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










Publication year range
1.
Adv Sci (Weinh) ; 11(20): e2307852, 2024 May.
Article in English | MEDLINE | ID: mdl-38477561

ABSTRACT

First-line treatment of multiple myeloma, a prevalent blood cancer lacking a cure, using anti-CD38 daratumumab antibody and lenalidomide is often inadequate due to relapse and severe side effects. To enhance drug safety and efficacy, an antibody-drug conjugate, TE-1146, comprising six lenalidomide drug molecules site-specifically conjugated to a reconfigured daratumumab to deliver cytotoxic lenalidomide to tumor cells is developed. TE-1146 is prepared using the HighDAR platform, which employs i) a maleimide-containing "multi-arm linker" to conjugate multiple drug molecules creating a drug bundle, and ii) a designed peptide with a Zn2+-binding cysteine at the C-termini of a reconfigured daratumumab for site-specific drug bundle conjugation. It is shown that TE-1146 remains intact and effectively enters CD38-expressing tumor cells, releasing lenalidomide, leading to enhanced cell-killing effects compared to lenalidomide/daratumumab alone or their combination. This reveals the remarkable potency of lenalidomide once internalized by myeloma cells. TE-1146 precisely delivers lenalidomide to target CD38-overexpressing tumor cells. In contrast, lenalidomide without daratumumab cannot easily enter cells, whereas daratumumab without lenalidomide relies on Fc-dependent effector functions to kill tumor cells.


Subject(s)
Antibodies, Monoclonal , Immunoconjugates , Lenalidomide , Multiple Myeloma , Multiple Myeloma/drug therapy , Humans , Immunoconjugates/pharmacology , Immunoconjugates/chemistry , Lenalidomide/pharmacology , Lenalidomide/therapeutic use , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Mice , Animals , Disease Models, Animal
2.
J Med Chem ; 66(15): 10604-10616, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37462154

ABSTRACT

Antibodies conjugated with diagnostic/therapeutic radionuclides are attractive options for inoperable cancers lacking accurate imaging methods and effective therapeutics, such as pancreatic cancer. Hence, we have produced an antibody radionuclide conjugate termed TE-1132 comprising a α-CA19-9 scFv-Fc that is site-specifically conjugated at each C-terminus to 3 DOTA chelators via a cysteine-containing peptide linker. The smaller scFv-Fc size facilitates diffusivity within solid tumors, whereas the chelator-to-antibody ratio of six enabled 177Lu-radiolabeled TE-1132 to exhibit high radioactivity up to 520 MBq/nmol. In mice bearing BxPC3 tumors, immuno-SPECT/CT imaging of [111In]In-TE-1132 and the biodistribution of [177Lu]Lu-TE-1132 showed selective tumor accumulation. Single and multiple doses of [177Lu]Lu-TE-1132 effectively inhibited the BxPC3 tumor growth and prolonged the survival of mice with no irreversible body weight loss or hematopoietic damage. The adequate pharmacokinetic parameters, prominent tumor accumulation, and efficacy with good safety in mice encourage the further investigation of theranostic TE-1132 for treating pancreatic cancer.


Subject(s)
Immunoconjugates , Pancreatic Neoplasms , Mice , Animals , Chelating Agents , CA-19-9 Antigen , Tissue Distribution , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Radiopharmaceuticals/pharmacokinetics , Cell Line, Tumor , Lutetium , Pancreatic Neoplasms
3.
Mol Pharm ; 14(11): 4113-4120, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28954509

ABSTRACT

Therapeutics reducing bone turnover, such as denosumab (Dmab), an anti-RANKL antibody, can provide treatments for patients with bone destruction. However, some patients with osteoporosis or localized primary bone tumors and many patients with various types of bone-metastatic cancer display unsatisfactory responses to Dmab. For achieving greater efficiency of RANKL neutralization in the bone microenvironment by enhancing the distribution of Dmab to the bone, we reengineered Dmab by fusing with single-chain variable fragments of an antibody specific for osteonectin (On), which is abundantly expressed in osseous tissues. The bispecific antibody, Dmab-FvOn, showed a similar activity as Dmab in inhibiting RANKL as examined in an osteoclast differentiation assay. When administered to mice, Dmab-FvOn was found to localize in increased proportions at the endosteum of the bone where osteonectin is abundant. Our study suggests that by linking anti-RANKL with an osteonectin-targeting moiety, a greater proportion of the therapeutic effector can be distributed in the bone. Future studies are needed to investigate whether the bispecific antibody can achieve higher therapeutic efficacy and lower toxicity.


Subject(s)
Osteonectin/metabolism , RANK Ligand/metabolism , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Cell Differentiation/drug effects , Denosumab/pharmacology , Mice , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteonectin/immunology , RANK Ligand/immunology
4.
Sci Rep ; 5: 11581, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26113483

ABSTRACT

Omalizumab, an anti-IgE antibody, used to treat severe allergic asthma and chronic idiopathic urticaria, binds to IgE in blood or membrane-bound on B lymphocytes but not to IgE bound to its high (FcεRI) or low (CD23) affinity receptor. Mutagenesis studies indicate overlapping FcεRI and omalizumab-binding sites in the Cε3 domain, but crystallographic studies show FcεRI and CD23-binding sites that are far apart, so how can omalizumab block IgE from binding both receptors? We report a 2.42-Šomalizumab-Fab structure, a docked IgE-Fc/omalizumab-Fab structure consistent with available experimental data, and the free energy contributions of IgE residues to binding omalizumab, CD23, and FcεRI. These results provide a structural and physical basis as to why omalizumab cannot bind receptor-bound IgE and why omalizumab-bound IgE cannot bind to CD23/FcεRI. They reveal the key IgE residues and their roles in binding omalizumab, CD23, and FcεRI.


Subject(s)
Antibodies, Anti-Idiotypic/chemistry , Immunoglobulin E/chemistry , Omalizumab/chemistry , Amino Acid Sequence , Anti-Asthmatic Agents/immunology , Anti-Asthmatic Agents/metabolism , Anti-Asthmatic Agents/therapeutic use , Antibodies, Anti-Idiotypic/metabolism , Antibodies, Anti-Idiotypic/therapeutic use , Asthma/drug therapy , Asthma/immunology , Binding Sites/genetics , Crystallography, X-Ray , Humans , Immunoglobulin E/metabolism , Models, Molecular , Mutation , Omalizumab/metabolism , Omalizumab/therapeutic use , Protein Binding , Protein Structure, Tertiary , Receptors, IgE/chemistry , Receptors, IgE/metabolism
5.
Nat Commun ; 5: 3139, 2014.
Article in English | MEDLINE | ID: mdl-24457896

ABSTRACT

IgE mediates hypersensitivity reactions responsible for most allergic diseases, which affect 20-40% of the population in developed countries. A 52-residue domain of membrane-bound IgE (mIgE) called CεmX is currently a target for developing therapeutic antibodies; however, its structure is unknown. Here we show that two antibodies with therapeutic potential in IgE-mediated allergic diseases, which can cause cytolytic effects on mIgE-expressing B lymphocytes and downregulate IgE production, target different conformations of an intrinsically disordered region (IDR) in the extracellular CεmX domain. We provide an important example of antibodies targeting an extracellular IDR of a receptor on the surface of intended target cells. We also provide fundamental structural characteristics unique to human mIgE, which may stimulate further studies to investigate whether other monoclonal antibodies (mAbs) targeting intrinsically disordered peptide segments or vaccine-like products targeting IDRs of a membrane protein can be developed.


