Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Eur J Nucl Med Mol Imaging ; 49(11): 3651-3667, 2022 09.
Article in English | MEDLINE | ID: mdl-35608703

ABSTRACT

PURPOSE: Fibroblast activation protein (FAP) is a membrane-bound protease that has limited expression in normal adult tissues but is highly expressed in the tumor microenvironment of many solid cancers. FAP-2286 is a FAP-binding peptide coupled to a radionuclide chelator that is currently being investigated in patients as an imaging and therapeutic agent. The potency, selectivity, and efficacy of FAP-2286 were evaluated in preclinical studies. METHODS: FAP expression analysis was performed by immunohistochemistry and autoradiography on primary human cancer specimens. FAP-2286 was assessed in biochemical and cellular assays and in in vivo imaging and efficacy studies, and was further evaluated against FAPI-46, a small molecule-based FAP-targeting agent. RESULTS: Immunohistochemistry confirmed elevated levels of FAP expression in multiple tumor types including pancreatic, breast, and sarcoma, which correlated with FAP binding by FAP-2286 autoradiography. FAP-2286 and its metal complexes demonstrated high affinity to FAP recombinant protein and cell surface FAP expressed on fibroblasts. Biodistribution studies in mice showed rapid and persistent uptake of 68Ga-FAP-2286, 111In-FAP-2286, and 177Lu-FAP-2286 in FAP-positive tumors, with renal clearance and minimal uptake in normal tissues. 177Lu-FAP-2286 exhibited antitumor activity in FAP-expressing HEK293 tumors and sarcoma patient-derived xenografts, with no significant weight loss. In addition, FAP-2286 maintained longer tumor retention and suppression in comparison to FAPI-46. CONCLUSION: In preclinical models, radiolabeled FAP-2286 demonstrated high tumor uptake and retention, as well as potent efficacy in FAP-positive tumors. These results support clinical development of 68Ga-FAP-2286 for imaging and 177Lu-FAP-2286 for therapeutic use in a broad spectrum of FAP-positive tumors.


Subject(s)
Gallium Radioisotopes , Sarcoma , Adult , Animals , Cell Line, Tumor , Fibroblasts , HEK293 Cells , Humans , Mice , Radionuclide Imaging , Tissue Distribution , Tumor Microenvironment
2.
J Nucl Med ; 63(3): 415-423, 2022 03.
Article in English | MEDLINE | ID: mdl-34168013

ABSTRACT

Fibroblast activation protein (FAP) is a promising target for diagnosis and therapy of numerous malignant tumors. FAP-2286 is the conjugate of a FAP-binding peptide, which can be labeled with radionuclides for theranostic applications. We present the first-in-humans results using 177Lu-FAP-2286 for peptide-targeted radionuclide therapy (PTRT). Methods: PTRT using 177Lu-FAP-2286 was performed on 11 patients with advanced adenocarcinomas of the pancreas, breast, rectum, or ovary after prior confirmation of uptake on 68Ga-FAP-2286 or 68Ga-FAPI-04 PET/CT. Results: Administration of 177Lu-FAP-2286 (5.8 ± 2.0 GBq; range, 2.4-9.9 GBq) was well tolerated, with no adverse symptoms or clinically detectable pharmacologic effects being noticed or reported in any of the patients. The whole-body effective dose was 0.07 ± 0.02 Gy/GBq (range, 0.04-0.1 Gy/GBq). The mean absorbed doses for kidneys and red marrow were 1.0 ± 0.6 Gy/GBq (range, 0.4-2.0 Gy/GBq) and 0.05 ± 0.02 Gy/GBq (range, 0.03-0.09 Gy/GBq), respectively. Significant uptake and long tumor retention of 177Lu-FAP-2286 resulted in high absorbed tumor doses, such as 3.0 ± 2.7 Gy/GBq (range, 0.5-10.6 Gy/GBq) in bone metastases. No grade 4 adverse events were observed. Grade 3 events occurred in 3 patients-1 with pancytopenia, 1 with leukocytopenia, and 1 with pain flare-up; 3 patients reported a pain response. Conclusion:177Lu-FAP-2286 PTRT, applied in a broad spectrum of cancers, was relatively well tolerated, with acceptable side effects, and demonstrated long retention of the radiopeptide. Prospective clinical studies are warranted.


Subject(s)
Adenocarcinoma , Positron Emission Tomography Computed Tomography , Feasibility Studies , Female , Gallium Radioisotopes , Humans , Peptides , Prospective Studies , Radioisotopes/therapeutic use , Tissue Distribution
3.
J Nucl Med ; 59(5): 809-814, 2018 05.
Article in English | MEDLINE | ID: mdl-29025990

ABSTRACT

Neurotensin receptor 1 (NTR1) is overexpressed in ductal pancreatic adenocarcinoma, which is still one of the deadliest cancers, with a very poor prognosis. Eligible patients were offered salvage radiopharmaceutical therapy with the novel NTR1 antagonist 177Lu-3BP-227. Methods: Six patients with confirmed ductal pancreatic adenocarcinoma who had exhausted all other treatment options received 177Lu-3BP-227 for evaluation of NTR1 expression in vivo. Three patients received treatment activities of 5.1-7.5 GBq. Results: Administration of 177Lu-3BP-227 was well tolerated by all patients. The kidneys were identified as the dose-limiting organ. The most severe adverse event was reversible grade 2 anemia. One patient achieved a partial response and experienced significant improvement of symptoms and quality of life. This patient survived 13 mo from diagnosis and 11 mo from the start of 177Lu-3BP-227 therapy. Conclusion: This initial report provides clinical evidence of the feasibility of treatment of ductal pancreatic adenocarcinoma using 177Lu-3BP-227.


Subject(s)
Adenocarcinoma/therapy , Carcinoma, Pancreatic Ductal/therapy , Lutetium/pharmacology , Pancreatic Neoplasms/therapy , Receptors, Neurotensin/antagonists & inhibitors , Adenocarcinoma/metabolism , Aged , Carcinoma, Pancreatic Ductal/metabolism , Combined Modality Therapy , Drug Design , Female , Humans , Injections, Intravenous , Kidney/radiation effects , Male , Middle Aged , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Quality of Life , Radiometry , Radionuclide Imaging , Radiopharmaceuticals , Retrospective Studies , Salvage Therapy , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Treatment Outcome
4.
J Nucl Med ; 58(6): 936-941, 2017 06.
Article in English | MEDLINE | ID: mdl-28254866

ABSTRACT

Increased expression of neurotensin receptor 1 (NTR1) has been shown in a large number of tumor entities such as pancreatic or colon carcinoma. Hence, this receptor is a promising target for diagnostic imaging and radioligand therapy. Using the favorable biodistribution data of the NTR1-targeting agent 111In-3BP-227, we investigated the therapeutic effect of its 177Lu-labeled analog on the tumor growth of NTR1-positive HT29 colon carcinoma xenografts. Methods: 3BP-227 was labeled with 177Lu. To assess its biodistribution properties, SPECT and CT scans of HT29-xenografted nude mice injected with 177Lu-3BP-227 were acquired, and ex vivo tissue activity was determined. To evaluate therapeutic efficacy, 2 groups of mice received the radiopharmaceutical in a median dose of either 165 MBq (129-232 MBq, n = 10) or 110 MBq (82-116 MBq, n = 10), whereas control mice were injected with vehicle (n = 10). Tumor sizes and body weights were monitored for up to 49 d. Renal function and histologic morphology were evaluated. Results: Whole-body SPECT/CT images allowed clear tumor visualization with low background activity and high tumor-to-kidney and -liver ratios. Ex vivo biodistribution data confirmed high and persistent uptake of 177Lu-3BP-227 in HT29 tumors (19.0 ± 3.6 vs. 2.7 ± 1.6 percentage injected dose per gram at 3 and 69 h after injection, respectively). The application of 177Lu-3BP-227 resulted in a distinct delay of tumor growth. Median tumor doubling time for controls was 5.5 d (interquartile range [IQR], 2.8-7.0), compared with 17.5 d (IQR, 5.5-22.5 d) for the 110-MBq and 41.0 d (IQR, 27.5-55.0) for the 165-MBg group. Compared with controls, median relative tumor volume at day 23 after injection was reduced by 55% (P = 0.034) in the 110-MBq and by 88% (P < 0.01) in the 165-MBq group. Renal histology and clinical chemistry results did not differ between radiotherapy groups and controls, suggesting absence of therapy-induced acute renal damage. Conclusion: These data demonstrate that the novel NTR1-targeting theranostic agent 3BP-227 is an effective and promising candidate for radioligand therapy, with a favorable preliminary safety profile and high potential for clinical translation.


