Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Int J Cancer ; 139(4): 916-27, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27037627

ABSTRACT

Chondroitin sulfate proteoglycan 4 (CSPG4) has been identified as a highly promising target antigen for immunotherapy of triple-negative breast cancer (TNBC). TNBC represents a highly aggressive heterogeneous group of tumors lacking expression of estrogen, progesterone and human epidermal growth factor receptor 2. TNBC is particularly prevalent among young premenopausal women. No suitable targeted therapies are currently available and therefore, novel agents for the targeted elimination of TNBC are urgently needed. Here, we present a novel cytolytic fusion protein (CFP), designated αCSPG4(scFv)-MAP, that consists of a high affinity CSPG4-specific single-chain antibody fragment (scFv) genetically fused to a functionally enhanced form of the human microtubule-associated protein (MAP) tau. Our data indicate that αCSPG4(scFv)-MAP efficiently targets CSPG4(+) TNBC-derived cell lines MDA-MB-231 and Hs 578T and potently inhibits their growth with IC50 values of ∼200 nM. Treatment with αCSPG(scFv)-MAP resulted in induction of the mitochondrial stress pathway by activation of caspase-9 as well as endonuclease G translocation to the nucleus, while induction of the caspase-3 apoptosis pathway was not detectable. Importantly, in vivo studies in mice bearing human breast cancer xenografts revealed efficient targeting to and accumulation of αCSPG4(scFv)-MAP at tumor sites resulting in prominent tumor regression. Taken together, this preclinical proof of concept study confirms the potential clinical value of αCSPG4(scFv)-MAP as a novel targeted approach for the elimination of CSPG4-positive TNBC.


Subject(s)
Antibodies, Monoclonal/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Membrane Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/genetics , Triple Negative Breast Neoplasms/metabolism , tau Proteins/metabolism , Animals , Biomarkers , Biomarkers, Tumor , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chondroitin Sulfate Proteoglycans/genetics , Disease Models, Animal , Female , Gene Expression , Humans , Membrane Proteins/genetics , Mice , Molecular Targeted Therapy , Protein Binding , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Tubulin/metabolism , Xenograft Model Antitumor Assays , tau Proteins/genetics
2.
BMC Biotechnol ; 16: 18, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26883813

ABSTRACT

BACKGROUND: In an earlier study we developed a unique strategy allowing us to specifically eliminate antigen-specific murine B cells via their distinct B cell receptors using a new class of fusion proteins. In the present work we elaborated our idea to demonstrate the feasibility of specifically addressing and eliminating human memory B cells. RESULTS: The present study reveals efficient adaptation of the general approach to selectively target and eradicate human memory B cells. In order to demonstrate the feasibility we engineered a fusion protein following the principle of recombinant immunotoxins by combining a model antigen (tetanus toxoid fragment C, TTC) for B cell receptor targeting and a truncated version of Pseudomonas aeruginosa exotoxin A (ETA') to induce apoptosis after cellular uptake. The TTC-ETA' fusion protein not only selectively bound to a TTC-reactive murine B cell hybridoma cell line in vitro but also to freshly isolated human memory B cells from immunized donors ex vivo. Specific toxicity was confirmed on an antigen-specific population of human CD27(+) memory B cells. CONCLUSIONS: This protein engineering strategy can be used as a generalized platform approach for the construction of therapeutic fusion proteins with disease-relevant antigens as B cell receptor-binding domains, offering a promising approach for the specific depletion of autoreactive B-lymphocytes in B cell-driven autoimmune diseases.


Subject(s)
Models, Immunological , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/metabolism , Tetanus Toxoid/metabolism , Animals , B-Lymphocytes/chemistry , B-Lymphocytes/metabolism , Cells, Cultured , Escherichia coli , HEK293 Cells , Humans , Hybridomas/immunology , Hybridomas/metabolism , Leukocytes, Mononuclear/metabolism , Mice , Receptors, Cell Surface/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tetanus Toxoid/chemistry , Tetanus Toxoid/genetics , Tetanus Toxoid/immunology
3.
Cancer Immunol Immunother ; 64(12): 1575-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26472728