Subject(s)
Immunoglobulin E/immunology , Amino Acid Sequence , Binding Sites, Antibody , Cell Membrane/immunology , Crystallography, X-Ray , Humans , Immunoglobulin E/chemistry , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Conformation
6.
Mol Immunol ; 53(3): 187-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22944457

ABSTRACT

Surface IgD and IgM doubly positive cells comprise the major population of B cells in the human immune system. The heavy chain of membrane-bound IgD (mδ) differs from that of IgD (δ) in that mδ contains a C-terminal membrane-anchor peptide. Our group previously proposed that the N-terminal extracellular segment of 27 aa residues of the membrane-anchor peptide of mδ, referred to as the mIg isotype-specific-δ (migis-δ) segment, may provide a unique antigenic site for isotype-specific targeting of mIgD(+) B cells. Here we report the preparation of mouse mAbs specific for human migis-δ. The mAbs bound to human migis-δ-containing recombinant proteins in an ELISA and to mIgD-expressing transfectants of a CHO cell line as analyzed by flow cytometry. MAb 20E6, which binds to an epitope toward the N-terminal of human migis-δ, could stain human B cell line MC116, which expressed mIgD and mIgM. MC116 cells could be induced to undergo apoptosis by treatment with 20E6 in the presence of a second crosslinking antibody. Chimeric 20E6 caused antibody-dependent cellular cytotoxicity of MC116 cells in the presence of human PBMCs as the source of effector cells. In cultures of PBMCs, 20E6 down-regulated the population of mIgD(+) B cells. The production of human IgM by transplanted MC116 cells in NOD-SCID (NOD.CB17-Prkdc(scid)/IcrCrlBltw) mice could be suppressed by 20E6. These results encourage further investigation of the potential of anti-migis-δ mAbs to control mIgD(+) B cells, when such a manipulation may alleviate a disease state.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , B-Lymphocytes/immunology , Immunoglobulin D/metabolism , Amino Acid Sequence , Animals , Antibody-Dependent Cell Cytotoxicity , Apoptosis/immunology , B-Lymphocytes/cytology , B-Lymphocytes/transplantation , CHO Cells , Cell Line , Cricetinae , Cricetulus , Epitopes, B-Lymphocyte/genetics , Humans , Immunoglobulin D/genetics , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Immunoglobulin delta-Chains/genetics , Immunoglobulin delta-Chains/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Surface Plasmon Resonance , Transplantation, Heterologous
7.
FEBS Lett ; 584(18): 4083-8, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20807533

ABSTRACT

Various plant developmental processes involve phytohormones such as cytokinins. Isopentenyltransferase (IPT) reaction is the key rate-limiting step in cytokinin biosynthesis that transfers the isopentenyl (iP) group from dimethylallyl diphosphate to the N6-amino group of adenine. Here, a series of diadenosine polyphosphates (Ap(n)A) were screened as possible substrates of IPT, among which diadenosine tetraphosphate, diadenosine pentaphosphate and diadenosine hexaphosphate showed higher affinity than did the authentic substrates ADP and ATP. In addition, formation of mono-isopentenyl Ap(n)A and di-isopentenyl Ap(n)A was observed. Judging by the existing biosynthetic and hydrolytic systems for Ap(n)A in plants, Ap(n)A and isopentenyl-Ap(n)A may occur in the plant cells, with functional importance.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Dinucleoside Phosphates/biosynthesis , Humulus/enzymology , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/genetics , Catalysis , Mutation , Protein Conformation
8.
Nucleic Acids Res ; 38(5): 1738-48, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20007608

ABSTRACT

Cytokinins are important plant hormones, and their biosynthesis most begins with the transfer of isopentenyl group from dimethylallyl diphosphate (DMAPP) to the N6-amino group of adenine by either adenylate isopentenyltransferase (AIPT) or tRNA-IPT. Plant AIPTs use ATP/ADP as an isopentenyl acceptor and bacterial AIPTs prefer AMP, whereas tRNA-IPTs act on specific sites of tRNA. Here, we present the crystal structure of an AIPT-ATP complex from Humulus lupulus (HlAIPT), which is similar to the previous structures of Agrobacterium AIPT and yeast tRNA-IPT. The enzyme is structurally homologous to the NTP-binding kinase family of proteins but forms a solvent-accessible channel that binds to the donor substrate DMAPP, which is directed toward the acceptor substrate ATP/ADP. When measured with isothermal titration calorimetry, some nucleotides displayed different binding affinities to HlAIPT with an order of ATP > dATP approximately ADP > GTP > CTP > UTP. Two basic residues Lys275 and Lys220 in HlAIPT interact with the beta and gamma-phosphate of ATP. By contrast, the interactions are absent in Agrobacterium AIPT because they are replaced by the acidic residues Asp221 and Asp171. Despite its structural similarity to the yeast tRNA-IPT, HlAIPT has evolved with a different binding strategy for adenylate.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Humulus/enzymology , Purine Nucleotides/metabolism , Pyrimidine Nucleotides/metabolism , Alkyl and Aryl Transferases/metabolism , Aspartic Acid/chemistry , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Purine Nucleotides/chemistry , Pyrimidine Nucleotides/chemistry , RNA, Transfer/metabolism , Rhizobium/enzymology , Substrate Specificity
9.
J Biol Chem ; 283(50): 35265-72, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18840608

ABSTRACT

Protein S-nitrosylation mediated by cellular nitric oxide (NO) plays a primary role in executing biological functions in cGMP-independent NO signaling. Although S-nitrosylation appears similar to Cys oxidation induced by reactive oxygen species, the molecular mechanism and biological consequence remain unclear. We investigated the structural process of S-nitrosylation of protein-tyrosine phosphatase 1B (PTP1B). We treated PTP1B with various NO donors, including S-nitrosothiol reagents and compound-releasing NO radicals, to produce site-specific Cys S-nitrosylation identified using advanced mass spectrometry (MS) techniques. Quantitative MS showed that the active site Cys-215 was the primary residue susceptible to S-nitrosylation. The crystal structure of NO donor-reacted PTP1B at 2.6 A resolution revealed that the S-NO state at Cys-215 had no discernible irreversibly oxidized forms, whereas other Cys residues remained in their free thiol states. We further demonstrated that S-nitrosylation of the Cys-215 residue protected PTP1B from subsequent H(2)O(2)-induced irreversible oxidation. Increasing the level of cellular NO by pretreating cells with an NO donor or by activating ectopically expressed NO synthase inhibited reactive oxygen species-induced irreversible oxidation of endogenous PTP1B. These findings suggest that S-nitrosylation might prevent PTPs from permanent inactivation caused by oxidative stress.


Subject(s)
Cysteine/chemistry , Oxygen/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Crystallography, X-Ray/methods , Enzyme Activation , Humans , Mass Spectrometry/methods , Nitric Oxide/chemistry , Nitrogen/chemistry , Oxidative Stress , Reactive Oxygen Species
10.
Proteins ; 66(4): 996-1003, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17173287

ABSTRACT

The P-loop-containing protein phos-phatases are important regulators in signal transduction. These enzymes have structural and functional similarity with a conserved sequence of Dx(25-41)HCxxGxxR(T/S) essential for catalysis. The singular protein tyrosine phosphatase (PTP) from archaeal Sulfolobus solfataricus is one of the smallest known PTPs with extreme thermostability. Here, we report the crystal structure of this phosphatase and its complexes with two tyrosyl phosphopeptides A-(p)Y-R and N-K-(p)Y-G-N. The structure suggests the minimal structural motif of the PTP family, having two variable sequences inserted between the beta2-beta3 and beta3-beta4 strands, respectively. The phosphate of both phosphopeptide substrates is bound to the P-loop through several hydrogen bonds. Comparison of several phosphatase-substrate complexes revealed that Gln135 on the Q-loop has different modes of recognition toward phosphopeptides. The substrate specificity of SsoPTP is primarily localized at the phosphotyrosine, suggesting that this phosphatase may be a prototypical PTP.


Subject(s)
Phosphopeptides/chemistry , Phosphopeptides/metabolism , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Sulfolobus solfataricus/enzymology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Ligands , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/genetics , Structural Homology, Protein , Substrate Specificity , Sulfolobus solfataricus/genetics
11.
Structure ; 12(11): 2015-24, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15530366

ABSTRACT

Various inositide phosphatases participate in the regulation of inositol polyphosphate signaling molecules. Plant phytases are phosphatases that hydrolyze phytate to less-phosphorylated myo-inositol derivatives and phosphate. The phytase from Selenomonas ruminantium shares no sequence homology with other microbial phytases. Its crystal structure revealed a phytase fold of the dual-specificity phosphatase type. The active site is located near a conserved cysteine-containing (Cys241) P loop. We also solved two other crystal forms in which an inhibitor, myo-inositol hexasulfate, is cocrystallized with the enzyme. In the "standby" and the "inhibited" crystal forms, the inhibitor is bound, respectively, in a pocket slightly away from Cys241 and at the substrate binding site where the phosphate group to be hydrolyzed is held close to the -SH group of Cys241. Our structural and mutagenesis studies allow us to visualize the way in which the P loop-containing phytase attracts and hydrolyzes the substrate (phytate) sequentially.


Subject(s)
6-Phytase/chemistry , Phytic Acid/metabolism , Selenomonas/enzymology , 6-Phytase/antagonists & inhibitors , 6-Phytase/genetics , 6-Phytase/metabolism , Amino Acid Sequence , Hydrolysis , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Conformation , Protein Folding , Sequence Homology, Amino Acid , Substrate Specificity
12.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 8): 1381-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15272160

ABSTRACT

The protein Sac7d belongs to a class of small chromosomal proteins from the hyperthermophilic archaeon Sulfolobus acidocaldarius. Two new crystal forms of Sac7d in complex with the DNA decamers CCTATATAGG and CCTACGTAGG were obtained and their structures were determined by molecular replacement. The refined models yielded R/Rfree values of 0.221/0.257 and 0.248/0.290 at 1.9 and 2.2 A resolution, respectively. The protein structures are similar to the previously determined structure of Sac7d-GCGATCGC (PDB code 1azp), but the DNA molecules are more bent overall, by 14-20 degrees. The relative positions of the Sac7d protein and the bound DNA also differ by rotations of 6-10 degrees and translations of 1.0-2.4 A. In addition to the water molecules in the central cavity, three additional conserved water molecules are found that mediate the protein-DNA interactions. The decamer DNA fragments form virtual double helices in the crystal, with a unit length of eight base pairs. The molecular packing of the new crystal forms differs from that of 1azp. The terminal nucleotides are opened up and form triple base pairs with other DNA molecules. Through lattice contacts, the Sac7d molecule also makes additional interactions with DNA, whereas only limited protein-protein interactions are seen.


Subject(s)
Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , Nucleic Acid Conformation , Sulfolobus acidocaldarius/chemistry , Base Sequence , Crystallography, X-Ray , Models, Molecular , Protein Structure, Tertiary , Sulfolobus acidocaldarius/metabolism , Temperature , Water/chemistry
13.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 12): 2265-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14646090

ABSTRACT

Octaprenyl pyrophosphate synthase (OPPs) catalyzes the condensation of five isopentenyl pyrophosphates with farnesyl pyrophosphate to generate C(40) octaprenyl pyrophosphate. The enzymes from the hyperthermophilic bacterium Thermotoga maritima and from the mesophilic Escherichia coli were expressed in E. coli and the recombinant proteins were purified and crystallized. The T. maritima OPPs crystals belong to space group P42(1)2, with unit-cell parameters a = b = 151.53, c = 69.72 A. The E. coli OPPs crystals belong to space group C222(1), with unit-cell parameters a = 247.66, b = 266.10, c = 157.93 A. Diffraction data were collected at 100 K using synchrotron radiation and an in-house X-ray source. Structure determination of T. maritima OPPs has been carried out using MIR data sets at 2.8 A resolution. The asymmetric unit contains one dimer. An initial model with 280 residues per subunit has been built and refined to 2.28 A resolution. It shows mostly helical structure and resembles that of avian farnesyl pyrophosphate synthase.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Escherichia coli/enzymology , Thermotoga maritima/enzymology , Alkyl and Aryl Transferases/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Crystallization , Crystallography, X-Ray , Dimerization , Escherichia coli/genetics , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Thermotoga maritima/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...