Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 27(2): 622-631, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33148666

ABSTRACT

PURPOSE: Mortality due to acute myeloid leukemia (AML) remains high, and the management of relapsed or refractory AML continues to be therapeutically challenging. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has provided a proof of concept for an ADC-based therapeutic for AML. Several other ADCs have since entered clinical development of AML, but have met with limited success. We sought to develop a next-generation ADC for AML with a wide therapeutic index (TI) that overcomes the shortcomings of previous generations of ADCs. EXPERIMENTAL DESIGN: We compared the TI of our novel CD33-targeted ADC platform with other currently available CD33-targeted ADCs in preclinical models of AML. Next, using this next-generation ADC platform, we performed a head-to-head comparison of two attractive AML antigens, CD33 and CD123. RESULTS: Our novel ADC platform offered improved safety and TI when compared with certain currently available ADC platforms in preclinical models of AML. Differentiation between the CD33- and CD123-targeted ADCs was observed in safety studies conducted in cynomolgus monkeys. The CD33-targeted ADC produced severe hematologic toxicity, whereas minimal hematologic toxicity was observed with the CD123-targeted ADC at the same doses and exposures. The improved toxicity profile of an ADC targeting CD123 over CD33 was consistent with the more restricted expression of CD123 in normal tissues. CONCLUSIONS: We optimized all components of ADC design (i.e., leukemia antigen, antibody, and linker-payload) to develop an ADC that has the potential to translate into an effective new therapy against AML.


Subject(s)
Gemtuzumab/therapeutic use , Immunoconjugates/therapeutic use , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Area Under Curve , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Gemtuzumab/immunology , Gemtuzumab/pharmacokinetics , HL-60 Cells , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Macaca fascicularis , Mice , Sialic Acid Binding Ig-like Lectin 3/immunology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
2.
Angew Chem Int Ed Engl ; 59(31): 12998-13003, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32285542

ABSTRACT

Preparative reactions that occur efficiently under dilute, buffered, aqueous conditions in the presence of biomolecules find application in ligation, peptide synthesis, and polynucleotide synthesis and sequencing. However, the identification of functional groups or reagents that are mutually reactive with one another, but unreactive with biopolymers and water, is challenging. Shown here are cobalt catalysts that react with alkenes under dilute, aqueous, buffered conditions and promote efficient cycloisomerization and formal Friedel-Crafts reactions. The constraining conditions of bioorthogonal chemistry are beneficial for reaction efficiency as superior conversion at low catalyst concentration is obtained and competent rates in dilute conditions are maintained. Efficiency at high dilution in the presence of buffer and nucleobases suggests that these reaction conditions may find broad application.


Subject(s)
Alkenes/chemistry , Water/chemistry , Catalysis , Cobalt/chemistry , Coordination Complexes/chemistry , Cyclization , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/chemical synthesis , Isomerism
3.
Mol Cancer Ther ; 18(11): 2008-2020, 2019 11.
Article in English | MEDLINE | ID: mdl-31434693

ABSTRACT

The restricted expression pattern of B-cell maturation antigen (BCMA) makes it an ideal tumor-associated antigen (TAA) for the treatment of myeloma. BCMA has been targeted by both CD3 bispecific antibody and antibody-drug conjugate (ADC) modalities, but a true comparison of modalities has yet to be performed. Here we utilized a single BCMA antibody to develop and characterize both a CD3 bispecific and 2 ADC formats (cleavable and noncleavable) and compared activity both in vitro and in vivo with the aim of generating an optimal therapeutic. Antibody affinity, but not epitope was influential in drug activity and hence a high-affinity BCMA antibody was selected. Both the bispecific and ADCs were potent in vitro and in vivo, causing dose-dependent cell killing of myeloma cell lines and tumor regression in orthotopic myeloma xenograft models. Primary patient cells were effectively lysed by both CD3 bispecific and ADCs, with the bispecific demonstrating improved potency, maximal cell killing, and consistency across patients. Safety was evaluated in cynomolgus monkey toxicity studies and both modalities were active based on on-target elimination of B lineage cells. Distinct nonclinical toxicity profiles were seen for the bispecific and ADC modalities. When taken together, results from this comparison of BCMA CD3 bispecific and ADC modalities suggest better efficacy and an improved toxicity profile might be achieved with the bispecific modality. This led to the advancement of a bispecific candidate into phase I clinical trials.


Subject(s)
Antibodies, Bispecific/administration & dosage , B-Cell Maturation Antigen/metabolism , CD3 Complex/immunology , Immunoconjugates/administration & dosage , Multiple Myeloma/drug therapy , Animals , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacology , Antibody Affinity , B-Cell Maturation Antigen/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/adverse effects , Immunoconjugates/pharmacology , Mice , Multiple Myeloma/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
4.
Bioorg Med Chem Lett ; 29(7): 943-947, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30655215

ABSTRACT

Novel neolymphostin-based antibody-drug conjugate (ADC) precursors were synthesized either through amide couplings between both cleavable and non-cleavable linkers and neolymphostin derivatives, or through Cu(I)-catalyzed acetylene-azide click cycloadditon between non-cleavable linkers and neolymphostin acetal derivatives. These precursors were site-specifically conjugated to cysteine mutant trastuzumab-A114C to provide neolymphostin-based ADCs. Preliminary in vitro data indicated that the corresponding ADCs were active against HER2-expressing tumor cell lines, thus providing a proof-of-concept for using neolymphostin as ADC-based anticancer agents.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Trastuzumab/pharmacology , Aminoquinolines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Mutation , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Proof of Concept Study , Pyrroles/chemical synthesis , Trastuzumab/genetics
5.
Proc Natl Acad Sci U S A ; 115(28): E6404-E6410, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29946037

ABSTRACT

DNA-encoded libraries (DEL)-based discovery platforms have recently been widely adopted in the pharmaceutical industry, mainly due to their powerful diversity and incredible number of molecules. In the two decades since their disclosure, great strides have been made to expand the toolbox of reaction modes that are compatible with the idiosyncratic aqueous, dilute, and DNA-sensitive parameters of this system. However, construction of highly important C(sp3)-C(sp3) linkages on DNA through cross-coupling remains unexplored. In this article, we describe a systematic approach to translating standard organic reactions to a DEL setting through the tactical combination of kinetic analysis and empirical screening with information captured from data mining. To exemplify this model, implementation of the Giese addition to forge high value C-C bonds on DNA was studied, which represents a radical-based synthesis in DEL.


Subject(s)
DNA/chemistry , Gene Library , Models, Molecular , Kinetics
6.
Mol Cancer Ther ; 17(1): 243-253, 2018 01.
Article in English | MEDLINE | ID: mdl-29054985

ABSTRACT

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) that has demonstrated clinical benefit for patients with HER2+ metastatic breast cancer; however, its clinical activity is limited by inherent or acquired drug resistance. The molecular mechanisms that drive clinical resistance to T-DM1, especially in HER2+ tumors, are not well understood. We used HER2+ cell lines to develop models of T-DM1 resistance using a cyclical dosing schema in which cells received T-DM1 in an "on-off" routine until a T-DM1-resistant population was generated. T-DM1-resistant N87 cells (N87-TM) were cross-resistant to a panel of trastuzumab-ADCs (T-ADCs) with non-cleavable-linked auristatins. N87-TM cells do not have a decrease in HER2 protein levels or an increase in drug transporter protein (e.g., MDR1) expression compared with parental N87 cells. Intriguingly, T-ADCs using auristatin payloads attached via an enzymatically cleavable linker overcome T-DM1 resistance in N87-TM cells. Importantly, N87-TM cells implanted into athymic mice formed T-DM1 refractory tumors that remain sensitive to T-ADCs with cleavable-linked auristatin payloads. Comparative proteomic profiling suggested enrichment in proteins that mediate caveolae formation and endocytosis in the N87-TM cells. Indeed, N87-TM cells internalize T-ADCs into intracellular caveolin-1 (CAV1)-positive puncta and alter their trafficking to the lysosome compared with N87 cells. T-DM1 colocalization into intracellular CAV1-positive puncta correlated with reduced response to T-DM1 in a panel of HER2+ cell lines. Together, these data suggest that caveolae-mediated endocytosis of T-DM1 may serve as a novel predictive biomarker for patient response to T-DM1. Mol Cancer Ther; 17(1); 243-53. ©2017 AACR.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Endocytosis/drug effects , Trastuzumab/therapeutic use , Animals , Antineoplastic Agents, Immunological/pharmacology , Caveolae , Drug Resistance, Neoplasm , Female , Humans , Male , Mice , Trastuzumab/pharmacology
7.
Bioconjug Chem ; 28(4): 1102-1114, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28151644

ABSTRACT

Antibody drug conjugates (ADCs) provide an efficacious and relatively safe means by which chemotherapeutic agents can be specifically targeted to cancer cells. In addition to the selection of antibody targets, ADCs offer a modular design that allows selection of ADC characteristics through the choice of linker chemistries, toxins, and conjugation sites. Many studies have indicated that release of toxins bound to antibodies via noncleavable linker chemistries relies on the internalization and intracellular trafficking of the ADC. While this can make noncleavable ADCs more stable in the serum, it can also result in lower efficacy when their respective targets are not internalized efficiently or are recycled back to the cell surface following internalization. Here, we show that a lysosomally targeted ADC against the protein APLP2 mediates cell killing, both in vitro and in vivo, more effectively than an ADC against Trop2, a protein with less efficient lysosomal targeting. We also engineered a bispecific ADC with one arm targeting HER2 for the purpose of directing the ADC to tumors, and the other arm targeting APLP2, whose purpose is to direct the ADC to lysosomes for toxin release. This proof-of-concept bispecific ADC demonstrates that this technology can be used to shift the intracellular trafficking of a constitutively recycled target by directing one arm of the antibody against a lysosomally delivered protein. Our data also show limitations of this approach and potential future directions for development.


Subject(s)
Drug Delivery Systems , Immunoconjugates/pharmacology , Lysosomes/metabolism , Transcytosis , Amyloid beta-Protein Precursor/immunology , Amyloid beta-Protein Precursor/therapeutic use , Animals , Antibodies, Bispecific/therapeutic use , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Immunoconjugates/metabolism , Mice, Nude , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/therapeutic use , Receptor, ErbB-2/immunology , Receptor, ErbB-2/therapeutic use
8.
Sci Transl Med ; 9(372)2017 01 11.
Article in English | MEDLINE | ID: mdl-28077676

ABSTRACT

Disease relapse after treatment is common in triple-negative breast cancer (TNBC), ovarian cancer (OVCA), and non-small cell lung cancer (NSCLC). Therapies that target tumor-initiating cells (TICs) should improve patient survival by eliminating the cells that can drive tumor recurrence and metastasis. We demonstrate that protein tyrosine kinase 7 (PTK7), a highly conserved but catalytically inactive receptor tyrosine kinase in the Wnt signaling pathway, is enriched on TICs in low-passage TNBC, OVCA, and NSCLC patient-derived xenografts (PDXs). To deliver a potent anticancer drug to PTK7-expressing TICs, we generated a targeted antibody-drug conjugate (ADC) composed of a humanized anti-PTK7 monoclonal antibody, a cleavable valine-citrulline-based linker, and Aur0101, an auristatin microtubule inhibitor. The PTK7-targeted ADC induced sustained tumor regressions and outperformed standard-of-care chemotherapy. Moreover, the ADC specifically reduced the frequency of TICs, as determined by serial transplantation experiments. In addition to reducing the TIC frequency, the PTK7-targeted ADC may have additional antitumor mechanisms of action, including the inhibition of angiogenesis and the stimulation of immune cells. Together, these preclinical data demonstrate the potential for the PTK7-targeted ADC to improve the long-term survival of cancer patients.


Subject(s)
Antibodies/therapeutic use , Cell Adhesion Molecules/chemistry , Immunoconjugates/therapeutic use , Neoplastic Stem Cells/drug effects , Receptor Protein-Tyrosine Kinases/chemistry , Aminobenzoates/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Clinical Trials as Topic , Female , Humans , Immunotherapy/methods , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Macaca fascicularis , Mice , Mice, Inbred NOD , Mice, SCID , Microtubules/chemistry , Neoplasm Recurrence, Local/drug therapy , Oligopeptides/therapeutic use , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Receptor Protein-Tyrosine Kinases/immunology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 15(11): 2698-2708, 2016 11.
Article in English | MEDLINE | ID: mdl-27582525

ABSTRACT

Trop-2, also known as TACSTD2, EGP-1, GA733-1, and M1S1, is frequently expressed on a variety of human carcinomas, and its expression is often associated with poor prognosis of the diseases. However, it is also present on the epithelium of several normal tissues. A comprehensively designed Trop-2-targeting antibody-drug conjugate (ADC), balancing both efficacy and toxicity, is therefore necessary to achieve clinical utility. To this end, we developed a cleavable Trop-2 ADC (RN927C) using a site-specific transglutaminase-mediated conjugation method and a proprietary microtubule inhibitor (MTI) linker-payload, PF-06380101. Robust in vitro cytotoxicity of RN927C was observed on a panel of Trop-2-expressing tumor cell lines, with IC50 generally in the subnanomolar range. As expected for an MTI-containing ADC, RN927C readily induced mitotic arrest of treated cells in vitro and in vivo, followed by subsequent cell death. The in vivo efficacy of RN927C was tested in multiple cell line and patient-derived xenograft tumor models, including pancreatic, lung, ovarian, and triple-negative breast tumor types. Single-dose administration of RN927C at 0.75 to 3 mg/kg was generally sufficient to induce sustained regression of Trop-2-expressing tumors and showed superior efficacy over standard treatment with paclitaxel or gemcitabine. Administration of RN927C in nonhuman primate toxicity studies resulted in target-mediated effects in skin and oral mucosa, consistent with Trop-2 expression in these epithelial tissues with minimal, non-dose limiting off-target toxicities. On the basis of the combined efficacy and safety results, RN927C is postulated to have a favorable therapeutic index for treatment of solid tumors. Mol Cancer Ther; 15(11); 2698-708. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Immunoconjugates/pharmacology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Stability , Female , Gene Expression , Humans , Immunoconjugates/chemistry , Lysosomes , Mice , Mitosis/drug effects , Mitosis/genetics , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Mol Cancer Ther ; 15(5): 958-70, 2016 05.
Article in English | MEDLINE | ID: mdl-26944918

ABSTRACT

The degree of stability of antibody-drug linkers in systemic circulation, and the rate of their intracellular processing within target cancer cells are among the key factors determining the efficacy of antibody-drug conjugates (ADC) in vivo Previous studies demonstrated the susceptibility of cleavable linkers, as well as auristatin-based payloads, to enzymatic cleavage in rodent plasma. Here, we identify Carboxylesterase 1C as the enzyme responsible for the extracellular hydrolysis of valine-citrulline-p-aminocarbamate (VC-PABC)-based linkers in mouse plasma. We further show that the activity of Carboxylesterase 1C towards VC-PABC-based linkers, and consequently the stability of ADCs in mouse plasma, can be effectively modulated by small chemical modifications to the linker. While the introduced modifications can protect the VC-PABC-based linkers from extracellular cleavage, they do not significantly alter the intracellular linker processing by the lysosomal protease Cathepsin B. The distinct substrate preference of the serum Carboxylesterase 1C offers the opportunity to modulate the extracellular stability of cleavable ADCs without diminishing the intracellular payload release required for ADC efficacy. Mol Cancer Ther; 15(5); 958-70. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Carbamates/chemistry , Citrulline/chemistry , Immunoconjugates/chemistry , Valine/chemistry , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biomarkers , Carboxylesterase/chemistry , Carboxylesterase/metabolism , Drug Design , Drug Stability , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Mice , Mice, Knockout , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Binding , Structure-Activity Relationship
11.
PLoS One ; 10(7): e0132282, 2015.
Article in English | MEDLINE | ID: mdl-26161543

ABSTRACT

The efficacy of an antibody-drug conjugate (ADC) is dependent on the properties of its linker-payload which must remain stable while in systemic circulation but undergo efficient processing upon internalization into target cells. Here, we examine the stability of a non-cleavable Amino-PEG6-based linker bearing the monomethyl auristatin D (MMAD) payload site-specifically conjugated at multiple positions on an antibody. Enzymatic conjugation with transglutaminase allows us to create a stable amide linkage that remains intact across all tested conjugation sites on the antibody, and provides us with an opportunity to examine the stability of the auristatin payload itself. We report a position-dependent degradation of the C terminus of MMAD in rodent plasma that has a detrimental effect on its potency. The MMAD cleavage can be eliminated by either modifying the C terminus of the toxin, or by selection of conjugation site. Both approaches result in improved stability and potency in vitro and in vivo. Furthermore, we show that the MMAD metabolism in mouse plasma is likely mediated by a serine-based hydrolase, appears much less pronounced in rat, and was not detected in cynomolgus monkey or human plasma. Clarifying these species differences and controlling toxin degradation to optimize ADC stability in rodents is essential to make the best ADC selection from preclinical models. The data presented here demonstrate that site selection and toxin susceptibility to mouse plasma degradation are important considerations in the design of non-cleavable ADCs, and further highlight the benefits of site-specific conjugation methods.


Subject(s)
Aminobenzoates/pharmacokinetics , Drug Carriers/pharmacokinetics , Oligopeptides/pharmacokinetics , Aminobenzoates/administration & dosage , Aminobenzoates/chemistry , Animals , Antibodies/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Stability , Female , HEK293 Cells , Humans , Macaca fascicularis , Mice, SCID , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Rats
13.
Bioconjug Chem ; 26(4): 650-9, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25643134

ABSTRACT

The systemic stability of the antibody-drug linker is crucial for delivery of an intact antibody-drug conjugate (ADC) to target-expressing tumors. Linkers stable in circulation but readily processed in the target cell are necessary for both safety and potency of the delivered conjugate. Here, we report a range of stabilities for an auristatin-based payload site-specifically attached through a cleavable valine-citrulline-p-aminobenzylcarbamate (VC-PABC) linker across various sites on an antibody. We demonstrate that the conjugation site plays an important role in determining VC-PABC linker stability in mouse plasma, and that the stability of the linker positively correlates with ADC cytotoxic potency both in vitro and in vivo. Furthermore, we show that the VC-PABC cleavage in mouse plasma is not mediated by Cathepsin B, the protease thought to be primarily responsible for linker processing in the lysosomal degradation pathway. Although the VC-PABC cleavage is not detected in primate plasma in vitro, linker stabilization in the mouse is an essential prerequisite for designing successful efficacy and safety studies in rodents during preclinical stages of ADC programs. The divergence of linker metabolism in mouse plasma and its intracellular cleavage offers an opportunity for linker optimization in the circulation without compromising its efficient payload release in the target cell.


Subject(s)
Aminobenzoates/chemistry , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Immunoconjugates/chemistry , Oligopeptides/chemistry , Pancreatic Neoplasms/drug therapy , Aminobenzoates/blood , Aminobenzoates/pharmacokinetics , Aminobenzoates/pharmacology , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Carbamates/chemistry , Cathepsin B/chemistry , Cathepsin B/metabolism , Cell Line, Tumor , Dipeptides/chemistry , Drug Delivery Systems/methods , Drug Stability , Female , Humans , Immunoconjugates/blood , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Mice , Mice, Nude , Models, Molecular , Oligopeptides/blood , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Structure-Activity Relationship , Xenograft Model Antitumor Assays
14.
Org Lett ; 16(16): 4146-9, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25069029

ABSTRACT

Reported here are procedures for a one-pot oxidation and rearrangement of propargylamines to synthesize enaminones, with supporting mechanistic studies. Also reported are the extended one-pot syntheses of pyrazoles, including celecoxib and various heterocyclic compounds.


Subject(s)
Pargyline/analogs & derivatives , Propylamines/chemistry , Pyrazoles/chemical synthesis , Sulfonamides/chemical synthesis , Catalysis , Celecoxib , Combinatorial Chemistry Techniques , Molecular Structure , Oxidation-Reduction , Pargyline/chemistry , Pyrazoles/chemistry , Sulfonamides/chemistry
15.
Proc Natl Acad Sci U S A ; 111(5): 1766-71, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24443552

ABSTRACT

Using an expanded genetic code, antibodies with site-specifically incorporated nonnative amino acids were produced in stable cell lines derived from a CHO cell line with titers over 1 g/L. Using anti-5T4 and anti-Her2 antibodies as model systems, site-specific antibody drug conjugates (NDCs) were produced, via oxime bond formation between ketones on the side chain of the incorporated nonnative amino acid and hydroxylamine functionalized monomethyl auristatin D with either protease-cleavable or noncleavable linkers. When noncleavable linkers were used, these conjugates were highly stable and displayed improved in vitro efficacy as well as in vivo efficacy and pharmacokinetic stability in rodent models relative to conventional antibody drug conjugates conjugated through either engineered surface-exposed or reduced interchain disulfide bond cysteine residues. The advantages of the oxime-bonded, site-specific NDCs were even more apparent when low-antigen-expressing (2+) target cell lines were used in the comparative studies. NDCs generated with protease-cleavable linkers demonstrated that the site of conjugation had a significant impact on the stability of these rationally designed prodrug linkers. In a single-dose rat toxicology study, a site-specific anti-Her2 NDC was well tolerated at dose levels up to 90 mg/kg. These experiments support the notion that chemically defined antibody conjugates can be synthesized in commercially relevant yields and can lead to antibody drug conjugates with improved properties relative to the heterogeneous conjugates formed by nonspecific chemical modification.


Subject(s)
Antibodies/metabolism , Immunoconjugates/metabolism , Pharmaceutical Preparations/chemical synthesis , Protein Engineering/methods , Animals , Antibodies/blood , Antibodies/chemistry , Antibodies/toxicity , Batch Cell Culture Techniques , CHO Cells , Cell Death/drug effects , Cell Line , Cricetinae , Cricetulus , Cysteine/metabolism , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/chemistry , Protein Stability/drug effects , Rats
16.
Chem Biol ; 20(2): 161-7, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23438745

ABSTRACT

Antibody drug conjugates (ADCs) are a therapeutic class offering promise for cancer therapy. The attachment of cytotoxic drugs to antibodies can result in an effective therapy with better safety potential than nontargeted cytotoxics. To understand the role of conjugation site, we developed an enzymatic method for site-specific antibody drug conjugation using microbial transglutaminase. This allowed us to attach diverse compounds at multiple positions and investigate how the site influences stability, toxicity, and efficacy. We show that the conjugation site has significant impact on ADC stability and pharmacokinetics in a species-dependent manner. These differences can be directly attributed to the position of the linkage rather than the chemical instability, as was observed with a maleimide linkage. With this method, it is possible to produce homogeneous ADCs and tune their properties to maximize the therapeutic window.


Subject(s)
Antibodies/chemistry , Antineoplastic Agents/chemistry , Immunoconjugates/chemistry , Animals , Antibodies/immunology , Half-Life , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/immunology , Mice , Neoplasms/drug therapy , Rats , Transglutaminases/metabolism , Tubulin Modulators/chemistry
17.
Mol Cancer Ther ; 12(1): 38-47, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23223830

ABSTRACT

Antibody-drug conjugates (ADC) represent a promising therapeutic modality for the clinical management of cancer. We sought to develop a novel ADC that targets 5T4, an oncofetal antigen expressed on tumor-initiating cells (TIC), which comprise the most aggressive cell population in the tumor. We optimized an anti-5T4 ADC (A1mcMMAF) by sulfydryl-based conjugation of the humanized A1 antibody to the tubulin inhibitor monomethylauristatin F (MMAF) via a maleimidocaproyl linker. A1mcMMAF exhibited potent in vivo antitumor activity in a variety of tumor models and induced long-term regressions for up to 100 days after the last dose. Strikingly, animals showed pathologic complete response in each model with doses as low as 3 mg antibody/kg dosed every 4 days. In a non-small cell lung cancer patient-derived xenograft model, in which 5T4 is preferentially expressed on the less differentiated tumor cells, A1mcMMAF treatment resulted in sustained tumor regressions and reduced TIC frequency. These results highlight the potential of ADCs that target the most aggressive cell populations within tumors, such as TICs. In exploratory safety studies, A1mcMMAF exhibited no overt toxicities when administered to cynomolgus monkeys at doses up to 10 mg antibody/kg/cycle × 2 and displayed a half-life of 5 days. The preclinical efficacy and safety data established a promising therapeutic index that supports clinical testing of A1mcMMAF.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Membrane Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/toxicity , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitory Concentration 50 , Macaca fascicularis , Male , Maximum Tolerated Dose , Membrane Glycoproteins/immunology , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Remission Induction , Tissue Distribution , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
18.
Bioorg Med Chem Lett ; 21(15): 4602-7, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21715165

ABSTRACT

An extension of our previously reported series of macrocyclic ortho-aminobenzamide Hsp90 inhibitors is reported. Addition of a second methyl group to the tether provided analogs that show increased potency in binding as well as cell-proliferation assays and, more importantly, are stable toward microsomes. We wish to disclose the discovery of a macrocycle which showed impressive biomarker activity 24-h post dosing and which demonstrated prolonged exposure in tumors. When studied in a lung cancer xenograft model, the compound demonstrated significant tumor size reduction.


Subject(s)
Antineoplastic Agents/chemistry , Benzamides/chemistry , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lung Neoplasms/drug therapy , Macrocyclic Compounds/chemistry , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Benzamides/pharmacokinetics , Benzamides/therapeutic use , Binding Sites , Biomarkers/metabolism , Drug Evaluation, Preclinical , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Nude , Microsomes, Liver/metabolism , Protein Structure, Tertiary , Rats , Transplantation, Heterologous
19.
Bioorg Med Chem Lett ; 21(12): 3627-31, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21605975

ABSTRACT

A novel series of macrocyclic ortho-aminobenzamide Hsp90 inhibitors is reported. In continuation of our research, heterocycle-containing tethers were explored with the intent to further improve potency and minimize hERG liabilities. This effort culminated in the discovery of compound 10, which efficiently suppressed proliferation of HCT116 and U87 cells. This compound showed prolonged Hsp90-inhibitory activity at least 24h post-administration consistent with elevated and prolonged exposure in the tumor. When studied in a xenograft model, the compound demonstrated significant suppression of tumor growth.


Subject(s)
Amines/chemical synthesis , Benzamides/chemical synthesis , Biomarkers, Tumor , Drug Discovery , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/pharmacology , Amines/chemistry , Amines/pharmacology , Animals , Benzamides/chemistry , Benzamides/pharmacology , Benzoquinones/chemical synthesis , Benzoquinones/chemistry , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Inhibitory Concentration 50 , Lactams, Macrocyclic/chemical synthesis , Lactams, Macrocyclic/chemistry , Lactams, Macrocyclic/pharmacology , Macrocyclic Compounds/chemistry , Mice , Models, Molecular , Molecular Structure , Neoplasms/drug therapy , Xenograft Model Antitumor Assays
20.
Bioorg Med Chem Lett ; 21(11): 3411-6, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21515049

ABSTRACT

A novel series of macrocyclic ortho-aminobenzamide Hsp90 inhibitors is reported. In continuation of our research in this area, macrocyclic amides and lactams were explored to reduce the risk of hERG liabilities. This effort culminated in the discovery of compound 38, which showed a favorable in vitro profile, and efficiently suppressed proliferation of several relevant cell lines. This compound showed prolonged Hsp90-inhibitory activity at least 24 h post-administration, consistent with elevated and prolonged exposure in the tumor.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers/metabolism , Drug Design , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/chemical synthesis , Lactams, Macrocyclic/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Humans , Inhibitory Concentration 50 , Lactams, Macrocyclic/chemistry , Models, Molecular , Molecular Structure , ortho-Aminobenzoates/chemical synthesis , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...