Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 90(9): 1863-70, 2004 May 04.
Article in English | MEDLINE | ID: mdl-15150594

ABSTRACT

Anti-MUC1 single-chain Fv (scFv) fragments generated from the humanised antibody huHMFG1 had adequate antigen-binding properties but very poor stability irrespective of the applied linker or domain orientation. Mutagenesis of heavy-chain framework residue V(H)-71, previously described as a key residue for maintaining the CDR-H2 main-chain conformation and thus important for antigen binding, markedly stabilised the scFv while having only a minor effect on the binding affinity of the molecule. Because of its improved stability, the engineered fragment exhibited immunoreactivity with tumour cells even after 7 days of incubation in human serum at 37 degrees C. It also showed, in contrast to the wild-type scFv, a concentration-dependent binding to the target antigen when displayed on phage. When fusing the scFv to the recombinant ribonuclease rapLRI, only the fusion protein generated with the stable mutant scFv was able to kill MUC1(+) tumour cells with an IC(50) of 80 nM. We expect this novel immunoenzyme to become a promising tool for the treatment of MUC1(+) malignancies.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibody Affinity , Membrane Proteins/immunology , Protein Engineering , Amino Acid Sequence , Animals , Computer Simulation , Enzyme Stability , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Models, Immunological , Mutation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Tumor Cells, Cultured
2.
Expert Opin Biol Ther ; 1(6): 995-1003, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11728231

ABSTRACT

The approval of antibodies for cancer treatment has provoked increased interest in the development of new and improved antibody-mediated therapies. This emerging approach centres on targeting CD22 on human B-cells with a monoclonal antibody (mAb). Anti-CD22 antibodies conjugated to a cytotoxic RNAse elicits potent and specific killing of the lymphoma cells in vitro and in human lymphoma models in severe combined immune deficiency (SCID) mice. RNA damage caused by RNAses could be an important alternative to standard DNA damaging chemotherapeutics. Moreover, targeted RNAses may overcome problems of toxicity and immunogenicity associated with plant- or bacterial toxin-containing immunotoxins.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Cell Adhesion Molecules , Lectins , Lymphoma, Non-Hodgkin/therapy , RNA/antagonists & inhibitors , Animals , Humans , Mice , Mice, SCID , Sialic Acid Binding Ig-like Lectin 2
3.
Crit Rev Oncol Hematol ; 39(1-2): 79-86, 2001.
Article in English | MEDLINE | ID: mdl-11418304

ABSTRACT

Targeting CD22 on human B-cells with a monoclonal antibody conjugated to a cytotoxic RNAse causes potent and specific killing of the lymphoma cells in vitro. This translates to anti-tumor effects in human lymphoma models in SCID mice. RNA damage caused by RNAses could be an important alternative to standard DNA damaging chemotherapeutics. Moreover, targeted RNAses may overcome problems of toxicity and immunogenicity associated with plant or bacterial toxin containing immunotoxins.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Cell Adhesion Molecules , Immunotoxins/therapeutic use , Lectins , Lymphoma, B-Cell/drug therapy , Ribonucleases/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Humans , Immunotoxins/chemistry , RNA/metabolism , Sialic Acid Binding Ig-like Lectin 2
5.
Blood ; 97(2): 528-35, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11154233

ABSTRACT

LL2, an anti-CD22 monoclonal antibody against B-cell lymphoma, was covalently linked to the amphibian ribonuclease, onconase, a member of the pancreatic RNase A superfamily. LL2 increased in vitro potency (10 000-fold) and specificity against human Daudi Burkitt lymphoma cells while decreasing systemic toxicity of onconase. Monensin further increased potency of LL2-onconase on Daudi cells (IC(50), 20 and 1.5 pM, absence and presence of monensin, respectively). A 1-hour exposure to LL2-onconase was sufficient to kill Daudi cells in culture. These favorable in vitro properties translated to significant antitumor activity against disseminated Daudi lymphoma in mice with severe combined immunodeficiency disease. In mice inoculated with tumor cells intraperitoneally (ip), LL2-onconase (100 microg 5 times ip every day) increased the life span of animals with minimal disease 200%. The life span of mice with advanced disseminated Daudi lymphoma (tumor cells inoculated intravenously) was increased 135%. Mice injected with LL2-onconase tolerated a dose as high as 300 mg/kg. Because both onconase and LL2 are in clinical trials as cancer therapeutics, the covalently linked agents should be considered for treatment of non-Hodgkin lymphoma.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cell Adhesion Molecules , Lectins , Ribonucleases/pharmacology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/toxicity , Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Death/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Stability , Female , Humans , Immunotoxins/pharmacology , Immunotoxins/therapeutic use , Immunotoxins/toxicity , Kinetics , Lymphoma, Non-Hodgkin/drug therapy , Mice , Mice, Inbred BALB C , Mice, SCID , Models, Animal , Neoplasm Transplantation , Pancreas/enzymology , Ribonucleases/therapeutic use , Ribonucleases/toxicity , Sialic Acid Binding Ig-like Lectin 2 , Survival Rate , Tumor Cells, Cultured
6.
Radiat Res ; 155(1 Pt 2): 171-174, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11121230

ABSTRACT

Angiogenesis defines the many steps involved in the growth and migration of endothelial cell-derived blood vessels. This process is necessary for the growth and metastasis of tumors, and considerable effort is being expended to find inhibitors of tumor angiogenesis. This usually involves screening of potential anti-angiogenic compounds on endothelial cells. To this end, two candidate anti-angiogenic RNA-damaging agents, onconase and (-4)rhEDN, were screened for their effects on endothelial cell proliferation using three distinct types of endothelial cells in culture: HPV-16 E6/E7-immortalized human umbilical vein endothelial cells (HUVECs), a Kras-transformed HPV-16 E6/E7 HUVEC (Rhim et al., Carcinogenesis 4, 673-681, 1998), and primary HUVECs. Onconase similarly inhibited proliferation in all three cell lines (IC(50) = 0.3-1.0 microM) while (-4)rhEDN was more effective on immortalized HUVEC cell lines (IC(50) = 0.02-0.06 microM) than on primary HUVECs (IC(50) > 0.1 microM). Differential sensitivity to these agents implies that more than one endothelial cell type must be used in proliferation assays to screen for novel anti-angiogenic compounds.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Egg Proteins/pharmacology , Endothelium, Vascular/drug effects , Proteins/pharmacology , Ribonuclease, Pancreatic/pharmacology , Ribonucleases/pharmacology , Animals , Cell Division/drug effects , Cell Line, Transformed , Cells, Cultured , Egg Proteins/toxicity , Endothelium, Vascular/cytology , Eosinophil-Derived Neurotoxin , Growth Inhibitors/pharmacology , Growth Inhibitors/toxicity , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Proteins/toxicity , RNA/drug effects , RNA/metabolism , Rana pipiens , Recombinant Proteins/pharmacology , Ribonuclease, Pancreatic/toxicity , Ribonucleases/toxicity
7.
Nucleic Acids Res ; 28(12): 2375-82, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10871370

ABSTRACT

A cDNA (2855 nt) encoding a putative cytotoxic ribonuclease (rapLR1) related to the antitumor protein onconase was cloned from a library derived from the liver of gravid female amphibian Rana pipiens. The cDNA was mainly comprised (83%) of 3' untranslated region (UTR). Secondary structure analysis predicted two unusual folding regions (UFRs) in the RNA 3' UTR. Two of these regions (711-1442 and 1877-2130 nt) contained remarkable, stalk-like, stem-loop structures greater than 38 and 12 standard deviations more stable than by chance, respectively. Secondary structure modeling demonstrated similar structures in the 3' UTRs of other species at low frequencies (0.01-0.3%). The size of the rapLR1 cDNA corresponded to the major hybridizing RNA cross-reactive with a genomic clone encoding onconase (3.6 kb). The transcript was found only in liver mRNA from female frogs. In contrast, immunoreactive onconase protein was detected only in oocytes. Deletion of the 3' UTR facilitated the in vitro translation of the rapLR1 cDNA. Taken together these results suggest that these unusual UFRs may affect mRNA metabolism and/or translation.


Subject(s)
3' Untranslated Regions , Liver/enzymology , RNA, Messenger/chemistry , RNA, Messenger/genetics , Ribonucleases/genetics , Animals , Base Sequence , DNA, Complementary , Female , Gene Library , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Oocytes/enzymology , Open Reading Frames , Protein Biosynthesis , Rana pipiens , Software , Transcription, Genetic
8.
Cancer Res ; 60(7): 1983-94, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10766189

ABSTRACT

Cytotoxic endoribonucleases (RNases) possess a potential for use in cancer therapy. However, the molecular determinants of RNase-induced cell death are not well understood. In this work, we identify such determinants of the cytotoxicity induced by onconase, an amphibian cytotoxic RNase. Onconase displayed a remarkable specificity for tRNA in vivo, leaving rRNA and mRNA apparently undamaged. Onconase-treated cells displayed apoptosis-associated cell blebbing, nuclear pyknosis and fragmentation (karyorrhexis), DNA fragmentation, and activation of caspase-3-like activity. The cytotoxic action of onconase correlated with inhibition of protein synthesis; however, we present evidence for the existence of a mechanism of onconase-induced apoptosis that is independent of inhibition of protein synthesis. The caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe) fluoromethyl ketone (zVADfmk), at concentrations that completely prevent apoptosis and caspase activation induced by ligation of the death receptor Fas, had only a partial protective effect on onconase-induced cell death. The proapoptotic activity of the p53 tumor suppressor protein and the Fas ligand/Fas/Fas-associating protein with death domain (FADD)/caspase-8 proapoptotic cascade were not required for onconase-induced apoptosis. Procaspases-9, -3, and -7 were processed in onconase-treated cells, suggesting the involvement of the mitochondrial apoptotic machinery in onconase-induced apoptosis. However, the onconase-induced activation of the caspase-9/caspase-3 cascade correlated with atypically little release of cytochrome c from mitochondria. In turn, the low levels of cytochrome c released from mitochondria correlated with a lack of detectable translocation of proapoptotic Bax from the cytosol onto mitochondria in response to onconase. This suggests the possibility of involvement of a different, potentially Bax- and cytochrome c-independent mechanism of caspase-9 activation in onconase-treated cells. As one possible mechanism, we demonstrate that procaspase-9 is released from mitochondria in onconase-treated cells. A detailed understanding of the molecular determinants of the cytotoxic action of onconase could provide means of positive or negative therapeutic modulation of the activity of this potent anticancer agent.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Egg Proteins/metabolism , Egg Proteins/toxicity , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/toxicity , Proto-Oncogene Proteins c-bcl-2 , RNA, Transfer/metabolism , Ribonucleases/metabolism , Ribonucleases/toxicity , Apoptosis/physiology , Cell Death/drug effects , Cell Survival/drug effects , Cycloheximide/toxicity , Cytochrome c Group/metabolism , Emetine/toxicity , HeLa Cells , Humans , Leucine/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Substrate Specificity , bcl-2-Associated X Protein
9.
Mol Cell Biol Res Commun ; 4(2): 122-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11170843

ABSTRACT

Onconase, an anticancer ribonuclease, damages cellular tRNA and causes caspase-dependent apoptosis in targeted cells (M. S. Iordanov, O. P. Ryabinina, J. Wong, T. H. Dinh, D. L. Newton, S. M. Rybak, and B. E. Magun. Cancer Res. 60, 1983-1994, 2000). The proapoptotic action of onconase depends on its RNase activity, but the molecular mechanisms leading to RNA damage-induced caspase activation are completely unknown. In this study, we have investigated whether onconase activates two signal-transduction pathways commonly stimulated by conventional chemo- and radiotherapy, namely the stress-activated protein kinase (SAPK) cascade and the pathway leading to the activation of nuclear factor-kappa B (NF-kappaB). We found that, in all cell types tested, onconase is a potent activator of SAPK1 (JNK1 and JNK2) and SAPK2 (p38 MAP kinase), but that it is incapable of activating NF-kappaB. Inhibition of p38 MAP kinase activity with a pharmacological inhibitor, SB203580, demonstrated that p38 MAP kinase is not required for onconase cytotoxicity. Using explanted fibroblasts from mice that contain targeted disruption of both jnk1 and jnk2 alleles, we found that JNKs are important mediators of onconase-induced cytotoxicity. Surprisingly, following the immortalization of these same cells with human papilloma virus (HPV16) gene products E6 and E7, additional proapoptotic pathways (exclusive of JNK) were provoked by onconase. Our results demonstrate that onconase may activate proapoptotic pathways in tumor cells that are not able to be accessed in normal cells. These results present the possibility that the cytotoxic activity of onconase in normal cells may be reduced by blocking the activity of JNKs.


Subject(s)
Apoptosis/drug effects , Calcium-Binding Proteins , Caspases/metabolism , Mitogen-Activated Protein Kinases/metabolism , RNA/metabolism , Cell Survival/drug effects , Enzyme Activation , Enzyme Inhibitors , Eosine Yellowish-(YS) , HeLa Cells/drug effects , Hematoxylin , Humans , Immunoblotting , Ligases/metabolism , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins , Phosphorylation , Synaptotagmins , Transfection , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/virology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases
10.
Methods Mol Med ; 25: 27-35, 2000.
Article in English | MEDLINE | ID: mdl-21318838

ABSTRACT

Immunotoxins based on human and humanized ribonuclease may have potential for cancer therapy while exhibiting less toxic side effects and stimulating less of an immune response in humans than immunotoxins based on plant and bacterial toxins (1). Both recombinant RNase fusion proteins (2-4 see also Chapter 6 , this volume) and chemical RNase conjugates have been made and characterized. The cytotoxic potential of targeted ribonuclease was first demonstrated with bovine RNase conjugated to transferrin or an antibody directed against the human transferrin receptor (5). Antibody RNase conjugates have also been shown to have potent anti-tumor activity against human glioma cells in athymic mice (6) and to enhance the activity of vincristine in mdr1 multidrug-resistant colon cancer cells in vitro and in vivo (7). Recently, RNase chemically conjugated to an antibody against CD22 was found to specifically kill Daudi lymphoma cells in cell culture at picomolar concentrations (IC(50), 10-50 pM) and to exhibit potent antitumor activity in SCID mice with disseminated Daudi lymphoma (unpublished data). Methods for linking RNase to specific cell binding ligands are described.

11.
Methods Mol Med ; 25: 77-95, 2000.
Article in English | MEDLINE | ID: mdl-21318842

ABSTRACT

Selective cytotoxicity is an important goal of specific drug targeting. Toward this end, toxins isolated primarily from higher plants and bacteria have been coupled to monoclonal antibodies (MAbs) and evaluated for their clinical efficacy in cancer, AIDS, and immunological diseases (1,2). Immune responses against murine monoclonal antibodies MAbs (3,4) and antitoxin antibodies have been detected in both animals and humans treated with immunotoxins (ITs) (5-7) and present a major obstacle to the successful application of this technology. Although development of humanized antibodies have alleviated some of these effects (8, and references therein), the toxins themselves remain a problem. Consequently, the identification of human proteins to be used as components of immunoconjugates is highly desirable.

