Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Breast Cancer Res ; 20(1): 43, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29859099

ABSTRACT

BACKGROUND: Proteolytic impairment of the Fc effector functions of therapeutic monoclonal antibodies (mAbs) can compromise their antitumor efficacy in the tumor microenvironment and may represent an unappreciated mechanism of host immune evasion. Pertuzumab is a human epidermal growth factor receptor 2 (HER2)-targeting antibody and has been widely used in the clinic in combination with trastuzumab for treatment of HER2-overexpressing breast cancer. Pertuzumab susceptibility to proteolytic hinge cleavage and its impact on the drug's efficacy has not been previously studied. METHODS: Pertuzumab was incubated with high and low HER2-expressing cancer cells and proteolytic cleavage in the lower hinge region was detected by western blotting. The single hinge cleaved pertuzumab (scIgG-P) was purified and evaluated for its ability to mediate antibody-dependent cellular cytotoxicity (ADCC) in vitro and anti-tumor efficacy in vivo. To assess the cleavage of trastuzumab (IgG-T) and pertuzumab (IgG-P) when simultaneously bound to the same cancer cell surface, F(ab')2 fragments of IgG-T or IgG-P were combined with the intact IgG-P and IgG-T, respectively, to detect scIgG generation by western blotting. RESULTS: Pertuzumab hinge cleavage occurred when the mAb was incubated with high HER2-expressing cancer cells. The hinge cleavage of pertuzumab caused a substantial loss of ADCC in vitro and reduced antitumor efficacy in vivo. The reduced ADCC function of scIgG-P was restored by an anti-hinge mAb specific for a cleavage site neoepitope. In addition, we constructed a protease-resistant version of the anti-hinge mAb that restored ADCC and the cell-killing functions of pertuzumab when cancer cells exressed a potent IgG hinge-cleaving protease. We also observed increased hinge cleavage of pertuzumab when combined with trastuzumab. CONCLUSION: The reduced Fc effector function of single hinge-cleaved pertuzumab can be restored by an anti-hinge mAb. The restoration effect indicated that immune function could be readily augmented when the damaged primary antibodies were bound to cancer cell surfaces. The anti-hinge mAb also restored Fc effector function to the mixture of proteolytically disabled trastuzumab and pertuzumab, suggesting a general therapeutic strategy to restore the immune effector function to protease-inactivated anticancer antibodies in the tumor microenvironment. The findings point to a novel tactic for developing breast cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Breast Neoplasms/drug therapy , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoglobulin Fc Fragments/adverse effects , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Mice , Proteolysis/drug effects , Receptor, ErbB-2/immunology , Tumor Escape/immunology , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
2.
J Immunol ; 194(9): 4379-86, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25795760

ABSTRACT

Trastuzumab has been used for the treatment of HER2-overexpressing breast cancer for more than a decade, but the mechanisms of action for the therapy are still being actively investigated. Ab-dependent cell-mediated cytotoxicity mediated by NK cells is well recognized as one of the key mechanisms of action for trastuzumab, but trastuzumab-mediated Ab-dependent cellular phagocytosis (ADCP) has not been established. In this study, we demonstrate that macrophages, by way of phagocytic engulfment, can mediate ADCP and cancer cell killing in the presence of trastuzumab. Increased infiltration of macrophages in the tumor tissue was associated with enhanced efficacy of trastuzumab whereas depletion of macrophages resulted in reduced antitumor efficacy in mouse xenograft tumor models. Among the four mouse FcγRs, FcγRIV exhibits the strongest binding affinity to trastuzumab. Knockdown of FcγRIV in mouse macrophages reduced cancer cell killing and ADCP activity triggered by trastuzumab. Consistently, an upregulation of FcγRIV expression by IFN-γ triggered an increased ADCP activity by trastuzumab. In an analogous fashion, IFN-γ priming of human macrophages increased the expression of FcγRIII, the ortholog of murine FcγRIV, and increased trastuzumab-mediated cancer cell killing. Thus, in two independent systems, the results indicated that activation of macrophages in combination with trastuzumab can serve as a therapeutic strategy for treating high HER2 breast cancer by boosting ADCP killing of cancer cells.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Macrophages/immunology , Macrophages/metabolism , Phagocytosis/drug effects , Phagocytosis/immunology , Receptor, ErbB-2/metabolism , Receptors, IgG/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Disease Models, Animal , Gene Expression , Heterografts , Humans , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Receptor, ErbB-2/genetics , Trastuzumab
3.
Gastroenterology ; 149(6): 1564-1574.e3, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26170138

