Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
2.
J Ocul Pharmacol Ther ; 38(7): 471-480, 2022 09.
Article in English | MEDLINE | ID: mdl-35537481

ABSTRACT

Purpose: To characterize the ocular toxicity of an antibody-drug conjugate (ADC), depatuxizumab mafodotin (Depatux-m), in nonclinical species and to evaluate the effects of drug-antibody ratios (DARs), variations of the ADC construct, and potential methods for mitigation of the corneal toxicity. Depatux-m contains the potent cytotoxic agent monomethyl auristatin F as the ADC payload. Methods: Depatux-m was administered intravenously to cynomolgus monkeys at doses up to 30 mg/kg and to mice up to 100 mg/kg. Ocular toxicity was evaluated by clinical ophthalmic examinations and histopathology. Potential mitigation was tested through agents to block target engagement and multiple topical ophthalmic treatments (antioxidant, vasoconstrictor, tear stimulant). Results: Effects primarily involved corneal epithelium and were dose-dependent with respect to onset, severity, and time to reversal in both monkeys and mice. On slit lamp biomicroscopy, the initial effect in monkeys was superficial multifocal punctate opacities (granularity), which migrated axially and were followed by pigmentation and multifocal punctate fluorescein staining. Microscopically, findings were characterized by single-cell necrosis, pigmentation, disordered basilar layer, and thinning of the corneal epithelium. Increased toxicity was associated with a higher DAR or more stably attached linker. Treatment with agents to block target engagement did not affect toxicity, and none of the topical treatments was successful. Conclusions: The corneal findings observed were similar to the effects described in clinical trials with Depatux-m and other ADCs. Collectively, these studies and available literature support the hypothesis that ADC-mediated toxicity is driven primarily by mechanism of action of the payload.


Subject(s)
Immunoconjugates , Toxic Optic Neuropathy , Animals , Antibodies, Monoclonal, Humanized/toxicity , Immunoconjugates/toxicity , Mice
3.
Mol Cancer Ther ; 17(5): 1039-1050, 2018 05.
Article in English | MEDLINE | ID: mdl-29592882

ABSTRACT

Antiangiogenic therapy is a clinically validated modality in cancer treatment. To date, all approved antiangiogenic drugs primarily inhibit the VEGF pathway. Delta-like ligand 4 (DLL4) has been identified as a potential drug target in VEGF-independent angiogenesis and tumor-initiating cell (TIC) survival. A dual-specific biologic targeting both VEGF and DLL4 could be an attractive strategy to improve the effectiveness of anti-VEGF therapy. ABT-165 was uniquely engineered using a proprietary dual-variable domain immunoglobulin (DVD-Ig) technology based on its ability to bind and inhibit both DLL4 and VEGF. In vivo, ABT-165 induced significant tumor growth inhibition compared with either parental antibody treatment alone, due, in part, to the disruption of functional tumor vasculature. In combination with chemotherapy agents, ABT-165 also induced greater antitumor response and outperformed anti-VEGF treatment. ABT-165 displayed nonlinear pharmacokinetic profiles in cynomolgus monkeys, with an apparent terminal half-life > 5 days at a target saturation dose. In a GLP monkey toxicity study, ABT-165 was well-tolerated at doses up to 200 mg/kg with non-adverse treatment-related histopathology findings limited to the liver and thymus. In summary, ABT-165 represents a novel antiangiogenic strategy that potently inhibits both DLL4 and VEGF, demonstrating favorable in vivo efficacy, pharmacokinetic, and safety profiles in preclinical models. Given these preclinical attributes, ABT-165 has progressed to a phase I study. Mol Cancer Ther; 17(5); 1039-50. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Glioblastoma/drug therapy , Immunoglobulins/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Glioblastoma/metabolism , Glioblastoma/pathology , HT29 Cells , Humans , Immunoglobulins/metabolism , Immunologic Factors/metabolism , Immunologic Factors/pharmacokinetics , Immunologic Factors/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Macaca fascicularis/metabolism , Membrane Proteins/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
4.
Mol Cancer Ther ; 17(4): 795-805, 2018 04.
Article in English | MEDLINE | ID: mdl-29483208

ABSTRACT

Depatuxizumab mafodotin (depatux-m, ABT-414) is a tumor-selective antibody drug conjugate (ADC) comprised of the anti-EGFR antibody ABT-806 and the monomethyl auristatin F (MMAF) warhead. Depatux-m has demonstrated promising clinical activity in glioblastoma multiforme (GBM) patients and is currently being evaluated in clinical trials in first-line and recurrent GBM disease settings. Depatux-m responses have been restricted to patients with amplified EGFR, highlighting the need for therapies with activity against tumors with nonamplified EGFR overexpression. In addition, depatux-m dosing has been limited by corneal side effects common to MMAF conjugates. We hypothesized that a monomethyl auristatin E (MMAE) ADC utilizing an EGFR-targeting antibody with increased affinity may have broader utility against tumors with more modest EGFR overexpression while mitigating the risk of corneal side effects. We describe here preclinical characterization of ABBV-221, an EGFR-targeting ADC comprised of an affinity-matured ABT-806 conjugated to MMAE. ABBV-221 binds to a similar EGFR epitope as depatux-m and retains tumor selectivity with increased binding to EGFR-positive tumor cells and greater in vitro potency. ABBV-221 displays increased tumor uptake and antitumor activity against wild-type EGFR-positive xenografts with a greatly reduced incidence of corneal side effects relative to depatux-m. ABBV-221 has similar activity as depatux-m against an EGFR-amplified GBM patient derived xenograft (PDX) model and is highly effective alone and in combination with standard-of-care temozolomide in an EGFRvIII-positive GBM xenograft model. Based on these results, ABBV-221 has advanced to a phase I clinical trial in patients with advanced solid tumors associated with elevated levels of EGFR. Mol Cancer Ther; 17(4); 795-805. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Glioblastoma/drug therapy , Immunoconjugates/pharmacology , Oligopeptides/chemistry , Animals , Antibodies, Monoclonal, Humanized/chemistry , Apoptosis , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunoconjugates/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...