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
1.
Mol Cancer Ther ; 23(4): 541-551, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38354416

ABSTRACT

Although microtubule inhibitors (MTI) remain a therapeutically valuable payload option for antibody-drug conjugates (ADC), some cancers do not respond to MTI-based ADCs. Efforts to fill this therapeutic gap have led to a recent expansion of the ADC payload "toolbox" to include payloads with novel mechanisms of action such as topoisomerase inhibition and DNA cross-linking. We present here the development of a novel DNA mono-alkylator ADC platform that exhibits sustained tumor growth suppression at single doses in MTI-resistant tumors and is well tolerated in the rat upon repeat dosing. A phosphoramidate prodrug of the payload enables low ADC aggregation even at drug-to-antibody ratios of 5:1 while still delivering a bystander-capable payload that is effective in multidrug resistant (MDR)-overexpressing cell lines. The platform was comparable in xenograft studies to the clinical benchmark DNA mono-alkylator ADC platform DGN459 but with a significantly better tolerability profile in rats. Thus, the activity and tolerability profile of this new platform make it a viable option for the development of ADCs.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Humans , Rats , Animals , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Alkylating Agents , Neoplasms/drug therapy , DNA/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology
2.
Bioorg Med Chem Lett ; 72: 128876, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35788036

ABSTRACT

Pyrrolobenzodiazepine (PBD) dimers are well-known highly potent antibody drug conjugate (ADC) payloads. The corresponding PBD monomers, in contrast, have received much less attention from the ADC community. We prepared several novel polyamide-linked PBD monomers and evaluated their utility as ADC payloads. The unconjugated polyamide-PBD hybrids exhibited potent antiproliferative activity (IC50 range: 10-11-10-8 M) against a variety of HER2-expressing cancer cell lines. Several peptide-linked variants of the lead compound were prepared and conjugated to trastuzumab to afford ADCs with drug-to-antibody (DAR) ratios ranging from 3 to 5. The ADCs exhibited antigen-dependent cytotoxicity in vitro and potently suppressed tumor xenograft growth in vivo in a target-dependent manner. Moreover, the ADCs were well-tolerated in both mouse and rat. This work demonstrates for the first time that PBD polyamide hybrids can serve as effective ADC payloads.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Animals , Antineoplastic Agents/pharmacology , Benzodiazepines , Cell Line, Tumor , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice , Nylons/pharmacology , Pyrroles , Rats , Xenograft Model Antitumor Assays
3.
Mol Cancer Ther ; 20(5): 885-895, 2021 05.
Article in English | MEDLINE | ID: mdl-33722857

ABSTRACT

After significant effort over the last 30 years, antibody-drug conjugates (ADC) have recently gained momentum as a therapeutic modality, and nine ADCs have been approved by the FDA to date, with additional ADCs in late stages of development. Here, we introduce dolaflexin, a novel ADC technology that overcomes key limitations of the most common ADC platforms with two key features: a higher drug-to-antibody ratio and a novel auristatin with a controlled bystander effect. The novel, cell permeable payload, auristatin F-hydroxypropylamide, undergoes metabolic conversion to the highly potent, but less cell permeable auristatin F to balance the bystander effect through drug trapping within target cells. We conducted studies in mice, rats, and cynomolgus monkeys to complement in vitro characterization and contrasted the performance of dolaflexin with regard to antitumor activity, pharmacokinetic properties, and safety in comparison with the ADC platform utilized in the approved ADC ado-trastuzumab emtansine (T-DM1). A HER2-targeted dolaflexin ADC was shown to have a much lower threshold of antigen expression for potent cell killing in vitro, was effective in vivo in tumors with low HER2 expression, and induced tumor regressions in a xenograft model that is resistant to T-DM1.


Subject(s)
Immunoconjugates/therapeutic use , Oligopeptides/therapeutic use , Polymers/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Immunoconjugates/pharmacology , Mice , Mice, SCID , Oligopeptides/pharmacology , Polymers/pharmacology
4.
Mol Cancer Ther ; 20(5): 896-905, 2021 05.
Article in English | MEDLINE | ID: mdl-33722858

ABSTRACT

Target selection for antibody-drug conjugates (ADC) frequently focuses on identifying antigens with differential expression in tumor and normal tissue, to mitigate the risk of on-target toxicity. However, this strategy restricts the possible target space. SLC34A2/NaPi2b is a sodium phosphate transporter expressed in a variety of human tumors including lung and ovarian carcinoma, as well as the normal tissues from which these tumors arise. Previous clinical trials with a NaPi2b targeting MMAE-ADCs have shown objective durable responses. However, the protein-based biomarker assay developed for use in that study was unable to discern a statistically significant relationship between NaPi2b protein expression and the probability of response. XMT-1536 is a NaPi2b targeting ADC comprised of a unique humanized antibody conjugated with 10-15 auristatin F- hydroxypropylamide (AF-HPA) payload molecules via the Dolaflexin platform. AF-HPA is a cell-permeable, antimitotic compound that is slowly metabolized intratumorally to an active, very low-permeable metabolite, auristatin F (AF), resulting in controlled bystander killing. We describe the preclinical in vitro and in vivo antitumor effects of XMT-1536 in models of ovarian and lung adenocarcinoma. Pharmacokinetic analysis showed approximately proportional increases in exposure in rat and monkey. Systemic free AF-HPA and AF concentrations were observed to be low in all animal species. Finally, we describe a unique IHC reagent, generated from a chimeric construct of the therapeutic antibody, that was used to derive a target expression and efficacy relationship in a series of ovarian primary xenograft cancer models.


Subject(s)
Antigens, Neoplasm/metabolism , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Oligopeptides/therapeutic use , Polymers/therapeutic use , Animals , Female , Humans , Immunoconjugates/pharmacology , Mice , Mice, SCID , Oligopeptides/pharmacology , Polymers/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...