Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nature ; 554(7692): 378-381, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29414946

ABSTRACT

Using a functional model of breast cancer heterogeneity, we previously showed that clonal sub-populations proficient at generating circulating tumour cells were not all equally capable of forming metastases at secondary sites. A combination of differential expression and focused in vitro and in vivo RNA interference screens revealed candidate drivers of metastasis that discriminated metastatic clones. Among these, asparagine synthetase expression in a patient's primary tumour was most strongly correlated with later metastatic relapse. Here we show that asparagine bioavailability strongly influences metastatic potential. Limiting asparagine by knockdown of asparagine synthetase, treatment with l-asparaginase, or dietary asparagine restriction reduces metastasis without affecting growth of the primary tumour, whereas increased dietary asparagine or enforced asparagine synthetase expression promotes metastatic progression. Altering asparagine availability in vitro strongly influences invasive potential, which is correlated with an effect on proteins that promote the epithelial-to-mesenchymal transition. This provides at least one potential mechanism for how the bioavailability of a single amino acid could regulate metastatic progression.


Subject(s)
Asparagine/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neoplasm Metastasis/pathology , Animals , Asparaginase/metabolism , Asparaginase/therapeutic use , Asparagine/deficiency , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/metabolism , Biological Availability , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mice , Neoplasm Invasiveness/pathology , Prognosis , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , Reproducibility of Results
3.
Biol Chem ; 403(5-6): 495-508, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35073465

ABSTRACT

Driven by the potential to broaden the target space of conventional monospecific antibodies, the field of multi-specific antibody derivatives is growing rapidly. The production and screening of these artificial proteins entails a high combinatorial complexity. Antibody-domain exchange was previously shown to be a versatile strategy to produce bispecific antibodies in a robust and efficient manner. Here, we show that the domain exchange reaction to generate hybrid antibodies also functions under physiological conditions. Accordingly, we modified the exchange partners for use in therapeutic applications, in which two inactive prodrugs convert into a product with additional functionalities. We exemplarily show the feasibility for generating active T cell bispecific antibodies from two inactive prodrugs, which per se do not activate T cells alone. The two complementary prodrugs harbor antigen-targeting Fabs and non-functional anti-CD3 Fvs fused to IgG-CH3 domains engineered to drive chain-exchange reactions between them. Importantly, Prodrug-Activating Chain Exchange (PACE) could be an attractive option to conditionally activate therapeutics at the target site. Several examples are provided that demonstrate the efficacy of PACE as a new principle of cancer immunotherapy in vitro and in a human xenograft model.


Subject(s)
Antibodies, Bispecific , Prodrugs , Humans , Immunotherapy , Prodrugs/pharmacology , T-Lymphocytes
4.
Nature ; 520(7547): 358-62, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25855289

ABSTRACT

Cancer metastasis requires that primary tumour cells evolve the capacity to intravasate into the lymphatic system or vasculature, and extravasate into and colonize secondary sites. Others have demonstrated that individual cells within complex populations show heterogeneity in their capacity to form secondary lesions. Here we develop a polyclonal mouse model of breast tumour heterogeneity, and show that distinct clones within a mixed population display specialization, for example, dominating the primary tumour, contributing to metastatic populations, or showing tropism for entering the lymphatic or vasculature systems. We correlate these stable properties to distinct gene expression profiles. Those clones that efficiently enter the vasculature express two secreted proteins, Serpine2 and Slpi, which were necessary and sufficient to program these cells for vascular mimicry. Our data indicate that these proteins not only drive the formation of extravascular networks but also ensure their perfusion by acting as anticoagulants. We propose that vascular mimicry drives the ability of some breast tumour cells to contribute to distant metastases while simultaneously satisfying a critical need of the primary tumour to be fed by the vasculature. Enforced expression of SERPINE2 and SLPI in human breast cancer cell lines also programmed them for vascular mimicry, and SERPINE2 and SLPI were overexpressed preferentially in human patients that had lung-metastatic relapse. Thus, these two secreted proteins, and the phenotype they promote, may be broadly relevant as drivers of metastatic progression in human cancer.


Subject(s)
Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Endothelium, Vascular/pathology , Neoplasm Metastasis/pathology , Animals , Anticoagulants/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Clone Cells/metabolism , Clone Cells/pathology , Disease Models, Animal , Disease Progression , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression Profiling , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis/genetics , Recurrence , Secretory Leukocyte Peptidase Inhibitor/metabolism , Sequence Analysis, DNA , Serpin E2/metabolism
5.
Biol Chem ; 400(3): 343-350, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30763031

ABSTRACT

A novel bispecific antibody format was applied to generate T cell-engaging antibodies. The TriFab format is a trivalent IgG-shaped entity composed of two Fab arms that bind to antigens on the surface of tumor cells, which are linked via flexible peptides to a CD3 binding moiety that replaces the CH2 domains of conventional IgGs. The distinctive feature of these T cell recruiting bispecifics is that their CD3 variable regions are incorporated between domains, rather than N- or C-terminally fused to an Fc or antibody fragments. T cell recruiting TriFabs resemble in size and shape, are expressed and show biophysical properties similar to regular IgGs. Transmission electron microscopy (TEM) demonstrates high flexibility of the cell surface binding arms as well as target antigen accessibility of the interspersed CD3 binding domain. Functional co-culturing assays of peripheral blood mononuclear cells (PBMCs) and different tumor cell lines (MCF7 and A431) revealed a dose-dependent T cell-mediated cytotoxicity that was induced by the TriFabs targeting either LeY or EGFR cell surface antigens.


Subject(s)
Antibodies, Bispecific/immunology , Immunoglobulin G/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Cell Survival/immunology , Coculture Techniques , Cytokines/biosynthesis , HEK293 Cells , Humans , Leukocytes, Mononuclear/immunology , MCF-7 Cells , Microscopy, Electron, Transmission , Neoplasms/pathology , Tumor Cells, Cultured
6.
MAbs ; 15(1): 2245111, 2023.
Article in English | MEDLINE | ID: mdl-37608616

ABSTRACT

Antibody-cytokine fusions targeted against tumor-associated antigens (TAAs) are promising cancer immunotherapy agents, with many such molecules currently undergoing clinical trials. However, due to the limited number of tumor-specific targets, on-target off-tumor effects can lead to systemic toxicity. Additionally, targeted cytokines can be scavenged by cytokine receptors on peripheral cells, decreasing tumor penetration. This study aims at overcoming these issues by engineering a platform for targeted conditionally active type I cytokines. Building on our previously reported PACE (Prodrug-Activating Chain Exchange) platform, we split the type I cytokine interleukin-4 (IL-4) to create two inactive IL-4 prodrugs, and fused these split IL-4 counterparts to the C-termini of antibody-like molecules that undergo proximity-induced chain exchange. In doing so, we developed IL-4 prodrugs that preferentially reconstitute into active IL-4 on target cells. We demonstrate that pre-assembled split IL-4 (without additional inactivation) retains activity and present two different strategies of splitting and inactivating IL-4. Using an IL-4 responsive cell-line, we show that IL-4 prodrugs are targeted to TAAs on target cells and regain activity upon chain exchange, primarily in a cis-activation setting. Furthermore, we demonstrate that split IL-4 complementation is also possible in a trans-activation setting, which opens up the possibility for activation of immune cells in the tumor vicinity. We demonstrate that targeted on-cell prodrug conversion is more efficient than nonspecific activation in-solution. Due to the structural similarity between IL-4 and other type I cytokines relevant in cancer immunotherapy such as IL-2, IL-15, and IL-21, cytokine-PACE may be expanded to develop a variety of targeted conditionally active cytokines for cancer immunotherapy.


Subject(s)
Neoplasms , Prodrugs , Humans , Cytokines , Interleukin-4 , Prodrugs/pharmacology , Neoplasms/therapy , Antigens, Neoplasm , Antibodies , Immunotherapy
7.
Comput Struct Biotechnol J ; 18: 1221-1227, 2020.
Article in English | MEDLINE | ID: mdl-32542108

ABSTRACT

Multispecific antibodies can be generated in different formats. More than two decades of R&D in the field of bispecific antibody engineering revealed that the design and choice of format can have a profound impact on the antibody functionality. This holds in particular true for entities that elicit (inter-)cellular processes such as receptor activation, receptor internalization, receptor clustering or the formation of immunological synapses between two cells. This review covers design parameters that influence the functionality of multispecific formats, with particular focus on T cell-recruiting bispecific antibodies. We describe formats that display the same size and domain sequences but a varying geometry. The structural composition of (artificial) immune synapses is reviewed and allows conclusions why some formats that share size and domain composition are more effective than others. To support the statement that the geometry matters, we present a recently designed antibody format that is characterized by its compact shape. The TriFab-Contorsbody consists of two tumor cell-targeting entities and one moiety for T cell recruitment. The unique barrel-like shape provides a 35-fold increase in potency compared to an IgG-like molecule with identical domain sequences.

8.
MAbs ; 11(8): 1402-1414, 2019.
Article in English | MEDLINE | ID: mdl-31526159

ABSTRACT

High specificity accompanied with the ability to recruit immune cells has made recombinant therapeutic antibodies an integral part of drug development. Here we present a generic approach to generate two novel IgG-derived antibody formats that are based on a modification of the CrossMab technology. MoAbs harbor two heavy chains (HCs) resulting in one binding entity and one fragment crystallizable region (Fc), whereas DuoMabs are composed of four HCs harboring two binding entities and two Fc regions linked at a disulfide-bridged hinge. The latter bivalent format is characterized by avidity-enhanced target cell binding while simultaneously increasing the 'Fc-load' on the surface. DuoMabs were shown to be producible in high yield and purity and bind to surface cells with affinities comparable to IgGs. The increased Fc load directed at the surface of target cells by DuoMabs modulates their antibody-dependent cell-mediated cytotoxicity competency toward target cells, making them attractive for applications that require or are modulated by FcR interactions.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Antibodies, Bispecific/chemistry , Antibodies, Monoclonal/chemistry , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry
9.
Cancer Genomics Proteomics ; 15(1): 17-39, 2018.
Article in English | MEDLINE | ID: mdl-29275360

ABSTRACT

Despite the approval of several molecular therapies in the last years, breast cancer-associated death ranks as the second highest in women. This is due to metastatic disease, which represents a challenge for treatment. A better understanding of the molecular mechanisms of metastasis is, therefore, of paramount importance. In this review we summarize the role of micro RNAs (miRs) involved in metastasis of breast cancer. We present an overview on metastasis-promoting, -suppressing and context-dependent miRs with both activities. We have categorized the corresponding miRs according to their target classes, interaction with stromal cells or exosomes. The pathways affected by individual miRs are outlined in regard to in vitro properties, activity in metastasis-related in vivo models and clinical significance. Current approaches that may be suitable for therapeutic inhibition or restauration of miR activity are outlined. Finally, we discuss the delivery bottlenecks which present as a major challenge in nucleic acid (miR)-based therapies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/secondary , MicroRNAs/physiology , Breast Neoplasms/therapy , Exosomes/metabolism , Female , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Stromal Cells/metabolism
10.
Sci Rep ; 7(1): 15480, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29133816

ABSTRACT

We have devised an effective and robust method for the characterization of gene-editing events. The efficacy of editing-mediated mono- and bi-allelic gene inactivation and integration events is quantified based on colony counts. The combination of diphtheria toxin (DT) and puromycin (PM) selection enables analyses of 10,000-100,000 individual cells, assessing hundreds of clones with inactivated genes per experiment. Mono- and bi-allelic gene inactivation is differentiated by DT resistance, which occurs only upon bi-allelic inactivation. PM resistance indicates integration. The robustness and generalizability of the method were demonstrated by quantifying the frequency of gene inactivation and cassette integration under different editing approaches: CRISPR/Cas9-mediated complete inactivation was ~30-50-fold more frequent than cassette integration. Mono-allelic inactivation without integration occurred >100-fold more frequently than integration. Assessment of gRNA length confirmed 20mers to be most effective length for inactivation, while 16-18mers provided the highest overall integration efficacy. The overall efficacy was ~2-fold higher for CRISPR/Cas9 than for zinc-finger nuclease and was significantly increased upon modulation of non-homologous end joining or homology-directed repair. The frequencies and ratios of editing events were similar for two different DPH genes (independent of the target sequence or chromosomal location), which indicates that the optimization parameters identified with this method can be generalized.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Minor Histocompatibility Antigens/genetics , Proteins/genetics , Tumor Suppressor Proteins/genetics , Alleles , Diphtheria Toxin/administration & dosage , Gene Knockout Techniques/methods , Genetic Vectors/genetics , Histidine/analogs & derivatives , Histidine/biosynthesis , Humans , MCF-7 Cells , Minor Histocompatibility Antigens/metabolism , Proteins/metabolism , Puromycin/administration & dosage , Transfection/methods , Transgenes/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL