Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 14(1): 90-103, 2024 01 12.
Article in English | MEDLINE | ID: mdl-37861452

ABSTRACT

The tumor-associated antigen STEAP1 is a potential therapeutic target that is expressed in most prostate tumors and at increased levels in metastatic castration-resistant prostate cancer (mCRPC). We developed a STEAP1-targeted XmAb 2+1 T-cell engager (TCE) molecule, AMG 509 (also designated xaluritamig), that is designed to redirect T cells to kill prostate cancer cells that express STEAP1. AMG 509 mediates potent T cell-dependent cytotoxicity of prostate cancer cell lines in vitro and promotes tumor regression in xenograft and syngeneic mouse models of prostate cancer in vivo. The avidity-driven activity of AMG 509 enables selectivity for tumor cells with high STEAP1 expression compared with normal cells. AMG 509 is the first STEAP1 TCE to advance to clinical testing, and we report a case study of a patient with mCRPC who achieved an objective response on AMG 509 treatment. SIGNIFICANCE: Immunotherapy in prostate cancer has met with limited success due to the immunosuppressive microenvironment and lack of tumor-specific targets. AMG 509 provides a targeted immunotherapy approach to engage a patient's T cells to kill STEAP1-expressing tumor cells and represents a new treatment option for mCRPC and potentially more broadly for prostate cancer. See related commentary by Hage Chehade et al., p. 20. See related article by Kelly et al., p. 76. This article is featured in Selected Articles from This Issue, p. 5.


Subject(s)
Antibodies, Bispecific , Prostatic Neoplasms, Castration-Resistant , Male , Mice , Animals , Humans , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , T-Lymphocytes , Immunotherapy , Antibodies, Bispecific/therapeutic use , Tumor Microenvironment , Antigens, Neoplasm , Oxidoreductases/therapeutic use
2.
Clin Cancer Res ; 25(13): 3921-3933, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30918018

ABSTRACT

PURPOSE: Despite advances in the treatment of multiple myeloma, new therapies are needed to induce more profound clinical responses. T-cell-redirected lysis triggered by bispecific antibodies recruiting T cells to cancer cells is a clinically validated mechanism of action against hematologic malignancies and CD38 is a tumor-associated antigen with near-universal expression in multiple myeloma. Thus, an anti-CD38/CD3 bispecific T-cell-recruiting antibody has the potential to be an effective new therapeutic for multiple myeloma. EXPERIMENTAL DESIGN: Anti-CD38/CD3 XmAb T-cell-recruiting antibodies with different affinities for CD38 and CD3 were assessed in vitro and in vivo for their redirected T-cell lysis activity against cancer cell lines, their lower levels of cytokine release, and their potency in the presence of high levels of soluble CD38. Select candidates were further tested in cynomolgus monkeys for B-cell depletion and cytokine release properties. RESULTS: AMG 424 was selected on the basis of its ability to kill cancer cells expressing high and low levels of CD38 in vitro and trigger T-cell proliferation, but with attenuated cytokine release. In vivo, AMG 424 induces tumor growth inhibition in bone marrow-invasive mouse cancer models and the depletion of peripheral B cells in cynomolgus monkeys, without triggering excessive cytokine release. The activity of AMG 424 against normal immune cells expressing CD38 is also presented. CONCLUSIONS: These findings support the clinical development of AMG 424, an affinity-optimized T-cell-recruiting antibody with the potential to elicit significant clinical activity in patients with multiple myeloma.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents, Immunological/therapeutic use , Cytokines/biosynthesis , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , ADP-ribosyl Cyclase 1/antagonists & inhibitors , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/adverse effects , Antibody Affinity/immunology , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD3 Complex/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Lymphocyte Activation/immunology , Macaca fascicularis , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays
3.
Methods ; 154: 38-50, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30366098

ABSTRACT

Bispecific monoclonal antibodies can bind two protein targets simultaneously and enable therapeutic modalities inaccessible by traditional mAbs. Bispecific formats containing a heterodimeric Fc region are of particular interest, as a heterodimeric Fc empowers both bispecificity and altered valencies while retaining the developability and druggability of a monoclonal antibody. We present a robust heterodimeric Fc platform, called the XmAb® bispecific platform, engineered for efficient development of bispecific antibodies and Fc fusions of multiple formats. First, we engineer a purification solution for proteins containing a heterodimeric Fc using engineered isoelectric point differences in the Fc region that enable straightforward purification of the heterodimeric species. Then, we combine this purification solution with a novel set of Fc substitutions capable of achieving heterodimer yields over 95% with little change in thermostability. Next, we illustrate the flexibility of our heterodimeric Fc with a case study in which a wide range of tumor-associated antigen × CD3 bispecifics are generated, differing in choice of tumor antigen, affinities for both tumor antigen and CD3, and tumor antigen valency. Finally, we present manufacturing data reinforcing the robustness of the heterodimeric Fc platform at scale.


Subject(s)
Antibodies, Bispecific , Antibodies, Monoclonal , Protein Engineering/methods , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Humans
4.
MAbs ; 6(2): 409-21, 2014.
Article in English | MEDLINE | ID: mdl-24492248

ABSTRACT

Antibody-dependent cell-mediated cytotoxicity (ADCC) has been suggested as an essential mechanism for the in vivo activity of cetuximab, an epidermal growth factor receptor (EGFR)-targeting therapeutic antibody. Thus, enhancing the affinity of human IgG1 antibodies to natural killer (NK) cell-expressed FcγRIIIa by glyco- or protein-engineering of their Fc portion has been demonstrated to improve NK cell-mediated ADCC and to represent a promising strategy to improve antibody therapy. However, human polymorphonuclear (PMN) effector cells express the highly homologous FcγRIIIb isoform, which is described to be ineffective in triggering ADCC. Here, non-fucosylated or protein-engineered anti-EGFR antibodies with optimized FcγRIIIa affinities demonstrated the expected benefit in NK cell-mediated ADCC, but did not mediate ADCC by PMN, which could be restored by FcγRIIIb blockade. Furthermore, eosinophils and PMN from paroxysmal nocturnal hemoglobinuria patients that expressed no or low levels of FcγRIIIb mediated effective ADCC with FcγRIII-optimized anti-EGFR antibody. Additional experiments with double FcγRIIa/FcγRIII-optimized constructs demonstrated enhanced PMN-mediated ADCC compared with single FcγRIII-optimized antibody. In conclusion, our data demonstrate that FcγRIIIb engagement impairs PMN-mediated ADCC activity of FcγRIII-optimized anti-EGFR antibodies, while further optimization of FcγRIIa binding significantly restores PMN recruitment.


Subject(s)
Antibodies, Monoclonal, Humanized/metabolism , Eosinophils/immunology , Hemoglobinuria, Paroxysmal/immunology , Hemoglobinuria, Paroxysmal/therapy , Immunoglobulin G/metabolism , Immunotherapy/methods , Neutrophils/immunology , Antibodies, Monoclonal, Humanized/genetics , Antibody Affinity/genetics , Antibody-Dependent Cell Cytotoxicity/genetics , Cells, Cultured , Cetuximab , Cytotoxicity, Immunologic/genetics , ErbB Receptors/immunology , Glycosylation , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Immunotherapy/trends , Polymorphism, Genetic , Protein Engineering , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, IgG/metabolism
5.
MAbs ; 5(3): 384-96, 2013.
Article in English | MEDLINE | ID: mdl-23549103

ABSTRACT

The CTLA4-Ig fusion proteins abatacept and belatacept are clinically proven immunosuppressants used for rheumatoid arthritis and renal transplant, respectively. Given that both biologics are typically administered chronically by infusion, a need exists for a next-generation CTLA4-Ig with more convenient dosing. We used structure-based protein engineering to optimize the affinity of existing CTLA4-Ig therapeutics for the ligands CD80 and CD86, and for the neonatal Fc receptor, FcRn. From a rationally designed library, we identified four substitutions that enhanced binding to human CD80 and CD86. Coupled with two IgG1 Fc substitutions that enhanced binding to human FcRn, these changes comprise the novel CTLA4-Ig fusion protein, XPro9523. Compared with abatacept, XPro9523 demonstrated 5.9-fold, 23-fold, and 12-fold increased binding to CD80, CD86, and FcRn, respectively; compared with belatacept, CD80, CD86, and FcRn binding increased 1.5-fold, 7.7-fold, and 11-fold, respectively. XPro9523 and belatacept suppressed human T cell proliferation and IL-2 production more potently than abatacept. XPro9523 also suppressed inflammation in the mouse collagen-induced arthritis model. In cynomolgus monkeys, XPro9523 saturated CD80 and CD86 more effectively than abatacept and belatacept, potently inhibited IgM and IgG immunization responses, and demonstrated longer half-life. Pharmacokinetic modeling of its increased potency and persistence suggests that, in humans, XPro9523 may demonstrate superior efficacy and dosing convenience compared with abatacept and belatacept.


Subject(s)
Arthritis, Experimental/therapy , Arthritis, Rheumatoid/therapy , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Graft Rejection/therapy , Histocompatibility Antigens Class I/metabolism , Immunoconjugates/metabolism , Protein Binding/drug effects , Receptors, Fc/metabolism , Recombinant Fusion Proteins/metabolism , Abatacept , Animals , Antibody Affinity , Antibody Formation/drug effects , B7-1 Antigen/immunology , B7-2 Antigen/immunology , Cells, Cultured , Female , Histocompatibility Antigens Class I/immunology , Humans , Immunoconjugates/genetics , Immunoconjugates/pharmacology , Immunosuppression Therapy , Kidney Transplantation , Lymphocyte Activation/drug effects , Macaca fascicularis , Male , Mice , Mice, Inbred DBA , Mutation/genetics , Protein Binding/immunology , Protein Engineering , Receptors, Fc/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Structure-Activity Relationship
6.
Immunol Lett ; 143(1): 34-43, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22305932

ABSTRACT

Mast cells and basophils play a central role in allergy, asthma, and anaphylaxis, as well as in non-allergic inflammatory, neurological and autoimmune diseases. Allergen-mediated cross-linking of IgE bound to FcεRI leads to cellular activation, and the low-affinity Fc receptor FcγRIIb is a key inhibitor of subsequent degranulation. FcγRIIb, when coengaged with FcεRI via allergen bound to IgE, stimulates ITIM domain-mediated inhibitory signaling that efficiently suppresses mast cell and basophil activation. To assess the therapeutic potential of directed coengagement of FcεRI and FcγRIIb in the absence of FcεRI crosslinking, we developed a fusion protein comprising the coupled Fc domains of murine IgE and human IgG1. As a key functional component of this tandem Fcε-Fcγ biologic, we engineered its IgG1 Fc domain to bind to human FcγRIIb with 100-fold enhanced affinity relative to native IgG1 Fc. Using mast cells from mice transgenic for human FcγRIIb, we show that this tandem Fc binds with high affinity to murine FcεRI and human FcγRIIb on mast cells, triggers phosphorylation of FcγRIIb, and inhibits FcεRI-dependent calcium mobilization. Control tandem Fc biologics containing a native IgG1 Fc domain or lacking binding to Fcγ receptors were markedly less active, demonstrating that the affinity-optimized tandem Fc can inhibit degranulation through stimulation of FcγRIIb signaling as well as through competition with allergen-IgE immune complex for FcεRI binding. We propose that in the context of a fully human tandem Fc biologic, high-affinity coengagement of FcεRI and FcγRIIb has potential as a novel therapy for allergy and other mast cell and basophil-mediated pathologies.


Subject(s)
Cell Degranulation , Immunoglobulin E/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Mast Cells/physiology , Receptors, IgG/immunology , Animals , Calcium/metabolism , Cell Differentiation , Humans , Mast Cells/cytology , Mice , Mice, Inbred C57BL , Phenotype , Phosphorylation , Recombinant Proteins/immunology
7.
J Allergy Clin Immunol ; 129(4): 1102-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22257644

ABSTRACT

BACKGROUND: Sequestration of IgE to prevent its binding to high-affinity IgE receptor FcεRI on basophils and mast cells is an effective therapy for allergic asthma. IgE production requires differentiation of activated IgE(+) B cells into plasma cells upon allergen sensitization. B-cell receptor signaling is suppressed by the inhibitory IgG Fc receptor FcγRIIb; therefore, we reasoned that a therapeutic antibody that coengages FcγRIIb and IgE B-cell receptor would not only sequester IgE but also suppress its production by blocking IgE(+) B-cell activation and differentiation to IgE-secreting plasma cells. OBJECTIVE: To explore the effects of IgE sequestration versus IgE suppression by comparing omalizumab to FcγRIIb-optimized anti-IgE antibodies in humanized mouse models of immunoglobulin production. METHODS: By using a murine anti-IgE antibody as a template, we humanized, increased IgE binding, and modified its Fc domain to increase affinity for FcγRIIb. We next compared effects of this antibody (XmAb7195) versus omalizumab on the secretion of IgE and other isotypes in human PBMC cultures and in PBMC-engrafted severe combined immunodeficiency mice. RESULTS: Relative to omalizumab, XmAb7195 has a 5-fold higher affinity for human IgE and more than 400-fold higher affinity for FcγRIIb. In addition to sequestering soluble IgE, XmAb7195 inhibited plasma cell differentiation and consequent human IgE production through coengagement of IgE B-cell receptor with FcγRIIb. In PBMC-engrafted mice, XmAb7195 reduced total human IgE (but not IgG or IgM) levels by up to 40-fold relative to omalizumab. CONCLUSION: XmAb7195 acts by IgE sequestration coupled with an FcγRIIb-mediated inhibitory mechanism to suppress the formation of IgE-secreting plasma cells and reduce both free and total IgE levels.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Immunoglobulin E/biosynthesis , Receptors, Antigen, B-Cell/antagonists & inhibitors , Receptors, IgE/antagonists & inhibitors , Receptors, IgG/antagonists & inhibitors , Animals , Anti-Allergic Agents/pharmacology , Antibodies, Anti-Idiotypic/blood , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/genetics , Antibody Affinity/immunology , Humans , Immunoglobulin E/metabolism , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin M/biosynthesis , Immunoglobulin M/blood , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/transplantation , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Omalizumab , Protein Binding/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, IgE/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism
8.
Blood ; 119(9): 2074-82, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22246035

ABSTRACT

HM1.24, an immunologic target for multiple myeloma (MM) cells, has not been effectively targeted with therapeutic monoclonal antibodies (mAbs). In this study, we investigated in vitro and in vivo anti-MM activities of XmAb5592, a humanized anti-HM1.24 mAb with Fc-domain engineered to significantly enhance FcγR binding and associated immune effector functions. XmAb5592 increased antibody-dependent cellular cytotoxicity (ADCC) several fold relative to the anti-HM1.24 IgG1 analog against both MM cell lines and primary patient myeloma cells. XmAb5592 also augmented antibody dependent cellular phagocytosis (ADCP) by macrophages. Natural killer (NK) cells became more activated by XmAb5592 than the IgG1 analog, evidenced by increased cell surface expression of granzyme B-dependent CD107a and MM cell lysis, even in the presence of bone marrow stromal cells. XmAb5592 potently inhibited tumor growth in mice bearing human MM xenografts via FcγR-dependent mechanisms, and was significantly more effective than the IgG1 analog. Lenalidomide synergistically enhanced in vitro ADCC against MM cells and in vivo tumor inhibition induced by XmAb5592. A single dose of 20 mg/kg XmAb5592 effectively depleted both blood and bone marrow plasma cells in cynomolgus monkeys. These results support clinical development of XmAb5592, both as a monotherapy and in combination with lenalidomide, to improve patient outcome of MM.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antigens, CD/immunology , Immunoglobulin Fc Fragments/immunology , Multiple Myeloma/therapy , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Degranulation , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Drug Synergism , Female , GPI-Linked Proteins/immunology , Humans , Killer Cells, Natural/immunology , Lenalidomide , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Depletion , Macaca fascicularis , Mice , Mice, SCID , Phagocytosis/drug effects , Phagocytosis/immunology , Plasma Cells/drug effects , Plasma Cells/immunology , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Xenograft Model Antitumor Assays
9.
MAbs ; 3(6): 546-57, 2011.
Article in English | MEDLINE | ID: mdl-22123055

ABSTRACT

Bispecific antibodies based on full-length antibody structures are more optimal than fragment-based formats because they benefit from the favorable properties of the Fc region. However, the homodimeric nature of Fc effectively imposes bivalent binding on all current full-length bispecific antibodies, an attribute that can result in nonspecific activation of cross-linked receptors. We engineered a novel bispecific format, referred to as mAb-Fv, that utilizes a heterodimeric Fc region to enable monovalent co-engagement of a second target antigen in a full-length context. mAb-Fv constructs co-targeting CD16 and CD3 were expressed and purified as heterodimeric species, bound selectively to their co-target antigens, and mediated potent cytotoxic activity by NK cells and T cells, respectively. The capacity to co-engage distinct target antigens simultaneously with different valencies is an improved feature for bispecific antibodies with promising therapeutic implications.


Subject(s)
Antibodies, Bispecific/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , CD3 Complex/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fragments/immunology , Receptors, IgG/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , CD3 Complex/genetics , CD3 Complex/metabolism , Dimerization , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/metabolism , Killer Cells, Natural/immunology , Mice , Models, Molecular , Protein Engineering/methods , Receptors, IgG/genetics , Receptors, IgG/metabolism , T-Lymphocytes/immunology
10.
J Mol Biol ; 396(5): 1474-90, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20045416

ABSTRACT

Fully human monoclonal antibodies (mAbs) derived from transgenic mice or human antibody libraries are the current state of the art for reducing the immunogenicity risk of antibody drugs. Here, we describe a novel method for generating fully human mAbs from nonhuman variable regions using information from the human germline repertoire. Central to our strategy is the rational engineering of residues within and proximal to CDRs and the V(H)/V(L) interface by iteratively exploring substitutions to the closest human germline sequences using semi-automated computational methods. Starting from the parent murine variable regions of three currently marketed mAbs targeting CD25, vascular endothelial growth factor, and tumor necrosis factor alpha, we have generated fully human antibodies with 59, 46, and 45 substitutions, respectively, compared to the parent murine sequences. A large number of these substitutions were in the CDRs, which are typically avoided in humanization methods. Antigen affinities of the fully human variants were comparable to the chimeric mAbs in each case. Furthermore, in vitro functional characterization indicated that all retain potency of the chimeric mAbs and have comparable activity to their respective marketed drugs daclizumab, bevacizumab, and infliximab. Based on local and global sequence identity, the sequences of our engineered mAbs are indistinguishable from those of fully human mAbs isolated from transgenic mice or human antibody libraries. This work establishes a simple rational engineering methodology for generating fully human antibody therapeutics from murine mAbs produced from standard hybridoma technology.


Subject(s)
Antibodies, Monoclonal/genetics , Immunoglobulin Variable Region/genetics , Protein Engineering/methods , Amino Acid Sequence , Amino Acid Substitution , Animals , Cells, Cultured , Complementarity Determining Regions/genetics , Humans , In Vitro Techniques , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Mice , Mice, Transgenic , Molecular Sequence Data , Peptide Library , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Homology, Amino Acid , Species Specificity , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors
11.
Science ; 301(5641): 1895-8, 2003 Sep 26.
Article in English | MEDLINE | ID: mdl-14512626

ABSTRACT

Tumor necrosis factor (TNF) is a key regulator of inflammatory responses and has been implicated in many pathological conditions. We used structure-based design to engineer variant TNF proteins that rapidly form heterotrimers with native TNF to give complexes that neither bind to nor stimulate signaling through TNF receptors. Thus, TNF is inactivated by sequestration. Dominant-negative TNFs represent a possible approach to anti-inflammatory biotherapeutics, and experiments in animal models show that the strategy can attenuate TNF-mediated pathology. Similar rational design could be used to engineer inhibitors of additional TNF superfamily cytokines as well as other multimeric ligands.


Subject(s)
Protein Engineering , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Substitution , Animals , Antigens, CD/metabolism , Apoptosis , Arthritis, Experimental/drug therapy , Biopolymers , Caspases/metabolism , Cell Line , Cell Nucleus/metabolism , Computer Simulation , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Galactosamine/pharmacology , HeLa Cells , Humans , Liver/drug effects , NF-kappa B/metabolism , Point Mutation , Rats , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Transcription Factor RelA , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...