ABSTRACT

BACKGROUND & AIMS: Many patients with inflammatory bowel disease (IBD) fail to respond to anti-tumor necrosis factor (TNF) agents such as infliximab and adalimumab, and etanercept is not effective for treatment of Crohn's disease. Activated matrix metalloproteinase 3 (MMP3) and MMP12, which are increased in inflamed mucosa of patients with IBD, have a wide range of substrates, including IgG1. TNF-neutralizing agents act in inflamed tissues; we investigated the effects of MMP3, MMP12, and mucosal proteins from IBD patients on these drugs. METHODS: Biopsy specimens from inflamed colon of 8 patients with Crohn's disease and 8 patients with ulcerative colitis, and from normal colon of 8 healthy individuals (controls), were analyzed histologically, or homogenized and proteins were extracted. We also analyzed sera from 29 patients with active Crohn's disease and 33 patients with active ulcerative colitis who were candidates to receive infliximab treatment. Infliximab, adalimumab, and etanercept were incubated with mucosal homogenates from patients with IBD or activated recombinant human MMP3 or MMP12 and analyzed on immunoblots or in luciferase reporter assays designed to measure TNF activity. IgG cleaved by MMP3 or MMP12 and antihinge autoantibodies against neo-epitopes on cleaved IgG were measured in sera from IBD patients who subsequently responded (clinical remission and complete mucosal healing) or did not respond to infliximab. RESULTS: MMP3 and MMP12 cleaved infliximab, adalimumab, and etanercept, releasing a 32-kilodalton Fc monomer. After MMP degradation, infliximab and adalimumab functioned as F(ab')2 fragments, whereas cleaved etanercept lost its ability to neutralize TNF. Proteins from the mucosa of patients with IBD reduced the integrity and function of infliximab, adalimumab, and etanercept. TNF-neutralizing function was restored after incubation of the drugs with MMP inhibitors. Serum levels of endogenous IgG cleaved by MMP3 and MMP12, and antihinge autoantibodies against neo-epitopes of cleaved IgG, were higher in patients who did not respond to treatment vs responders. CONCLUSIONS: Proteolytic degradation may contribute to the nonresponsiveness of patients with IBD to anti-TNF agents.


Subject(s)
Biological Factors/metabolism , Immunoglobulin G/metabolism , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Proteolysis/drug effects , Tumor Necrosis Factor-alpha/metabolism , Adalimumab/metabolism , Antibodies, Monoclonal, Humanized/metabolism , Biological Factors/pharmacology , Biopsy , Case-Control Studies , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Colitis, Ulcerative/metabolism , Colon/immunology , Colon/metabolism , Colon/pathology , Crohn Disease/drug therapy , Crohn Disease/immunology , Crohn Disease/metabolism , Epitopes/metabolism , Etanercept/metabolism , Female , Humans , Immunoblotting/methods , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Infliximab/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Male , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 3/metabolism , Middle Aged
4.
J Biol Chem ; 288(43): 30843-54, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-23986451

ABSTRACT

Molecularly engineered antibodies with fit-for-purpose properties will differentiate next generation antibody therapeutics from traditional IgG1 scaffolds. One requirement for engineering the most appropriate properties for a particular therapeutic area is an understanding of the intricacies of the target microenvironment in which the antibody is expected to function. Our group and others have demonstrated that proteases secreted by invasive tumors and pathological microorganisms are capable of cleaving human IgG1, the most commonly adopted isotype among monoclonal antibody therapeutics. Specific cleavage in the lower hinge of IgG1 results in a loss of Fc-mediated cell-killing functions without a concomitant loss of antigen binding capability or circulating antibody half-life. Proteolytic cleavage in the hinge region by tumor-associated or microbial proteases is postulated as a means of evading host immune responses, and antibodies engineered with potent cell-killing functions that are also resistant to hinge proteolysis are of interest. Mutation of the lower hinge region of an IgG1 resulted in protease resistance but also resulted in a profound loss of Fc-mediated cell-killing functions. In the present study, we demonstrate that specific mutations of the CH2 domain in conjunction with lower hinge mutations can restore and sometimes enhance cell-killing functions while still retaining protease resistance. By identifying mutations that can restore either complement- or Fcγ receptor-mediated functions on a protease-resistant scaffold, we were able to generate a novel protease-resistant platform with selective cell-killing functionality.


Subject(s)
Antibodies, Monoclonal , Antibody-Dependent Cell Cytotoxicity , Binding Sites, Antibody , Protein Engineering , Proteolysis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Binding Sites, Antibody/genetics , Binding Sites, Antibody/immunology , Cell Line , Humans , Immunoglobulin G , Receptors, IgG/genetics , Receptors, IgG/immunology
5.
Proteins ; 82(8): 1656-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24638881

ABSTRACT

The functional role of human antihinge (HAH) autoantibodies in normal health and disease remains elusive, but recent evidence supports their role in the host response to IgG cleavage by proteases that are prevalent in certain disorders. Characterization and potential exploitation of these HAH antibodies has been hindered by the absence of monoclonal reagents. 2095-2 is a rabbit monoclonal antibody targeting the IdeS-cleaved hinge of human IgG1. We have determined the crystal structure of the Fab of 2095-2 and its complex with a hinge analog peptide. The antibody is selective for the C-terminally cleaved hinge ending in G236 and this interaction involves an uncommon disulfide in VL CDR3. We probed the importance of the disulfide in VL CDR3 through engineering variants. We identified one variant, QAA, which does not require the disulfide for biological activity or peptide binding. The structure of this variant offers a starting point for further engineering of 2095-2 with the same specificity, but lacking the potential manufacturing liability of an additional disulfide. Proteins 2014; 82:1656-1667. © 2014 Wiley Periodicals, Inc.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Amino Acid Sequence , Animals , Crystallography, X-Ray , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Molecular Docking Simulation , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Protein Conformation , Proteolysis , Rabbits
6.
J Immunol ; 189(11): 5457-66, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23105143

ABSTRACT

Tumor-associated macrophages (TAMs) have been shown to promote tumor progression, and increased TAM infiltration often correlates with poor prognosis. However, questions remain regarding the phenotype of macrophages within the tumor and their role in mAb-dependent cytotoxicity. This study demonstrates that whereas TAMs have protumor properties, they maintain Fc-dependent anti-tumor function. CD11b(+)CD14(+) TAMs isolated from primary human breast tumors expressed activating FcγRs. To model breast cancer TAMs in vitro, conditioned medium from breast cancer cells was used to drive human peripheral monocyte differentiation into macrophages. Tumor-conditioned macrophages were compared with in vitro derived M1 and M2a macrophages and were found to promote tumor cell invasion and express M2a markers, confirming their protumor potential. However, unlike M2a macrophages, tumor-conditioned macrophages expressed FcγRs and phagocytosed tumor cells in the presence of a tumor Ag-targeting mAb, unmasking an underappreciated tumoricidal capacity of TAMs. In vivo macrophage depletion reduced the efficacy of anti-CD142 against MDA-MB-231 xenograft growth and metastasis in SCID/beige mice, implicating a critical role for macrophages in Fc-dependent cell killing. M-CSF was identified in tumor-conditioned media and shown to be capable of differentiating macrophages with both pro- and anti-tumor properties. These results highlight the plasticity of TAMs, which are capable of promoting tumor progression and invasion while still retaining tumoricidal function in the presence of tumor-targeting mAbs.


Subject(s)
Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Macrophages/immunology , Phagocytosis , Receptors, IgG/immunology , Animals , Breast Neoplasms/pathology , CD11b Antigen/immunology , Cell Movement/drug effects , Cell Proliferation , Culture Media, Conditioned/pharmacology , Disease Progression , Female , Humans , Immunophenotyping , Lipopolysaccharide Receptors/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, SCID , Neoplasm Invasiveness/immunology , Neoplasm Transplantation , Primary Cell Culture
7.
Breast Cancer Res ; 14(4): R116, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22873525

