Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5842, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992037

ABSTRACT

Activating interferon responses with STING agonists (STINGa) is a current cancer immunotherapy strategy, and therapeutic modalities that enable tumor-targeted delivery via systemic administration could be beneficial. Here we demonstrate that tumor cell-directed STING agonist antibody-drug-conjugates (STINGa ADCs) activate STING in tumor cells and myeloid cells and induce anti-tumor innate immune responses in in vitro, in vivo (in female mice), and ex vivo tumor models. We show that the tumor cell-directed STINGa ADCs are internalized into myeloid cells by Fcγ-receptor-I in a tumor antigen-dependent manner. Systemic administration of STINGa ADCs in mice leads to STING activation in tumors, with increased anti-tumor activity and reduced serum cytokine elevations compared to a free STING agonist. Furthermore, STINGa ADCs induce type III interferons, which contribute to the anti-tumor activity by upregulating type I interferon and other key chemokines/cytokines. These findings reveal an important role for type III interferons in the anti-tumor activity elicited by STING agonism and provide rationale for the clinical development of tumor cell-directed STINGa ADCs.


Subject(s)
Immunity, Innate , Immunoconjugates , Interferons , Membrane Proteins , Animals , Membrane Proteins/agonists , Membrane Proteins/immunology , Immunity, Innate/drug effects , Female , Humans , Mice , Cell Line, Tumor , Immunoconjugates/pharmacology , Immunoconjugates/administration & dosage , Interferons/metabolism , Interferon Lambda , Neoplasms/immunology , Neoplasms/drug therapy , Interferon Type I/immunology , Cytokines/metabolism , Myeloid Cells/immunology , Myeloid Cells/drug effects , Immunotherapy/methods , Mice, Inbred C57BL , Receptors, IgG/agonists , Receptors, IgG/metabolism , Receptors, IgG/immunology
2.
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
3.
Mol Cancer Ther ; 23(1): 84-91, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37774393

ABSTRACT

Key defining attributes of an antibody-drug conjugate (ADC) include the choice of the targeting antibody, linker, payload, and the drug-to-antibody ratio (DAR). Historically, most ADC platforms have used the same DAR for all targets, regardless of target characteristics. However, recent studies and modeling suggest that the optimal DAR can depend on target expression level and intratumoral heterogeneity, target internalization and trafficking, and characteristics of the linker and payload. An ADC platform that enables DAR optimization could improve the success rate of clinical candidates. Here we report a systematic exploration of DAR across a wide range, by combining THIOMAB protein engineering technology with Dolasynthen, an auristatin-based platform with monomeric and trimeric variants. This approach enabled the generation of homogeneous, site-specific ADCs spanning a discrete range of DARs 2, 4, 6, 12, and 18 by conjugation of trastuzumab IgG1 THIOMAB constructs with 1, 2, or 3 engineered cysteines to monomeric or trimeric Dolasynthen. All ADCs had physicochemical properties that translated to excellent in vivo pharmacology. Following a single dose of ADCs in a HER2 xenograft model with moderate antigen expression, our data demonstrated comparable pharmacokinetics for the conjugates across all DARs and dose-dependent efficacy of all test articles. These results demonstrate that the Dolasynthen platform enables the generation of ADCs with a broad range of DAR values and with comparable physiochemical, pharmacologic, and pharmacokinetics profiles; thus, the Dolasynthen platform enables the empirical determination of the optimal DAR for a clinical candidate for a given target.


Subject(s)
Immunoconjugates , Humans , Immunoconjugates/chemistry , Xenograft Model Antitumor Assays , Trastuzumab/pharmacology , Trastuzumab/chemistry , Receptor, ErbB-2/metabolism , Cysteine
4.
J Med Chem ; 66(15): 10715-10733, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37486969

ABSTRACT

While STING agonists have proven to be effective preclinically as anti-tumor agents, these promising results have yet to be translated in the clinic. A STING agonist antibody-drug conjugate (ADC) could overcome current limitations by improving tumor accessibility, allowing for systemic administration as well as tumor-localized activation of STING for greater anti-tumor activity and better tolerability. In line with this effort, a STING agonist ADC platform was identified through systematic optimization of the payload, linker, and scaffold based on multiple factors including potency and specificity in both in vitro and in vivo evaluations. The platform employs a potent non-cyclic dinucleotide STING agonist, a cleavable ester-based linker, and a hydrophilic PEG8-bisglucamine scaffold. A tumor-targeted ADC built with the resulting STING agonist platform induced robust and durable anti-tumor activity and demonstrated high stability and favorable pharmacokinetics in nonclinical species.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Humans , Immunoconjugates/pharmacokinetics , Antibodies, Monoclonal , Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy
5.
Mol Cancer Ther ; 22(9): 999-1012, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37294948

