Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Signal ; 17(844): eadn6052, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38980922

ABSTRACT

Inhibitors of the transforming growth factor-ß (TGF-ß) pathway are potentially promising antifibrotic therapies, but nonselective simultaneous inhibition of all three TGF-ß homologs has safety liabilities. TGF-ß1 is noncovalently bound to a latency-associated peptide that is, in turn, covalently bound to different presenting molecules within large latent complexes. The latent TGF-ß-binding proteins (LTBPs) present TGF-ß1 in the extracellular matrix, and TGF-ß1 is presented on immune cells by two transmembrane proteins, glycoprotein A repetitions predominant (GARP) and leucine-rich repeat protein 33 (LRRC33). Here, we describe LTBP-49247, an antibody that selectively bound to and inhibited the activation of TGF-ß1 presented by LTBPs but did not bind to TGF-ß1 presented by GARP or LRRC33. Structural studies demonstrated that LTBP-49247 recognized an epitope on LTBP-presented TGF-ß1 that is not accessible on GARP- or LRRC33-presented TGF-ß1, explaining the antibody's selectivity for LTBP-complexed TGF-ß1. In two rodent models of kidney fibrosis of different etiologies, LTBP-49247 attenuated fibrotic progression, indicating the central role of LTBP-presented TGF-ß1 in renal fibrosis. In mice, LTBP-49247 did not have the toxic effects associated with less selective TGF-ß inhibitors. These results establish the feasibility of selectively targeting LTBP-bound TGF-ß1 as an approach for treating fibrosis.


Subject(s)
Extracellular Matrix , Fibrosis , Latent TGF-beta Binding Proteins , Transforming Growth Factor beta1 , Transforming Growth Factor beta1/metabolism , Animals , Humans , Latent TGF-beta Binding Proteins/metabolism , Latent TGF-beta Binding Proteins/antagonists & inhibitors , Extracellular Matrix/metabolism , Mice , Male , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/drug therapy , Disease Progression , Kidney/pathology , Kidney/metabolism , Kidney/drug effects , Mice, Inbred C57BL
2.
Cell ; 186(25): 5620-5637.e16, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38065082

ABSTRACT

Colorectal cancer exhibits dynamic cellular and genetic heterogeneity during progression from precursor lesions toward malignancy. Analysis of spatial multi-omic data from 31 human colorectal specimens enabled phylogeographic mapping of tumor evolution that revealed individualized progression trajectories and accompanying microenvironmental and clonal alterations. Phylogeographic mapping ordered genetic events, classified tumors by their evolutionary dynamics, and placed clonal regions along global pseudotemporal progression trajectories encompassing the chromosomal instability (CIN+) and hypermutated (HM) pathways. Integrated single-cell and spatial transcriptomic data revealed recurring epithelial programs and infiltrating immune states along progression pseudotime. We discovered an immune exclusion signature (IEX), consisting of extracellular matrix regulators DDR1, TGFBI, PAK4, and DPEP1, that charts with CIN+ tumor progression, is associated with reduced cytotoxic cell infiltration, and shows prognostic value in independent cohorts. This spatial multi-omic atlas provides insights into colorectal tumor-microenvironment co-evolution, serving as a resource for stratification and targeted treatments.


Subject(s)
Colorectal Neoplasms , Microsatellite Instability , Tumor Microenvironment , Humans , Chromosomal Instability/genetics , Colorectal Neoplasms/pathology , Gene Expression Profiling , p21-Activated Kinases/genetics , Phylogeny , Mutation , Disease Progression , Prognosis
3.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37328286

ABSTRACT

BACKGROUND: Immune exclusion (IE) where tumors deter the infiltration of immune cells into the tumor microenvironment has emerged as a key mechanism underlying immunotherapy resistance. We recently reported a novel role of discoidin domain-containing receptor 1 (DDR1) in promoting IE in breast cancer and validated its critical role in IE using neutralizing rabbit monoclonal antibodies (mAbs) in multiple mouse tumor models. METHODS: To develop a DDR1-targeting mAb as a potential cancer therapeutic, we humanized mAb9 with a complementarity-determining region grafting strategy. The humanized antibody named PRTH-101 is currently being tested in a Phase 1 clinical trial. We determined the binding epitope of PRTH-101 from the crystal structure of the complex between DDR1 extracellular domain (ECD) and the PRTH-101 Fab fragment with 3.15 Å resolution. We revealed the underlying mechanisms of action of PRTH-101 using both cell culture assays and in vivo study in a mouse tumor model. RESULTS: PRTH-101 has subnanomolar affinity to DDR1 and potent antitumor efficacy similar to the parental rabbit mAb after humanization. Structural information illustrated that PRTH-101 interacts with the discoidin (DS)-like domain, but not the collagen-binding DS domain of DDR1. Mechanistically, we showed that PRTH-101 inhibited DDR1 phosphorylation, decreased collagen-mediated cell attachment, and significantly blocked DDR1 shedding from the cell surface. Treatment of tumor-bearing mice with PRTH-101 in vivo disrupted collagen fiber alignment (a physical barrier) in the tumor extracellular matrix (ECM) and enhanced CD8+ T cell infiltration in tumors. CONCLUSIONS: This study not only paves a pathway for the development of PRTH-101 as a cancer therapeutic, but also sheds light on a new therapeutic strategy to modulate collagen alignment in the tumor ECM for enhancing antitumor immunity.


Subject(s)
Antibodies, Monoclonal , Discoidin Domain Receptor 1 , Neoplasms , Animals , Mice , Collagen/metabolism , Discoidin Domain Receptor 1/metabolism , Extracellular Matrix/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Microenvironment , Antibodies, Monoclonal/pharmacology
4.
Int J Toxicol ; 40(3): 226-241, 2021.
Article in English | MEDLINE | ID: mdl-33739172

ABSTRACT

Checkpoint inhibitors offer a promising immunotherapy strategy for cancer treatment; however, due to primary or acquired resistance, many patients do not achieve lasting clinical responses. Recently, the transforming growth factor-ß (TGFß) signaling pathway has been identified as a potential target to overcome primary resistance, although the nonselective inhibition of multiple TGFß isoforms has led to dose-limiting cardiotoxicities. SRK-181 is a high-affinity, fully human antibody that selectively binds to latent TGFß1 and inhibits its activation. To support SRK-181 clinical development, we present here a comprehensive preclinical assessment of its pharmacology, pharmacokinetics, and safety across multiple species. In vitro studies showed that SRK-181 has no effect on human platelet function and does not induce cytokine release in human peripheral blood. Four-week toxicology studies with SRK-181 showed that weekly intravenous administration achieved sustained serum exposure and was well tolerated in rats and monkeys, with no treatment-related adverse findings. The no-observed-adverse-effect levels levels were 200 mg/kg in rats and 300 mg/kg in monkeys, the highest doses tested, and provide a nonclinical safety factor of up to 813-fold (based on Cmax) above the phase 1 starting dose of 80 mg every 3 weeks. In summary, the nonclinical pharmacology, pharmacokinetic, and toxicology data demonstrate that SRK-181 is a selective inhibitor of latent TGFß1 that does not produce the nonclinical toxicities associated with nonselective TGFß inhibition. These data support the initiation and safe conduct of a phase 1 trial with SRK-181 in patients with advanced cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Neoplasm Metastasis/drug therapy , Transforming Growth Factor beta1/adverse effects , Transforming Growth Factor beta1/therapeutic use , Animals , Cells, Cultured/drug effects , Disease Models, Animal , Humans , Immunotherapy/methods , Macaca fascicularis , Rats
5.
Sci Transl Med ; 12(536)2020 03 25.
Article in English | MEDLINE | ID: mdl-32213632

ABSTRACT

Despite breakthroughs achieved with cancer checkpoint blockade therapy (CBT), many patients do not respond to anti-programmed cell death-1 (PD-1) due to primary or acquired resistance. Human tumor profiling and preclinical studies in tumor models have recently uncovered transforming growth factor-ß (TGFß) signaling activity as a potential point of intervention to overcome primary resistance to CBT. However, the development of therapies targeting TGFß signaling has been hindered by dose-limiting cardiotoxicities, possibly due to nonselective inhibition of multiple TGFß isoforms. Analysis of mRNA expression data from The Cancer Genome Atlas revealed that TGFΒ1 is the most prevalent TGFß isoform expressed in many types of human tumors, suggesting that TGFß1 may be a key contributor to primary CBT resistance. To test whether selective TGFß1 inhibition is sufficient to overcome CBT resistance, we generated a high-affinity, fully human antibody, SRK-181, that selectively binds to latent TGFß1 and inhibits its activation. Coadministration of SRK-181-mIgG1 and an anti-PD-1 antibody in mice harboring syngeneic tumors refractory to anti-PD-1 treatment induced profound antitumor responses and survival benefit. Specific targeting of TGFß1 was also effective in tumors expressing more than one TGFß isoform. Combined SRK-181-mIgG1 and anti-PD-1 treatment resulted in increased intratumoral CD8+ T cells and decreased immunosuppressive myeloid cells. No cardiac valvulopathy was observed in a 4-week rat toxicology study with SRK-181, suggesting that selectively blocking TGFß1 activation may avoid dose-limiting toxicities previously observed with pan-TGFß inhibitors. These results establish a rationale for exploring selective TGFß1 inhibition to overcome primary resistance to CBT.


Subject(s)
Neoplasms , Transforming Growth Factor beta/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes , Cardiotoxicity , Cell Line, Tumor , Humans , Mice , Neoplasms/drug therapy , Rats , Signal Transduction
6.
J Biol Chem ; 288(45): 32314-32325, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24047894

ABSTRACT

Natalizumab antibody to α4-integrins is used in therapy of multiple sclerosis and Crohn's disease. A crystal structure of the Fab bound to an α4 integrin ß-propeller and thigh domain fragment shows that natalizumab recognizes human-mouse differences on the circumference of the ß-propeller domain. The epitope is adjacent to but outside of a ligand-binding groove formed at the interface with the ß-subunit ßI domain and shows no difference in structure when bound to Fab. Competition between Fab and the ligand vascular cell adhesion molecule (VCAM) for binding to cell surface α4ß1 shows noncompetitive antagonism. In agreement, VCAM docking models suggest that binding of domain 1 of VCAM to α4-integrins is unimpeded by the Fab, and that bound Fab requires a change in orientation between domains 1 and 2 of VCAM for binding to α4ß1. Mapping of species-specific differences onto α4ß1 and α4ß7 shows that their ligand-binding sites are highly conserved. Skewing away from these conserved regions of the epitopes recognized by current therapeutic function-blocking antibodies has resulted in previously unanticipated mechanisms of action.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Neutralizing/chemistry , Immunoglobulin Fab Fragments/chemistry , Integrin alpha4/chemistry , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/metabolism , Crystallography, X-Ray , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Integrin alpha4/genetics , Integrin alpha4/metabolism , Integrin alpha4beta1/antagonists & inhibitors , Integrin alpha4beta1/chemistry , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Mice , Molecular Docking Simulation , Natalizumab , Protein Structure, Quaternary , Protein Structure, Tertiary , Vascular Cell Adhesion Molecule-1/chemistry , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
7.
J Biol Chem ; 287(45): 38244-53, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22988250

ABSTRACT

We study a mechanism by which dimerization of the EGF receptor (EGFR) cytoplasmic domain is transmitted to the ectodomain. Therapeutic and other small molecule antagonists to the kinase domain that stabilize its active conformation, but not those that stabilize an inactive conformation, stabilize ectodomain dimerization. Inhibitor-induced dimerization requires an asymmetric kinase domain interface associated with activation. EGF and kinase inhibitors stimulate formation of identical dimer interfaces in the EGFR transmembrane domain, as shown by disulfide cross-linking. Disulfide cross-linking at an interface in domain IV in the ectodomain was also stimulated similarly; however, EGF but not inhibitors stimulated cross-linking in domain II. Inhibitors similarly induced noncovalent dimerization in nearly full-length, detergent-solubilized EGFR as shown by gel filtration. EGFR ectodomain deletion resulted in spontaneous dimerization, whereas deletion of exons 2-7, in which extracellular domains III and IV are retained, did not. In EM, kinase inhibitor-induced dimers lacked any well defined orientation between the ectodomain monomers. Fab of the therapeutic antibody cetuximab to domain III confirmed a variable position and orientation of this domain in inhibitor-induced dimers but suggested that the C termini of domain IV of the two monomers were in close proximity, consistent with dimerization in the transmembrane domains. The results provide insights into the relative energetics of intracellular and extracellular dimerization in EGFR and have significance for physiologic dimerization through the asymmetric kinase interface, bidirectional signal transmission in EGFR, and mechanism of action of therapeutics.


Subject(s)
ErbB Receptors/chemistry , Phosphotransferases/chemistry , Protein Multimerization , Protein Structure, Tertiary , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Cross-Linking Reagents/chemistry , Disulfides/chemistry , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gefitinib , Humans , Microscopy, Electron , Mutation , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/metabolism , Protein Conformation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology
8.
FASEB J ; 26(8): 3412-20, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22601780

ABSTRACT

Developmental endothelial cell locus-1 (Del-1) glycoprotein is secreted by endothelial cells and a subset of macrophages. Del-1 plays a regulatory role in vascular remodeling and functions in innate immunity through interaction with integrin α(V)ß(3). Del-1 contains 3 epidermal growth factor (EGF)-like repeats and 2 discoidin-like domains. An Arg-Gly-Asp (RGD) motif in the second EGF domain (EGF2) mediates adhesion by endothelial cells and phagocytes. We report the crystal structure of its 3 EGF domains. The RGD motif of EGF2 forms a type II' ß turn at the tip of a long protruding loop, dubbed the RGD finger. Whereas EGF2 and EGF3 constitute a rigid rod via an interdomain calcium ion binding site, the long linker between EGF1 and EGF2 lends considerable flexibility to EGF1. Two unique O-linked glycans and 1 N-linked glycan locate to the opposite side of EGF2 from the RGD motif. These structural features favor integrin binding of the RGD finger. Mutagenesis data confirm the importance of having the RGD motif at the tip of the RGD finger. A database search for EGF domain sequences shows that this RGD finger is likely an evolutionary insertion and unique to the EGF domain of Del-1 and its homologue milk fat globule-EGF 8.


Subject(s)
Amino Acid Motifs , Carrier Proteins/chemistry , Integrins/metabolism , Oligopeptides/chemistry , Calcium-Binding Proteins , Carrier Proteins/metabolism , Cell Adhesion Molecules , Epidermal Growth Factor/chemistry , Fucose/metabolism , Glycosylation , HEK293 Cells , Humans , Models, Molecular , Protein Structure, Tertiary
9.
J Biol Chem ; 286(49): 42115-42122, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-21965670

ABSTRACT

The activation of α/ß heterodimeric integrins is the result of highly coordinated rearrangements within both subunits. The molecular interactions between the two subunits, however, remain to be characterized. In this study, we use the integrin α(L)ß(2) to investigate the functional role of the C-linker polypeptide that connects the C-terminal end of the inserted (I) domain with the ß-propeller domain on the α subunit and is located at the interface with the ßI domain of the ß chain. We demonstrate that shortening of the C-linker by eight or more amino acids results in constitutively active α(L)ß(2) in which the αI domain is no longer responsive to the regulation by the ßI domain. Despite this intersubunit uncoupling, both I domains remain individually sensitive to intrasubunit conformational changes induced by allosteric modulators. Interestingly, the length and not the sequence of the C-linker appears to be critical for its functionality in α/ß intersubunit communication. Using two monoclonal antibodies (R7.1 and CBR LFA-1/1) we further demonstrate that shortening of the C-linker results in the gradual loss of combinational epitopes that require both the αI and ß-propeller domains for full reactivity. Taken together, our findings highlight the role of the C-linker as a spring-like element that allows relaxation of the αI domain in the resting state and controlled tension of the αI domain during activation, exerted by the ß chain.


Subject(s)
Integrin alpha1/chemistry , Allosteric Site , Animals , Antibodies, Monoclonal/chemistry , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Dimerization , Humans , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Microscopy, Fluorescence/methods , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Transfection
10.
EMBO J ; 30(23): 4712-27, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21946563

ABSTRACT

Lymphocyte activation triggers adhesiveness of lymphocyte function-associated antigen-1 (LFA-1; integrin α(L)ß(2)) for intercellular adhesion molecules (ICAMs) on endothelia or antigen-presenting cells. Whether the activation signal, after transmission through multiple domains to the ligand-binding αI domain, results in affinity changes for ligand has been hotly debated. Here, we present the first comprehensive measurements of LFA-1 affinities on T lymphocytes for ICAM-1 under a broad array of activating conditions. Only a modest increase in affinity for soluble ligand was detected after activation by chemokine or T-cell receptor ligation, conditions that primed LFA-1 and robustly induced lymphocyte adhesion to ICAM-1 substrates. By stabilizing well-defined LFA-1 conformations by Fab, we demonstrate the absolute requirement of the open LFA-1 headpiece for adhesiveness and high affinity. Interaction of primed LFA-1 with immobilized but not soluble ICAM-1 triggers energy-dependent affinity maturation of LFA-1 to an adhesive, high affinity state. Our results lend support to the traction or translational motion dependence of integrin activation.


Subject(s)
Cell Adhesion/physiology , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1 , Protein Binding/physiology , Chemokine CXCL12/metabolism , HEK293 Cells , Humans , Intercellular Adhesion Molecule-1/immunology , K562 Cells , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Models, Biological , Protein Conformation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/metabolism
11.
Glycobiology ; 16(11): 1033-44, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16877748

ABSTRACT

Intercellular adhesion molecule-1 (ICAM-1) is a heavily N-glycosylated transmembrane protein comprising five extracellular Ig-like domains. The soluble isoform of ICAM-1 (sICAM-1), consisting of its extracellular part, is elevated in the cerebrospinal fluid of patients with severe brain trauma. In mouse astrocytes, recombinant mouse sICAM-1 induces the production of the CXC chemokine macrophage inflammatory protein-2 (MIP-2). MIP-2 induction is glycosylation dependent, as it is strongly enhanced when sICAM-1 carries sialylated, complex-type N-glycans as synthesized by wild-type Chinese hamster ovary (CHO) cells. The present study was aimed at elucidating the N-glycosylation of mouse sICAM-1 expressed in wild-type CHO cells with regard to sialylation, N-glycan profile, and N-glycosylation sites. Ion-exchange chromatography and matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry (MS) of the released N-glycans showed that sICAM-1 mostly carried di- and trisialylated complex-type N-glycans with or without one fucose. In some sialylated N-glycans, one N-acetylneuraminic acid was replaced by N-glycolylneuraminic acid, and approximately 4% carried a higher number of sialic acid residues than of antennae. The N-glycosylation sites of mouse sICAM-1 were analyzed by MALDI-Fourier transform ion cyclotron resonance (FTICR)-MS and nanoLC-ESI-FTICR-MS of tryptic digests of mouse sICAM-1 expressed in the Lec1 mutant of CHO cells. All nine consensus sequences for N-glycosylation were found to be glycosylated. These results show that the N-glycans that enhance the MIP-2-inducing activity of mouse sICAM-1 are mostly di- and trisialylated complex-type N-glycans including a small fraction carrying more sialic acid residues than antennae and that the nine N-glycosylation sites of mouse sICAM-1 are all glycosylated.


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , Polysaccharides/metabolism , Animals , CHO Cells , Consensus Sequence , Cricetinae , Cricetulus , Glycosylation , Intercellular Adhesion Molecule-1/genetics , Mass Spectrometry , Mice , Mutation , N-Acetylneuraminic Acid/metabolism , Protein Isoforms/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
J Biol Chem ; 279(34): 35201-9, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15201278

ABSTRACT

Intercellular adhesion molecule-1 (ICAM-1) occurs as both a membrane and a soluble, secreted glycoprotein (sICAM-1). ICAM-1 on endothelial cells mediates leukocyte adhesion by binding to leukocyte function associated antigen-1 (LFA-1) and macrophage antigen-1 (Mac-1). Recombinant mouse sICAM-1 induces the production of macrophage inflammatory protein-2 (MIP-2) in mouse astrocytes by a novel LFA-1- and Mac-1-independent mechanism. Here we showed that N-glycan structures of sICAM-1 influence its ability to induce MIP-2 production. sICAM-1 expressed in Chinese hamster ovary (CHO) cells was a more potent inducer of MIP-2 production than sICAM-1 expressed in HEK 293 cells, suggesting that posttranslational modification of sICAM-1 could influence its signaling activity. To explore the roles of glycosylation in sICAM-1 activity, we expressed sICAM-1 in mutant CHO cell lines differing in glycosylation, including Lec2, Lec8, and Lec1 as well as in CHO cells cultured in the presence of the alpha-mannosidase-I inhibitor kifunensine. Signaling activity of sICAM-1 lacking sialic acid was reduced 3-fold compared with sICAM-1 from CHO cells. The activity of sICAM-1 lacking both sialic acid and galactose was reduced 12-fold, whereas the activity of sICAM-1 carrying only high mannose-type N-glycans was reduced 12-26-fold. sICAM-1 glycoforms carrying truncated glycans retained full ability to bind to LFA-1 on leukocytes. Thus, sialylated and galactosylated complex-type N-glycans strongly enhanced the ability of sICAM-1 to induce MIP-2 production in astrocytes but did not alter its binding to LFA-1 on leukocytes. Glycosylation could therefore serve as a means to regulate specifically the signaling function of sICAM-1 in vivo.


Subject(s)
Astrocytes/metabolism , Intercellular Adhesion Molecule-1/metabolism , Signal Transduction , Animals , CHO Cells , Cell Line , Chemokine CXCL2 , Cricetinae , Female , Glycosylation , Humans , Intercellular Adhesion Molecule-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Monokines/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Protein Processing, Post-Translational
13.
Breast Cancer Res Treat ; 76(2): 157-66, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12452453

ABSTRACT

Overexpression of the anti-apoptotic proteins bcl-2 and bcl-xL is implicated in breast cancer development, tumor progression and drug resistance. Here we describe the use of the bcl-2/bcl-xL bispecific antisense oligonucleotide 4625 to sensitize breast carcinoma cells to anti-cancer drugs routinely used in breast cancer therapy. MCF7 cells were treated with oligonucleotide 4625, doxorubicin, paclitaxel or cyclophosphamide alone, or with combinations of oligonucleotide and the anti-cancer drugs. As measured in cell viability assays, treatment with the various combinations reduced the number of viable MCF7 cells more effectively than treatment with the single drugs alone. Treatment with a sequence control oligonucleotide did not affect cell viability. All combination treatments induced apoptosis as demonstrated by the appearance of massive nuclear condensation in a high proportion of the cells. To further characterize the interaction between 4625 and doxorubicin, paclitaxel or cyclophosphamide, the median-effect method was used. In MCF7 cells all combinations resulted in potent synergistic effects over a broad range of toxicity with combination indices ranging from 0.8 to 0.1. Similarly, strong synergistic interactions between oligonucleotide 4625 and the anti-cancer drugs were also observed in cultures of the breast carcinoma cell line MDA-MB-231. Our data suggest the use of 4625 as a potent adjuvant in breast cancer chemotherapy.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cyclophosphamide/pharmacology , Doxorubicin/pharmacology , Oligonucleotides, Antisense/pharmacology , Paclitaxel/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Down-Regulation , Drug Resistance, Neoplasm , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oligonucleotides , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Cells, Cultured , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL