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










Publication year range
1.
Bioconjug Chem ; 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809610

ABSTRACT

Targeted protein degradation is an innovative therapeutic strategy to selectively eliminate disease-causing proteins. Exemplified by proteolysis-targeting chimeras (PROTACs), they have shown promise in overcoming drug resistance and targeting previously undruggable proteins. However, PROTACs face challenges, such as low oral bioavailability and limited selectivity. The recently published PROxAb Shuttle technology offers a solution enabling the targeted delivery of PROTACs using antibodies fused with PROTAC-binding domains derived from camelid single-domain antibodies (VHHs). Here, a modular approach to quickly generate PROxAb Shuttles by enzymatically coupling PROTAC-binding VHHs to off-the-shelf antibodies was developed. The resulting conjugates retained their target binding and internalization properties, and incubation with BRD4-targeting PROTACs resulted in formation of defined PROxAb-PROTAC complexes. These complexes selectively induced degradation of the BRD4 protein, resulting in cytotoxicity specifically to cells expressing the antibody's target. The chemoenzymatic approach described herein provides a versatile and efficient solution for generating antibody-VHH conjugates for targeted protein degradation applications, but it could also be used to combine antibodies and VHH binders to generate bispecific antibodies for further applications.

2.
Bioconjug Chem ; 34(12): 2221-2233, 2023 12 20.
Article in English | MEDLINE | ID: mdl-38054705

ABSTRACT

A crucial design feature for the therapeutic success of antibody-drug conjugates (ADCs) is the linker that connects the antibody with the drug. Linkers must be stable in circulation and efficiently release the drug inside the target cell, thereby having a fundamental impact on ADC pharmacokinetics and efficacy. The variety of enzymatically cleavable linkers applied in ADCs is limited, and some are believed to be associated with unwanted side effects due to the expression of cleavage-mediating enzymes in nonmalignant cells. Based on a bioinformatic screen of lysosomal enzymes, we identified α-l-iduronidase (IduA) as an interesting candidate for ADC linker cleavage because of its low expression in normal tissues and its overexpression in several tumor types. In the present study, we report a novel IduA-cleavable ADC linker using exatecan and duocarmycin as payloads. We showed the functionality of our linker system in cleavage assays using recombinant IduA or cell lysates and compared it to established ADC linkers. Subsequently, we coupled iduronide-exatecan via interchain cysteines or iduronide-duocarmycin via microbial transglutaminase (mTG) to an anti-CEACAM5 (aCEA5) antibody. The generated iduronide-exatecan ADC showed high serum stability and similar target-dependent tumor cell killing in the subnanomolar range but reduced toxicity on nonmalignant cells compared to an analogous cathepsin B-activatable valine-citrulline-exatecan ADC. Finally, in vivo antitumor activity could be demonstrated for an IduA-cleavable duocarmycin ADC. The presented results emphasize the potential of iduronide linkers for ADC development and represent a tool for further balancing out tumor selectivity and safety.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Immunoconjugates/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/metabolism , Iduronidase , Duocarmycins , Antibodies, Monoclonal , Cell Line, Tumor
3.
Drug Discov Today ; 27(10): 103311, 2022 10.
Article in English | MEDLINE | ID: mdl-35787480

ABSTRACT

Antibody-drug conjugates (ADCs) are on the brink of widespread use for the targeted treatment of cancer. ADCs manage the toxicity of drugs with unacceptable narrow therapeutic windows by guiding highly toxic compounds to the target cells, thereby sparing healthy cells. In this review, we describe approved ADCs and discuss their modes of action, together with medicinal chemical aspects, to evaluate the potential for improvement and to combat tumor-acquired resistance. A recent research focus has centered on the stimulation of immune responses to induce immunogenic cell death and the influence on the tumor microenvironment to enhance bystander effects.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Tumor Microenvironment
4.
J Pharm Sci ; 110(12): 3776-3785, 2021 12.
Article in English | MEDLINE | ID: mdl-34363839