ABSTRACT

Antibody-drug conjugates (ADC) achieve targeted drug delivery to a tumor and have demonstrated clinical success in many tumor types. The activity and safety profile of an ADC depends on its construction: antibody, payload, linker, and conjugation method, as well as the number of payload drugs per antibody [drug-to-antibody ratio (DAR)]. To allow for ADC optimization for a given target antigen, we developed Dolasynthen (DS), a novel ADC platform based on the payload auristatin hydroxypropylamide, that enables precise DAR-ranging and site-specific conjugation. We used the new platform to optimize an ADC that targets B7-H4 (VTCN1), an immune-suppressive protein that is overexpressed in breast, ovarian, and endometrial cancers. XMT-1660 is a site-specific DS DAR 6 ADC that induced complete tumor regressions in xenograft models of breast and ovarian cancer as well as in a syngeneic breast cancer model that is refractory to PD-1 immune checkpoint inhibition. In a panel of 28 breast cancer PDXs, XMT-1660 demonstrated activity that correlated with B7-H4 expression. XMT-1660 has recently entered clinical development in a phase I study (NCT05377996) in patients with cancer.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Immunoconjugates , Humans , Female , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antibodies , Cell Line, Tumor , Xenograft Model Antitumor Assays
6.
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
7.
Chembiochem ; 18(7): 613-617, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28140508

ABSTRACT

The enzyme-substrate complex is inherently transient, rendering its detection difficult. In our framework designed for bisubstrate systems-isotope-labeled, activity-based identification and tracking (IsoLAIT)-the common substrate, such as S-adenosyl-l-methionine (AdoMet) for methyltransferases, is replaced by an analogue (e.g., S-adenosyl-l-vinthionine) that, as a probe, creates a tightly bound [enzyme⋅substrate⋅probe] complex upon catalysis by thiopurine-S-methyltransferase (TPMT, EC 2.1.1.67). This persistent complex is then identified by native mass spectrometry from the cellular milieu without separation. Furthermore, the probe's isotope pattern flags even unknown substrates and enzymes. IsoLAIT is broadly applicable for other enzyme systems, particularly those catalyzing group transfer and with multiple substrates, such as glycosyltransferases and kinases.


Subject(s)
Methyltransferases/chemistry , Molecular Probes/analysis , S-Adenosylhomocysteine/analogs & derivatives , S-Adenosylmethionine/chemistry , Carbon Radioisotopes , Escherichia coli/metabolism , Isotope Labeling , Mass Spectrometry , Methyltransferases/metabolism , Molecular Probes/chemistry , Nitrogen Radioisotopes , S-Adenosylhomocysteine/chemistry , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Substrate Specificity
8.
J Am Chem Soc ; 138(9): 2877-80, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26901520

ABSTRACT

Identifying an enzyme's substrates is essential to understand its function, yet it remains challenging. A fundamental impediment is the transient interactions between an enzyme and its substrates. In contrast, tight binding is often observed for multisubstrate-adduct inhibitors due to synergistic interactions. Extending this venerable concept to enzyme-catalyzed in situ adduct formation, unknown substrates were affinity-captured by an S-adenosyl-methionine (AdoMet, SAM)-dependent methyltransferase (MTase). Specifically, the electrophilic methyl sulfonium (alkyl donor) in AdoMet is replaced with a vinyl sulfonium (Michael acceptor) in S-adenosyl-vinthionine (AdoVin). Via an addition reaction, AdoVin and the nucleophilic substrate form a covalent bisubstrate-adduct tightly complexed with thiopurine MTase (2.1.1.67). As such, an unknown substrate was readily identified from crude cell lysates. Moreover, this approach is applicable to other systems, even if the enzyme is unknown.


Subject(s)
Ethionine/analogs & derivatives , Methyltransferases/chemistry , Methyltransferases/metabolism , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Chromatography, High Pressure Liquid , Click Chemistry , Ethionine/chemistry , Ethionine/metabolism , Humans , Spectrophotometry, Ultraviolet , Substrate Specificity
9.
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
10.
J Sulphur Chem ; 36(2): 135-144, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-26005494

ABSTRACT

Selenium is an essential micronutrient in humans due to the important roles of the selenocysteine-containing selenoproteins. Organoselenium metabolites are generally found to be substrates for the biochemical pathways of their sulfur analogs, and the redox chemistry of selenomethionine and some other metabolites have been previously reported. We now report the first synthesis and characterization of Se-adenosylselenohomocysteine selenoxide (SeAHO) prepared via hydrogen peroxide oxidation of Se-adenosylselenohomocysteine (SeAH). The selenoxide SeAHO, in contrast to its corresponding sulfoxide S-adenosylhomocysteine (SAHO), can form hydrate, has an electrostatic interaction between the α-amino acid moiety and the highly polar selenoxide functional group, and readily oxidizes glutathione (GSH) and cysteine thiols.

SELECTION OF CITATIONS
SEARCH DETAIL
...