Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2033: 53-65, 2019.
Article in English | MEDLINE | ID: mdl-31332747

ABSTRACT

Random conjugation of chemical linkers to endogenous lysines or cysteines within an antibody yields a heterogeneous mixture of conjugates with various drug-to-antibody ratios. One approach for generating homogeneous antibody conjugates utilizes enzymatic transfer of payloads onto a specific glycan or amino acid residue. Microbial transglutaminase (MTG) is an enzyme that catalyzes the formation of a stable isopeptide bond between a glutamine and a lysine. We have previously identified and reported several sites throughout the antibody structure where an engineered lysine is sufficient for transfer of a glutamine-based substrate onto the antibody. Whereas other enzymatic transfer strategies typically require significant antibody engineering to either modify the N-glycans or introduce a multi-amino acid enzyme recognition site, the lower contextual specificity of MTG for lysines allows just a single lysine point mutation in an antibody to be efficiently transamidated. Here we describe the molecular positioning of these single engineered lysine residues and the conjugation conditions for producing homogeneous antibody conjugates exemplified using azido- and auristatin F-based acyl donor substrates.


Subject(s)
Glutamine/genetics , Immunoconjugates/genetics , Lysine/genetics , Protein Engineering/methods , Transglutaminases/chemistry , Antibodies/genetics , Antibodies/immunology , Cysteine/genetics , Humans , Immunoconjugates/immunology , Streptomyces/enzymology , Substrate Specificity
2.
Mol Cancer Ther ; 17(12): 2665-2675, 2018 12.
Article in English | MEDLINE | ID: mdl-30262588

ABSTRACT

Microtubule-targeting agents (MTA) have been investigated for many years as payloads for antibody-drug conjugates (ADC). In many cases, these ADCs have shown limited benefits due to lack of efficacy or significant toxicity, which has spurred continued investigation into novel MTA payloads for next-generation ADCs. In this study, we have developed ADCs using the MTA eribulin, a derivative of the macrocyclic polyether natural product halichondrin B, as a payload. Eribulin ADCs demonstrated in vitro potency and specificity using various linkers and two different conjugation approaches. MORAb-202 is an investigational agent that consists of the humanized anti-human folate receptor alpha (FRA) antibody farletuzumab conjugated via reduced interchain disulfide bonds to maleimido-PEG2-valine-citrulline-p-aminobenzylcarbamyl-eribulin at a drug-to-antibody ratio of 4.0. MORAb-202 displayed preferable biophysical properties and broad potency across a number of FRA-positive tumor cell lines as well as demonstrated improved specificity in vitro compared with farletuzumab conjugated with a number of other MTA payloads, including MMAE, MMAF, and the reducible maytansine linker-payload sulfo-SPDB-DM4. A single-dose administration of MORAb-202 in FRA-positive human tumor cell line xenograft and patient-derived tumor xenograft models elicited a robust and durable antitumor response. These data support further investigation of MORAb-202 as a potential new treatment modality for FRA-positive cancers, using the novel MTA eribulin as a payload.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Folate Receptor 1/antagonists & inhibitors , Furans/pharmacology , Immunoconjugates/pharmacology , Ketones/pharmacology , Microtubules/metabolism , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line, Tumor , Female , Folate Receptor 1/metabolism , Furans/chemistry , Humans , Immunoconjugates/chemistry , Ketones/chemistry , Mice, SCID , Polyethylene Glycols/chemistry , Treatment Outcome
3.
Bioconjug Chem ; 28(9): 2471-2484, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28820579

ABSTRACT

The use of microbial transglutaminase (MTG) to produce site-specific antibody-drug conjugates (ADCs) has thus far focused on the transamidation of engineered acyl donor glutamine residues in an antibody based on the hypothesis that the lower specificity of MTG for acyl acceptor lysines may result in the transamidation of multiple native lysine residues, thereby yielding heterogeneous products. We investigated the utilization of native IgG lysines as acyl acceptor sites for glutamine-based acyl donor substrates. Of the approximately 80 lysines in multiple recombinant IgG monoclonal antibodies (mAbs), none were transamidated. Because recombinant mAbs lack the C-terminal Lys447 due to cleavage by carboxypeptidase B in the production cell host, we explored whether blocking the cleavage of Lys447 by the addition of a C-terminal amino acid could result in transamidation of Lys447 by a variety of acyl donor substrates. MTG efficiently transamidated Lys447 in the presence of any nonacidic, nonproline amino acid residue at position 448. Lysine scanning mutagenesis throughout the antibody further revealed several transamidation sites in both the heavy- and light-chain constant regions. Additionally, scanning mutagenesis of the hinge region in a Fab' fragment revealed sites of transamidation that were not reactive in the context of the full-length mAb. Here, we demonstrate the utility of single lysine substitutions and the C-terminal Lys447 for engineering efficient acyl acceptor sites suitable for site-specific conjugation to a range of glutamine-based acyl donor substrates.


Subject(s)
Amino Acid Substitution , Immunoconjugates/metabolism , Immunoglobulin G/metabolism , Lysine/metabolism , Streptomyces/enzymology , Transglutaminases/metabolism , Amino Acid Sequence , HEK293 Cells , Humans , Immunoconjugates/chemistry , Immunoconjugates/genetics , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Lysine/chemistry , Lysine/genetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Protein Engineering , Substrate Specificity
4.
MAbs ; 9(6): 907-915, 2017.
Article in English | MEDLINE | ID: mdl-28541812

ABSTRACT

The prevailing techniques used to generate antibody-drug conjugates (ADCs) involve random conjugation of the linker-drug to multiple lysines or cysteines in the antibody. Engineering natural and non-natural amino acids into an antibody has been demonstrated to be an effective strategy to produce homogeneous ADC products with defined drug-to-antibody ratios. We recently reported an efficient residue-specific conjugation technology (RESPECT) where thiol-reactive payloads can be efficiently conjugated to a native unpaired cysteine in position 80 (C80) of rabbit light chains. Deimmunizing the rabbit variable domains through humanization is necessary to reduce the risk of anti-drug antibody responses in patients. However, we found that first-generation humanized RESPECT ADCs showed high levels of aggregation and low conjugation efficiency. We correlated these negative properties to the phenylalanine at position 83 present in most human variable kappa frameworks. When position 83 was substituted with selected amino acids, conjugation was restored and aggregation was reduced to levels similar to the chimeric ADC. This engineering strategy allows for development of second-generation humanized RESPECT ADCs with desirable biopharmaceutical properties.

5.
Cancer Biol Ther ; 18(5): 347-357, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28394698

ABSTRACT

The conjugation of toxins, dyes, peptides, or proteins to monoclonal antibodies is often performed via free thiol groups generated by either partial reduction methods or engineering free cysteine residues into the antibody sequence. Antibodies from the rabbit Oryctolagus cuniculus have an additional intrachain disulfide bond, whereby the light chain variable kappa domain is bridged to the constant kappa region between cysteine residues at positions 80 and 171, respectively. Chimerization of rabbit antibodies with human constant domains allows for the generation of a free thiol group at the light chain position 80 (C80) that can be used for site-specific conjugation. An efficient process for the purification and simultaneous removal of cysteinylation at the C80 site was developed. The unpaired C80 was shown to be efficiently conjugated using several different maleimido-based ligands. REsidue SPEcific Conjugation Technology (RESPECT) antibody-drug conjugates prepared using rabbit-human chimeric anti-human mesothelin rabbit antibodies and maleimido-PEG2-auristatin conjugated to C80 were shown to be highly potent and specific in vitro and effective in vivo in reduction of tumor growth in a highly aggressive mesothelin-expressing xenograft tumor model.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoconjugates/immunology , Neoplasms/drug therapy , Aminobenzoates/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/therapeutic use , Cysteine/chemistry , Cysteine/immunology , Humans , Immunoconjugates/therapeutic use , Mesothelin , Mice , Neoplasms/immunology , Oligopeptides/immunology , Rabbits , Trastuzumab/immunology , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem Lett ; 26(8): 2092-7, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26951751

ABSTRACT

(-)-Agelastatin A (AglA, 1), a member of the pyrrole-aminoimidazole marine alkaloid (PAI) family, possesses a unique tetracyclic structure and is one of the most potent anticancer PAIs isolated to date. In efforts to expand the SAR of these agents and delineate sites that tolerate modification while retaining activity, we synthesized several derivatives and tested their anticancer activity. The cytotoxic effects of these derivatives were measured against several cancer cell lines including cervical cancer (HeLa), epidermoid carcinoma (A431), ovarian (Igrov and Ovcar3), osteosarcoma (SJSA1), acute T cell leukemia (A3), epidermoid carcinoma (A431) in addition to primary human chronic lymphocytic leukemia (CLL) cells. New positions for modification of AglA and new substitutions were explored leading to novel derivatives, 14-chloro AglA (3) and 14-methyl AglA (12), that retained activity toward various cancer cell lines with decreased toxicity toward B- and T-cells. The SAR data informed the synthesis of a trifunctional probe bearing an alkyne and a diazirine potentially useful for cellular target identification.


Subject(s)
Alkaloids/chemistry , Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Molecular Probes/chemical synthesis , Molecular Probes/pharmacology , Oxazolidinones/chemistry , Oxazolidinones/pharmacology , Alkaloids/chemical synthesis , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Probes/chemistry , Molecular Structure , Oxazolidinones/chemical synthesis , Structure-Activity Relationship
7.
Cancer Biol Ther ; 14(11): 1032-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24025360

ABSTRACT

Because of its high mortality rate, ovarian cancer is a leading cause of death among women and a highly unmet medical need. New therapeutic agents that are effective and well tolerated are needed and cancer antigen-specific monoclonal antibodies that have direct pharmacologic effects or can stimulate immunological responses represent a promising class of agents for the treatment of this disease. The human folate receptor α (FOLR1), which is overexpressed in ovarian cancer but largely absent in normal tissues, appears to play a role in the transformed phenotype in ovarian cancer, cisplatin sensitivity, and growth in depleted folate conditions and therefore has potential as a target for passive immunotherapy. The anti-FOLR1 monoclonal antibody MORAb-003 (farletuzumab) was previously shown to elicit antibody dependent cellular cytotoxicity (ADCC) and inhibit tumor growth of human tumor xenografts in nude mice. Because of its promising preclinical profile, farletuzumab has been evaluated in clinical trials as a potential therapeutic agent for ovarian cancer. In this report, we demonstrated that farletuzumab's antitumor effect against an experimental model of ovarian cancer is mediated by its ADCC activity.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/pharmacology , Folate Receptor 1/metabolism , Ovarian Neoplasms/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Receptors, IgG/metabolism
8.
Nucl Med Biol ; 38(8): 1119-27, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21741258

ABSTRACT

INTRODUCTION: Radiolabeling of a monoclonal antibody (mAb) with a metallic radionuclide requires the conjugation of a bifunctional chelator to the mAb. The conjugation, however, can alter the physical and immunological properties of the mAb, consequently affecting its tumor-targeting pharmacokinetics. In this study, we investigated the effect of the amount of 2-(p-isothiocyanatobenzyl)-cyclohexyl-diethylenetriamine-pentaacetic acid (CHX-A″) conjugated to MORAb-009, a mAb directed against mesothelin, and the effect of MORAb dose on the biodistribution of (111)In-labeled MORAb-009. METHODS: We used nude mice bearing the A431/K5 tumor as a mesothelin-positive tumor model and the A431 tumor as a mesothelin-negative control. To find the optimal level of CHX-A″ conjugation, CHX-A″-MORAb-009 conjugates with 2.4, 3.5 and 5.5 CHX-A″ molecules were investigated. To investigate the effect of injected MORAb-009 dose on neutralizing the shed mesothelin in the circulation, biodistribution studies were performed after the intravenous co-injection of (111)In-labeled MORAb-009 (2.4 CHX-A″/MORAb-009) with three different doses: 0.2, 2 and 30 µg of MORAb-009. RESULTS: The tumor uptake in A431/K5 tumor was four times higher than that in A431 tumor, indicating that the tumor uptake in A431/K5 was mesothelin mediated. The conjugate with 5.5 CHX-A″ showed a lower isoelectric point (pI) and lower immunoreactivity (IR) than the 2.4 CHX-A″ conjugate. These differences were reflected in the biodistribution of the (111)In label. The (111)In-labeled MORAb-009 conjugated with 2.4 CHX-A″ produced higher tumor uptake and lower liver and spleen uptakes than the 5.5 CHX-A″ conjugate. The biodistribution studies also revealed that the tumor uptake was significantly affected by the injected MORAb-009 dose and tumor size. The 30-µg dose produced higher tumor uptake than the 0.2- and 2-µg doses, whereas the 30-µg dose produced lower liver and spleen uptakes than the 0.2-µg dose. CONCLUSION: This study demonstrates that the number of chelate conjugation and the injected dose are two important parameters to achieve high tumor and low non-target organ uptake of (111)In-labeled MORAb-009. This study also suggests that the injected dose of mAb could be individualized based on the tumor size or the blood level of shed antigen in a patient to achieve the ideal tumor-to-organ radioactivity ratios.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Chelating Agents/pharmacokinetics , GPI-Linked Proteins/metabolism , Indium Radioisotopes/pharmacokinetics , Isothiocyanates/pharmacokinetics , Neoplasms, Experimental/metabolism , Pentetic Acid/analogs & derivatives , Animals , Antibodies, Monoclonal/administration & dosage , Dose-Response Relationship, Radiation , Liver/metabolism , Mesothelin , Mice , Mice, Nude , Pentetic Acid/pharmacokinetics , Spleen/metabolism , Tissue Distribution
9.
J Surg Res ; 165(1): 38-45, 2011 Jan.
Article in English | MEDLINE | ID: mdl-19552923