14.
Exp Cell Res ; 247(1): 220-32, 1999 Feb 25.
Article in English | MEDLINE | ID: mdl-10047464

ABSTRACT

Microinjection of Onconase or RNase A into NIH/3T3 cells was used to study the intracellular actions of these two proteins. Onconase preferentially killed actively growing cells in both microinjection and cell culture experiments. Moreover, agents that increased the number of cells in S phase such as serum or microinjected signal transduction mediators (Ras, protein kinase C, and mitogen-activated protein kinase) enhanced Onconase cytotoxicity. Conversely, agents that decreased these proliferative pathways (dibutyryl cAMP and protein kinase A) correspondingly diminished Onconase cytotoxicity in microinjection experiments. These results were also mimicked in cell culture experiments since log-phase v-ras-transformed NIH/3T3 cells were more sensitive to Onconase (IC50 of 7 microg/ml) than parental NIH/3T3 fibroblasts (IC50 of 40 microg/ml). Based on those data we postulated that Onconase-mediated cell death in NIH/3T3 cells was related to events occurring at two or more points in the cell cycle preferentially associated with late G1/S and S phases. In contrast, quiescent NIH/3T3 cells were more sensitive to microinjected RNase A than log phase cells and positive mediators of proliferative signal transduction did not enhance RNase A-mediated cytotoxicity. Taken together, these results demonstrate that these two RNases use different pathways and/or mechanisms to elicit cytotoxic responses in NIH/3T3 cells. Predictions formulated from these studies can be tested for relevance to RNase actions in different target tumor cells.


Subject(s)
3T3 Cells/drug effects , Cell Cycle/drug effects , Ribonucleases/toxicity , 3T3 Cells/enzymology , Animals , Annexin A5/pharmacology , Cattle , Cell Death/drug effects , Cell Line, Transformed , Cell Transformation, Viral , Culture Media, Conditioned , Culture Media, Serum-Free , Drug Synergism , Egg Proteins/toxicity , Extracellular Space/enzymology , Interphase/drug effects , Mice , Microinjections , Oncogene Protein p21(ras)/physiology , Ribonuclease, Pancreatic/toxicity
15.
J Immunol Methods ; 231(1-2): 159-67, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10648935

ABSTRACT

Antibodies fused to human enzymes offer an alternative to specifically targeting tumors with antibodies linked to plant or bacterial toxins. Since large amounts of these reagents can be administered without eliciting non-specific toxicities, efficient methods of production are needed. The goal of this work was to express a complex immunoenzyme fusion protein (immunotoxin) in the mammary gland of transgenic mice. A chimeric mouse/human antibody directed against the human transferrin receptor (E6) was fused at its CH2 domain to the gene for a human angiogenic ribonuclease, angiogenin (Ang). It was expressed in the mammary gland of mice and secreted into mouse milk. Expression levels in milk were approximately 0.8 g/l. The chimeric protein retained antibody binding activity and protein synthesis inhibitory activity equivalent to that of free Ang. It was specifically cytotoxic to human tumor cells in vitro.


Subject(s)
Immunoglobulin G/biosynthesis , Mammary Glands, Animal/metabolism , Neoplasm Proteins/biosynthesis , Receptors, Transferrin/immunology , Ribonuclease, Pancreatic/biosynthesis , Animals , Female , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Mice, Transgenic , Milk , Neoplasm Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Ribonuclease, Pancreatic/genetics , Tumor Cells, Cultured
16.
J Natl Cancer Inst ; 90(23): 1787-91, 1998 Dec 02.
Article in English | MEDLINE | ID: mdl-9839518

ABSTRACT

BACKGROUND: Preparations of human chorionic gonadotropin (hCG) have been shown to exhibit anti-Kaposi's sarcoma (KS) activity, but the identity of the responsible agent(s) remains controversial. One candidate agent is an eosinophil-derived neurotoxin (EDN)-like polypeptide that contaminates preparations of hCG. We have genetically engineered a unique form of hEDN, which is a ribonuclease, and have evaluated the cytotoxic effects of the recombinant protein on KS Y-1 cells and on cells of other cancer types. METHODS: The amino-terminus of hEDN was extended by four amino acid residues, corresponding to the proximal part of the hEDN signal peptide (serine, leucine, histidine, and valine; positions -4 to -1, respectively), by use of the polymerase chain reaction and an hEDN complementary DNA. The recombinant protein was isolated from bacterial inclusion bodies. The cytotoxic activity of this hEDN variant, (-4)rhEDN, was tested on KS Y-1 cells and human glioma, melanoma, breast carcinoma, and renal carcinoma cells. RESULTS: Approximately half of the anti-KS activity in a crude commercial preparation of hCG was associated with a polypeptide that reacted with anti-recombinant-hEDN (rhEDN) polyclonal antibodies. Although rhEDN protein displayed little cytotoxicity against KS Y-1 cells (IC50 [50% inhibition concentration] = >100 microg/mL), (-4)rhEDN markedly inhibited cell viability (IC50 = 6 microg/mL). Neither version of rhEDN inhibited the viability of other tested human cancer cell types. CONCLUSIONS: A four amino acid extension of the amino-terminus of rhEDN confers cytotoxicity against KS Y-1 cells in vitro. Design of the (-4)rhEDN variant was based on the sequence of a natural human protein associated with hCG. Our results suggest that (-4)rhEDN is one of the agents in hCG responsible for anti-KS activity. A purified molecule is thus available for in vitro and in vivo mechanistic and, possibly, future clinical studies.


Subject(s)
Antineoplastic Agents/therapeutic use , Proteins/therapeutic use , Ribonuclease, Pancreatic/therapeutic use , Ribonucleases , Sarcoma, Kaposi/drug therapy , Blotting, Western , Breast Neoplasms/drug therapy , DNA, Complementary/chemical synthesis , Eosinophil-Derived Neurotoxin , Genes, Synthetic , Histidine/genetics , Humans , Kidney Neoplasms/drug therapy , Leucine/genetics , Polymerase Chain Reaction/methods , Recombinant Proteins/therapeutic use , Sarcoma, Kaposi/metabolism , Serine/genetics , Tumor Cells, Cultured/drug effects , Valine/genetics
17.
Biochemistry ; 37(15): 5173-83, 1998 Apr 14.
Article in English | MEDLINE | ID: mdl-9548748

ABSTRACT

Onconase is a cytotoxic ribonuclease with antitumor properties. A semisynthetic gene encoding the entire protein sequence was constructed by fusing oligonucleotides coding for the first 15 and the last 6 of the 104 amino acids to a genomic clone that encoded the remaining amino acid residues [Newton, D. L., et al. (1997) Protein Eng. 10, 463-470]. The resulting protein product expressed in Escherichia coli exhibited little enzymatic or cytotoxic activity due to the unprocessed N-terminal Met amino acid residue. In this study, we demonstrate that modification of the 5'-region of the gene to encode [Met-(-1)]Ser or [Met-(-1)]Tyr instead of the native pyroglutamate results in recombinant onconase derivatives with restored activities. [Met-(-1)]rOnc(E1S) was more active than [Met-(-1)]rOnc(E1Y) in all assays tested. Consistent with the action of native onconase, [Met-(-1)]rOnc(E1S) was a potent inhibitor of protein synthesis in the cell-free rabbit reticulocyte lysate assay, degrading tRNA at concentrations that correlated with inhibition of protein synthesis. An interesting difference between the recombinant onconase derivatives and the native protein was their susceptibility to inhibition by the major intracellular RNase inhibitor, PRI (onconase is refractory to PRI inhibition). [Met-(-1)]rOnc(E1S) and [Met-(-1)]rOnc(E1Y) inhibited protein synthesis in intact SF539 neuroblastoma cells with IC50's very similar to that of onconase (IC50 3.5, 10, and 10 microg/mL after 1 day and 0.16, 0.35, and 2.5 microg/mL after 5 days for onconase, [Met-(-1)]rOnc(E1S), and [Met-(-1)]rOnc(E1Y), respectively). Similar to that of onconase, cytotoxic activity of the recombinant derivatives was potentiated by monensin, NH4Cl, and retinoic acid. Brefeldin A completely blocked the enhancement of cytotoxicity caused by retinoic acid with all three proteins. Thus, drug-induced alterations of the intracellular trafficking of the recombinant derivatives also resembles that of onconase. Stability studies as assessed in serum-containing medium in the presence or absence of cells at 37 degreesC showed that the recombinant proteins were as stable to temperature and cell culture conditions as the native protein. Therefore, exchanging the Glu amino acid residue at the amino terminus of onconase with an amino acid residue containing a hydroxyl group produces recombinant proteins with ribonuclease and cytotoxic properties similar to native onconase.


Subject(s)
Antineoplastic Agents/pharmacology , Egg Proteins/pharmacology , Protein Synthesis Inhibitors/pharmacology , Ribonucleases/pharmacology , Animals , Cell-Free System/drug effects , Dose-Response Relationship, Drug , Drug Design , Egg Proteins/genetics , Humans , Protein Biosynthesis/drug effects , Protein Engineering , RNA, Transfer/metabolism , Rabbits , Recombinant Proteins/pharmacology , Reticulocytes , Ribonucleases/genetics , Structure-Activity Relationship , Substrate Specificity , Tumor Cells, Cultured/drug effects
18.
Gene Ther ; 5(1): 65-75, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9536266

ABSTRACT

An HIV-1-based expression vector has been constructed that produces protective genes tightly regulated by HIV-1 Tat and Rev proteins. The vector contains either a single protective gene (HIV-1 gag dominant negative mutant (delta-gag)) or a combination of two different protective genes (delta-gag and eosinophil-derived neurotoxin (EDN), a human ribonuclease) which are expressed from a dicistronic mRNA. After stable transfection of CEM T cells and following challenge with HIV-1, viral production was completely inhibited in cells transduced with the vector producing both delta-gag and EDN and delayed in cells producing delta-gag alone. In addition, cotransfection of HeLa-Tat cells with an infectious HIV-1 molecular clone and either protective vector demonstrated that the HIV-1 packaging signals present in the constructs were functional and allowed the efficient assembly of the protective RNAs into HIV-1 virions, thus potentially transmitting protection to the HIV-1 target cells.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , HIV Infections/therapy , HIV-1/physiology , Ribonucleases , Transfection , Blotting, Northern , Eosinophil-Derived Neurotoxin , Gene Expression Regulation , Gene Products, gag , Gene Products, rev , Gene Products, tat , HIV-1/genetics , HeLa Cells , Humans , Neurotoxins/genetics , Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic , Virus Replication , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
19.
Immunotechnology ; 3(2): 127-36, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9237097

ABSTRACT

BACKGROUND: Immunotoxins based on plant and bacterial proteins are usually very immunogenic. Human ribonucleases could provide an alternative basis for the construction of less immunogenic reagents. Two members of the human RNase family, angiogenin and eosinophil-derived neurotoxin (EDN), have been fused to a single chain antibody against the transferrin receptor, which is known to be internalised by endocytosis. The fusion proteins proved to be very efficient inhibitors of protein synthesis using various cell lines. It is not yet known whether the side effects of angiogenin and EDN will compromise their potential use as immunotoxins. OBJECTIVES: The goal of this work was to construct a human immunotoxin with no harmful side effects. Bovine pancreatic ribonuclease has been shown to be as potent as ricin at abolishing protein synthesis on injection into oocytes. We therefore decided to clone its human analogue, which is fairly ubiquitous and per se non-toxic. An immunofusion of human pancreatic RNase with a single chain antibody against the transferrin receptor was tested for its ability to inhibit protein synthesis in three different human tumor cell lines. STUDY DESIGN: DNA coding for the human pancreatic RNase was cloned partially from a human fetal brain cDNA library and then completed by PCR using a human placental cDNA library as a template. The RNase gene was then fused with a DNA coding for an single chain antibody against the transferrin receptor (CD71). After expressing the fusion protein in E. coli, the gene product was isolated from inclusion bodies and tested for cytotoxicity. RESULTS: This fusion protein inhibited the protein synthesis of three human tumor cell lines derived from a melanoma, a renal carcinoma and a breast carcinoma, with IC50s of 8, 5 and 10 nM, respectively. These values were comparable with those using a similar fusion protein constructed with eosinophil derived neurotoxin (EDN) as the toxic moiety (IC50s of 8, 1.2 and 3 nM, respectively). The slightly lower activities of the human pancreatic RNase-scFv (pancRNase-scFv) with two of the cell lines suggests that fewer molecules are reaching the cytoplasmic compartment, since it was twice as active as EDN-scFv in inhibiting the protein synthesis of a rabbit reticulocyte lysate. CONCLUSION: These results demonstrate that the human pancreatic RNase, which is expected to have a very low immunogenic potential in humans with no inherent toxicity, may be a potent cytotoxin for tumor cells after antibody targeting.


Subject(s)
Cytotoxicity, Immunologic , Immunotoxins/immunology , Immunotoxins/toxicity , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/toxicity , Ribonuclease, Pancreatic/immunology , Ribonuclease, Pancreatic/toxicity , Animals , Cell-Free System/chemistry , Cloning, Molecular , Genes , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/genetics , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunotoxins/genetics , Mice , Plasmids/chemical synthesis , Polymerase Chain Reaction , Protein Synthesis Inhibitors/pharmacology , Receptors, Transferrin/genetics , Receptors, Transferrin/immunology , Recombinant Fusion Proteins/genetics , Ribonuclease, Pancreatic/genetics , Tumor Cells, Cultured
20.
Protein Eng ; 10(4): 463-70, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9194172

ABSTRACT

Onconase is a cytotoxic ribonuclease with antitumor properties. A semisynthetic gene encoding the entire protein sequence was constructed by fusing oligonucleotides coding for the first 15 and last six of the 104 amino acid residues to a genomic clone that encoded the remaining amino acid residues. Additionally, the 15 N-terminal amino acid residues of onconase were replaced with the first 21 amino acid residues of the homologous human RNase, eosinophil-derived neurotoxin, EDN. Two versions of the hybrid EDN-onconase protein were cloned, expressed and purified. The chimera that contained a glycine in lieu of the aspartic acid present in native onconase (position 26 in the chimera) exhibited enzymatic activity more characteristic of EDN than native onconase and was considerably more active with respect to both RNase activity and cellular cytotoxicity than recombinant onconase. In contrast to native or recombinant onconase, the EDN chimera was recognized by anti-EDN polyclonal antibodies, demonstrating that the chimera also shared structural antigenic determinants to the human enzyme. These results demonstrate that a chimeric ribonuclease has cytotoxicity comparable to onconase in two out of four cell lines tested. The implications with regard to cancer therapy are presented.


Subject(s)
Antineoplastic Agents/metabolism , Egg Proteins/genetics , Neurotoxins/genetics , Ribonucleases/genetics , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Cloning, Molecular , Drug Screening Assays, Antitumor , Egg Proteins/chemistry , Egg Proteins/metabolism , Egg Proteins/therapeutic use , Electrophoresis, Polyacrylamide Gel , Eosinophil-Derived Neurotoxin , Humans , Methionine/chemistry , Methionine/metabolism , Models, Chemical , Models, Molecular , Molecular Sequence Data , Neurotoxins/chemistry , Neurotoxins/metabolism , Neurotoxins/therapeutic use , Rana pipiens , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Ribonucleases/chemistry , Ribonucleases/metabolism , Ribonucleases/therapeutic use , Software , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...