Subject(s)
Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/radiotherapy , Lutetium/therapeutic use , Molecular Targeted Therapy/methods , Receptors, Neurotensin/antagonists & inhibitors , Theranostic Nanomedicine/methods , Animals , Apoptosis/radiation effects , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , HT29 Cells , Humans , Mice , Mice, Nude , Radiopharmaceuticals/therapeutic use , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome
5.
J Nucl Med ; 57(7): 1120-3, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26940767

ABSTRACT

UNLABELLED: Neurotensin receptor-1 (NTR1) is a promising target for diagnostic imaging and targeted radionuclide therapy. The aim of this study was to evaluate the biodistribution profiles of a series of newly developed diarylpyrazole-based NTR1 antagonists regarding their suitability as diagnostic and potentially radiotherapeutic agents. METHODS: 3BP-227, 3BP-228, and 3BP-483 were labeled with (111)In and injected intravenously into NTR1-positive HT29 xenograft-bearing nude mice. At 3, 6, 12, and 24 h after administration, SPECT/CT images were acquired or mice were sacrificed for ex vivo determination of tissue-associated radioactivity. RESULTS: High-contrast tumor visualization in SPECT/CT images was achieved using the 3 compounds of this study. Ex vivo biodistribution studies confirmed a high and persistent tumor uptake, peaking at 6 h after injection for (111)In-3BP-227 (8.4 ± 3.1 percentage injected dose per gram [%ID/g]) and at 3 h after injection for (111)In-3BP-228 (10.2 ± 5.3 %ID/g) and (111)In-3BP-483 (1.9 ± 0.8 %ID/g). Tumor-to-normal-tissue ratios obtained with (111)In-3BP-227 and (111)In-3BP-228 were consistently greater than 1. CONCLUSION: On the basis of the superior biodistribution profile compared with previously reported radiolabeled NTR1 ligands, (111)In-3BP-227 is an ideal candidate for further development as a theranostic tracer.


Subject(s)
Receptors, Neurotensin/antagonists & inhibitors , Theranostic Nanomedicine/methods , Animals , Cell Line, Tumor , Humans , Indium Radioisotopes , Isotope Labeling , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Tissue Distribution , Tomography, Emission-Computed, Single-Photon
6.
J Biol Chem ; 289(3): 1732-41, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24275667

ABSTRACT

Tissue factor pathway inhibitor (TFPI) is a Kunitz-type protease inhibitor that inhibits activated factor X (FXa) via a slow-tight binding mechanism and tissue factor-activated FVII (TF-FVIIa) via formation of a quaternary FXa-TFPI-TF-FVIIa complex. Inhibition of TFPI enhances coagulation in hemophilia models. Using a library approach, we selected and subsequently optimized peptides that bind TFPI and block its anticoagulant activity. One peptide (termed compound 3), bound with high affinity to the Kunitz-1 (K1) domain of TFPI (Kd ∼1 nM). We solved the crystal structure of this peptide in complex with the K1 of TFPI at 2.55-Å resolution. The structure of compound 3 can be segmented into a N-terminal anchor; an Ω-shaped loop; an intermediate segment; a tight glycine-loop; and a C-terminal α-helix that is anchored to K1 at its reactive center loop and two-stranded ß-sheet. The contact surface has an overall hydrophobic character with some charged hot spots. In a model system, compound 3 blocked FXa inhibition by TFPI (EC50 = 11 nM) and inhibition of TF-FVIIa-catalyzed FX activation by TFPI (EC50 = 2 nM). The peptide prevented transition from the loose to the tight FXa-TFPI complex, but did not affect formation of the loose FXa-TFPI complex. The K1 domain of TFPI binds and inhibits FVIIa and the K2 domain similarly inhibits FXa. Because compound 3 binds to K1, our data show that K1 is not only important for FVIIa inhibition but also for FXa inhibition, i.e. for the transition of the loose to the tight FXa-TFPI complex. This mode of action translates into normalization of coagulation of hemophilia plasmas. Compound 3 thus bears potential to prevent bleeding in hemophilia patients.


Subject(s)
Coagulants/chemistry , Factor VIIa/chemistry , Factor Xa/chemistry , Lipoproteins/antagonists & inhibitors , Peptides/chemistry , Blood Coagulation/drug effects , Coagulants/chemical synthesis , Coagulants/metabolism , Coagulants/therapeutic use , Factor VIIa/metabolism , Factor Xa/metabolism , Hemophilia A/drug therapy , Hemophilia A/metabolism , Hemorrhage/drug therapy , Hemorrhage/metabolism , Humans , Lipoproteins/chemistry , Lipoproteins/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Peptides/therapeutic use , Protein Structure, Secondary , Protein Structure, Tertiary
7.
Med Microbiol Immunol ; 200(2): 85-97, 2011 May.
Article in English | MEDLINE | ID: mdl-20931340

ABSTRACT

Monoclonal antibody (MAb) 2c, specific for glycoprotein B of herpes simplex virus (HSV), had been shown to mediate clearance of infection from the mucous membranes of mice, thereby completely inhibiting mucocutaneous inflammation and lethality, even in mice depleted of both CD4(+) and CD8(+) cells. Additionally, ganglionic infection was highly restricted. In vitro, MAb 2c exhibits a potent complement-independent neutralising activity against HSV type 1 and 2, completely inhibits the viral cell-to-cell spread as well as the syncytium formation induced by syncytial HSV strains (Eis-Hübinger et al. in Intervirology 32:351-360, 1991; Eis-Hübinger et al. in J Gen Virol 74:379-385, 1993). Here, we describe the mapping of the epitope for MAb 2c. The antibody was found to recognise a discontinuous epitope comprised of the HSV type 1 glycoprotein B residues 299 to 305 and one or more additional discontinuous regions that can be mimicked by the sequence FEDF. Identification of the epitope was confirmed by loss of antibody binding to mutated glycoprotein B with replacement of the epitopic key residues, expressed in COS-1 cells. Similarly, MAb 2c was not able to neutralise HSV mutants with altered key residues, and MAb 2c was ineffective in mice inoculated with such mutants. Interestingly, identification and fine-mapping of the discontinuous epitope was not achieved by binding studies with truncated glycoprotein B variants expressed in COS cells but by peptide scanning with synthetic overlapping peptides and peptide key motif analysis. Reactivity of MAb 2c was immensely increased towards a peptide composed of the glycoprotein B residues 299 to 305, a glycine linker, and a C-terminal FEDF motif. If it could be demonstrated that antibodies of the specificity and bioactivity of MAb 2c can be induced by the epitope or a peptide mimicking the epitope, strategies for active immunisation might be conceivable.


Subject(s)
Antibodies, Monoclonal/immunology , Epitope Mapping , Herpes Simplex/prevention & control , Herpesvirus 1, Human/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/immunology , Antibody Specificity , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , Epitopes/chemistry , Epitopes/genetics , Female , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/pathogenicity , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
9.
Methods Mol Biol ; 524: 145-67, 2009.
Article in English | MEDLINE | ID: mdl-19377943

ABSTRACT

Information at the amino acid level about the epitopes of proteins recognized by antibodies or antibody fragments is important for their use as biological and diagnostic tools, therapeutic molecules, and for understanding molecular recognition events in general. The use of chemically prepared arrays of short peptides has emerged as a powerful tool to identify and characterize antibody epitopes. In this chapter the SPOT synthesis technique is described in detail. In addition, three different types of peptide libraries and their applications are described: protein sequence-derived scans of overlapping peptides (peptide scans) used to locate epitopes within the protein sequence, truncation libraries used to identify the minimal peptide length required for antibody binding, and complete substitutional analyses to identify the key residues important for antibody binding.


Subject(s)
Epitope Mapping/methods , Peptide Library , Acetylation , Amino Acid Sequence , Animals , Antibodies/immunology , Humans , Luminescent Measurements , Membranes, Artificial , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Peptides/immunology , Protein Array Analysis , Protein Binding
10.
Methods Mol Biol ; 524: 203-11, 2009.
Article in English | MEDLINE | ID: mdl-19377946

ABSTRACT

Identification of antibody binding peptides may be based on the primary structure of the protein antigens used to raise the antibodies (knowledge- or sequence-based approach). This involves scanning the entire sequence of the antigen with overlapping peptides (peptide scan), which are then probed for binding to the respective antibody. If a natural protein binding partner is not known, one has to use combinatorial synthetic libraries with peptide mixtures, randomly generated chemically synthesized libraries of single individual sequences, or biologically produced libraries (e.g., phage display libraries, see Chapter "Epitope Mapping Using Phage Display Peptide Libraries"). This chapter describes chemically synthesized combinatorial, as well as randomly generated peptide libraries, collectively called de novo approaches, and their application for antibody epitope mapping.


Subject(s)
Antibodies/immunology , Epitope Mapping/methods , Peptide Library , Animals , Humans , Interleukin-10/immunology
11.
J Mol Recognit ; 20(3): 185-96, 2007.
Article in English | MEDLINE | ID: mdl-17486692

ABSTRACT

Nogo-A is a potent inhibitor of axonal outgrowth in the central nervous system of adult mammals, where it is expressed as a membrane protein on oligodendrocytes and in myelin. Here we describe an attempt to identify linear peptide epitopes in its sequence that are responsible for the interaction either with the Nogo receptor (NgR) or with the neutralizing monoclonal antibody IN-1. Analysis of an array of immobilized overlapping 15 mer peptides covering the entire amino acid sequence of human Nogo-A (1192 residues) revealed a single epitope with prominent binding activity both towards the recombinant NgR and the IN-1 F(ab) fragment. Further truncation and substitution analysis yielded the minimal epitope sequence 'IKxLRRL' (x not equal to P), which occurs within the so-called Nogo66 region (residues 1054-1120) of Nogo-A. The bacterially produced Nogo66 fragment exhibited binding activity both for the recombinant NgR and for the IN-1 F(ab) fragment on the Western blot as well as in ELISA. Unexpectedly, the synthetic epitope peptide and the recombinant Nogo66 showed cross-reactivity with the 8-18C5 F(ab) fragment, which is directed against myelin oligodendrocyte glycoprotein (MOG) as a structurally unrelated target. On the other hand, the recombinant N-terminal domain of Nogo-A (residues 334-966) was shown to specifically interact on the Western blot and in an ELISA with the IN-1 F(ab) fragment but not with the recombinant NgR, which is in agreement with previous results. Hence, our data suggest that there is a distinct binding site for the Nogo receptor in the Nogo66 region of Nogo-A, whereas its interaction with NgR is less specific than anticipated before. Although there probably exists a non-linear epitope for the neutralizing antibody IN-1 in the N-terminal region of Nogo-A, which is likely to be accessible from outside the cell, a previously postulated second binding site for NgR in this region (called Nogo-A-24) remains elusive.


Subject(s)
Antibodies, Monoclonal/immunology , Combinatorial Chemistry Techniques , Epitope Mapping , Immunoglobulin Fab Fragments/immunology , Myelin Proteins/immunology , Receptors, Cell Surface/immunology , Binding Sites, Antibody/genetics , Binding Sites, Antibody/immunology , Blotting, Western , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Epitopes , Fluorescence , GPI-Linked Proteins , Humans , Myelin Proteins/metabolism , Nogo Proteins , Nogo Receptor 1 , Peptide Fragments/immunology , Peptide Fragments/metabolism , Plasmids , Protein Engineering , Receptors, Cell Surface/metabolism
12.
Protein Pept Lett ; 13(8): 829-33, 2006.
Article in English | MEDLINE | ID: mdl-17073730

ABSTRACT

We report the stepwise transformation of a linear peptide epitope recognized by the anti-transforming growth factor alpha monoclonal antibody Tab2 into peptomers and finally into peptoid analogs. The key experiment in this study is the substitution analysis in which each position of the peptide is exchanged by a set of different peptoid building blocks resulting in a peptidomimetic array. After probing the array toward antibody binding, the best binding peptomer spots were selected and subjected to a successive transformation. The best peptoid found in this study has a K(D) of 200 nM when binding to Tab2, which is only 8-fold higher than the starting peptide. Moreover, this approach permits to ask directly questions about the transformation of peptide lead structures into non-peptidic compounds in the context of protein recognition.


Subject(s)
Peptides/chemistry , Peptoids/chemical synthesis , Antibodies, Monoclonal/metabolism , Binding, Competitive , Epitopes/chemistry , Epitopes/immunology , Kinetics , Molecular Structure , Peptides/metabolism , Peptoids/chemistry , Protein Binding , Transforming Growth Factor alpha/chemistry , Transforming Growth Factor alpha/immunology
15.
Mol Cancer Ther ; 3(11): 1439-50, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542783

ABSTRACT

OBJECTIVE: Prostate cancer cells secrete the unique protease human glandular kallikrein 2 (hK2) that represents a target for proteolytic activation of cytotoxic prodrugs. The objective of this study was to identify hK2-selective peptide substrates that could be coupled to a cytotoxic analogue of thapsigargin, a potent inhibitor of the sarcoplasmic/endoplasmic reticulum calcium ATPase pump that induces cell proliferation-independent apoptosis through dysregulation of intracellular calcium levels. METHODS: To identify peptide sequence requirements for hK2, a combination of membrane-bound peptides (SPOT analysis) and combinatorial chemistry using fluorescence-quenched peptide substrates was used. Peptide substrates were then coupled to 8-O-(12[L-leucinoylamino]dodecanoyl)-8-O-debutanoylthapsigargin (L12ADT), a potent analogue of thapsigargin, to produce a prodrug that was then characterized for hK2 hydrolysis, plasma stability, and in vitro cytotoxicity. RESULTS: Both techniques indicated that a peptide with two arginines NH2-terminal of the scissile bond produced the highest rates of hydrolysis. A lead peptide substrate with the sequence Gly-Lys-Ala-Phe-Arg-Arg (GKAFRR) was hydrolyzed by hK2 with a Km of 26.5 micromol/L, kcat of 1.09 s(-1), and a kcat/Km ratio of 41,132 s(-1) mol/L(-1). The GKAFRR-L12ADT prodrug was rapidly hydrolyzed by hK2 and was stable in plasma, whereas the GKAFRR-L peptide substrate was unstable in human plasma. The hK2-activated thapsigargin prodrug was not activated by cathepsin B, cathepsin D, and urokinase but was an excellent substrate for plasmin. The GKAFRR-L12ADT was selectively cytotoxic in vitro to cancer cells in the presence of enzymatically active hK2. CONCLUSION: The hK2-activated thapsigargin prodrug represents potential novel targeted therapy for prostate cancer.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Peptide Library , Prodrugs/metabolism , Prodrugs/pharmacology , Prostatic Neoplasms/pathology , Tissue Kallikreins/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cell Line, Tumor , Drug Evaluation, Preclinical , Humans , Hydrolysis , Male , Mice , Molecular Structure , Peptides/chemistry , Peptides/metabolism , Prodrugs/chemistry , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Substrate Specificity , Thapsigargin/analogs & derivatives , Thapsigargin/chemistry , Thapsigargin/metabolism , Thapsigargin/pharmacology , Tissue Kallikreins/blood , Trypsin/metabolism
16.
FASEB J ; 18(7): 893-5, 2004 May.
Article in English | MEDLINE | ID: mdl-15033921

ABSTRACT

Tumor necrosis factor (TNF)-alpha converting enzyme (TACE) is responsible for the ectodomain release of various membrane proteins by proteolytic cleavage in close proximity to the cell membrane. Despite the wide spectrum of possible substrates, selective cleavage can be achieved by substrate cross-linking. To explore the underlying mechanism, we studied the TACE-mediated shedding of CD30. Whereas the constitutive release of the soluble ectodomain of CD30 (sCD30) from the lymphoma cell line Karpas 299 was enhanced by most anti-CD30 antibodies, it was inhibited by antibodies Ber-H2 and Ki-4. On the basis of the recognized epitopes, shedding seemed to depend on the availability of the cysteine-rich domains (CRD) 2 and 5 of the CD30 ectodomain. CRD2 and 5 have almost identical amino acid sequences and are localized distant from the TACE-targeted cleavage site. Soluble CD30, the product of this enzyme reaction, did not inhibit, but on the contrary, it stimulated CD30 shedding in a CRD2/5-dependent manner. This process could also be induced by CRD2/5-derived peptides but not by a CRD1-derived control peptide. This example of a product-activation was CD30 selective since other TACE substrates such as TNFR1 or TNF-alpha were not affected. These data suggest that CD30 shedding is stimulated by an elevated local availability of CRD2 or 5, possibly by forming a docking station for the releasing enzyme through substrate aggregation.


Subject(s)
Ki-1 Antigen/chemistry , ADAM Proteins , ADAM17 Protein , Amino Acid Motifs , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , COS Cells , Cell Line, Tumor/metabolism , Chlorocebus aethiops , Cysteine/chemistry , Epitopes/immunology , Hodgkin Disease/pathology , Humans , Hydrophobic and Hydrophilic Interactions , Immunoglobulin Fab Fragments/immunology , Ki-1 Antigen/immunology , Ki-1 Antigen/metabolism , Lymphoma, Non-Hodgkin/pathology , Metalloendopeptidases/metabolism , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Solubility , Structure-Activity Relationship , Substrate Specificity , Transfection
18.
Proc Natl Acad Sci U S A ; 100(7): 4072-7, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12646696

ABSTRACT

A kinase-anchoring proteins (AKAPs) coordinate cAMP-mediated signaling by binding and localizing cAMP-dependent protein kinase (PKA), using an amphipathic helical docking motif. Peptide disruptors of PKA localization that mimic this helix have been used successfully to assess the involvement of PKA in specific signaling pathways. However, these peptides were developed as disruptors for the type II regulatory subunit (RII) even though both RI and RII isoforms can bind to AKAPs and have discrete functions. To evaluate the effects of each localized isoform, we designed peptides that specifically bind to either RI or RII. Using a peptide array, we have defined the minimal binding sequence of dual specific-AKAP 2 (d-AKAP2), which binds tightly to both RI and RII. Side-chain requirements for affinity and isoform specificity were evaluated by using a peptide substitution array where each position along the A kinase binding domain of d-AKAP2 was substituted by the other 19 l-amino acids. This array comprises 513 single-site substitution analogs of the d-AKAP2 sequence. Peptides containing single and multiple mutations were evaluated in a quantitative fluorescence binding assay and a cell-based colocalization assay. This strategy has allowed us to design peptides with high affinity (K(D) = 1-2 nM) and high specificity for RIalpha versus RIIalpha. These isoform-specific peptides will be invaluable tools to evaluate functional differences between localized RI and RII PKA and are RIalpha-specific disruptors. This array-based analysis also provides a foundation for biophysical analysis of this docking motif.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Peptides/chemistry , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cattle , Cloning, Molecular , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Dimerization , Humans , Mice , Molecular Sequence Data , Peptides/chemical synthesis , Protein Subunits/chemistry , Protein Subunits/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion
19.
Vaccine ; 20(31-32): 3685-94, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12399196

ABSTRACT

T helper cell-recognized epitopes were determined in chitinase of Onchocerca volvulus, a vaccine candidate protein. The proliferation of splenic T cells of mice immunized with recombinant protein was tested with a library of chitinase-peptides of 16 amino acids with termini overlapping by 12 amino acids, and a library of "designer peptides", i.e. sequences identified with three epitope-predicting algorithms. Fourteen epitope-bearing stretches were identified with the peptides of the overlapping library. Testing of the designer peptides partially confirmed these data and revealed additional epitopes. Five clusters of epitopes were identified for the creation of peptide or minigene DNA vaccines with good potency and potential range of MHC allele presentation.


Subject(s)
Chitinases/chemistry , Chitinases/immunology , Epitopes, T-Lymphocyte/immunology , Onchocerca volvulus/immunology , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Antigens, Helminth/biosynthesis , Antigens, Helminth/chemistry , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Chitinases/biosynthesis , Chitinases/genetics , Cloning, Molecular , DNA, Complementary/genetics , DNA, Helminth/genetics , Epitopes, T-Lymphocyte/genetics , H-2 Antigens/genetics , Helminth Proteins/biosynthesis , Helminth Proteins/chemistry , Helminth Proteins/genetics , Helminth Proteins/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Onchocerca volvulus/enzymology , Peptides/chemistry , Peptides/genetics , Peptides/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocyte Subsets/immunology , Vaccines, Synthetic/genetics
20.
Biol Chem ; 383(6): 1011-20, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12222675

ABSTRACT

The ectodomain of the human transferrin receptor (TfR) is released as soluble TfR into the blood by cleavage within a stalk. The major cleavage site is located C-terminally of Arg-100; alternative cleavage sites are also present. Since the cleavage process is still unclear, we looked for proteases involved in TfR ectodomain release. In the supernatant of U937 histiocytic cells we detected alternatively cleaved TfR (at Glu-110). In membrane fractions of these cells we identified two distinct proteolytic activities responsible for TfR cleavage within the stalk at either Val-108 or Lys-95. Both activities could be inhibited by serine protease inhibitors, but not by inhibitors of any other class of proteases. Protein purification yielded a 28 kDa protein that generated the Val-108 terminus. The protease activity could be ascribed to neutrophil elastase according to the substrate specificity determined by amino acid substitution analysis of synthetic peptides, an inhibitor profile, the size of the protease and the use of specific antibodies. The results of analogous experiments suggest that the second activity is represented by another serine protease, cathepsin G. Thus, membrane-associated forms of neutrophil elastase and cathepsin G may be involved in alternative TfR shedding in U937 cells.


Subject(s)
Cathepsins/metabolism , Leukocyte Elastase/metabolism , Receptors, Transferrin/metabolism , Amino Acid Sequence , Biotin/metabolism , Cathepsin G , Cell Line , Cell-Free System , Cells, Cultured , Culture Media , Electrochemistry , Electrophoresis, Polyacrylamide Gel , Endopeptidases/analysis , Endopeptidases/isolation & purification , Humans , Immunohistochemistry , Leukemia/metabolism , Membranes/enzymology , Molecular Sequence Data , Protein Conformation , Receptors, Transferrin/chemistry , Serine Endopeptidases
SELECTION OF CITATIONS
SEARCH DETAIL
...