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










Publication year range
1.
Front Immunol ; 15: 1293883, 2024.
Article in English | MEDLINE | ID: mdl-38455057

ABSTRACT

Fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF) and systemic scleroderma (SSc), are commonly associated with high morbidity and mortality, thereby representing a significant unmet medical need. Interleukin 11 (IL11)-mediated cell activation has been identified as a central mechanism for promoting fibrosis downstream of TGFß. IL11 signaling has recently been reported to promote fibroblast-to-myofibroblast transition, thus leading to various pro-fibrotic phenotypic changes. We confirmed increased mRNA expression of IL11 and IL11Rα in fibrotic diseases by OMICs approaches and in situ hybridization. However, the vital role of IL11 as a driver for fibrosis was not recapitulated. While induction of IL11 secretion was observed downstream of TGFß signaling in human lung fibroblasts and epithelial cells, the cellular responses induced by IL11 was quantitatively and qualitatively inferior to that of TGFß at the transcriptional and translational levels. IL11 blocking antibodies inhibited IL11Rα-proximal STAT3 activation but failed to block TGFß-induced profibrotic signals. In summary, our results challenge the concept of IL11 blockade as a strategy for providing transformative treatment for fibrosis.


Subject(s)
Interleukin-11 , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/metabolism , Signal Transduction , Fibrosis , Myofibroblasts/metabolism
3.
Mol Pharm ; 17(1): 50-58, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31742408

ABSTRACT

DNA-targeting indolinobenzodiazepine dimer (IGN) payloads are used in several clinical-stage antibody-drug conjugates. IGN drugs alkylate DNA through the single imine moiety present in the dimer in contrast to the pyrrolobenzodiazepine dimer drugs, such as talirine and tesirine, which contain two imine moieties per dimer and cross-link DNA. This study explored the mechanism of binding of IGN to DNA in cells and to synthetic duplex and hairpin oligonucleotides. New, highly sensitive IGN-DNA binding enzyme-linked immunosorbent assay methods were developed using biotinylated IGN analogues (monoimine, diimine, and diamine IGNs) and digoxigenin-labeled duplex oligonucleotides, which allowed the measurement of drug-DNA adducts in viable cells at concentrations below IC50. Furthermore, the release of free drug from the IGN-DNA adduct upon treatment with nuclease ex vivo was tested under physiological conditions. The monoimine IGN drug formed a highly stable adduct with DNA in cells, with stability similar to that of the diimine drug analogue. Both monoimine and diimine IGN-DNA adducts released free drugs upon DNA cleavage by nuclease at 37 °C, although more free drug was released from the monoimine compared to the diimine adduct, which presumably was partly cross-linked. The strong binding of the monoimine IGN drug to duplex DNA results from both the noncovalent IGN-DNA interaction and the covalent bond formation between the 2-amino group of a guanine residue and the imine moiety in IGN.


Subject(s)
Antineoplastic Agents/chemistry , Benzodiazepines/chemistry , DNA Adducts/chemistry , DNA/chemistry , Immunoconjugates/pharmacology , Indoles/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA Adducts/metabolism , Dimerization , Enzyme-Linked Immunospot Assay , Humans , Imines/chemistry , Immunoconjugates/administration & dosage , Oligonucleotides/chemistry , Pyrroles/chemistry
4.
Mol Pharm ; 16(12): 4817-4825, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31609629

ABSTRACT

Although peptide linkers are used in multiple clinical-stage ADCs, there are only few reports on optimizing peptide linkers for efficient lysosomal proteolysis and for stability in circulation. We screened multiple dipeptide linkers for efficiency of proteolysis and compared them to the dipeptide linkers currently being evaluated in the clinic: Val-Cit, Val-Ala, and Ala-Ala. Lead dipeptide linkers selected from the initial screen were incorporated into ADCs with indolinobenzodiazepine dimer (IGN) payloads to evaluate cellular processing, in vitro cytotoxic activity, plasma stability, and in vivo efficacy. ADCs with several dipeptide linkers bearing l-amino acids showed faster lysosomal processing in target cancer cells compared to the l-Ala-l-Ala linked ADC. These variances in linker processing rates did not result in different in vitro and in vivo activities among peptide linker ADCs, presumably due to accumulation of threshold cytotoxic catabolite levels for ADCs of several peptide linkers in the cell lines and xenografts tested. ADCs with l-amino acid dipeptide linkers exhibited superior in vitro cytotoxic potencies in multiple cell lines compared to an ADC with a d-Ala-d-Ala dipeptide linker and an ADC with a noncleavable linker. This work adds to the toolbox of stable, lysosomally cleavable peptide linkers for ADCs.


Subject(s)
Antibodies/chemistry , Biopolymers/chemistry , Dipeptides/chemistry , Immunoconjugates/chemistry , Lysosomes/metabolism , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, SCID , Molecular Structure , Xenograft Model Antitumor Assays
5.
ACS Med Chem Lett ; 10(8): 1211-1215, 2019 Aug 08.
Article in English | MEDLINE | ID: mdl-31413807

ABSTRACT

Indolinobenzodiazepine DNA alkylators (IGNs) are the cytotoxic payloads in antibody-drug conjugates (ADCs) currently undergoing Phase I clinical evaluation (IMGN779, IMGN632, and TAK164). These ADCs possess linkers that have been incorporated into a central substituted phenyl spacer. Here, we present an alternative strategy for the IGNs, linking through a carbamate at the readily available N-10 amine present in the monoimine containing dimer. As a result, we have designed a series of N-10 linked IGN ADCs with a wide range of in vitro potency and tolerability, which may allow us to better match an IGN with a particular target based on the potential dosing needs.

6.
Mol Cancer Ther ; 17(3): 650-660, 2018 03.
Article in English | MEDLINE | ID: mdl-29440292

ABSTRACT

Tumor-selective delivery of cytotoxic agents in the form of antibody-drug conjugates (ADCs) is now a clinically validated approach for cancer treatment. In an attempt to improve the clinical success rate of ADCs, emphasis has been recently placed on the use of DNA-cross-linking pyrrolobenzodiazepine compounds as the payload. Despite promising early clinical results with this class of ADCs, doses achievable have been low due to systemic toxicity. Here, we describe the development of a new class of potent DNA-interacting agents wherein changing the mechanism of action from a cross-linker to a DNA alkylator improves the tolerability of the ADC. ADCs containing the DNA alkylator displayed similar in vitro potency, but improved bystander killing and in vivo efficacy, compared with those of the cross-linker. Thus, the improved in vivo tolerability and antitumor activity achieved in rodent models with ADCs of the novel DNA alkylator could provide an efficacious, yet safer option for cancer treatment. Mol Cancer Ther; 17(3); 650-60. ©2018 AACR.


Subject(s)
Immunoconjugates/pharmacology , Intercalating Agents/pharmacology , Neoplasms/drug therapy , Therapeutic Index, Drug , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cross-Linking Reagents/chemistry , DNA/genetics , DNA/metabolism , Drug Design , Humans , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Mice , Neoplasms/pathology , Tumor Burden/drug effects
7.
Mol Cancer Ther ; 15(8): 1870-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27216304

ABSTRACT

The promise of tumor-selective delivery of cytotoxic agents in the form of antibody-drug conjugates (ADC) has now been realized, evidenced by the approval of two ADCs, both of which incorporate highly cytotoxic tubulin-interacting agents, for cancer therapy. An ongoing challenge remains in identifying potent agents with alternative mechanisms of cell killing that can provide ADCs with high therapeutic indices and favorable tolerability. Here, we describe the development of a new class of potent DNA alkylating agents that meets these objectives. Through chemical design, we changed the mechanism of action of our novel DNA cross-linking agent to a monofunctional DNA alkylator. This modification, coupled with linker optimization, generated ADCs that were well tolerated in mice and demonstrated robust antitumor activity in multiple tumor models at doses 1.5% to 3.5% of maximally tolerated levels. These properties underscore the considerable potential of these purpose-created, unique DNA-interacting conjugates for broadening the clinical application of ADC technology. Mol Cancer Ther; 15(8); 1870-8. ©2016 AACR.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Immunoconjugates/pharmacology , Animals , Antineoplastic Agents, Alkylating/chemistry , Bystander Effect , Cell Line, Tumor , Cell Survival/drug effects , DNA/chemistry , DNA/metabolism , DNA Adducts , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Immunoconjugates/chemistry , Mice , Molecular Structure , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 15(6): 1311-20, 2016 06.
Article in English | MEDLINE | ID: mdl-27197308

ABSTRACT

A triglycyl peptide linker (CX) was designed for use in antibody -: drug conjugates (ADC), aiming to provide efficient release and lysosomal efflux of cytotoxic catabolites within targeted cancer cells. ADCs comprising anti-epithelial cell adhesion molecule (anti-EpCAM) and anti-EGFR antibodies with maytansinoid payloads were prepared using CX or a noncleavable SMCC linker (CX and SMCC ADCs). The in vitro cytotoxic activities of CX and SMCC ADCs were similar for several cancer cell lines; however, the CX ADC was more active (5-100-fold lower IC50) than the SMCC ADC in other cell lines, including a multidrug-resistant line. Both CX and SMCC ADCs showed comparable MTDs and pharmacokinetics in CD-1 mice. In Calu-3 tumor xenografts, antitumor efficacy was observed with the anti-EpCAM CX ADC at a 5-fold lower dose than the corresponding SMCC ADC in vivo Similarly, the anti-EGFR CX ADC showed improved antitumor activity over the respective SMCC conjugate in HSC-2 and H1975 tumor models; however, both exhibited similar activity against FaDu xenografts. Mechanistically, in contrast with the charged lysine-linked catabolite of SMCC ADC, a significant fraction of the carboxylic acid catabolite of CX ADC could be uncharged in the acidic lysosomes, and thus diffuse out readily into the cytosol. Upon release from tumor cells, CX catabolites are charged at extracellular pH and do not penetrate and kill neighboring cells, similar to the SMCC catabolite. Overall, these data suggest that CX represents a promising linker option for the development of ADCs with improved therapeutic properties. Mol Cancer Ther; 15(6); 1311-20. ©2016 AACR.


Subject(s)
Epithelial Cell Adhesion Molecule/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Immunoconjugates/administration & dosage , Maytansine/chemistry , Neoplasms/drug therapy , Peptides/chemical synthesis , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Maximum Tolerated Dose , Mice , Mice, SCID , Peptides/chemistry , Peptides/pharmacokinetics , Xenograft Model Antitumor Assays
9.
Bioconjug Chem ; 26(11): 2261-78, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26355774

ABSTRACT

Antibody anilino maytansinoid conjugates (AaMCs) have been prepared in which a maytansinoid bearing an aniline group was linked through the aniline amine to a dipeptide, which in turn was covalently attached to a desired monoclonal antibody. Several such conjugates were prepared utilizing different dipeptides in the linkage including Gly-Gly, l-Val-l-Cit, and all four stereoisomers of the Ala-Ala dipeptide. The properties of AaMCs could be altered by the choice of dipeptide in the linker. Each of the AaMCs, except the AaMC bearing a d-Ala-d-Ala peptide linker, displayed more bystander killing in vitro than maytansinoid ADCs that utilize disulfide linkers. In mouse models, the anti-CanAg AaMC bearing a d-Ala-l-Ala dipeptide in the linker was shown to be more efficacious against heterogeneous HT-29 xenografts than maytansinoid ADCs that utilize disulfide linkers, while both types of the conjugates displayed similar tolerabilities.


Subject(s)
Aniline Compounds/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Immunoconjugates/chemistry , Maytansine/chemistry , Aniline Compounds/pharmacokinetics , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Maytansine/pharmacokinetics , Maytansine/therapeutic use , Mice , Neoplasms/drug therapy
10.
Mol Cancer Ther ; 14(7): 1605-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25904506

ABSTRACT

A majority of ovarian and non-small cell lung adenocarcinoma cancers overexpress folate receptor α (FRα). Here, we report the development of an anti-FRα antibody-drug conjugate (ADC), consisting of a FRα-binding antibody attached to a highly potent maytansinoid that induces cell-cycle arrest and cell death by targeting microtubules. From screening a large panel of anti-FRα monoclonal antibodies, we selected the humanized antibody M9346A as the best antibody for targeted delivery of a maytansinoid payload into FRα-positive cells. We compared M9346A conjugates with various linker/maytansinoid combinations, and found that a conjugate, now denoted as IMGN853, with the N-succinimidyl 4-(2-pyridyldithio)-2-sulfobutanoate (sulfo-SPDB) linker and N(2')-deacetyl-N(2')-(4-mercapto-4-methyl-1-oxopentyl)-maytansine (DM4) exhibited the most potent antitumor activity in several FRα-expressing xenograft tumor models. The level of expression of FRα on the surface of cells was a major determinant in the sensitivity of tumor cells to the cytotoxic effect of the conjugate. Efficacy studies of IMGN853 in xenografts of ovarian cancer and non-small cell lung cancer cell lines and of a patient tumor-derived xenograft model demonstrated that the ADC was highly active against tumors that expressed FRα at levels similar to those found on a large fraction of ovarian and non-small cell lung cancer patient tumors, as assessed by immunohistochemistry. IMGN853 displayed cytotoxic activity against FRα-negative cells situated near FRα-positive cells (bystander cytotoxic activity), indicating its ability to eradicate tumors with heterogeneous expression of FRα. Together, these findings support the clinical development of IMGN853 as a novel targeted therapy for patients with FRα-expressing tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Folate Receptor 1/antagonists & inhibitors , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Bystander Effect/drug effects , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Female , Folate Receptor 1/immunology , Humans , Immunoconjugates/immunology , Maytansine/analogs & derivatives , Maytansine/immunology , Maytansine/pharmacology , Mice, Nude , Mice, SCID , Molecular Targeted Therapy/methods , Neoplasms/immunology , Neoplasms/metabolism , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/immunology
11.
Mol Pharm ; 12(6): 1752-61, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25738394

ABSTRACT

A new, sensitive ELISA method has been developed which measures catabolites in cells and media upon processing of antibody-drug conjugates (ADCs) by target cancer cells. This ELISA method, exemplified for maytansinoid ADCs, uses competitive inhibition by a maytansinoid analyte of the binding of biotinylated antimaytansine antibody to an immobilized BSA-maytansinoid conjugate. Synthetic standards of several maytansinoid catabolites derived from ADCs with different linkers were tested and showed similar inhibition curves, with an EC50 of about 0.1 nM (0.03 pmol in an assay volume of 0.25 mL). This high sensitivity allowed quantification of catabolites from a methanolic cell extract and from the medium, generated from an ADC in 1 day using only about 1 million cells. The processing of anti-EpCAM and anti-CanAg ADCs with noncleavable linker (SMCC-DM1), disulfide linker (SPDB-DM4), and charged sulfonate-bearing disulfide linker (sulfo-SPDB-DM4), each containing an average of about four maytansinoid molecules per antibody, were compared in colon cancer cell lines (COLO 205 and HT-29). An 8-10-fold higher total level of catabolite was observed for anti-CanAg ADCs than for anti-EpCAM ADCs upon processing by COLO 205 cells, consistent with a higher cell-surface expression of CanAg. In a multidrug resistant HCT-15 colon cancer cell line, the anti-EpCAM-SPDB-DM4 linker conjugate was not cytotoxic and showed a significantly lower level of catabolite within cells compared to that in medium, presumably due to Pgp-mediated efflux of the nonpolar DM4 catabolite. In contrast, sulfo-SPDB-DM4 and SMCC-DM1 linker conjugates were cytotoxic, which correlated with higher amounts of catabolites found within the HCT-15 cells relative to amounts in medium. In a nonmultidrug resistant HT-29 cell line, the anti-EpCAM-SPDB-DM4 linker conjugate was cytotoxic, with most of the catabolite found in cells and little in the medium. In conclusion, this highly sensitive ELISA method for measurement of ADC catabolite is convenient for screening multiple ADC parameters such as linkers and antibodies in a number of cell lines, does not require concentration of sample or extraction of media, and is complementary to other reported methods such as radiolabeling of ADCs or mass spectrometry.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Immunoconjugates/metabolism , Cell Line, Tumor , Cell Survival/drug effects , HT29 Cells , Humans , Immunoconjugates/adverse effects , Immunoconjugates/chemistry , Maytansine/chemistry , Reproducibility of Results
12.
Chem Commun (Camb) ; 47(38): 10752-4, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21874179

ABSTRACT

A novel pathway for ex vivo maytansinoid release from thioether linked antibody maytansinoid conjugates (AMCs) upon incubation in human plasma has been identified. A thioether succinimide-linked AMC can undergo chemical oxidation followed by sulfoxide elimination under mild aqueous conditions (pH 5.5-7.5, 37 °C). Oxidized thioether-linked AMCs exhibit high, target-specific cytotoxicity toward cancer cells.


Subject(s)
Antibodies/chemistry , Immunoconjugates/chemistry , Maytansine/chemistry , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Immunoconjugates/blood , Immunoconjugates/toxicity , Maleimides/chemistry , Oxidation-Reduction , Polyethylene Glycols/chemistry , Sulfenic Acids/chemistry
13.
J Med Chem ; 54(10): 3606-23, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21517041

ABSTRACT

The synthesis and biological evaluation of hydrophilic heterobifunctional cross-linkers for conjugation of antibodies with highly cytotoxic agents are described. These linkers contain either a negatively charged sulfonate group or a hydrophilic, noncharged PEG group in addition to an amine-reactive N-hydroxysuccinimide (NHS) ester and sulfhydryl reactive termini. These hydrophilic linkers enable conjugation of hydrophobic organic molecule drugs, such as a maytansinoid, at a higher drug/antibody ratio (DAR) than hydrophobic SPDB and SMCC linkers used earlier without triggering aggregation or loss of affinity of the resulting conjugate. Antibody-maytansinoid conjugates (AMCs) bearing these sulfonate- or PEG-containing hydrophilic linkers were, depending on the nature of the targeted cells, equally to more cytotoxic to antigen-positive cells and equally to less cytotoxic to antigen-negative cells than conjugates made with SPDB or SMCC linkers and thus typically displayed a wider selectivity window, particularly against multidrug resistant (MDR) cancer cell lines in vitro and tumor xenograft models in vivo.


Subject(s)
Antibodies/chemistry , Immunoconjugates/chemistry , Maytansine/chemistry , Animals , Chemistry, Pharmaceutical/methods , Drug Design , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Mice , Models, Chemical , Neoplasm Transplantation , Polyethylene Glycols/chemistry , Succinimides/chemistry , Sulfones/chemistry
14.
Bioconjug Chem ; 22(4): 717-27, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21425776

ABSTRACT

In this report, we describe the synthesis of a panel of disulfide-linked huC242 (anti-CanAg) antibody maytansinoid conjugates (AMCs), which have varying levels of steric hindrance around the disulfide bond, in order to investigate the relationship between stability to reduction of the disulfide linker and antitumor activity of the conjugate in vivo. The conjugates were first tested for stability to reduction by dithiothreitol in vitro and for plasma stability in CD1 mice. It was found that the conjugates having the more sterically hindered disulfide linkages were more stable to reductive cleavage of the maytansinoid in both settings. When the panel of conjugates was tested for in vivo efficacy in two human colon cancer xenograft models in SCID mice, it was found that the conjugate with intermediate disulfide bond stability having two methyl groups on the maytansinoid side of the disulfide bond and no methyl groups on the linker side of the disulfide bond (huC242-SPDB-DM4) displayed the best efficacy. The ranking of in vivo efficacies of the conjugates was not predicted by their in vitro potencies, since all conjugates were highly active in vitro, including a huC242-SMCC-DM1 conjugate with a noncleavable linkage which showed only marginal activity in vivo. These data suggest that factors in addition to intrinsic conjugate potency and conjugate half-life in plasma influence the magnitude of antitumor activity observed for an AMC in vivo. We provide evidence that bystander killing of neighboring nontargeted tumor cells by diffusible cytotoxic metabolites produced from target cell processing of disulfide-linked antibody-maytansinoid conjugates may be one additional factor contributing to the activity of these conjugates in vivo.


Subject(s)
Antibodies/chemistry , Antineoplastic Agents/chemistry , Carbon/chemistry , Colonic Neoplasms/drug therapy , Disulfides/chemistry , Maytansine/chemistry , Animals , Antibodies/blood , Antibodies/pharmacology , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacology , Colonic Neoplasms/metabolism , Disulfides/blood , Disulfides/pharmacology , Humans , Maytansine/blood , Maytansine/pharmacology , Mice , Mice, Inbred Strains , Mice, SCID , Molecular Conformation , Xenograft Model Antitumor Assays
15.
Bioconjug Chem ; 22(4): 728-35, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21391620

ABSTRACT

Antibody-maytansinoid conjugates (AMCs) are targeted chemotherapeutic agents consisting of a potent microtubule-depolymerizing maytansinoid (DM1 or DM4) attached to lysine residues of a monoclonal antibody (mAb) using an uncleavable thioether linker or a stable disulfide linker. Most of the administered dose of an antibody-based therapeutic is slowly catabolized by the liver and other tissues of the reticuloendothelial system. Maytansinoids released from an AMC during this catabolic process could potentially be a source of toxicity. To investigate this, we isolated and identified liver metabolites in mice for three different [(3)H]AMCs with structures similar to those currently undergoing evaluation in the clinic. We then synthesized each metabolite to confirm the identification and assessed their cytotoxic potencies when added extracellularly. We found that the uncleavable mAb-SMCC-[(3)H]DM1 conjugate was degraded to a single major maytansinoid metabolite, lysine-SMCC-[(3)H]DM1, that was nearly 50-fold less cytotoxic than maytansine. The two disulfide-linked conjugates, mAb-SPP-[(3)H]DM1 and mAb-SPDB-[(3)H]DM4, were also found to be catabolized to the analogous lysine-linked maytansinoid metabolites. However, subsequent reduction, S-methylation, and NADPH-dependent oxidation steps in the liver yielded the corresponding S-methyl sulfoxide and S-methyl sulfone derivatives. The cytotoxic potencies of the oxidized maytansinoids toward several human carcinoma cell lines were found to be 5- to 50-fold less potent than maytansine. Our results suggest that liver plays an important role in the detoxification of both cleavable and uncleavable AMCs.


Subject(s)
Antibodies, Monoclonal/metabolism , Drug Design , Liver/metabolism , Maytansine/metabolism , Animals , Antibodies, Monoclonal/chemistry , Female , Liver/chemistry , Maytansine/analogs & derivatives , Maytansine/chemistry , Mice , Mice, Inbred Strains , Molecular Structure
16.
Cancer Res ; 70(6): 2528-37, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20197459

ABSTRACT

Conjugation of cytotoxic compounds to antibodies that bind to cancer-specific antigens makes these drugs selective in killing cancer cells. However, many of the compounds used in such antibody-drug conjugates (ADC) are substrates for the multidrug transporter MDR1. To evade the MDR1-mediated resistance, we conjugated the highly cytotoxic maytansinoid DM1 to antibodies via the maleimidyl-based hydrophilic linker PEG(4)Mal. Following uptake into target cells, conjugates made with the PEG(4)Mal linker were processed to a cytotoxic metabolite that was retained by MDR1-expressing cells better than a metabolite of similar conjugates prepared with the nonpolar linker N-succinimidyl-4-(maleimidomethyl)cyclohexane-1-carboxylate (SMCC). In accord, PEG(4)Mal-linked conjugates were more potent in killing MDR1-expressing cells in culture. In addition, PEG(4)Mal-linked conjugates were markedly more effective in eradicating MDR1-expressing human xenograft tumors than SMCC-linked conjugates while being tolerated similarly, thus showing an improved therapeutic index. This study points the way to the development of ADCs that bypass multidrug resistance.


Subject(s)
Immunotoxins/pharmacology , Maytansine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Epithelial Cell Adhesion Molecule , Female , Humans , Immunotoxins/chemistry , Immunotoxins/pharmacokinetics , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Maleimides/chemistry , Maytansine/chemistry , Maytansine/pharmacokinetics , Maytansine/pharmacology , Mice , Mice, SCID , Polyethylene Glycols/chemistry
17.
Bioconjug Chem ; 21(1): 84-92, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19891424

ABSTRACT

Antibody-drug conjugates (ADCs) are designed to eradicate cancer cells that express the target antigen on their cell surface. A key component of an ADC is the linker that covalently connects the cytotoxic agent to the antibody. Several antibody-maytansinoid conjugates prepared with disulfide-based linkers such as those targeting the CanAg antigen have been shown to display more activity in preclinical mouse xenograft models than corresponding conjugates prepared with uncleavable thioether-based linkers. To investigate how the linker influences delivery and activation of antibody-maytansinoid conjugates, we isolated and characterized the [(3)H]maytansinoids from CanAg-positive tumor tissues following a single intravenous administration of 300 microg/kg (based on maytansinoid dose) of anti-CanAg antibody (huC242)-(3)H-maytansinoid conjugates prepared with cleavable disulfide linkers and an uncleavable thioether linker. We identified three target-dependent tumor metabolites of the disulfide-linked huC242-SPDB-DM4, namely, lysine-N(epsilon)-SPDB-DM4, DM4, and S-methyl-DM4. We found similar metabolites for the less hindered disulfide-linked huC242-SPP-DM1 conjugate with the exception that no S-methyl-DM1 was detected. The sole metabolite of the uncleavable thioether-linked huC242-SMCC-DM1 was lysine-N(epsilon)-SMCC-DM1. The AUC for the metabolites of huC242-SMCC-DM1 at the tumor over 7 d was about 2-fold greater than the corresponding AUC for the metabolites of the disulfide-linked conjugates. The lipophilic metabolites of the disulfide-linked conjugates were found to be nearly 1000 times more cytotoxic than the more hydrophilic lysine-N(epsilon)-linker-maytansinoids in cell-based viability assays when added extracellularly. The cell killing properties associated with the lipophilic metabolites of the disulfide-linked conjugates (DM4 and S-methyl-DM4, and DM1) provide an explanation for the superior in vivo efficacy that is often observed with antibody-maytansinoid conjugates prepared with disulfide-based linkers in xenograft mouse models.


Subject(s)
Antibodies/metabolism , Disulfides/chemistry , Immunoconjugates/metabolism , Immunoconjugates/therapeutic use , Maytansine/metabolism , Neoplasms/metabolism , Sulfides/chemistry , Animals , Antibodies/chemistry , Antibodies/immunology , Antibodies/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Maytansine/chemistry , Maytansine/immunology , Maytansine/therapeutic use , Mice , Mice, SCID , Neoplasms/drug therapy , Neoplasms/immunology , Xenograft Model Antitumor Assays
18.
Methods Mol Biol ; 525: 445-67, xiv, 2009.
Article in English | MEDLINE | ID: mdl-19252846

ABSTRACT

Conjugates of antibodies with cytotoxic agents offer a targeted therapeutic strategy against cancer cells expressing target antigens. Several antibodies against various cancer cell-surface antigens have been conjugated with different cytotoxic agents that inhibit essential cellular targets such as microtubules or DNA. Antibody-cytotoxic agent conjugates (ACCs) against several types of cancer are currently in advanced stages of clinical trials and one, gemtuzumab ozogamicin (Mylotarg), is approved for the treatment of acute myeloid leukemia. The linker group connecting the antibody to the cytotoxic agent is an important feature of the ACC, modulating the release of the active cytotoxic agent in the targeted cell. Several linker strategies employed for ACCs in current clinical trials include cleavable linkers with disulfide, hydrazone, lysosomal protease-substrate groups, and non-cleavable linkers. This chapter describes the methods of preparation of conjugates of antibodies with small-molecule cytotoxic agents (maytansinoids, calicheamicin, and auristatins) bearing different linkers. Methods to evaluate the in vitro cytotoxicity and in vivo anti-tumor efficacy of ACC are described in brief. Analytical methods are described to evaluate the mechanism of cellular processing of the ACCs with different linkers and the generation of the active metabolites.


Subject(s)
Antibodies/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Molecular Biology/methods , Animals , Antibodies/chemistry , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line, Tumor , Disulfides/metabolism , Drug Screening Assays, Antitumor , Glycosylation/drug effects , Humans , Mass Spectrometry , Mice
19.
Ann Surg Oncol ; 14(10): 2838-46, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17653802

ABSTRACT

BACKGROUND: Colorectal carcinomas (CRC) express high levels of insulin-like growth factor-I/II (IGF-I/II) and the receptor (IGF-IR). We hypothesized that selective inhibition of IGF-IR would inhibit hepatic growth of human CRC in mice. METHODS: Human CRC cells were treated in vitro with anti-IGF-IR monoclonal antibody (MoAB) with and without oxaliplatin to assess cytotoxicity. The effect of anti-IGF-IR MoAB on IGF-I-induced vascular endothelial growth factor (VEGF) production in human CRC cells was assessed by Northern blot and ELISA. We injected human CRC cells intrahepatically in nude mice, and then administered anti-IGF-IR MoAB with and without oxaliplatin. We delayed treatment in one group until large hepatic tumors were present. We assessed tumors for apoptosis, proliferation, and angiogenesis. RESULTS: Anti-IGF-IR MoAB and oxaliplatin inhibited CRC cell growth in vitro and combination treatment was even more effective. IGF-I stimulation of CRC cells resulted in significant upregulation of VEGF and this was completely inhibited by pretreatment with anti-IGF-IR MoAB. Anti-IGF-IR MoAB significantly inhibited hepatic growth of tumors in mice. Anti-IGF-IR MoAB plus oxaliplatin led to a significantly greater inhibition of tumor growth. Anti-IGF-IR MoAB plus oxaliplatin was just as effective at inhibiting growth of larger, more advanced liver tumors. Anti-IGF-IR MoAB, alone and in combination with oxaliplatin, led to a significant increase in tumor cell apoptosis, and a significant inhibition of tumor cell proliferation and angiogenesis. CONCLUSIONS: These findings suggest that IGF-IR is a potential target for therapy in patients with advanced CRC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/pathology , Liver Neoplasms, Experimental/secondary , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Northern , Enzyme-Linked Immunosorbent Assay , HT29 Cells/drug effects , HT29 Cells/pathology , HT29 Cells/transplantation , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Organoplatinum Compounds/pharmacology , Oxaliplatin , Vascular Endothelial Growth Factor A/metabolism
20.
J Child Neurol ; 21(12): 1067-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17156700

ABSTRACT

Three normal children with headache occurring only with exertion were advised to try "head cooling" (eg, immersion of the head in cold water, cold water poured over the head, application of a cold, wet towel or ice pack) at the onset of headache. The patients were followed up quarterly as outpatients, and the effectiveness of head cooling in terms of the frequency of headaches, intensity (interference with play), duration, and side effects was assessed over 18 months.


Subject(s)
Body Temperature/physiology , Cold Temperature , Exercise/physiology , Headache Disorders, Primary/etiology , Headache Disorders, Primary/therapy , Hypothermia, Induced/methods , Adolescent , Carotid Artery, External/physiopathology , Child , Head/physiopathology , Headache Disorders, Primary/physiopathology , Humans , Male , Sports , Treatment Outcome , Vasoconstriction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...