Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Bioconjug Chem ; 31(1): 93-103, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31747250

ABSTRACT

Antibody-drug conjugates have elicited great interest recently as targeted chemotherapies for cancer. Recent preclinical and clinical data have continued to raise questions about optimizing the design of these complex therapeutics. Biochemical methods for site-specific antibody conjugation have been a design feature of recent clinical ADCs, and preclinical reports suggest that site-specifically conjugated ADCs generically offer improved therapeutic indices (i.e., the fold difference between efficacious and maximum tolerated doses). Here we present the results of a systematic preclinical comparison of ADCs embodying the DNA-alkylating linker-payload DGN549 generated with both heterogeneous lysine-directed and site-specific cysteine-directed conjugation chemistries. Importantly, the catabolites generated by each ADC are the same regardless of the conjugation format. In two different model systems evaluated, the site-specific ADC showed a therapeutic index benefit. However, the therapeutic index benefit is different in each case: both show evidence of improved tolerability, though with different magnitudes, and in one case significant efficacy improvement is also observed. These results support our contention that conjugation chemistry of ADCs is best evaluated in the context of a particular antibody, target, and linker-payload, and ideally across multiple disease models.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Benzodiazepines/therapeutic use , Immunoconjugates/therapeutic use , Lysine/therapeutic use , Neoplasms/drug therapy , Oxindoles/therapeutic use , Animals , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Benzodiazepines/adverse effects , Benzodiazepines/chemistry , Benzodiazepines/pharmacokinetics , Cell Line, Tumor , Female , Humans , Immunoconjugates/adverse effects , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Lysine/adverse effects , Lysine/chemistry , Lysine/pharmacokinetics , Mice , Mice, SCID , Oxindoles/adverse effects , Oxindoles/chemistry , Oxindoles/pharmacokinetics , Therapeutic Index
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.
Bioorg Med Chem Lett ; 29(17): 2455-2458, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31350125

ABSTRACT

Antibody-drug conjugates (ADCs) incorporating potent indolinobenzodiazepine (IGN) DNA alkylators as the cytotoxic payload are currently undergoing clinical evaluation. The optimized design of these payloads consists of an unsymmetrical dimer possessing both an imine and an amine effectively eliminating DNA crosslinking and demonstrating improved tolerability in mice. Here we present an alternate approach to generating DNA alkylating ADCs by linking the IGN monomer with a biaryl system which has a high DNA binding affinity to potentially enhance tolerability. These BIA ADCs were found to be highly cytotoxic in vitro and demonstrated potent antitumor activity in vivo.


Subject(s)
Alkylating Agents/chemistry , Drug Design , Immunoconjugates/chemistry , Animals , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Cell Survival/drug effects , DNA/metabolism , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice , Mice, SCID , Neoplasms/drug therapy , Neoplasms/pathology , Structure-Activity Relationship , Transplantation, Heterologous
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.
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
6.
Proc Natl Acad Sci U S A ; 109(30): 11993-8, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22778432

ABSTRACT

Standard genetic approaches allow the production of protein composites by fusion of polypeptides in head-to-tail fashion. Some applications would benefit from constructions that are genetically impossible, such as the site-specific linkage of proteins via their N or C termini, when a remaining free terminus is required for biological activity. We developed a method for the production of N-to-N and C-to-C dimers, with full retention of the biological activity of both fusion partners and without inflicting chemical damage on the proteins to be joined. We use sortase A to install on the N or C terminus of proteins of interest the requisite modifications to execute a strain-promoted copper-free cycloaddition and show that the ensuing ligation proceeds efficiently. Applied here to protein-protein fusions, the method reported can be extended to connecting proteins with any entity of interest.


Subject(s)
Carbon/chemistry , Multiprotein Complexes/chemistry , Nitrogen/chemistry , Protein Engineering/methods , Aminoacyltransferases/metabolism , Animals , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Flow Cytometry , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred BALB C , Models, Chemical , Molecular Structure , Multiprotein Complexes/pharmacokinetics , Protein Multimerization
7.
Chembiochem ; 14(3): 343-52, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23335262

ABSTRACT

Protein ubiquitylation controls many cellular pathways, and timely removal of ubiquitin by deubiquitylating enzymes (DUBs) is essential to govern these different functions. To map endogenous expression of individual DUBs as well as that of any interacting proteins, we developed a catch-and-release ubiquitin probe. Ubiquitin was equipped with an activity-based warhead and a cleavable linker attached to a biotin affinity-handle through tandem site-specific modification, in which we combined intein chemistry with sortase-mediated ligation. The resulting probe is cell-impermeable and was therefore delivered to the cytosol of perfringolysin O (PFO)-permeabilized cells. This allowed us to retrieve and identify 34 DUBs and their interacting partners. We also noted the expression, in host cells infected with Chlamydia trachomatis, of two additional DUBs. Furthermore, we retrieved and identified chlamydial DUB1 (ChlaDUB1) and DUB2 (ChlaDUB2), demonstrating by experiment that ChlaDUB2, the presence and activity of which had not been detected in infected cells, is in fact expressed during the course of infection.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia Infections/metabolism , Endopeptidases/metabolism , Molecular Probes/metabolism , Ubiquitin/chemistry , Aminoacyltransferases/metabolism , Azo Compounds/chemistry , Bacterial Proteins/analysis , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Biotin/chemistry , Biotin/metabolism , Chlamydia Infections/pathology , Chlamydia trachomatis/enzymology , Cysteine Endopeptidases/metabolism , Endopeptidases/analysis , Endopeptidases/genetics , HEK293 Cells , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Humans , Hydrazones/chemistry , Levulinic Acids/chemistry , Molecular Probes/chemistry , Ubiquitin/metabolism , Ubiquitin-Specific Proteases , Ubiquitination
8.
Mol Cancer Ther ; 21(7): 1047-1059, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35511740

ABSTRACT

ADAM metallopeptidase domain 9 (ADAM9) is a member of the ADAM family of multifunctional, multidomain type 1 transmembrane proteins. ADAM9 is overexpressed in many cancers, including non-small cell lung, pancreatic, gastric, breast, ovarian, and colorectal cancer, but exhibits limited expression in normal tissues. A target-unbiased discovery platform based on intact tumor and progenitor cell immunizations, followed by an IHC screen, led to the identification of anti-ADAM9 antibodies with selective tumor-versus-normal tissue binding. Subsequent analysis revealed anti-ADAM9 antibodies were efficiently internalized and processed by tumor cells making ADAM9 an attractive target for antibody-drug conjugate (ADC) development. Here, we describe the preclinical evaluation of IMGC936, a novel ADC targeted against ADAM9. IMGC936 is comprised of a high-affinity humanized antibody site-specifically conjugated to DM21-C, a next-generation linker-payload that combines a maytansinoid microtubule-disrupting payload with a stable tripeptide linker, at a drug antibody ratio of approximately 2.0. In addition, the YTE mutation (M252Y/S254T/T256E) was introduced into the CH2 domain of the antibody Fc to maximize in vivo plasma half-life and exposure. IMGC936 exhibited cytotoxicity toward ADAM9-positive human tumor cell lines, as well as bystander killing, potent antitumor activity in human cell line-derived xenograft and patient-derived xenograft tumor models, and an acceptable safety profile in cynomolgus monkeys with favorable pharmacokinetic properties. Our preclinical data provide a strong scientific rationale for the further development of IMGC936 as a therapeutic candidate for the treatment of ADAM9-positive cancers. A first-in-human study of IMGC936 in patients with advanced solid tumors has been initiated (NCT04622774).


Subject(s)
Immunoconjugates , ADAM Proteins , Cell Line, Tumor , Heterografts , Humans , Immunoconjugates/chemistry , Membrane Proteins/genetics , Xenograft Model Antitumor Assays
9.
J Struct Biol ; 168(1): 125-36, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19406246

ABSTRACT

We present a universal mimetic approach of the prehairpin intermediate of gp41, which represents the active drug target for fusion inhibitors of HIV (human immunodeficiency virus) and SIV (simian immunodeficiency virus) based on membrane anchored lipopeptides. For this purpose, we have in situ coupled terminal cysteine-modified peptides originating from the NHR of SIV and HIV to a maleimide-functionalized DOPC bilayer and monitored the interactions with potential antagonists of the trimer-of-hairpin conformation C34 and T20 peptides by means of atomic force microscopy and ellipsometry. FT-IR analysis in conjugation with CD-spectroscopy of hydrated N36-lipopeptides, incorporated in multilamellar bilayer stacks was employed to investigate peptide conformation prior to antagonist binding. In contrast to solution studies substantial secondary structure formation of S-N36 after in situ coupling to the bilayer was found. We could show that S-N36-lipopeptide-aggregates in bilayers were selectively able to bind T20 or the corresponding C-peptides (C34) and similar results could be achieved by using H-N36 lipopeptides. It was found that T20 binding to coiled coil S-N36 lipopeptide assemblies was fully reversible at elevated temperatures, while T20 binds irreversibly to H-N36 bundles.


Subject(s)
HIV Envelope Protein gp41/chemistry , HIV/metabolism , Lipid Bilayers/chemistry , Lipopeptides/chemistry , Simian Immunodeficiency Virus/metabolism , Circular Dichroism , HIV Envelope Protein gp41/metabolism , HIV Fusion Inhibitors , Microscopy, Atomic Force , Models, Biological , Spectroscopy, Fourier Transform Infrared
10.
J Am Chem Soc ; 131(31): 10800-1, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19610623

ABSTRACT

The unique reactivity of two sortase enzymes, SrtA(staph) from Staphylococcus aureus and SrtA(strep) from Streptococcus pyogenes, is exploited for site-specific labeling of a single polypeptide with different labels at its N and C termini. SrtA(strep) is used to label the protein's C terminus at an LPXTG site with a fluorescently labeled dialanine nucleophile. Selective N-terminal labeling of proteins containing N-terminal glycine residues is achieved using SrtA(staph) and LPXT derivatives. The generality of N-terminal labeling with SrtA(staph) is demonstrated by near-quantitative labeling of multiple protein substrates with excellent site specificity.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Fluorescent Dyes/chemistry , Peptides/chemistry , Alanine , Amino Acid Sequence , Binding Sites , Staphylococcus aureus/enzymology , Streptococcus pyogenes/enzymology , Substrate Specificity
11.
ACS Med Chem Lett ; 10(8): 1193-1197, 2019 Aug 08.
Article in English | MEDLINE | ID: mdl-31413805

ABSTRACT

Antibody-drug conjugates (ADCs) that incorporate potent indolinobenzodiazepine DNA alkylators as the payload component are currently undergoing clinical evaluation. In one ADC design, the payload molecules are linked to the antibody through a peptidase-labile l-Ala-l-Ala linker. In order to determine the role of amino acid stereochemistry on antitumor activity and tolerability, we incorporated l- and d-alanyl groups in the dipeptide, synthesized all four diastereomers, and prepared and tested the corresponding ADCs. Results of our preclinical evaluation showed that the l-Ala-l-Ala configuration provided the ADC with the highest therapeutic index (antitumor activity vs toxicity).

12.
J Phys Chem B ; 112(28): 8250-6, 2008 Jul 17.
Article in English | MEDLINE | ID: mdl-18563929

ABSTRACT

Lipid bilayers consisting of lipids with terminally perfluoroalkylated chains have remarkable properties. They exhibit increased stability and phase-separated nanoscale patterns in mixtures with nonfluorinated lipids. In order to understand the bilayer properties that are responsible for this behavior, we have analyzed the structure of solid-supported bilayers composed of 1,2-dipalmitoyl- sn-glycero-3-phosphocholine (DPPC) and of a DPPC analogue with 6 terminal perfluorinated methylene units (F6-DPPC). Polarized attenuated total reflection Fourier-transform infrared spectroscopy indicates that for F6-DPPC, the tilt of the lipid acyl chains to the bilayer normal is increased to 39 degrees as compared to 21 degrees for native DPPC, for both lipids in the gel phase. This substantial increase of the tilt angle is responsible for a decrease of the bilayer thickness from 5.4 nm for DPPC to 4.5 nm for F6-DPPC, as revealed by temperature-controlled imaging ellipsometry on microstructured lipid bilayers and solution atomic force microscopy. During the main phase transition from the gel to the fluid phase, both the relative bilayer thickness change and the relative area change are substantially smaller for F6-DPPC than for DPPC. In light of these structural and thermotropic data, we propose a model in which the higher acyl-chain tilt angle in F6-DPPC is the result of a conformational rearrangement to minimize unfavorable fluorocarbon-hydrocarbon interactions in the center of the bilayer due to chain staggering.


Subject(s)
1,2-Dipalmitoylphosphatidylcholine/chemistry , Fluorine Compounds/chemistry , Lipid Bilayers/chemistry , Temperature , Microscopy, Atomic Force , Molecular Structure , Spectroscopy, Fourier Transform Infrared
13.
Blood Adv ; 2(8): 848-858, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29661755

ABSTRACT

The outlook for patients with refractory/relapsed acute myeloid leukemia (AML) remains poor, with conventional chemotherapeutic treatments often associated with unacceptable toxicities, including severe infections due to profound myelosuppression. Thus there exists an urgent need for more effective agents to treat AML that confer high therapeutic indices and favorable tolerability profiles. Because of its high expression on leukemic blast and stem cells compared with normal hematopoietic stem cells and progenitors, CD123 has emerged as a rational candidate for molecularly targeted therapeutic approaches in this disease. Here we describe the development and preclinical characterization of a CD123-targeting antibody-drug conjugate (ADC), IMGN632, that comprises a novel humanized anti-CD123 antibody G4723A linked to a recently reported DNA mono-alkylating payload of the indolinobenzodiazepine pseudodimer (IGN) class of cytotoxic compounds. The activity of IMGN632 was compared with X-ADC, the ADC utilizing the G4723A antibody linked to a DNA crosslinking IGN payload. With low picomolar potency, both ADCs reduced viability in AML cell lines and patient-derived samples in culture, irrespective of their multidrug resistance or disease status. However, X-ADC exposure was >40-fold more cytotoxic to the normal myeloid progenitors than IMGN632. Of particular note, IMGN632 demonstrated potent activity in all AML samples at concentrations well below levels that impacted normal bone marrow progenitors, suggesting the potential for efficacy in AML patients in the absence of or with limited myelosuppression. Furthermore, IMGN632 demonstrated robust antitumor efficacy in multiple AML xenograft models. Overall, these findings identify IMGN632 as a promising candidate for evaluation as a novel therapy in AML.


Subject(s)
Immunoconjugates/therapeutic use , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/drug therapy , Animals , Cell Line, Tumor , Cell Survival/drug effects , Hematopoietic Stem Cells/drug effects , Heterografts , Humans , Immunoconjugates/immunology , Mice , Neoplastic Stem Cells/drug effects , Tumor Cells, Cultured
14.
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
15.
Curr Opin Chem Biol ; 10(6): 576-83, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17055332

ABSTRACT

Chemical entities designed to noncovalently interact with predetermined partners have fashioned a new paradigm in chemical biology. Fluorocarbons are extremely promising as supramolecular synthons toward these objectives. Bioorthogonal noncovalent interactions provide a way to modulate self-assembled systems in environments where such control has hitherto not been possible. Fluorocarbons have now found applications in self-assembly as well as proteomics, biomolecule purification and in the creation of microarray platforms. Other self-assembly motifs with similar attributes might be exploited using the same general approach.


Subject(s)
Fluorocarbons/chemistry , Cell Adhesion , Proteomics
16.
Cancer Discov ; 7(9): 1030-1045, 2017 09.
Article in English | MEDLINE | ID: mdl-28526733

ABSTRACT

Despite an improving therapeutic landscape, significant challenges remain in treating the majority of patients with advanced ovarian or renal cancer. We identified the cell-cell adhesion molecule cadherin-6 (CDH6) as a lineage gene having significant differential expression in ovarian and kidney cancers. HKT288 is an optimized CDH6-targeting DM4-based antibody-drug conjugate (ADC) developed for the treatment of these diseases. Our study provides mechanistic evidence supporting the importance of linker choice for optimal antitumor activity and highlights CDH6 as an antigen for biotherapeutic development. To more robustly predict patient benefit of targeting CDH6, we incorporate a population-based patient-derived xenograft (PDX) clinical trial (PCT) to capture the heterogeneity of response across an unselected cohort of 30 models-a novel preclinical approach in ADC development. HKT288 induces durable tumor regressions of ovarian and renal cancer models in vivo, including 40% of models on the PCT, and features a preclinical safety profile supportive of progression toward clinical evaluation.Significance: We identify CDH6 as a target for biotherapeutics development and demonstrate how an integrated pharmacology strategy that incorporates mechanistic pharmacodynamics and toxicology studies provides a rich dataset for optimizing the therapeutic format. We highlight how a population-based PDX clinical trial and retrospective biomarker analysis can provide correlates of activity and response to guide initial patient selection for first-in-human trials of HKT288. Cancer Discov; 7(9); 1030-45. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 920.