ABSTRACT

Appropriate selection of conjugation sites and conjugation technologies is now widely accepted as crucial for the success of antibody-drug conjugates (ADCs). Herein, we present ADCs conjugated by different conjugation methods to different conjugation positions being systematically characterized by multiple in vitro assays as well as in vivo pharmacokinetic (PK) analyses in transgenic Tg276 mice. Conjugation to cysteines, genetically introduced at positions N325, L328, S239, D265, and S442, was compared to enzymatic conjugation via microbial transglutaminase (mTG) either to C-terminal light (LC) or heavy chain (HC) recognition motifs or to endogenous position Q295 of a native antibody. All conjugations yielded homogeneous DAR 2 ADCs with similar hydrophobicity, thermal stability, human neonatal Fc receptor (huFcRn) binding, and serum stability properties, but with pronounced differences in their PK profiles. mTG-conjugated ADC variants conjugated either to Q295 or to LC recognition motifs showed superior PK behavior. Within the panel of engineered cysteine variants L328 showed a similar PK profile compared to previously described S239 but superior PK compared to S442, D265, and N325. While all positions were first tested with trastuzumab, L328 and mTG LC were further evaluated with additional antibody scaffolds derived from clinically evaluated monoclonal antibodies (mAb). Based on PK analyses, this study confirms the newly described position L328 as favorable site for cysteine conjugation, comparable to the well-established engineered cysteine position S239, and emphasizes the favorable position Q295 of native antibodies and the tagged LC antibody variant for enzymatic conjugations via mTG. In addition, hemizygous Tg276 mice are evaluated as an adequate model for ADC pharmacokinetics, facilitating the selection of suitable ADC candidates early in the drug discovery process.


Subject(s)
Antineoplastic Agents, Immunological , Immunoconjugates , Animals , Antibodies, Monoclonal/chemistry , Antineoplastic Agents, Immunological/chemistry , Cysteine/chemistry , Immunoconjugates/chemistry , Mice , Trastuzumab/chemistry
6.
Bioconjug Chem ; 31(4): 1070-1076, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32134638

ABSTRACT

Site-specific bioconjugation technologies are frequently employed to generate homogeneous antibody-drug conjugates (ADCs) and are generally considered superior to stochastic approaches like lysine coupling. However, most of the technologies developed so far require undesired manipulation of the antibody sequence or its glycan structures. Herein, we report the successful engineering of microbial transglutaminase enabling efficient, site-specific conjugation of drug-linker constructs to position HC-Q295 of native, fully glycosylated IgG-type antibodies. ADCs generated via this approach demonstrate excellent stability in vitro as well as strong efficacy in vitro and in vivo. As it employs different drug-linker structures and several native antibodies, our study additionally proves the broad applicability of this approach.


Subject(s)
Immunoconjugates/metabolism , Protein Engineering , Transglutaminases/genetics , Transglutaminases/metabolism , Binding Sites , Streptomyces/enzymology , Transglutaminases/chemistry
7.
Nature ; 520(7549): 697-701, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25901683

ABSTRACT

TP53, a well-known tumour suppressor gene that encodes p53, is frequently inactivated by mutation or deletion in most human tumours. A tremendous effort has been made to restore p53 activity in cancer therapies. However, no effective p53-based therapy has been successfully translated into clinical cancer treatment owing to the complexity of p53 signalling. Here we demonstrate that genomic deletion of TP53 frequently encompasses essential neighbouring genes, rendering cancer cells with hemizygous TP53 deletion vulnerable to further suppression of such genes. POLR2A is identified as such a gene that is almost always co-deleted with TP53 in human cancers. It encodes the largest and catalytic subunit of the RNA polymerase II complex, which is specifically inhibited by α-amanitin. Our analysis of The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE) databases reveals that POLR2A expression levels are tightly correlated with its gene copy numbers in human colorectal cancer. Suppression of POLR2A with α-amanitin or small interfering RNAs selectively inhibits the proliferation, survival and tumorigenic potential of colorectal cancer cells with hemizygous TP53 loss in a p53-independent manner. Previous clinical applications of α-amanitin have been limited owing to its liver toxicity. However, we found that α-amanitin-based antibody-drug conjugates are highly effective therapeutic agents with reduced toxicity. Here we show that low doses of α-amanitin-conjugated anti-epithelial cell adhesion molecule (EpCAM) antibody lead to complete tumour regression in mouse models of human colorectal cancer with hemizygous deletion of POLR2A. We anticipate that inhibiting POLR2A will be a new therapeutic approach for human cancers containing such common genomic alterations.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Genes, p53/genetics , Tumor Suppressor Protein p53/deficiency , Alpha-Amanitin/adverse effects , Alpha-Amanitin/chemistry , Alpha-Amanitin/pharmacology , Alpha-Amanitin/therapeutic use , Animals , Antibodies/chemistry , Antibodies/immunology , Antigens, Neoplasm/immunology , Catalytic Domain , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Databases, Genetic , Disease Models, Animal , Epithelial Cell Adhesion Molecule , Female , Gene Deletion , Gene Dosage/genetics , Humans , Immunoconjugates/adverse effects , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Mice , Protein Subunits/chemistry , Protein Subunits/deficiency , Protein Subunits/genetics , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase II/chemistry , RNA Polymerase II/deficiency , RNA Polymerase II/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
8.
BMC Mol Biol ; 15: 7, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24694320

ABSTRACT

BACKGROUND: Many studies of the eukaryotic transcription mechanism and its regulation rely on in vitro assays. Conventional RNA polymerase II transcription assays are based on radioactive labelling of the newly synthesized RNA. Due to the inefficient in vitro transcription, the detection of the RNA involving purification and gel electrophoresis is laborious and not always quantitative. RESULTS: Herein, we describe a new, non-radioactive, robust and reproducible eukaryotic in vitro transcription assay that has been established in our laboratory. Upon transcription, the newly synthesized RNA is directly detected and quantified using the QuantiGene assay. Alternatively, the RNA can be purified and a primer extension followed by PCR detection or qPCR quantification can be performed. When applied to assess the activity of RNA polymerase II inhibitors, this new method allowed an accurate estimation of their relative potency. CONCLUSIONS: Our novel assay provides a non-radioactive alternative to a standard in vitro transcription assay that allows for sensitive detection and precise quantification of the newly transcribed, unlabelled RNA and is particularly useful for quantification of strong transcriptional inhibitors like α-amanitin. Moreover, the method can be easily adapted to quantify the reaction yield and the transcription efficiency of other eukaryotic in vitro systems, thus providing a complementary tool for the field of transcriptional research.


Subject(s)
Polymerase Chain Reaction/methods , RNA Polymerase II/genetics , Transcription, Genetic/genetics , Oligonucleotides/genetics , RNA/genetics
9.
Methods Mol Biol ; 1045: 51-70, 2013.
Article in English | MEDLINE | ID: mdl-23913141

ABSTRACT

Toxin payloads, or drugs, are the crucial components of therapeutic antibody-drug conjugates (ADCs). This review will give an introduction on the requirements that make a toxic compound suitable to be used in an antitumoral ADC and will summarize the structural and mechanistic features of four drug families that yielded promising results in preclinical and clinical studies.


Subject(s)
Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Pharmaceutical Preparations/chemistry , Animals , Antineoplastic Agents/chemistry , Humans , Immunoconjugates/chemistry
10.
Beilstein J Org Chem ; 8: 2072-84, 2012.
Article in English | MEDLINE | ID: mdl-23209542

ABSTRACT

Phallotoxins inhibit the dynamics of microfilaments in cells and lead to apoptosis. Due to poor cellular uptake these effects cannot be studied in live cells, even at millimolar toxin concentrations, nor can phalloidin be used for the elimination of tumor cells. Uptake is greatly enhanced by conjugation of phallotoxins to either lipophilic or polycationic moieties, such as oleic acid, polylysine, or Tat-peptide. These conjugates were lethally toxic for cells, e.g., mouse fibroblasts or Jurkat leukemia cells, in the micromolar range. Uptake into cells starts with the attachment of the toxin conjugates to the plasma membrane, followed by endocytosis and, in most cases, cleavage of the toxin from the carrier. Interestingly, the internalization rate of phalloidin into cells was also significantly increased by the fluorescent moiety tetramethylrhodaminyl, as well as by high molecular weight methoxy-polyethyleneglycol, two compounds unknown so far for their uptake-mediating activity. Conjugation to carriers as investigated in this work will allow the use of phallotoxins in experimental cell biology and possibly in tumor therapy. The findings obtained with phallotoxins could be applied also to the family of amatoxins, where α-amanitin, for example, when conjugated to oleic acid was more than 100-fold more toxic for cells than the native toxin. This suggests the possibility of a more general use of the moieties examined here to enhance the uptake of hydrophilic peptides, or drugs, into live cells.

11.
J Natl Cancer Inst ; 104(8): 622-34, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22457476

ABSTRACT

BACKGROUND: Human epithelial cell adhesion molecule (EpCAM) is overexpressed in many cancers. Anti-EpCAM antibodies have shown promise in preclinical studies, but showed no tumor regression in a recent phase II clinical trial. Therefore, we generated a novel anti-EpCAM antibody-drug conjugate and assessed whether it showed enhanced antitumor effects. METHODS: Chemical cross-linking was conducted to covalently conjugate α-amanitin, a toxin known to inhibit DNA transcription, with chiHEA125, a chimerized anti-EpCAM monoclonal antibody, to generate the antibody-drug conjugate α-amanitin-glutarate-chiHEA125 (chiHEA125-Ama). Antiproliferative activity of chiHEA125-Ama was tested in human pancreatic (BxPc-3 and Capan-1), colorectal (Colo205), breast (MCF-7), and bile duct (OZ) cancer cell lines in vitro using [(3)H]-thymidine incorporation assay. Antitumor activity of chiHEA125-Ama was assessed in vivo in immunocompromised mice bearing subcutaneous human BxPc-3 pancreatic carcinoma xenograft tumors (n = 66 mice). Cell proliferation and apoptosis were evaluated in xenograft tumors by immunohistochemistry. All statistical tests were two-sided. RESULTS: In all cell lines, chiHEA125-Ama reduced cell proliferation (mean half maximal inhibitory concentration [IC(50)] = 2.5 × 10(-10) to 5.4 × 10(-12) M). A single dose of chiHEA125-Ama inhibited BxPc-3 xenograft tumor growth (chiHEA125 [control, n = 4 mice] vs. chiHEA125-Ama [n = 6 mice], dose of 15 mg/kg with respect to IgG and 50 µg/kg with respect to α-amanitin, mean relative increase in tumor volume on day 16 = 884% vs. -79%, difference = 963%, 95% CI = 582% to 1344%, P = .019). Two higher doses of chiHEA125-Ama (100 µg/kg with respect to α-amanitin), administered 1 week apart (n = 10 mice per group), led to complete tumor regression in nine of 10 (90%) mice compared with chiHEA125, during the observation period of 16 days; increased apoptosis and reduced cell proliferation were observed in mice treated with chiHEA125-Ama. CONCLUSION: This preclinical study suggests that anti-EpCAM antibody conjugates with α-amanitin have the potential to be highly effective therapeutic agents for pancreatic carcinomas and various EpCAM-expressing malignancies.


Subject(s)
Alpha-Amanitin/pharmacology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Carcinoma/drug therapy , Cell Adhesion Molecules/metabolism , Enzyme Inhibitors/pharmacology , Immunoconjugates/pharmacology , Pancreatic Neoplasms/drug therapy , Alpha-Amanitin/immunology , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Antineoplastic Agents/immunology , Biomarkers, Tumor/immunology , Carcinoma/immunology , Carcinoma/pathology , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Chimera , Colonic Neoplasms/drug therapy , Enzyme Inhibitors/immunology , Epithelial Cell Adhesion Molecule , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/immunology , Immunohistochemistry , Liver/enzymology , Mice , Mice, Inbred NOD , Mice, SCID , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
12.
Stem Cells ; 27(4): 909-19, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19353519

ABSTRACT

Mesenchymal stem cells (MSC) display unique suppressive properties on T-cell immunity, thus representing an attractive vehicle for the treatment of conditions associated with harmful T-cell responses such as organ-specific autoimmunity and graft-versus-host disease. Toll-like receptors (TLR) are primarily expressed on antigen-presenting cells and recognize conserved pathogen-derived components. Ligation of TLR activates multiple innate and adaptive immune response pathways to eliminate and protect against invading pathogens. In this work, we show that TLR expressed on human bone marrow-derived MSC enhanced the immunosuppressive phenotype of MSC. Immunosuppression mediated by TLR was dependent on the production of immunosuppressive kynurenines by the tryptophan-degrading enzyme indoleamine-2,3-dioxygenase-1 (IDO1). Induction of IDO1 by TLR involved an autocrine interferon (IFN)-beta signaling loop, which was dependent on protein kinase R (PKR), but independent of IFN-gamma. These data define a new role for TLR in MSC immunobiology, which is to augment the immunosuppressive properties of MSC in the absence of IFN-gamma rather than inducing proinflammatory immune response pathways. PKR and IFN-beta play a central, previously unidentified role in orchestrating the production of immunosuppressive kynurenines by MSC.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-beta/immunology , Mesenchymal Stem Cells/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , eIF-2 Kinase/immunology , Blotting, Western , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-beta/metabolism , Kynurenine/biosynthesis , Kynurenine/immunology , Lymphocyte Culture Test, Mixed , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/metabolism , eIF-2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...