ABSTRACT

Immunotoxins are fusion proteins that combine a targeting component such as an antibody fragment or ligand with a cytotoxic effector component that induces apoptosis in specific cell populations displaying the corresponding antigen or receptor. Human cytolytic fusion proteins (hCFPs) are less immunogenic than conventional immunotoxins because they contain human pro-apoptotic enzymes as effectors. However, one drawback of hCFPs is that target cells can protect themselves by expressing endogenous inhibitor proteins. Inhibitor-resistant enzyme mutants that maintain their cytotoxic activity are therefore promising effector domain candidates. We recently developed potent variants of the human ribonuclease angiogenin (Ang) that were either more active than the wild-type enzyme or less susceptible to inhibition because of their lower affinity for the ribonuclease inhibitor RNH1. However, combining the mutations was unsuccessful because although the enzyme retained its higher activity, its susceptibility to RNH1 reverted to wild-type levels. We therefore used molecular dynamic simulations to determine, at the atomic level, why the affinity for RNH1 reverted, and we developed strategies based on the introduction of further mutations to once again reduce the affinity of Ang for RNH1 while retaining its enhanced activity. We were able to generate a novel Ang variant with remarkable in vitro cytotoxicity against HL-60 cells and pro-inflammatory macrophages. We also demonstrated the pro-apoptotic potential of Ang-based hCFPs on cells freshly isolated from leukemia patients.


Subject(s)
Leukemia/pathology , Macrophages/drug effects , Ribonuclease, Pancreatic/genetics , Apoptosis , Carrier Proteins/metabolism , Cell Survival/drug effects , Cytotoxins/genetics , Cytotoxins/toxicity , Flow Cytometry , HL-60 Cells , Humans , Macrophages/cytology , Macrophages/immunology , Molecular Dynamics Simulation , Mutation , Protein Binding/genetics , Ribonuclease, Pancreatic/toxicity
5.
Bioconjug Chem ; 20(5): 1010-5, 2009 May 20.
Article in English | MEDLINE | ID: mdl-19388673

ABSTRACT

Recombinant antibodies are promising tools for a wide range of bioanalytical and medical applications. However, the chemical modification of such molecules can be challenging, which limits their broader utilization. Here we describe a universal method for the site-specific labeling of antibody fragments and protein ligands by genetically fusing them to an engineered version of the human DNA-repair enzyme O(6)-alkyllguanine DNA alkyltransferase (AGT), known as SNAP-Tag (1-3) . Substrates containing O(6)-benzylguanine are covalently bound to the fusion proteins via a stable thioether bond in a rapid and highly specific self-labeling reaction. The coupling is site-directed, allowing the design and synthesis of antibody conjugates with predefined stoichiometry. We cloned a series of ligand SNAP-Tag fusion proteins and expressed them in HEK 293T cells. The antibody/ligand-fusions were characterized by labeling with different fluorophores, labeling with biotin, or by coupling them to fluorescent nanobeads, followed by analysis by flow cytometry and confocal microscopy. All ligands retained their original antigen-binding properties when fused to the SNAP-Tag. The combination of recombinant antibodies or protein ligands with the SNAP-Tag facilitates simple and efficient covalent modification with a broad range of substrates, thus providing a useful and advantageous alternative to existing coupling strategies.


Subject(s)
O(6)-Methylguanine-DNA Methyltransferase/genetics , Protein Engineering/methods , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , Animals , Binding Sites , Biotin/metabolism , CD30 Ligand/metabolism , CHO Cells , Cricetinae , Cricetulus , Flow Cytometry , Fluorescent Dyes/metabolism , Humans , Ki-1 Antigen/immunology , Ligands , Mice , Microscopy, Confocal , Microspheres , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Silicon Dioxide/chemistry , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Staining and Labeling , Substrate Specificity
6.
J Neuroimmunol ; 195(1-2): 28-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18280586

ABSTRACT

We report the construction of a fusion protein comprising the extracellular domain of myelin oligodendrocyte glycoprotein (MOG) and a truncated version of Pseudomonas aeruginosa exotoxin A (ETA'). The chimeric immunotoxin targeted MOG-reactive B-lymphocytes by binding selectively to the appropriate receptors, leading to internalization and apoptosis of the target cells. The functionality of the immunotoxin was tested on a MOG-sensitive murine hybridoma cell line and ex vivo on freshly isolated splenocytes from transgenic IgH(MOG) mice. These data demonstrate, for the first time, the specific cytotoxicity of a MOG-containing recombinant immunotoxin expressed in bacteria towards MOG-reactive B-lymphocytes.


Subject(s)
B-Lymphocytes/drug effects , Lymphocyte Depletion , Myelin-Associated Glycoprotein/genetics , Recombinant Fusion Proteins/toxicity , B-Lymphocytes/immunology , Cell Line, Transformed , Cell Proliferation , Cell Survival , Dose-Response Relationship, Immunologic , Flow Cytometry/methods , Humans , Hybridomas , Myelin Proteins , Myelin-Associated Glycoprotein/toxicity , Myelin-Oligodendrocyte Glycoprotein , Recombinant Fusion Proteins/chemistry , Transfection
7.
Biomedicines ; 6(1)2018 Mar 05.
Article in English | MEDLINE | ID: mdl-29510557

ABSTRACT

Targeted cancer therapy includes, amongst others, antibody-based delivery of toxic payloads to selectively eliminate tumor cells. This payload can be either a synthetic small molecule drug composing an antibody-drug conjugate (ADC) or a cytotoxic protein composing an immunotoxin (IT). Non-human cytotoxic proteins, while potent, have limited clinical efficacy due to their immunogenicity and potential off-target toxicity. Humanization of the cytotoxic payload is essential and requires harnessing of potent apoptosis-inducing human proteins with conditional activity, which rely on targeted delivery to contact their substrate. Ribonucleases are attractive candidates, due to their ability to induce apoptosis by abrogating protein biosynthesis via tRNA degradation. In fact, several RNases of the pancreatic RNase A superfamily have shown potential as anti-cancer agents. Coupling of a human RNase to a humanized antibody or antibody derivative putatively eliminates the immunogenicity of an IT (now known as a human cytolytic fusion protein, hCFP). However, RNases are tightly regulated in vivo by endogenous inhibitors, controlling the ribonucleolytic balance subject to the cell's metabolic requirements. Endogenous inhibition limits the efficacy with which RNase-based hCFPs induce apoptosis. However, abrogating the natural interaction with the natural inhibitors by mutation has been shown to significantly enhance RNase activity, paving the way toward achieving cytolytic potency comparable to that of bacterial immunotoxins. Here, we review the immunoRNases that have undergone preclinical studies as anti-cancer therapeutic agents.

8.
PLoS One ; 12(7): e0180305, 2017.
Article in English | MEDLINE | ID: mdl-28704435

ABSTRACT

The antigen-specific targeting of autoreactive B cells via their unique B cell receptors (BCRs) is a novel and promising alternative to the systemic suppression of humoral immunity. We generated and characterized cytolytic fusion proteins based on an existing immunotoxin comprising tetanus toxoid fragment C (TTC) as the targeting component and the modified Pseudomonas aeruginosa exotoxin A (ETA') as the cytotoxic component. The immunotoxin was reconfigured to replace ETA' with either the granzyme B mutant R201K or MAPTau as human effector domains. The novel cytolytic fusion proteins were characterized with a recombinant human lymphocytic cell line developed using Transpo-mAb™ technology. Genes encoding a chimeric TTC-reactive immunoglobulin G were successfully integrated into the genome of the precursor B cell line REH so that the cells could present TTC-reactive BCRs on their surface. These cells were used to investigate the specific cytotoxicity of GrB(R201K)-TTC and TTC-MAPTau, revealing that the serpin proteinase inhibitor 9-resistant granzyme B R201K mutant induced apoptosis specifically in the lymphocytic cell line. Our data confirm that antigen-based fusion proteins containing granzyme B (R201K) are suitable candidates for the depletion of autoreactive B cells.


Subject(s)
B-Lymphocytes/cytology , Cell Engineering/methods , Granzymes/genetics , Tetanus Toxoid/immunology , B-Lymphocytes/metabolism , Cell Line, Tumor , Granzymes/metabolism , Humans , Immunotoxins/metabolism , Receptors, Antigen, B-Cell/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/metabolism , Tetanus Toxoid/metabolism
9.
Protein Sci ; 25(8): 1451-60, 2016 08.
Article in English | MEDLINE | ID: mdl-27110669

ABSTRACT

Targeted human cytolytic fusion proteins (hCFPs) represent a new generation of immunotoxins (ITs) for the specific targeting and elimination of malignant cell populations. Unlike conventional ITs, hCFPs comprise a human/humanized target cell-specific binding moiety (e.g., an antibody or a fragment thereof) fused to a human proapoptotic protein as the cytotoxic domain (effector domain). Therefore, hCFPs are humanized ITs expected to have low immunogenicity. This reduces side effects and allows long-term application. The human ribonuclease angiogenin (Ang) has been shown to be a promising effector domain candidate. However, the application of Ang-based hCFPs is largely hampered by the intracellular placental ribonuclease inhibitor (RNH1). It rapidly binds and inactivates Ang. Mutations altering Ang's affinity for RNH1 modulate the cytotoxicity of Ang-based hCFPs. Here we perform in total 2.7 µs replica-exchange molecular dynamics simulations to investigate some of these mutations-G85R/G86R (GGRRmut ), Q117G (QGmut ), and G85R/G86R/Q117G (GGRR/QGmut ). GGRRmut turns out to perturb greatly the overall Ang-RNH1 interactions, whereas QGmut optimizes them. Combining QGmut with GGRRmut compensates the effects of the latter. Our results explain the in vitro finding that, while Ang GGRRmut -based hCFPs resist RNH1 inhibition remarkably, Ang WT- and Ang QGmut -based ones are similarly sensitive to RNH1 inhibition, whereas Ang GGRR/QGmut -based ones are only slightly resistant. This work may help design novel Ang mutants with reduced affinity for RNH1 and improved cytotoxicity.


Subject(s)
Antineoplastic Agents/chemistry , Carrier Proteins/chemistry , Immunotoxins/chemistry , Recombinant Fusion Proteins/chemistry , Ribonuclease, Pancreatic/chemistry , Binding Sites , Carrier Proteins/genetics , Gene Expression , Humans , Immunotoxins/genetics , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Engineering , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Fusion Proteins/genetics , Ribonuclease, Pancreatic/genetics
10.
Cancer Lett ; 372(2): 201-9, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-26806809

ABSTRACT

Triple-negative breast cancer (TNBC) is associated with poor prognosis and high prevalence among young premenopausal women. Unlike in other breast cancer subtypes, no targeted therapy is currently available. Overexpression of epithelial cell adhesion molecule (EpCAM) in 60% of TNBC tumors correlates with poorer prognosis and is associated with cancer stem cell phenotype. Thus, selective elimination of EpCAM(+) TNBC tumor cells is of clinical importance. Therefore, we constructed a fully human targeted cytolytic fusion protein, designated GbR201K-αEpCAM(scFv), in which an EpCAM-selective single-chain antibody fragment (scFv) is genetically fused to a granzyme B (Gb) mutant with reduced sensitivity to its natural inhibitor serpin B9. In vitro studies confirmed its specific binding, internalization and cytotoxicity toward a panel of EpCAM-expressing TNBC cells. Biodistribution kinetics and tumor-targeting efficacy using MDA-MB-468 cells in a human TNBC xenograft model in mice revealed selective accumulation of GbR201K-αEpCAM(scFv) in the tumors after i.v. injection. Moreover, treatment of tumor-bearing mice demonstrated a prominent inhibition of tumor growth of up to 50 % in this proof-of-concept study. Taken together, our results indicate that GbR201K-αEpCAM(scFv) is a promising novel targeted therapeutic for the treatment of TNBC.


Subject(s)
Apoptosis/drug effects , Cell Adhesion Molecules/antagonists & inhibitors , Granzymes/pharmacology , Immunotherapy/methods , Immunotoxins/pharmacology , Single-Chain Antibodies/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Tumor Burden/drug effects , Animals , Antibody Specificity , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cell Adhesion Molecule , Female , Granzymes/genetics , Granzymes/pharmacokinetics , HEK293 Cells , Humans , Immunotoxins/genetics , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Mice, Inbred BALB C , Mice, Nude , Mutation , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacokinetics , Tissue Distribution , Transfection , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
11.
J Immunother ; 38(3): 85-95, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25710248

ABSTRACT

Human cytolytic fusion proteins (hCFPs) are therapeutically efficacious recombinant polypeptides comprising a target cell-specific binding component and a human effector domain that induces apoptosis. Compared with former generations of immunotoxins, which contain immunogenic cytotoxic domains derived from bacteria or plants, hCFPs contain solely human proteins that do not induce an immune response, thus avoiding the development of neutralizing antibodies. Here, we investigated the suitability of human angiogenin (Ang) mutants as effector domains. We engineered 3 different Ang variants that outperformed the wild-type enzyme by replacing amino acid residues with key roles in the protein's catalytic activity and its interaction with the ribonuclease inhibitor RNH1. The cytotoxic potential of these mutants was compared with wild-type Ang by fusing each to the CD64-specific single-chain variable fragment H22. All hCFPs were successfully expressed in HEK293T cells and purified from the cell culture supernatant by immobilized metal ion affinity chromatography. The Ang mutant-based hCFPs showed normal binding activity towards human interferon-γ-stimulated CD64 HL-60 cells and activated human macrophages isolated from peripheral blood mononuclear cells, but increased cytotoxicity based on reduced affinity towards RNH1 and higher ribonucleolytic activity.


Subject(s)
Mutant Proteins , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/toxicity , Ribonuclease, Pancreatic/genetics , Apoptosis/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Survival/drug effects , Gene Expression , Genetic Variation , HL-60 Cells , Humans , Immunotoxins/pharmacology , Kinetics , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Protein Binding , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Ribonuclease, Pancreatic/metabolism , Substrate Specificity
12.
Curr Pharm Des ; 19(30): 5429-36, 2013.
Article in English | MEDLINE | ID: mdl-23431985

ABSTRACT

Although current cancer treatment strategies are highly aggressive, they are often not effective enough to destroy the collectivity of malignant cells. The residual tumor cells that survived the first-line treatment may continue to proliferate or even metastasize. Therefore, the development of novel more effective strategies to specifically eliminate also single cancer cells is urgently needed. In this respect, the development of antibody-based therapeutics, in particular example immunotoxins, has attracted broad interest. Since the internalization of immunotoxins is essential for their cytotoxic effectivity, it is of crucial importance to study their internalization behavior to assess the potential for their therapeutic use. In this study, we determined the internalization behavior of four different single-chain fragments variable (scFv) when binding to the corresponding target antigen as expressed on solid or non-solid tumor cell lines. The scFvs were recombinantly fused to the SNAP-tag, an engineered variant of the human repair enzyme O(6)-alkylguanine-DNA alkyltransferase that covalently reacts with benzylguanine derivatives. Since a large number of highly sensitive organic fluorescent dyes are already available or can easily be derivatized to react with the self-labeling SNAP-tag, this system provides versatile applications for imaging of intraand extracellular compartments of living cells. The fusion proteins were coupled to SNAP-surface(®) Alexa Fluor(®) 488 or SNAP-surface(®) Alexa Fluor(®) 647 and binding as well as internalization was monitored by flow cytometry and confocal microscopy, respectively. Depending on the respective target antigen, we could distinguish between slow and rapid internalization behavior. Moreover, we detected increased internalization rate for bivalent scFv constructs. Our approach allows for rapid and early stage evaluation of the internalization characteristics of new antibodies designated for further therapeutic development.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Molecular Imaging/methods , Neoplasms/metabolism , Animals , Antibodies , Cloning, Molecular , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Neoplasms/drug therapy , Protein Binding
13.
J Allergy Clin Immunol ; 116(4): 910-5, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16210069

ABSTRACT

BACKGROUND: With the exception of antigen-specific immunotherapy, current treatments for atopic diseases provide only symptomatic relief. Because of the increasing incidence of such diseases, the development of novel strategies and concepts for the treatment of allergies is urgently needed. OBJECTIVE: Here we present a new approach for the treatment of atopic diseases. The strategy is comparable to the application of immunotoxins in cancer therapy, in which a cytotoxic peptide is coupled to a cancer cell-specific antibody fragment or ligand. In the case of so-called allergen toxins (ATs), the target cell-specific moiety is an allergen or allergen-derived fragment, which should be bound only by allergen-reactive cells. After receptor-mediated internalization, allergen-specific cells are killed, and the allergic pathogenesis is interrupted. METHODS: Proof of the AT principle was shown by using a human ex vivo system in which EBV was used to transform human B cells specific for the timothy grass pollen allergen Phl p 5b. The AT is composed of the major B-cell and T-cell epitopes of the Phl p 5b (P5) allergen fused to a truncated form of the highly toxic Pseudomonas aeruginosa exotoxin A (ETA'). RESULTS: Allergen-specific and nonspecific B cells were challenged with P5-ETA', but only the Phl p 5b-reactive B cells showed selective binding and cytotoxicity. CONCLUSION: This approach represents an initial step toward a novel therapeutic strategy in the treatment of atopic diseases.


Subject(s)
Allergens , B-Lymphocytes/immunology , B-Lymphocytes/virology , Herpesvirus 4, Human/immunology , Hypersensitivity, Immediate/immunology , Immunotoxins/pharmacology , Recombinant Fusion Proteins/pharmacology , Allergens/genetics , Allergens/immunology , Cell Transformation, Viral , Cytotoxicity, Immunologic , Humans , Hypersensitivity, Immediate/therapy , Immunotoxins/genetics , Immunotoxins/immunology , In Vitro Techniques , Phleum/genetics , Phleum/immunology , Plant Proteins/genetics , Plant Proteins/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Ribonucleases/genetics , Ribonucleases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL