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










Publication year range
1.
Mol Cancer Ther ; 20(6): 1112-1120, 2021 06.
Article in English | MEDLINE | ID: mdl-33722856

ABSTRACT

Calicheamicin antibody-drug conjugates (ADCs) are effective therapeutics for leukemias with two recently approved in the United States: Mylotarg (gemtuzumab ozogamicin) targeting CD33 for acute myeloid leukemia and Besponsa (inotuzumab ozogamicin) targeting CD22 for acute lymphocytic leukemia. Both of these calicheamicin ADCs are heterogeneous, aggregation-prone, and have a shortened half-life due to the instability of the acid-sensitive hydrazone linker in circulation. We hypothesized that we could improve upon the heterogeneity, aggregation, and circulation stability of calicheamicin ADCs by directly attaching the thiol of a reduced calicheamicin to an engineered cysteine on the antibody via a disulfide bond to generate a linkerless and traceless conjugate. We report herein that the resulting homogeneous conjugates possess minimal aggregation and display high in vivo stability with 50% of the drug remaining conjugated to the antibody after 21 days. Furthermore, these calicheamicin ADCs are highly efficacious in mouse models of both solid tumor (HER2+ breast cancer) and hematologic malignancies (CD22+ non-Hodgkin lymphoma). Safety studies in rats with this novel calicheamicin ADC revealed an increased tolerability compared with that reported for Mylotarg. Overall, we demonstrate that applying novel linker chemistry with site-specific conjugation affords an improved, next-generation calicheamicin ADC.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Calicheamicins/therapeutic use , Immunoconjugates/therapeutic use , Animals , Antibiotics, Antineoplastic/pharmacology , Calicheamicins/pharmacology , Disease Models, Animal , Humans , Immunoconjugates/pharmacology , Mice
2.
Mol Pharm ; 16(9): 3926-3937, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31287952

ABSTRACT

Antibody-drug conjugates are an emerging class of cancer therapeutics constructed from monoclonal antibodies conjugated with small molecule effectors. First-generation molecules of this class often employed heterogeneous conjugation chemistry, but many site-specifically conjugated ADCs have been described recently. Here, we undertake a systematic comparison of ADCs made with the same antibody and the same macrocyclic maytansinoid effector but conjugated either heterogeneously at lysine residues or site-specifically at cysteine residues. Characterization of these ADCs in vitro reveals generally similar properties, including a similar catabolite profile, a key element in making a meaningful comparison of conjugation chemistries. In a mouse model of cervical cancer, the lysine-conjugated ADC affords greater efficacy on a molar payload basis. Rather than making general conclusions about ADCs conjugated by a particular chemistry, we interpret these results as highlighting the complexity of ADCs and the interplay between payload class, linker chemistry, target antigen, and other variables that determine efficacy in a given setting.


Subject(s)
Antibodies, Monoclonal/chemistry , Cysteine/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Lysine/chemistry , Maytansine/immunology , Uterine Cervical Neoplasms/drug therapy , Animals , Cell Survival/drug effects , Female , HeLa Cells , Humans , Immunoconjugates/administration & dosage , Injections, Intravenous , Mice , Mice, SCID , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Bioconjug Chem ; 28(10): 2538-2548, 2017 10 18.
Article in English | MEDLINE | ID: mdl-28885827

ABSTRACT

The incorporation of cysteines into antibodies by mutagenesis allows for the direct conjugation of small molecules to specific sites on the antibody via disulfide bonds. The stability of the disulfide bond linkage between the small molecule and the antibody is highly dependent on the location of the engineered cysteine in either the heavy chain (HC) or the light chain (LC) of the antibody. Here, we explore the basis for this site-dependent stability. We evaluated the in vivo efficacy and pharmacokinetics of five different cysteine mutants of trastuzumab conjugated to a pyrrolobenzodiazepine (PBD) via disulfide bonds. A significant correlation was observed between disulfide stability and efficacy for the conjugates. We hypothesized that the observed site-dependent stability of the disulfide-linked conjugates could be due to differences in the attachment site cysteine thiol pKa. We measured the cysteine thiol pKa using isothermal titration calorimetry (ITC) and found that the variants with the highest thiol pKa (LC K149C and HC A140C) were found to yield the conjugates with the greatest in vivo stability. Guided by homology modeling, we identified several mutations adjacent to LC K149C that reduced the cysteine thiol pKa and, thus, decreased the in vivo stability of the disulfide-linked PBD conjugated to LC K149C. We also present results suggesting that the high thiol pKa of LC K149C is responsible for the sustained circulation stability of LC K149C TDCs utilizing a maleimide-based linker. Taken together, our results provide evidence that the site-dependent stability of cys-engineered antibody-drug conjugates may be explained by interactions between the engineered cysteine and the local protein environment that serves to modulate the side-chain thiol pKa. The influence of cysteine thiol pKa on stability and efficacy offers a new parameter for the optimization of ADCs that utilize cysteine engineering.


Subject(s)
Cysteine/chemistry , Immunoconjugates/chemistry , Benzodiazepines/chemistry , Drug Stability , Immunoconjugates/genetics , Maleimides/chemistry , Models, Molecular , Mutation , Protein Conformation , Pyrroles/chemistry
4.
Bioconjug Chem ; 28(5): 1371-1381, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28388844

ABSTRACT

Antibody-drug conjugates (ADCs) are being actively pursued as a treatment option for cancer following the regulatory approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla). ADCs consist of a cytotoxic agent conjugated to a targeting antibody through a linker. The two approved ADCs (and most ADCs now in the clinic that use a microtubule disrupting agent as the payload) are heterogeneous conjugates with an average drug-to-antibody ratio (DAR) of 3-4 (potentially ranging from 0 to 8 for individual species). Ado-trastuzumab emtansine employs DM1, a semisynthetic cytotoxic payload of the maytansinoid class, which is conjugated via lysine residues of the antibody to an average DAR of 3.5. To understand the effect of DAR on the preclinical properties of ADCs using maytansinoid cytotoxic agents, we prepared a series of conjugates with a cleavable linker (M9346A-sulfo-SPDB-DM4 targeting folate receptor α (FRα)) or an uncleavable linker (J2898A-SMCC-DM1 targeting the epidermal growth factor receptor (EGFR)) with varying DAR and evaluated their biochemical characteristics, in vivo stability, efficacy, and tolerability. For both formats, a series of ADCs with DARs ranging from low (average of ∼2 and range of 0-4) to very high (average of 10 and range of 7-14) were prepared in good yield with high monomer content and low levels of free cytotoxic agent. The in vitro potency consistently increased with increasing DAR at a constant antibody concentration. We then characterized the in vivo disposition of these ADCs. Pharmacokinetic analysis showed that conjugates with an average DAR below ∼6 had comparable clearance rates, but for those with an average DAR of ∼9-10, rapid clearance was observed. Biodistribution studies in mice showed that these 9-10 DAR ADCs rapidly accumulate in the liver, with maximum localization for this organ at 24-28% percentage injected dose per gram (%ID/g) compared with 7-10% for lower-DAR conjugates (all at 2-6 h post-injection). Our preclinical findings on tolerability and efficacy suggest that maytansinoid conjugates with DAR ranging from 2 to 6 have a better therapeutic index than conjugates with very high DAR (∼9-10). These very high DAR ADCs suffer from decreased efficacy, likely due to faster clearance. These results support the use of DAR 3-4 for maytansinoid ADCs but suggest that the exploration of lower or higher DAR may be warranted depending on the biology of the target antigen.


Subject(s)
Antibodies, Monoclonal/immunology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Immunoconjugates/pharmacokinetics , Maytansine/pharmacokinetics , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Female , Humans , Immunoconjugates/pharmacology , KB Cells , Maytansine/pharmacology , Mice , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Drug Metab Dispos ; 44(12): 1958-1962, 2016 12.
Article in English | MEDLINE | ID: mdl-27683653

ABSTRACT

Pyrrolobenzodiazepine (PBD)-dimer is a DNA minor groove alkylator, and its CD22 THIOMAB antibody drug conjugate (ADC) demonstrated, through a disulfide linker, an efficacy in tumor reduction for more than 7 weeks with minimal body weight loss in xenograft mice after a single 0.5-1 mg/kg i.v. dose. The DNA alkylation was investigated here in tumors and healthy organs of mice to understand the sustained efficacy and tolerability. The experimental procedures included the collection of tumors and organ tissues of xenograft mice treated with the ADC followed by DNA isolation/hydrolysis/quantitation and payload recovery from reversible DNA alkylation. PBD-dimer formed a considerable amount of adducts with tissue DNA, representing approximately 98% (at 24 hours), and 99% (at 96 hours) of the total PBD-dimer in tumors, and 78-89% in liver and lung tissues, suggesting highly efficient covalent binding of the released PBD-dimer to tissue DNA. The amount of PBD-DNA adducts in tumor tissues was approximately 24-fold (at 24 hours) and 70-fold (at 96 hours) greater than the corresponding amount of adducts in liver and lung tissues. In addition, the DNA alkylation levels increased 3-fold to 4-fold from 24 to 96 hours in tumors [41/106 base pairs (bp) at 96 hours] but remained at the same level (1/106 bp) in livers and lungs. These results support the typical target-mediated cumulative uptake of ADC into tumors and payload release that offers an explanation for its sustained antitumor efficacy. In addition, the low level of DNA alkylation in normal tissues is consistent with the tolerability observed in mice.


Subject(s)
Alkylation/physiology , Antibodies/metabolism , Benzodiazepines/metabolism , DNA/metabolism , Pyrroles/metabolism , Animals , DNA Adducts/metabolism , Heterografts/metabolism , Immunoconjugates/metabolism , Liver/metabolism , Lung/metabolism , Mice , Neoplasms/metabolism
6.
Drug Metab Dispos ; 44(9): 1517-23, 2016 09.
Article in English | MEDLINE | ID: mdl-27417182

ABSTRACT

Despite recent technological advances in quantifying antibody drug conjugate (ADC) species, such as total antibody, conjugated antibody, conjugated drug, and payload drug in circulation, the correlation of their exposures with the efficacy of ADC outcomes in vivo remains challenging. Here, the chemical structures and concentrations of intratumor catabolites were investigated to better understand the drivers of ADC in vivo efficacy. Anti-CD22 disulfide-linked pyrrolobenzodiazepine (PBD-dimer) conjugates containing methyl- and cyclobutyl-substituted disulfide linkers exhibited strong efficacy in a WSU-DLCL2 xenograft mouse model, whereas an ADC derived from a cyclopropyl linker was inactive. Total ADC antibody concentrations and drug-to-antibody ratios (DAR) in circulation were similar between the cyclobutyl-containing ADC and the cyclopropyl-containing ADC; however, the former afforded the release of the PBD-dimer payload in the tumor, but the latter only generated a nonimmolating thiol-containing catabolite that did not bind to DNA. These results suggest that intratumor catabolite analysis rather than systemic pharmacokinetic analysis may be used to better explain and predict ADC in vivo efficacy. These are good examples to demonstrate that the chemical nature and concentration of intratumor catabolites depend on the linker type used for drug conjugation, and the potency of the released drug moiety ultimately determines the ADC in vivo efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Benzodiazepines/pharmacokinetics , Immunoconjugates/pharmacokinetics , Neoplasms/metabolism , Pyrroles/pharmacokinetics , Animals , Antibodies, Monoclonal, Humanized/chemistry , Benzodiazepines/chemistry , Female , Immunoconjugates/chemistry , Mice , Mice, SCID , Pyrroles/chemistry , Xenograft Model Antitumor Assays
7.
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
8.
Bioconjug Chem ; 27(7): 1588-98, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27174129

ABSTRACT

Antibody-drug conjugates (ADCs) have become a widely investigated modality for cancer therapy, in part due to the clinical findings with ado-trastuzumab emtansine (Kadcyla). Ado-trastuzumab emtansine utilizes the Ab-SMCC-DM1 format, in which the thiol-functionalized maytansinoid cytotoxic agent, DM1, is linked to the antibody (Ab) via the maleimide moiety of the heterobifunctional SMCC linker. The pharmacokinetic (PK) data for ado-trastuzumab emtansine point to a faster clearance for the ADC than for total antibody. Cytotoxic agent release in plasma has been reported with nonmaytansinoid, cysteine-linked ADCs via thiol-maleimide exchange, for example, brentuximab vedotin. For Ab-SMCC-DM1 ADCs, however, the main catabolite reported is lysine-SMCC-DM1, the expected product of intracellular antibody proteolysis. To understand these observations better, we conducted a series of studies to examine the stability of the thiol-maleimide linkage, utilizing the EGFR-targeting conjugate, J2898A-SMCC-DM1, and comparing it with a control ADC made with a noncleavable linker that lacked a thiol-maleimide adduct (J2898A-(CH2)3-DM). We employed radiolabeled ADCs to directly measure both the antibody and the ADC components in plasma. The PK properties of the conjugated antibody moiety of the two conjugates, J2898A-SMCC-DM1 and J2898A-(CH2)3-DM (each with an average of 3.0 to 3.4 maytansinoid molecules per antibody), appear to be similar to that of the unconjugated antibody. Clearance values of the intact conjugates were slightly faster than those of the Ab components. Furthermore, J2898A-SMCC-DM1 clears slightly faster than J2898A-(CH2)3-DM, suggesting that there is a fraction of maytansinoid loss from the SMCC-DM1 ADC, possibly through a thiol-maleimide dependent mechanism. Experiments on ex vivo stability confirm that some loss of maytansinoid from Ab-SMCC-DM1 conjugates can occur via thiol elimination, but at a slower rate than the corresponding rate of loss reported for thiol-maleimide links formed at thiols derived by reduction of endogenous cysteine residues in antibodies, consistent with expected differences in thiol-maleimide stability related to thiol pKa. These findings inform the design strategy for future ADCs.


Subject(s)
Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Lysine/chemistry , Maleimides/chemistry , Maytansine/chemistry , Animals , Drug Stability , Mice , Structure-Activity Relationship
9.
MAbs ; 8(3): 513-23, 2016.
Article in English | MEDLINE | ID: mdl-26752675

ABSTRACT

Antibody-drug conjugates (ADCs) are of great interest as targeted cancer therapeutics. Preparation of ADCs for early stage screening is constrained by purification and biochemical analysis techniques that necessitate burdensome quantities of antibody. Here we describe a method, developed for the maytansinoid class of ADCs, enabling parallel conjugation of antibodies in 96-well format. The method utilizes ∼ 100 µg of antibody per well and requires <5 µg of ADC for characterization. We demonstrate the capabilities of this system using model antibodies. We also provide multiple examples applying this method to early-stage screening of maytansinoid ADCs. The method can greatly increase the throughput with which candidate ADCs can be screened in cell-based assays, and may be more generally applicable to high-throughput preparation and screening of different types of protein conjugates.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Neoplasm/pharmacology , Immunoconjugates/pharmacology , Maytansine/pharmacology , Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neoplasm/immunology , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Humans , Immunoconjugates/immunology , Neoplasms/immunology
10.
Cancer Chemother Pharmacol ; 74(5): 969-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25186956

ABSTRACT

PURPOSE: Trastuzumab emtansine (T-DM1), an antibody-drug conjugate (ADC) comprised of trastuzumab linked to the antimitotic agent DM1, has shown promising results in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer. Investigations of the mechanisms of the action of ADCs, including T-DM1, have been primarily descriptive or semiquantitative. However, quantitative pharmacokinetic/pharmacodynamic (PK/PD) analysis may provide insights into their complex behavior. The analyses described herein applied PK/PD modeling to nonclinical studies of maytansinoid conjugates. METHODS: The maytansinoid conjugates T-DM1 and T-SPP-DM1, with thioether and disulfide linkers, respectively, were tested in mouse efficacy, PK, and tumor uptake studies. (3)[H]DM1-bearing ADCs were used to facilitate the quantitation of the ADCs in plasma, as well as ADC and ADC catabolites in tumors. Three mechanistic PK/PD models were used to characterize plasma ADC, tumor ADC, and tumor catabolite concentrations. Tumor catabolite concentrations were used to fit tumor response. Model parameters were estimated using R software and nonlinear least squares regression. RESULTS: Plasma ADC-associated DM1 concentrations of T-DM1 decreased more slowly than those of T-SPP-DM1, likely due to slower DM1 release. A comparison of the mechanistic models found that the best model allowed catabolism and catabolite exit rates to differ between ADCs, that T-DM1 exhibited both faster tumor catabolism and catabolite exit rate from tumors than T-SPP-DM1; findings inconsistent with expected behavior based on the physicochemical nature of the respective catabolites. Tumor catabolite concentrations adequately described tumor response with both ADCs showing similar potency. CONCLUSION: Mechanistic PK/PD studies described herein provided results that confirmed and challenged current hypotheses, and suggested new areas of investigation.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Maytansine/analogs & derivatives , Models, Biological , Neoplasms/drug therapy , Neoplasms/metabolism , Ado-Trastuzumab Emtansine , Algorithms , Animals , Antibodies, Monoclonal, Humanized/blood , Area Under Curve , Cell Line, Tumor , Female , Humans , Maytansine/blood , Maytansine/pharmacokinetics , Mice, Nude , Trastuzumab , Treatment Outcome , Tritium
11.
Blood ; 122(20): 3500-10, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24002446

ABSTRACT

CD37 has gathered renewed interest as a therapeutic target in non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia (CLL); however, CD37-directed antibody-drug conjugates (ADCs) have not been explored. Here, we identified a novel anti-CD37 antibody, K7153A, with potent in vitro activity against B-cell lines through multiple mechanisms including apoptosis induction, antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and complement-dependent cytotoxicity. The antibody was conjugated to the maytansinoid, DM1, a potent antimicrotubule agent, via the thioether linker, N-succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC), and the resulting ADC, IMGN529, retained the intrinsic antibody activities and showed enhanced cytotoxic activity from targeted payload delivery. In lymphoma cell lines, IMGN529 induced G2/M cell cycle arrest after internalization and lysosomal processing to lysine-N(ε)-SMCC-DM1 as the sole intracellular maytansinoid metabolite. IMGN529 was highly active against subcutaneous B-cell tumor xenografts in severe combined immunodeficient mice with comparable or better activity than rituximab, a combination of cyclophosphamide, vincristine, and prednisone, or bendamustine. In human blood cells, CD37 is expressed in B cells at similar levels as CD20, and IMGN529 resulted in potent and specific depletion of normal and CLL B cells. These results support evaluation of the CD37-targeted ADC, IMGN529, in clinical trials in patients with B-cell malignancies including NHL and CLL.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antigens, Neoplasm/immunology , B-Lymphocytes/drug effects , Immunotoxins/therapeutic use , Maytansine/analogs & derivatives , Molecular Targeted Therapy , Tetraspanins/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , B-Lymphocytes/pathology , Bendamustine Hydrochloride , Cell Line, Tumor/drug effects , Cyclophosphamide/administration & dosage , Cytotoxicity, Immunologic/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Immunotoxins/immunology , Immunotoxins/pharmacology , Maytansine/administration & dosage , Maytansine/pharmacology , Maytansine/therapeutic use , Mice , Mice, SCID , Nitrogen Mustard Compounds/therapeutic use , Prednisone/administration & dosage , Rituximab , Vincristine/administration & dosage , Xenograft Model Antitumor Assays
12.
AAPS J ; 14(4): 799-805, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22875610

ABSTRACT

The concept of treating cancer with antibody-drug conjugates (ADCs) has gained momentum with the favorable activity and safety of trastuzumab emtansine (T-DM1), SAR3419, and lorvotuzumab mertansine (IMGN901). All three ADCs utilize maytansinoid cell-killing agents which target tubulin and suppress microtubule dynamics. Each ADC utilizes a different optimized chemical linker to attach the maytansinoid to the antibody. Characterizing the absorption, distribution, metabolism, and excretion (ADME) of these ADCs in preclinical animal models is important to understanding their efficacy and safety profiles. The ADME properties of these ADCs in rodents were inferred from studies with radio-labeled ADCs prepared with nonbinding antibodies since T-DM1, SAR3419, IMGN901 all lack cross-reactivity with rodent antigens. For studies exploring tumor localization and activation in tumor-bearing mice, tritium-labeled T-DM1, SAR3419, and IMGN901 were utilized. The chemical nature of the linker was found to have a significant impact on the ADME properties of these ADCs-particularly on the plasma pharmacokinetics and observed catabolites in tumor and liver tissues. Despite these differences, T-DM1, SAR3419, and IMGN901 were all found to facilitate efficient deliveries of active maytansinoid catabolites to the tumor tissue in mouse xenograft models. In addition, all three ADCs were effectively detoxified during hepatobiliary elimination in rodents.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Maytansine/analogs & derivatives , Neoplasms/drug therapy , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Drug Evaluation, Preclinical , Humans , Maytansine/adverse effects , Maytansine/pharmacokinetics , Mice , Neoplasms/pathology , Tissue Distribution , Trastuzumab , Tubulin Modulators/adverse effects , Tubulin Modulators/pharmacokinetics , Xenograft Model Antitumor Assays
13.
Mol Cancer Ther ; 11(5): 1133-42, 2012 May.
Article in English | MEDLINE | ID: mdl-22408268

ABSTRACT

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate consisting of the anti-HER2 antibody trastuzumab linked via a nonreducible thioether linker to the maytansinoid antitubulin agent DM1. T-DM1 has shown favorable safety and efficacy in patients with HER2-positive metastatic breast cancer. In previous animal studies, T-DM1 exhibited better pharmacokinetics (PK) and slightly more efficacy than several disulfide-linked versions. The efficacy findings are unique, as other disulfide-linked antibody-drug conjugates (ADC) have shown greater efficacy than thioether-linked designs. To explore this further, the in vitro and in vivo activity, PK, and target cell activation of T-DM1 and the disulfide-linked T-SPP-DM1 were examined. Both ADCs showed high in vitro potency, with T-DM1 displaying greater potency in two of four breast cancer cell lines. In vitro target cell processing of T-DM1 and T-SPP-DM1 produced lysine-N(ε)-MCC-DM1, and lysine-N(ε)-SPP-DM1 and DM1, respectively; in vivo studies confirmed these results. The in vitro processing rates for the two conjugate to their respective catabolites were similar. In vivo, the potencies of the conjugates were similar, and T-SPP-DM1 had a faster plasma clearance than T-DM1. Slower T-DM1 clearance translated to higher overall tumor concentrations (conjugate plus catabolites), but unexpectedly, similar levels of tumor catabolite. These results indicate that, although the ADC linker can have clear impact on the PK and the chemical nature of the catabolites formed, both linkers seem to offer the same payload delivery to the tumor.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Maytansine/analogs & derivatives , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Maytansine/chemistry , Maytansine/metabolism , Maytansine/pharmacokinetics , Maytansine/pharmacology , Mice , Mice, Nude , Receptor, ErbB-2/metabolism , Trastuzumab , Xenograft Model Antitumor Assays
14.
J Med Chem ; 55(2): 766-82, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22148292

ABSTRACT

The synthesis and biological evaluation of phosphate prodrugs of analogues of 1 (CC-1065) and their conjugates with antibodies are described. The phosphate group on the 1,2,9,9a-tetrahydrocyclopropa[c]benz[e]indol-4-one (CBI) portion of the compounds confers enhanced solubility and stability in aqueous solutions. In the presence of phosphatases, these compounds convert into active DNA-alkylating agents. The synthesis of the prodrugs was achieved sequentially through coupling of CBI with a bis-indolyl moiety, followed by attachment of a thiol-containing linker, and conversion of the hydroxyl group of CBI into a phosphate prodrug. The linkers incorporated into the prodrugs enable conjugation to an antibody via either a stable disulfide or thioether bond, in aqueous buffer solutions containing as little as 5% organic cosolvent, resulting in exclusively monomeric and stable antibody-cytotoxic prodrug conjugates. Two disulfide-containing linkers differing in the degree of steric hindrance were used in antibody conjugates to test the effect of different rates of intracellular disulfide cleavage and effector release on biological activity. The prodrugs can be converted to the active cytotoxic compounds through the action of endogenous phosphatases. Antibody-prodrug conjugates displayed potent antigen-selective cytotoxic activity in vitro and antitumor activity in vivo.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents, Alkylating/chemical synthesis , Indoles/chemical synthesis , Organophosphorus Compounds/chemical synthesis , Prodrugs/chemical synthesis , Animals , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Drug Stability , Duocarmycins , Female , Humans , Indoles/chemistry , Indoles/pharmacology , Mice , Mice, SCID , Neoplasm Transplantation , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Solubility , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous
15.
J Org Chem ; 75(14): 4769-77, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20545375

ABSTRACT

Squalene synthase catalyzes the conversion of two molecules of (E,E)-farnesyl diphosphate to squalene via the cyclopropylcarbinyl intermediate, presqualene diphosphate (PSPP). Since this novel reaction constitutes the first committed step in sterol biosynthesis, there has been considerable interest and research on the stereochemistry and mechanism of the process and in the design of selective inhibitors of the enzyme. This paper reports the synthesis and characterization of five racemic and two enantiopure aziridine analogues of PSPP and the evaluation of their potencies as inhibitors of recombinant yeast squalene synthase. The key aziridine-2-methanol intermediates (6-OH, 7-OH, and 8-OH) were obtained by N-alkylations or by an N-acylation-reduction sequence of (+/-)-, (2R,3S)-, and (2S,3R)-2,3-aziridinofarnesol (9-OH) protected as tert-butyldimethylsilyl ethers. S(N)2 displacements of the corresponding methanesulfonates with pyrophosphate and methanediphosphonate anions afforded aziridine 2-methyl diphosphates and methanediphosphonates bearing N-undecyl, N-bishomogeranyl, and N-(alpha-methylene)bishomogeranyl substituents as mimics for the 2,6,10-trimethylundeca-2,5,9-trienyl side chain of PSPP. The 2R,3S diphosphate enantiomer bearing the N-bishomogeranyl substituent corresponding in absolute stereochemistry to PSPP proved to be the most potent inhibitor (IC(50) 1.17 +/- 0.08 muM in the presence of inorganic pyrophosphate), a value 4-fold less than that of its 2S,3R stereoisomer. The other aziridine analogues bearing the N-(alpha-methylene)bishomogeranyl and N-undecyl substituents, and the related methanediphosphonates, exhibited lower affinities for recombinant squalene synthase.


Subject(s)
Aziridines/chemistry , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Polyisoprenyl Phosphates/chemistry , Sesquiterpenes/chemistry , Squalene/chemistry , Catalysis , Farnesyl-Diphosphate Farnesyltransferase/chemistry , Farnesyl-Diphosphate Farnesyltransferase/metabolism , Kinetics , Molecular Structure , Stereoisomerism
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.
J Am Chem Soc ; 130(6): 1966-71, 2008 Feb 13.
Article in English | MEDLINE | ID: mdl-18198872

ABSTRACT

Four reactions--chain elongation, cyclopropanation, branching, and cyclobutanation--are used in nature to join isoprenoid units for construction of the carbon skeletons for over 55,000 naturally occurring isoprenoid compounds. Those molecules produced by chain elongation have head-to-tail (regular) carbon skeletons, while those from cyclopropanation, branching, or cyclobutanation have non-head-to-tail (irregular) skeletons. Although wild type enzymes have not been identified for the branching and cyclobutanation reactions, chimeric proteins constructed from farnesyl diphosphate synthase (chain elongation) and chrysanthemyl diphosphate synthase (cyclopropanation) catalyze all four of the known isoprenoid coupling reactions to give a mixture of geranyl diphosphate (chain elongation), chrysanthemyl diphosphate (cyclopropanation), lavandulyl diphosphate (branching), and maconelliyl and planococcyl diphosphate (cyclobutanation). Replacement of the hydrogen atoms at C1 or C2 or hydrogen atoms in the methyl groups of dimethylallyl diphosphate by deuterium alters the distribution of the cyclopropanation, branching, and cyclobutanation products through primary and secondary kinetic isotope effects on the partitioning steps of common carbocationic intermediates. These experiments establish the sequence in which the intermediates are formed and indicate that enzyme-mediated control of the carbocationic rearrangement and elimination steps determines the distribution of products.


Subject(s)
Cyclobutanes/metabolism , Cyclopropanes/metabolism , Terpenes/metabolism , Artemisia/enzymology , Cyclobutanes/chemistry , Cyclopropanes/chemistry , Geranyltranstransferase/chemistry , Geranyltranstransferase/metabolism , Molecular Conformation , Terpenes/chemistry
20.
Science ; 316(5821): 73-6, 2007 Apr 06.
Article in English | MEDLINE | ID: mdl-17412950

ABSTRACT

The carbon skeletons of over 55,000 naturally occurring isoprenoid compounds are constructed from four fundamental coupling reactions: chain elongation, cyclopropanation, branching, and cyclobutanation. Enzymes that catalyze chain elongation and cyclopropanation are well studied, whereas those that catalyze branching and cyclobutanation are unknown. We have catalyzed the four reactions with chimeric proteins generated by replacing segments of a chain-elongation enzyme with corresponding sequences from a cyclopropanation enzyme. Stereochemical and mechanistic considerations suggest that the four coupling enzymes could have evolved from a common ancestor through relatively small changes in the catalytic site.


Subject(s)
Geranyltranstransferase/metabolism , Terpenes/metabolism , Amino Acid Sequence , Artemisia/enzymology , Catalysis , Catalytic Domain , Chrysanthemum cinerariifolium/enzymology , Cyclopropanes/chemistry , Evolution, Molecular , Geranyltranstransferase/chemistry , Geranyltranstransferase/genetics , Kinetics , Molecular Conformation , Molecular Sequence Data , Molecular Structure , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Stereoisomerism , Terpenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...