ABSTRACT

BACKGROUND: Glucagon-like peptide-1 (GLP-1) has insulinomimetic, insulinotropic, and antiapoptotic properties that may make it a useful adjunct to reperfusion therapy for myocardial infarction (MI); however, GLP-1 has a short plasma half-life. Fusion of GLP-1 to human transferrin (GLP-1-Tf) significantly prolongs drug half-life. MATERIALS AND METHODS: We tested the ability of single dose GLP-1-Tf to limit myocardial ischemia (30 min)/reperfusion (180 min) injury in rabbits. Nineteen animals were untreated controls. The pre-ischemic group (n=10) was given 10mg/kg of GLP-1-Tf 12 h before ischemia. Immediately after reperfusion, the post-ischemic group (n=10) received GLP-1-Tf (10 mg/kg) and the Tf group (n=4) received transferrin alone. RESULTS: Infarct size as a percentage of the area at risk was 59.1% ± 1.3%, 45.7% ± 1.9%, 44.1% ± 3.3%, 59.7% ± 2.0% in the control group, pre-ischemic group, post-ischemic group, and Tf group, respectively (P<0.05 for both GLP-1-Tf treatments group versus control). GLP-1-Tf reduced the apoptotic index from 4.67% ± 0.40% in the control group to 3.15% ± 0.46% in the pre-ischemic group and to 2.66% ± 0.40% in the post-ischemic group (P<0.05 for both GLP-1-Tf treatments versus control). The size of the wall motion abnormality and ejection fraction was significantly improved in the post-ischemic group relative to the control group. Serum GLP-1 levels were 239.8 ± 25.7 µg/mL in the post-ischemic group, 27.9 ± 5.8 µg/mL in the pre-ischemic group, and undetectable in the control group. CONCLUSION: GLP-1-Tf limits myocardial reperfusion injury whether given prior to the onset of ischemia or given at reperfusion. GLP-1-Tf may also limit myocardial stunning at high serum levels of the drug.


Subject(s)
Glucagon-Like Peptide 1/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Transferrin/therapeutic use , Animals , Apoptosis/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Echocardiography , Glucagon-Like Peptide 1/blood , Hemodynamics/drug effects , Myocytes, Cardiac/pathology , Rabbits , Recombinant Fusion Proteins/therapeutic use
10.
Tumour Biol ; 27(3): 122-32, 2006.
Article in English | MEDLINE | ID: mdl-16612145

ABSTRACT

OBJECTIVE: Antibodies to CA 125 have been used to predict relapse of ovarian cancer, but have performed poorly as therapeutic agents. One rationale for this is antibody binding to circulating shed antigen. Our aim in this study was to develop antibodies to human CA 125 that have enhanced selectivity for the cell-associated form of the antigen. METHODS: Monoclonal antibodies were raised to a recombinant fragment of CA 125 that included sequence proximal to the putative membrane attachment site. Antibodies were characterized in terms of their binding site, affinity and selectivity for cell-associated CA 125. RESULTS: In assays using patient-derived CA 125, a subset of high-affinity (KD <5 nM) monoclonal antibodies demonstrated a 10- to greater than 200-fold increase in selectivity for cell-associated CA 125 when compared with controls. Based on mapping of the various monoclonal antibodies obtained, it was determined that shedding of CA 125 most likely occurs in the most C-terminal repeat domain. CONCLUSION: Results from competition analysis using patient-derived shed antigen predict that the antibodies described in this study may have significantly enhanced tumor-targeting properties when compared with existing antibodies to CA 125 in a tumor environment having high concentrations (>10,000 CA 125 units) of shed CA 125.


Subject(s)
Antibodies, Monoclonal/immunology , CA-125 Antigen/analysis , CA-125 Antigen/immunology , Neoplasm Recurrence, Local/immunology , Ovarian Neoplasms/immunology , Animals , Binding, Competitive , Blotting, Western , CA-125 Antigen/genetics , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Protein Structure, Tertiary , Recombinant Proteins/immunology
11.
Gynecol Oncol ; 102(3): 462-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16434088

ABSTRACT

OBJECTIVE: 776.1 is a monoclonal antibody prepared against the human ovarian cancer antigen CA 125 that demonstrates preferential binding to the cell-associated form of the antigen and has shown promising results as an yttrium-90-labeled antibody in pre-clinical studies examining the effects on tumor growth in a murine xenograft model of human ovarian cancer. The purpose of the present study was to examine the effects of combined therapy with [90Y-DOTA]776.1 and paclitaxel compared with monotherapy with either agent on the growth of OVCAR-3 xenografts in nude mice. METHODS: Mice bearing OVCAR-3 xenografts were treated with paclitaxel alone, 50 microCi or 150 microCi [90Y-DOTA]776.1 alone, or a combination of both treatments. Control groups were included which consisted of a nonspecific antibody, MOPC-21, labeled to a similar degree, administered as monotherapy or in combination with paclitaxel. The effects of administration of radioimmunotherapy prior to or following chemotherapy were also examined. RESULTS: Treatment with paclitaxel and [90Y-DOTA]776.1 had a synergistic anti-tumor effect on the growth of OVCAR-3 xenografts. Synergy was only observed when a tumor-specific antibody was used in radioimmunotherapy. While no difference in tumor growth was observed with order of dosing, reduced toxicity was seen when paclitaxel was administered prior to radioimmunotherapy. CONCLUSION: The combination of radioimmunotherapy using an anti-CA 125 monoclonal antibody and chemotherapy with paclitaxel was shown to be effective in an in vivo model of ovarian cancer and may hold promise as a treatment regimen for patients with ovarian cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CA-125 Antigen/immunology , Carcinoma/drug therapy , Carcinoma/radiotherapy , Ovarian Neoplasms/therapy , Paclitaxel/administration & dosage , Radioimmunotherapy , Yttrium Radioisotopes/therapeutic use , Animals , Female , Humans , Immunoconjugates/therapeutic use , Mice , Mice, Nude , Neoplasm Transplantation , Radiation-Sensitizing Agents/administration & dosage , Transplantation, Heterologous , Xenograft Model Antitumor Assays
12.
Cancer Biother Radiopharm ; 20(6): 589-602, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16398611

ABSTRACT

776.1 is a murine IgG1 monoclonal antibody to the human ovarian cancer antigen CA 125 that has the unique property of having a clear preference for binding to the cell-associated form of the antigen. We have examined the tumor localization properties and efficacy of 776.1 in a subcutaneous OVCAR-3 xenograft mouse model of human ovarian cancer. Biodistribution experiments using (125)I-labeled 776.1 demonstrated a peak uptake in tumors at 72 hours postinjection, with an average of 17.7% of injected dose per gram localized to the tumor. Little uptake in other organs was observed. Further experiments using CA 125-transfected syngeneic tumors, as well as an immunoprecipitation assay using human chimeric 776.1, both clearly demonstrated that 776.1 localizes to the tumor in a CA 125-dependent manner. DOTA-776.1 (1,4,7,10-tetraazacyclododecane-N,N',N",N'" tetraacetic acid-conjugated 776.1) was labeled with (90)Y and used in efficacy studies. [(90)Y-DOTA]776.1 at a single dose of 150 microCi was able to mediate efficient reduction of tumor growth, with regression observed in a subset of animals for a period ranging from 3 to 48 days, equivalent to 3 weekly administrations of cisplatin at 6 mg/kg. No significant regression was observed in groups receiving [(90)Y-DOTA]MOPC-21 control antibody at any dose. These results suggest that 776.1 may be a promising radioimmunotherapeutic agent for the treatment of human ovarian cancer.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Ovarian Neoplasms/radiotherapy , Radioimmunotherapy/methods , Animals , CA-125 Antigen/analysis , Female , Humans , Immunoglobulin G/metabolism , Immunoglobulin G/therapeutic use , Iodine Radioisotopes/pharmacokinetics , Iodine Radioisotopes/therapeutic use , Mice , Tissue Distribution , Transplantation, Heterologous , Yttrium Radioisotopes/pharmacokinetics , Yttrium Radioisotopes/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...