Subject(s)
Antineoplastic Agents/therapeutic use , Cadherins/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cadherins/genetics , Cadherins/metabolism , Female , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Macaca fascicularis , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Rats , Xenograft Model Antitumor Assays
17.
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
19.
Integr Biol (Camb) ; 4(2): 220-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22200052

ABSTRACT

The number of life-threatening fungal infections has risen in immunocompromised patients, and identification of the rules that govern an appropriate immune response is essential to develop better diagnostics and targeted therapeutics. The outer cell wall component on pathogenic fungi consists of ß-1,3-glucan, and Dectin-1, a pattern recognition receptor present on the cell surface of innate immune cells, binds specifically to this carbohydrate. A barrier in understanding the exact immunological response to pathogen-derived carbohydrate epitopes is the presence of multiple types of carbohydrate moieties on fungal cell walls. To dissect the immunological mechanisms used to recognize pathogens, a system of "fungal like particles" was developed that consisted of polystyrene beads, which mimicked the three dimensional shape of the fungus, coated covalently with purified ß-1,3-glucan derived from Saccharomyces cerevisiae. The morphology of the ß-1,3-glucan layer was examined by immunofluorescence, flow cytometery, and immuno-transmission electron microscopy. The covalent linkages of the ß-1,3-glucan to the polystyrene surface were stable after subjecting the beads to detergents. By pre-treating ß-1,3-glucan beads with laminarinase, a specific ß-1,3-gluconase, the reactivity of the anti-ß-1,3-glucan antibody was abrogated in comparison to treatment with proteinase K indicating that the coating of these beads was predominantly ß-1,3-glucan. TNF-α was also measured by stimulating bone-marrow derived macrophages with the ß-1,3-glucan beads, and showed a dose dependent response compared to soluble ß-glucan, insoluble ß-1,3-glucan, uncoated beads, and soluble ß-1,3-glucan mixed with uncoated beads. Finally, ß-1,3-glucan beads were incubated with GFP-Dectin-1 expressing macrophages and imaged using confocal microscopy. ß-1,3-beads were taken up within minutes and retained Dectin-1 recruitment to the phagosome as compared to uncoated beads. These data describe a unique fungal-like particle system that will permit immunologists to probe the critical steps in early recognition of pathogen-derived fungal carbohydrate antigens by innate immune cells.


Subject(s)
Immunity, Innate/immunology , Lectins, C-Type/immunology , Saccharomyces cerevisiae/immunology , beta-Glucans/immunology , Animals , Cell Line , Cell Wall/immunology , Cell Wall/ultrastructure , Cellulases/metabolism , Flow Cytometry , Humans , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microspheres , Surface Properties , Tumor Necrosis Factor-alpha/immunology
20.
Biochemistry ; 46(5): 1402-11, 2007 Feb 06.
Article in English | MEDLINE | ID: mdl-17260970

ABSTRACT

The E2 protein of papillomavirus is the key regulator of viral transcription and replication. Dimerization, which takes place via its conserved C-terminal DNA-binding domain (DBD), is critical for these functions. The presence and conservation of two histidines (H290 and H320) at or near the dimer interface suggests the importance of their roles in protein structure and stability that was explored by mutating them to neutral alanine. The H290A mutant but not the H320A mutant showed a significant change in the secondary as well as tertiary structure, as monitored by far- and near-UV circular dichroism and fluorescence. We show that the wild-type DBD was more stable than either of the two histidine mutants at pH 7.5 but that the order of stability changed with pH, where, at pH 4.5, the H290A mutant was most stable. Although H290 is important for pH dependence of the stability, it is not critical for dimerization or folding. The determination of pKa values for the solvent-exposed histidine residues shows that the surface properties of the protein change with pH, suggesting different interactions that can be made by the protein in response to cellular acidification. Moreover, identification of residues crucial for E2 stability will help in the design of modified proteins with desired characteristics.


Subject(s)
DNA-Binding Proteins/chemistry , Histidine , Human papillomavirus 16/chemistry , Oncogene Proteins, Viral/chemistry , Amino Acid Substitution , Binding Sites , Conserved Sequence , Dimerization , Humans , Hydrogen-Ion Concentration , Protein Conformation , Protein Denaturation , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL