In summary, high CD74 expression levels in tumors, nanomolar cellular potency, and significant anti-tumor in DLBCL and MCL xenograft models support the ongoing clinical study of STRO-001 in patients with B-cell NHL.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Lymphoma, Non-Hodgkin , Multiple Myeloma , Animals , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Multiple Myeloma/pathology , Lymphoma, Non-Hodgkin/drug therapy , Cell Line, Tumor
2.
Eur J Med Chem ; 249: 115123, 2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36708677

ABSTRACT

The alkylindole (AI), WIN55212-2, modulates the activity of several proteins, including cannabinoid receptors 1 and 2 (CB1R, CB2R), and at least additional G protein-coupled receptor (GPCR) that remains uncharacterized with respect to its molecular identity and pharmacological profile. Evidence suggests that such AI-sensitive GPCRs are expressed by the human kidney cell line HEK293. We synthesized fourteen novel AI analogues and evaluated their activities at AI-sensitive GPCRs using [35S]GTPγS and [3H]WIN55212-2 binding in HEK293 cell membranes, and performed in silico pharmacophore modeling to identify characteristics that favor binding to AI-sensitive GPCRs versus CB1R/CB2R. Compounds 10 and 12 stimulated [35S]GTPγS binding (EC50s = 3.5 and 1.1 nM, respectively), and this response was pertussis toxin-sensitive, indicating that AI-sensitive GPCRs couple to Gi/o proteins. Five AI analogues reliably distinguished two binding sites that correspond to the high and low affinity state of AI-sensitive GPCRs coupled or not to G proteins. In silico pharmacophore modeling suggest 3 characteristics that favor binding to AI-sensitive GPCRs versus CB1R/CB2R: 1) an s-cis orientation of the two aromatic rings in AI analogues, 2) a narrow dihedral angle between the carbonyl group and the indole ring plane [i.e., O-C(carbonyl)-C3-C2] and 3) the presence of a carbonyl oxygen. The substituted alkylindoles reported here represent novel chemical tools to study AI-sensitive GPCRs.


Subject(s)
Cannabinoids , Humans , Cannabinoids/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate) , HEK293 Cells , Receptors, G-Protein-Coupled/metabolism , Receptor, Cannabinoid, CB2 , Receptor, Cannabinoid, CB1 , Receptors, Cannabinoid/metabolism
3.
Mol Cancer Ther ; 22(2): 155-167, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36459691

ABSTRACT

STRO-002 is a novel homogeneous folate receptor alpha (FolRα) targeting antibody-drug conjugate (ADC) currently being investigated in the clinic as a treatment for ovarian and endometrial cancers. Here, we describe the discovery, optimization, and antitumor properties of STRO-002. STRO-002 was generated by conjugation of a novel cleavable 3-aminophenyl hemiasterlin linker-warhead (SC239) to the nonnatural amino acid para-azidomethyl-L-phenylalanine incorporated at specific positions within a high affinity anti-FolRα antibody using Sutro's XpressCF+, which resulted in a homogeneous ADC with a drug-antibody ratio (DAR) of 4. STRO-002 binds to FolRα with high affinity, internalizes rapidly into target positive cells, and releases the tubulin-targeting cytotoxin 3-aminophenyl hemiasterlin (SC209). SC209 has reduced potential for drug efflux via P-glycoprotein 1 drug pump compared with other tubulin-targeting payloads. While STRO-002 lacks nonspecific cytotoxicity toward FolRα-negative cell lines, bystander killing of target negative cells was observed when cocultured with target positive cells. STRO-002 is stable in circulation with no change in DAR for up to 21 days and has a half-life of 6.4 days in mice. A single dose of STRO-002 induced significant tumor growth inhibition in FolRα-expressing xenograft models and patient-derived xenograft models. In addition, combination treatment with carboplatin or Avastin further increased STRO-002 efficacy in xenograft models. The potent and specific preclinical efficacy of STRO-002 supports clinical development of STRO-002 for treating patients with FolRα-expressing cancers, including ovarian, endometrial, and non-small cell lung cancer. Phase I dose escalation for STRO-002 is in progress in ovarian cancer and endometrial cancer patients (NCT03748186 and NCT05200364).


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Endometrial Neoplasms , Immunoconjugates , Lung Neoplasms , Female , Humans , Animals , Mice , Immunoconjugates/chemistry , Tubulin/metabolism , Folate Receptor 1 , Antineoplastic Agents/pharmacology , Endometrial Neoplasms/drug therapy , Cell Line, Tumor , Xenograft Model Antitumor Assays
4.
Nat Chem Biol ; 18(10): 1065-1075, 2022 10.
Article in English | MEDLINE | ID: mdl-35788181

ABSTRACT

Aldehyde dehydrogenases (ALDHs) are promising cancer drug targets, as certain isoforms are required for the survival of stem-like tumor cells. We have discovered selective inhibitors of ALDH1B1, a mitochondrial enzyme that promotes colorectal and pancreatic cancer. We describe bicyclic imidazoliums and guanidines that target the ALDH1B1 active site with comparable molecular interactions and potencies. Both pharmacophores abrogate ALDH1B1 function in cells; however, the guanidines circumvent an off-target mitochondrial toxicity exhibited by the imidazoliums. Our lead isoform-selective guanidinyl antagonists of ALDHs exhibit proteome-wide target specificity, and they selectively block the growth of colon cancer spheroids and organoids. Finally, we have used genetic and chemical perturbations to elucidate the ALDH1B1-dependent transcriptome, which includes genes that regulate mitochondrial metabolism and ribosomal function. Our findings support an essential role for ALDH1B1 in colorectal cancer, provide molecular probes for studying ALDH1B1 functions and yield leads for developing ALDH1B1-targeting therapies.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Aldehyde Dehydrogenase/chemistry , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase, Mitochondrial/genetics , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Aldehydes , Colonic Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Guanidines , Humans , Molecular Probes , Proteome/genetics
5.
Chembiochem ; 20(3): 394-407, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30395379

ABSTRACT

The bacterial second messenger cyclic diguanosine monophosphate (c-di-GMP) is a key regulator of cellular motility, the cell cycle, and biofilm formation with its resultant antibiotic tolerance, which can make chronic infections difficult to treat. Therefore, diguanylate cyclases, which regulate the spatiotemporal production of c-di-GMP, might be attractive drug targets for control of biofilm formation that is part of chronic infections. We present a FRET-based biochemical high-throughput screening approach coupled with detailed structure-activity studies to identify synthetic small-molecule modulators of the diguanylate cyclase DgcA from Caulobacter crescentus. We identified a set of seven small molecules that regulate DgcA enzymatic activity in the low-micromolar range. Subsequent structure-activity studies on selected scaffolds revealed a remarkable diversity of modulatory behavior, including slight chemical substitutions that reverse the effects from allosteric enzyme inhibition to activation. The compounds identified represent new chemotypes and are potentially developable into chemical genetic tools for the dissection of c-di-GMP signaling networks and alteration of c-di-GMP-associated phenotypes. In sum, our studies underline the importance of detailed mechanism-of-action studies for inhibitors of c-di-GMP signaling and demonstrate the complex interplay between synthetic small molecules and the regulatory mechanisms that control the activity of diguanylate cyclases.


Subject(s)
Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Phosphorus-Oxygen Lyases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Allosteric Regulation/drug effects , Caulobacter crescentus/enzymology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli Proteins/metabolism , Molecular Structure , Phosphorus-Oxygen Lyases/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
6.
Eur J Med Chem ; 161: 416-432, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30384045

ABSTRACT

Antibody-drug conjugates (ADCs) represent a new dimension of anticancer chemotherapeutics, with warheads to date generally involving either antitubulin or DNA-directed agents to achieve low-to sub-nanomolar potency. However, other potent cytotoxins working by different pharmacological mechanisms are under investigation, such as α,ß-epoxyketone based proteasome inhibitors. These proteasome active agents are an emerging class of anticancer drug that possesses ultra-potent cytotoxicity to some cancer cell lines. The carmaphycins are representatives of this latter class that we isolated and characterized from a marine cyanobacterium, and these as well as several synthetic analogues exhibit this level of potency. In the current work, we investigated the use of these highly potent cytotoxic compounds as warheads in the design of novel ADCs. We designed and synthesized a library of carmaphycin B analogues that contain amine handles, enabling their attachment to an antibody linker. The basicity of these incorporated amine handles was shown to strongly affect their cytotoxic properties. Linear amines resulted in the greatest reduction in cytotoxicity whereas less basic aromatic amines retained potent activity as demonstrated by a 4-sulfonylaniline derivative. These investigations resulted in identifying the P2 residue in the carmaphycins as the most suitable site for linker attachment point, and hence, we synthesized a highly potent analogue of carmaphycin B that contained a 4-sulfonylaniline handle as an attachment point for the linker antibody.


Subject(s)
Amines/pharmacology , Aniline Compounds/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Oligopeptides/pharmacology , Proteasome Inhibitors/pharmacology , Amines/chemistry , Aniline Compounds/chemistry , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/chemistry , Structure-Activity Relationship
7.
J Nat Prod ; 81(12): 2630-2637, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30485098

ABSTRACT

1,4-Benzodioxane lignans are a class of bioactive compounds that have received much attention through the years. Herein research pertaining to both 1,4-benzodioxane flavonolignans and 1,4-benzodioxane neolignans is presented. A novel synthesis of both traditional 1,4-benzodioxane flavonolignans and 3-deoxyflavonolignans is described. The antiviral and cytotoxic activities of 1,4-benzodioxane neolignans were then investigated; eusiderins A, B, G, and M, deallyl eusiderin A, and nitidanin, which contain the 1,4-benzodioxane motif but lack the chromanone motif found in the known antiviral flavonolignans, were tested. Notably, it was found that all eusiderin 1,4-benzodioxane neolignans exhibited greater antiviral activity than the potent and well-known silybin flavonolignans. While most modifications of the C-1' side chain did not significantly alter the cytotoxicity or antiviral activity, eusiderin M and nitidanin, which contain an allylic alcohol side chain, had lower cytotoxicity. All the eusiderins had similar antiviral activities, with eusiderin B having the best selectivity index. These results show that the chromanone moiety of the flavonolignans is not essential for bioactivity.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Flavonolignans/chemical synthesis , Flavonolignans/pharmacology , Lignans/chemical synthesis , Lignans/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Cell Line, Tumor , Hepacivirus/drug effects , Humans , Molecular Structure , Silybin/chemistry
8.
Mol Pharm ; 15(5): 2054-2059, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29569925

ABSTRACT

Antibody-drug conjugates (ADCs) are antigen-targeted therapeutics that employ antibodies to deliver potent, cytotoxic effectors to cells with potentially high specificity. While promising clinical results have been achieved, significant pitfalls remain including internalization of ADCs in nontargeted cells expressing target antigen, which can limit therapeutic windows. Novel ADC linkers that are cleaved selectively in cancer cells but not in normal cells could minimize collateral damage caused by ADC uptake in nontargeted tissues. Here, we describe a prototypical ADC linker based on an Fe(II)-reactive 1,2,4-trioxolane scaffold (TRX) that by itself has demonstrated tumor-selective activity in preclinical cancer models. We prepared TRX-linked ADCs by site-selective conjugation to two sites in trastuzumab and compared their activity in Her2 positive and negative cells to ADC controls based on established linker chemistry. Our results confirm that the TRX moiety efficiently releases its payload following ADC uptake, affording picomolar potencies in antigen-positive cells. We also identified a destabilizing interaction between these initial TRX linkers and nearby antibody residues and suggest an approach to improve upon these prototypical designs.


Subject(s)
Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Immunoconjugates/chemistry , Iron/chemistry , Animals , Antigens/chemistry , Cell Line, Tumor , Mammals , Receptor, ErbB-2/metabolism , Trastuzumab/chemistry
9.
Oncotarget ; 9(102): 37700-37714, 2018 Dec 28.
Article in English | MEDLINE | ID: mdl-30701025

ABSTRACT

STRO-001 is a site-specific, predominantly single-species, fully human, aglycosylated anti-CD74 antibody-drug conjugate incorporating a non-cleavable linker-maytansinoid warhead with a drug-antibody ratio of 2 which was produced by a novel cell-free antibody synthesis platform. We examined the potential pharmacodynamics and anti-tumor effects of STRO-001 in multiple myeloma (MM). CD74 expression was assessed in MM cell lines and primary bone marrow (BM) MM biopsies. CD74 mRNA was detectable in CD138+ enriched plasma cells from 100% (892/892) of patients with newly diagnosed MM. Immunohistochemistry confirmed CD74 expression in 35/36 BM biopsies from patients with newly diagnosed and relapsed/refractory MM. Cytotoxicity assays demonstrated nanomolar STRO-001 potency in 4/6 MM cell lines. In ARP-1 and MM.1S tumor-bearing mice, repeat STRO-001 dosing provided significant antitumor activity with eradication of malignant hCD138+ BM plasma cells and prolonged survival. In a luciferase-expressing MM.1S xenograft model, dose-dependent STRO-001 efficacy was confirmed using bioluminescent imaging and BM tumor burden quantification. Consistent with the intended pharmacodynamic effect, STRO-001 induced dose-responsive, reversible B-cell and monocyte depletion in cynomolgus monkeys, up to a maximum tolerated 10 mg/kg, with no evidence of off-target toxicity. Collectively, these data suggest that STRO-001 is a promising therapeutic agent for the treatment of MM.

10.
Pharm Res ; 32(11): 3480-93, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25511917

ABSTRACT

Antibody drug conjugates (ADCs) have progressed from hypothesis to approved therapeutics in less than 30 years, and the technologies available to modify both the antibodies and the cytotoxic drugs are expanding rapidly. For reasons well reviewed previously, the field is trending strongly toward homogeneous, defined antibody conjugation. In this review we present the antibody and small molecule chemistries that are currently used and being explored to develop specific, homogenous ADCs.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoconjugates/chemistry , Protein Engineering , Small Molecule Libraries/chemistry , Technology, Pharmaceutical/methods , Animals , Drug Design , Humans
11.
Curr Drug Targets ; 13(3): 338-51, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22206256

ABSTRACT

Type III Secretion Systems (T3SSs) are highly organized multi-protein nanomachines which translocate effector proteins from the bacterial cytosol directly into host cells. These systems are required for the pathogenesis of a wide array of Gram-negative bacterial pathogens, and thus have attracted attention as potential antibacterial drug targets. A decade of research has enabled the identification of natural products, conventional small molecule drug-like structures, and proteins that inhibit T3SSs. The mechanism(s) of action and molecular target(s) of the majority of these inhibitors remain to be determined. At the same time, structural biology methods are providing an increasingly detailed picture of the functional arrangement of the T3SS component proteins. The confluence of these two research areas may ultimately identify non-classical drug targets and facilitate the development of novel therapeutics.


Subject(s)
Anti-Bacterial Agents/metabolism , Drug Delivery Systems , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Drug Delivery Systems/methods , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/immunology , Escherichia coli Proteins/metabolism , Humans , Type III Secretion Systems
12.
Bioorg Med Chem Lett ; 19(5): 1340-3, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19195888

ABSTRACT

Disruption of protein-protein interactions by small molecules is achievable but presents significant hurdles for effective compound design. In earlier work we identified a series of thiazolidinone inhibitors of the bacterial type III secretion system (T3SS) and demonstrated that this scaffold had the potential to be expanded into molecules with broad-spectrum anti-Gram negative activity. We now report on one series of thiazolidinone analogs in which the heterocycle is presented as a dimer at the termini of a series of linkers. Many of these dimers inhibited the T3SS-dependent secretion of a virulence protein at concentrations lower than that of the original monomeric compound identified in our screen.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Secretory Pathway/physiology , Thiazolidinediones/chemical synthesis , Anti-Bacterial Agents/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Secretory Pathway/drug effects , Thiazolidinediones/pharmacology , Type III Secretion Systems , Virulence/drug effects , Virulence/physiology
13.
Nucleosides Nucleotides Nucleic Acids ; 27(12): 1282-300, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19003573

ABSTRACT

The bacterial second messenger cyclic bis-(3'-5')-diguanylic acid (c-di-GMP) regulates diverse Gram-negative bacterial virulence functions. The pathways that control, or are controlled by, c-di-GMP suggest that c-di-GMP signaling systems may encompass potential drug targets. It is presently undetermined, however, whether up- or down-modulation of c-di-GMP signaling would be the desired therapeutic state. We addressed potential drug target validation by synthesizing nonhydrolysable carbamate analogs of both the cyclic dinucleotide and the acyclic (seco) dinucleotide. A molecular docking simulation of the carbamate isostere suggests that this analog is capable of assuming the correct conformation and pose at a c-di-GMP binding site.


Subject(s)
Carbamates/chemistry , Cyclic GMP/analogs & derivatives , Cyclic GMP/chemical synthesis , Cyclic GMP/chemistry , Dimerization , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure
14.
J Med Chem ; 51(22): 7065-74, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-18947223

ABSTRACT

Diverse species of pathogenic Gram-negative bacteria use secretion systems to export a variety of protein toxins and virulence factors that help establish and maintain infection. Disruption of such secretion systems is a potentially effective therapeutic strategy. We developed a high-throughput screen and identified a tris-aryl substituted 2-imino-5-arylidenethiazolidin-4-one, compound 1, as an inhibitor of the type III secretion system. Expansion of this chemotype enabled us to define the essential pharmacophore for type III secretion inhibition by this structural class. A synthetic diversity set helped us identify N-3 as the most permissive locus and led to the design of a panel of novel N-3-dipeptide-modified congeners with improved activity and physiochemical properties. We now report on the synthesis of these compounds, including a novel solid phase approach to the rapid generation of the dipeptide-thiazolidinone hybrids, and their in vitro characterization as inhibitors of type III secretion in Salmonella enterica serovar Typhimurium.


Subject(s)
Anti-Bacterial Agents/pharmacology , Salmonella typhimurium/drug effects , Salmonella typhimurium/metabolism , Thiazolidines/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Drug Design , Microbial Sensitivity Tests , Molecular Structure , Salmonella typhimurium/pathogenicity , Stereoisomerism , Thiazolidines/chemical synthesis , Thiazolidines/chemistry
15.
Cell Host Microbe ; 4(4): 325-36, 2008 Oct 16.
Article in English | MEDLINE | ID: mdl-18854237

ABSTRACT

Bacterial virulence mechanisms are attractive targets for antibiotic development because they are required for the pathogenesis of numerous global infectious disease agents. The bacterial secretion systems used to assemble the surface structures that promote adherence and deliver protein virulence effectors to host cells could comprise one such therapeutic target. In this study, we developed and performed a high-throughput screen of small molecule libraries and identified one compound, a 2-imino-5-arylidene thiazolidinone that blocked secretion and virulence functions of a wide array of animal and plant Gram-negative bacterial pathogens. This compound inhibited type III secretion-dependent functions, with the exception of flagellar motility, and type II secretion-dependent functions, suggesting that its target could be an outer membrane component conserved between these two secretion systems. This work provides a proof of concept that compounds with a broad spectrum of activity against Gram-negative bacterial secretion systems could be developed to prevent and treat bacterial diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Gram-Negative Bacteria/drug effects , Membrane Transport Proteins/metabolism , Thiazolidines/pharmacology , Virulence Factors/metabolism , Animals , Anti-Bacterial Agents/isolation & purification , Bacterial Adhesion/drug effects , Drug Evaluation, Preclinical , Gram-Negative Bacteria/pathogenicity , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Molecular Structure , Plant Leaves/microbiology , Thiazolidines/isolation & purification , Nicotiana/microbiology , Virulence
16.
Antimicrob Agents Chemother ; 51(8): 2820-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17526758

ABSTRACT

The p-nitroaromatic antibiotic chloramphenicol has been used extensively to treat life-threatening infections due to Haemophilus influenzae and Neisseria meningitidis; its mechanism of action is the inhibition of protein synthesis. We found that during incubation with H. influenzae cells and lysates, chloramphenicol is converted to a 4-aminophenyl allylic alcohol that lacks antibacterial activity. The allylic alcohol moiety undergoes facile re-addition of water to restore the 1,3-diol, as well as further dehydration driven by the aromatic amine to form the iminoquinone. Several Neisseria species and most chloramphenicol-susceptible Haemophilus species, but not Escherichia coli or other gram-negative or gram-positive bacteria we examined, were also found to metabolize chloramphenicol. The products of chloramphenicol metabolism by species other than H. influenzae have not yet been characterized. The strains reducing the antibiotic were chloramphenicol susceptible, indicating that the pathway does not appear to mediate chloramphenicol resistance. The role of this novel nitroreductase pathway in the physiology of H. influenzae and Neisseria species is unknown. Further understanding of the H. influenzae chloramphenicol reduction pathway will contribute to our knowledge of the diversity of prokaryotic nitroreductase mechanisms.


Subject(s)
Anti-Bacterial Agents/metabolism , Chloramphenicol/metabolism , Haemophilus influenzae/enzymology , Nitroreductases/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Chloramphenicol/pharmacology , Haemophilus/classification , Haemophilus/drug effects , Haemophilus/enzymology , Haemophilus/growth & development , Haemophilus influenzae/drug effects , Haemophilus influenzae/growth & development , Humans , Microbial Sensitivity Tests , Neisseria/classification , Neisseria/drug effects , Neisseria/enzymology , Neisseria/growth & development , Oxidation-Reduction , Substrate Specificity
17.
Cancer Lett ; 247(1): 130-6, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-16677756

ABSTRACT

Preventing peritoneal implantation of ovarian carcinoma cells could prolong patient remission and survival. CA125 is expressed on most ovarian cancer cells and was reported to be a ligand of mesothelin, a peritoneal protein. We developed a cell adhesion assay with CA125-expresser ovarian cancer cells and human mesothelin-transfected cells and we confirmed that CA125 and mesothelin mediate cell attachment. We also showed that this assay supplies a high-throughput screening system for reagents able to block CA125/mesothelin-dependent cell attachment with a sensitive quantitative readout. We finally demonstrated that a mesothelin chimeric protein and anti-CA125 antibodies block CA125/mesothelin-dependent cell attachment.


Subject(s)
CA-125 Antigen/metabolism , Cell Adhesion , Drug Screening Assays, Antitumor , Membrane Glycoproteins/metabolism , Ovarian Neoplasms/pathology , Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor , CA-125 Antigen/immunology , Cell Line, Tumor , Female , GPI-Linked Proteins , Humans , Membrane Glycoproteins/genetics , Mesothelin , Peritoneal Neoplasms/prevention & control , Peritoneal Neoplasms/secondary , Transfection
18.
Bioconjug Chem ; 17(3): 831-40, 2006.
Article in English | MEDLINE | ID: mdl-16704224

ABSTRACT

A beta-glucuronide-based linker for attaching cytotoxic agents to monoclonal antibodies (mAbs) was designed and evaluated. We employed the cytotoxic auristatin derivatives MMAE (1a) and MMAF (1b) and doxorubicin propyloxazoline (DPO, 2) to give the beta-glucuronide drug-linkers 9a, 9b, and 17, respectively. Cysteine-quenched derivatives of 9b and 17 were determined to be substrates for E. coli beta-glucuronidase, resulting in facile drug release. The beta-glucuronide MMAF drug-linker 9b was highly stable in rat plasma with an extrapolated half-life of 81 days. Each drug-linker when conjugated to mAbs c1F6 (anti-CD70) and cAC10 (anti-CD30) gave monomeric antibody-drug conjugates (ADCs) with as many as eight drugs per mAb and had high levels of immunologically specific cytotoxic activity on cancer cell lines. cAC10-9a displayed pronounced antitumor activity in a subcutaneous Karpas 299 lymphoma tumor model. A single dose treatment led to cures in all animals at the 0.5 mg/kg dose level and above, and the conjugate was well tolerated at 100 mg/kg. In mice with subcutaneous renal cell carcinoma xenografts, the MMAF conjugate c1F6-9b was tolerated at 25 mg/kg and efficacious at 0.75 mg/kg. These results demonstrate that the beta-glucuronide linker system is an effective strategy for targeting cytotoxic agents providing ADCs with high degrees of efficacy at well-tolerated doses.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Cross-Linking Reagents/chemical synthesis , Glucuronides/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cross-Linking Reagents/chemistry , Female , Glucuronides/chemical synthesis , Mice , Mice, SCID , Molecular Structure , Neoplasms/immunology , Neoplasms/pathology , Structure-Activity Relationship
19.
Antimicrob Agents Chemother ; 50(6): 2178-84, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16723580

ABSTRACT

LpxC [UDP-3-O-(R-3-hydroxymyristoyl)-GlcNAc deacetylase] is a metalloamidase that catalyzes the first committed step in the biosynthesis of the lipid A component of lipopolysaccharide. A previous study (H. R. Onishi, B. A. Pelak, L. S. Gerckens, L. L. Silver, F. M. Kahan, M. H. Chen, A. A. Patchett, S. M. Galloway, S. A. Hyland, M. S. Anderson, and C. R. H. Raetz, Science 274:980-982, 1996) identified a series of synthetic LpxC-inhibitory molecules that were bactericidal for Escherichia coli. These molecules did not inhibit the growth of Pseudomonas aeruginosa and were therefore not developed further as antibacterial drugs. The inactivity of the LpxC inhibitors for P. aeruginosa raised the possibility that LpxC activity might not be essential for all gram-negative bacteria. By placing the lpxC gene of P. aeruginosa under tight control of an arabinose-inducible promoter, we demonstrated the essentiality of LpxC activity for P. aeruginosa. It was found that compound L-161,240, the most potent inhibitor from the previous study, was active against a P. aeruginosa construct in which the endogenous lpxC gene was inactivated and in which LpxC activity was supplied by the lpxC gene from E. coli. Conversely, an E. coli construct in which growth was dependent on the P. aeruginosa lpxC gene was resistant to the compound. The differential activities of L-161,240 against the two bacterial species are thus the result primarily of greater potency toward the E. coli enzyme rather than of differences in the intrinsic resistance of the bacteria toward antibacterial compounds due to permeability or efflux. These data validate P. aeruginosa LpxC as a target for novel antibiotic drugs and should help direct the design of inhibitors against clinically important gram-negative bacteria.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Anti-Bacterial Agents/pharmacology , Pseudomonas aeruginosa/drug effects , Amidohydrolases/chemistry , Amidohydrolases/genetics , Amidohydrolases/isolation & purification , Amino Acid Motifs , Amino Acid Sequence , Catalysis , Conserved Sequence , Escherichia coli/enzymology , Escherichia coli/genetics , Genes, Bacterial , Histidine/chemistry , Lipid A/biosynthesis , Molecular Sequence Data , Molecular Structure , Mutation , Oxazoles/chemistry , Oxazoles/pharmacology , Phenylalanine/chemistry , Promoter Regions, Genetic , Pseudomonas aeruginosa/genetics , Reproducibility of Results , Sequence Homology, Amino Acid , Zinc/chemistry
20.
J Med Chem ; 48(5): 1344-58, 2005 Mar 10.
Article in English | MEDLINE | ID: mdl-15743178

ABSTRACT

Antibody-drug conjugates (ADCs) were prepared consisting of DNA minor groove binder drugs (MGBs) attached to monoclonal antibodies (mAbs) through peptide linkers designed to release drugs inside the lysosomes of target cells. The site of linker attachment on the MGB was at the 5-position on the B-ring, since model studies showed that attachment of an electron-withdrawing group (i.e., acyl, carbamoyl) at this position increased the stability of the molecule. Because of the hydrophobic nature of the MGBs, several measures were required to overcome their tendencies to induce mAb aggregation upon conjugation. This is exemplified in the series of ADCs containing the amino-CBI drug 1. Initial adducts were prepared using the peptide sequence valine-citrulline, attached to a self-immolative para-aminobenzyl carbamate spacer. The resulting ADCs were completely aggregated. Removal of the self-immolative spacer, introduction of a more hydrophilic valine-lysine sequence, and incorporation of a tetraethyleneglycol unit between the mAb and the peptide resulted in conjugates that were nonaggregated, even with as many as eight drugs per mAb. These results were extended to include the hydroxy aza-CBI drug 2, which was linked to the valine-lysine sequence through a para-aminobenzyl ether self-immolative spacer. The resulting mAb conjugates were monomeric and released the hydroxy aza-CBI drug upon treatment with human cathepsin B. In vitro cytotoxicity assays established that the mAb-MGB drug conjugates were highly cytotoxic and effected immunologically specific cell kill at subsaturating doses. The results provide a general strategy for MGB prodrug design and illustrate the importance of linker hydrophilicity in making nonaggregated, active mAb-MGB conjugates.


Subject(s)
Antibodies, Monoclonal/chemistry , Antineoplastic Agents, Alkylating/chemical synthesis , DNA/chemistry , Dipeptides/chemistry , Immunoconjugates/chemistry , Indoles/chemical synthesis , Animals , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacology , Aza Compounds/chemical synthesis , Aza Compounds/chemistry , Aza Compounds/pharmacology , Cathepsin B/chemistry , Cell Line, Tumor , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Drug Design , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Indoles/pharmacology , Mice , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...