ABSTRACT

INTRODUCTION: Recent studies reported that human IgG antibodies are susceptible to specific proteolytic cleavage in their lower hinge region, and the hinge cleavage results in a loss of Fc-mediated effector functions. Trastuzumab is a humanized IgG1 therapeutic monoclonal antibody for the treatment of HER2-overexpressing breast cancers, and its mechanisms of action consist of inhibition of HER2 signaling and Fc-mediated antibody-dependent cellular cytotoxicity (ADCC). The objective of this study is to investigate the potential effect of proteinase hinge cleavage on the efficacy of trastuzumab using both a breast cancer cell culture method and an in vivo mouse xenograft tumor model. METHODS: Trastuzumab antibody was incubated with a panel of human matrix metalloproteinases, and proteolytic cleavage in the lower hinge region was detected using both western blotting and mass spectrometry. Single hinge cleaved trastuzumab (scIgG-T) was purified and evaluated for its ability to mediate ADCC and inhibition of breast cancer cell proliferation in vitro as well as anti-tumor efficacy in the mouse xenograft tumor model. Infiltrated immune cells were detected in tumor tissues by immunohistochemistry. RESULTS: scIgG-T retains HER2 antigen binding activity and inhibits HER2-mediated downstream signaling and cell proliferation in vitro when compared with the intact trastuzumab. However, scIgG-T lost Fc-mediated ADCC activity in vitro, and had significantly reduced anti-tumor efficacy in a mouse xenograft tumor model. Immunohistochemistry showed reduced immune cell infiltration in tumor tissues treated with scIgG-T when compared with those treated with the intact trastuzumab, which is consistent with the decreased ADCC mediated by scIgG-T in vitro. CONCLUSION: Trastuzumab can be cleaved by matrix metalloproteinases within the lower hinge. scIgG-T exhibited a significantly reduced anti-tumor efficacy in vivo due to the weakened immune effector function such as ADCC. The results suggest that the lower hinge cleavage of trastuzumab can occur in the tumor microenvironment where matrix metalloproteinases often have high levels of expression and scIgG-T might compromise its anti-tumor efficacy in the clinic. However, further studies are needed to validate these hypotheses in the clinical setting.


Subject(s)
Antibodies, Monoclonal, Humanized/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line , Disease Models, Animal , Female , Humans , Matrix Metalloproteinases/metabolism , Mice , Molecular Sequence Data , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Protein Binding , Proteolysis , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptors, IgG/metabolism , Trastuzumab , Xenograft Model Antitumor Assays
8.
Proc Natl Acad Sci U S A ; 106(42): 17864-9, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19815504

ABSTRACT

The successful elimination of pathogenic cells and microorganisms by the humoral immune system relies on effective interactions between host immunoglobulins and Fc gamma receptors on effector cells, in addition to the complement system. Essential Ig motifs that direct those interactions reside within the conserved IgG lower hinge/CH2 interface. We noted that a group of tumor-related and microbial proteases cleaved human IgG1s in that region, and the "nick" of just one of the heavy chains profoundly inhibited IgG1 effector functions. We focused on IgG1 monoclonal antibodies (mAbs) since IgG1 is the most abundant human subclass and demonstrates robust Fc-mediated effector functions. The loss of Fc-mediated cell killing activities was correlated with diminished binding to the Fc gamma family of receptors, but a similar decrease in affinity was not observed toward the FcRn receptor that maintains IgG in circulation. Endogenous human IgG cleavage products of comparable size to mAbs with the single cleavage were detected by Western blot analysis in synovial fluid from patients with rheumatoid arthritis and in breast carcinoma extracts. Their detection is problematic under physiological conditions, since there is no loss of structure, and antigen-binding capability is unaffected. These findings suggest that within the hostile proteolytic microenvironments associated with many diseases, key effector functions of host IgGs, or therapeutic Abs, may be compromised.


Subject(s)
Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Peptide Hydrolases/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , Bacterial Proteins/metabolism , Binding Sites , Breast Neoplasms/enzymology , Cell Membrane/immunology , Female , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/metabolism , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Receptors, IgG/metabolism , Serine Endopeptidases/metabolism , Staphylococcus aureus/enzymology , Streptococcus pyogenes/enzymology
9.
Commun Biol ; 5(1): 960, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36104515

ABSTRACT

Natural killer (NK) cells mediate antibody dependent cytotoxic killing of cancer cells via cross-linking FcγR on NK cells with IgG-Fc. Studies have shown that the single-hinge cleaved IgGs (scIgGs) have dysfunctional Fc and failed engagement with FcγRs on immune cells. However, little is known about how scIgGs impact on antitumor immunity in the tumor microenvironment. In this study, we revealed a significant association of tumor scIgGs with tumor progression and poor outcomes of breast cancer patients (n = 547). Using multiple mouse tumor models, we demonstrated that tumor scIgGs reduced NK cell cytotoxic activities and resulted in aggressive tumor progression. We further showed that an anti-hinge specific monoclonal antibody (AHA) rescued the dysfunctional Fc in scIgGs by providing a functional Fc and restored NK cell cytotoxic activity. These findings point to a novel immunotherapeutic strategy to enhance Fc engagement with FcγRs for activation of anticancer immunity.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Immunoglobulin G , Killer Cells, Natural , Mice , Neoplastic Processes , Tumor Microenvironment
10.
ScientificWorldJournal ; 11: 1153-67, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21623461

ABSTRACT

Human anti-IgG hinge (HAH) autoantibodies constitute a class of immunoglobulins that recognize cryptic epitopes in the hinge region of antibodies exposed after proteolytic cleavage, but do not bind to the intact IgG counterpart. Detailed molecular characterizations of HAH autoantibodies suggest that they are, in some cases, distinct from natural autoantibodies that arise independent of antigenic challenge. Multiple studies have attempted to define the specificity of HAH autoantibodies, which were originally detected as binding to fragments possessing C-terminal amino acid residues exposed in either the upper or lower hinge regions of IgGs. Numerous investigators have provided information on the isotype profiles of the HAH autoantibodies, as well as correlations among protease cleavage patterns and HAH autoantibody reactivity. Several biological functions have been attributed to HAH autoantibodies, ranging from house-cleaning functions to an immunosuppressive role to restoring function to cleaved IgGs. In this review, we discuss both the historic and current literature regarding HAH autoantibodies in terms of their origins, specificity, and proposed biological relevance.


Subject(s)
Antibodies, Anti-Idiotypic/physiology , Autoantibodies/physiology , Models, Immunological , Antibodies, Anti-Idiotypic/immunology , Autoantibodies/immunology , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Hinge Exons/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin G/metabolism
11.
J Immunol ; 181(5): 3183-92, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18713989

ABSTRACT

A number of proteases of potential importance to human physiology possess the ability to selectively degrade and inactivate Igs. Proteolytic cleavage within and near the hinge domain of human IgG1 yielded products including Fab and F(ab')(2) possessing full Ag binding capability but absent several functions needed for immune destruction of cellular pathogens. In parallel experiments, we showed that the same proteolytically generated Fabs and F(ab')(2)s become self-Ags that were widely recognized by autoantibodies in the human population. Binding analyses using various Fab and F(ab')(2), as well as single-chain peptide analogues, indicated that the autoantibodies targeted the newly exposed sequences where proteases cleave the hinge. The point of cleavage may be less of a determinant for autoantibody binding than the exposure of an otherwise cryptic stretch of hinge sequence. It was noted that the autoantibodies possessed an unusually high proportion of the IgG3 isotype in contrast to Abs induced against foreign immunogens in the same human subjects. In light of the recognized potency of IgG3 effector mechanisms, we adopted a functional approach to determine whether human anti-hinge (HAH) autoantibodies could reconstitute the (missing) Fc region effector functions to Fab and F(ab')(2). Indeed, in in vitro cellular assays, purified HAH autoantibodies restored effector functions to F(ab')(2) in both Ab-dependent cellular cytotoxicity and complement-dependent cytotoxicity assays. The results indicate that HAH autoantibodies selectively bind to proteolytically cleaved IgGs and can thereby provide a surrogate Fc domain to reconstitute cell lytic functions.


Subject(s)
Autoantibodies/immunology , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/metabolism , Peptide Hydrolases/metabolism , Antigen-Antibody Complex , Autoantibodies/metabolism , Autoantigens , Binding Sites, Antibody , Humans , Immunoglobulin Fab Fragments/metabolism
12.
Mol Immunol ; 45(7): 1837-46, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18157932

ABSTRACT

A comparative in vitro survey of physiologically relevant human and microbial proteinases defined a number of enzymes that induced specific hinge domain cleavage in human IgG1. Several of these proteinases have been associated with tumor growth, inflammation, and infection. A majority of the identified proteinases converted IgG to F(ab')(2), and a consistent feature of their action was a transient accumulation of a single-cleaved intermediate (scIgG). The scIgG resulted from the relatively rapid cleavage of the first hinge domain heavy chain, followed by a slower cleavage of the second chain to separate the Fc domain from F(ab')(2). Major sites of enzymatic cleavage were identified or confirmed from the mass of the F(ab')(2) or Fab fragments and/or the amino-terminal amino acid sequence of the Fc for each enzyme including human matrix metalloproteinases (MMPs) 3 and 12, human cathepsin G, human neutrophil elastase (Fab), staphylococcal glutamyl endopeptidase I and streptococcal immunoglobulin-degrading enzyme (IdeS). The cleavage sites in IgG1 by MMP-3, cathepsin G and IdeS were used to guide the synthesis of peptide analogs containing the corresponding carboxy-termini to be used as immunogens in rabbits. Rabbit antibodies were successfully generated that showed selective binding to different human F(ab')(2)s and other hinge-cleavage fragments, but not to intact IgG. In Western blotting studies of synovial fluids from individuals with rheumatoid arthritis, the rabbit antibodies yielded patterns consistent with the presence of endogenous IgG fragments including F(ab')(2) and the single-cleaved IgG intermediate. The detection in synovial fluid of IgG fragments similar to those observed in the in vitro biochemical studies suggests that proteolysis of IgG may contribute to localized immune dysfunction in inflammatory environments.


Subject(s)
Arthritis, Rheumatoid/immunology , Bacteria/enzymology , Immunoglobulin Fragments/immunology , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Protein Processing, Post-Translational , Synovial Fluid/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Humans , Hydrogen-Ion Concentration , Immunoglobulin Fragments/isolation & purification , Immunoglobulin G/chemistry , Molecular Sequence Data , Peptides/immunology , Rabbits
13.
Oncoimmunology ; 7(9): e1480300, 2018.
Article in English | MEDLINE | ID: mdl-30228951

ABSTRACT

The host immune system adopts multiple mechanisms involving antibodies to confront cancer cells. Accordingly, anti-tumor mAbs have become mainstays in cancer treatment. However, neither host immunity nor mAb therapies appear capable of controlling tumor growth in all cases. Structural instability of IgG was overlooked as a factor contributing to immunosuppression in the tumor microenvironment. Recently, physiological proteinases were identified that disable IgG immune effector functions. Evidence shows that these proteinases cause localized IgG impairment by selective cleavage of a single IgG peptide bond in the hinge-region. The recognition of IgG cleavage in the tumor microenvironment provides alternatives for tumor immunotherapy.

14.
Transplantation ; 84(3): 308-15, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17700154

ABSTRACT

BACKGROUND: Tissue factor (TF) expression on islets can result in an instant blood-mediated inflammatory reaction (IBMIR) that contributes to early islet loss. We tested whether peritransplant protection of islets from IBMIR with a monoclonal anti-TF antibody (CNTO859) would enhance engraftment in our nonhuman primate marginal mass model. METHODS: Each of six pairs of cynomolgus monkeys (CM) with streptozotocin-induced diabetes was closely matched for metabolic control and was transplanted with 5,000 IEQ/kg allogeneic, ABO-compatible islets from the same donor under the cover of steroid-free immunosuppression. For each pair, experimental animals received islets cultured with 20 microg/mL anti-TF and were dosed with 6 mg/kg anti-TF intravenously, 10-25 min before islet infusion; control monkeys received an equal number of islets from the same preparation cultured without anti-TF and no in vivo treatment. RESULTS: Early fasting C-peptide (CP) values were different between (P<0.01), but not within, pairs and correlated with in vitro functional capacity of islets as assessed by perifusion (r=0.60; P=0.022). Compared to their matched controls, experimental animals had decreased posttransplant markers of coagulation, higher fasting CP levels (1 month posttransplant and end of study) and prolonged graft function. CONCLUSIONS: These data suggest that pretreatment of islets and the recipient with anti-TF may limit the effects of IBMIR, thereby enhancing islet engraftment and survival.


Subject(s)
Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Thromboplastin/physiology , Transplantation Tolerance/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Survival/drug effects , Diabetes Mellitus, Experimental/surgery , Dose-Response Relationship, Drug , Fibrinolysis/immunology , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Macaca fascicularis , Models, Biological , Streptozocin , Thromboplastin/drug effects
15.
J Phys Chem B ; 110(50): 25308-13, 2006 Dec 21.
Article in English | MEDLINE | ID: mdl-17165976

ABSTRACT

We report on the use of femtosecond electron diffraction to resolve the dynamics of electron-phonon relaxation in silicon. Nanofabricated free-standing membranes of polycrystalline silicon were excited below the damage threshold with 387 nm light at a fluence of 5.6 mJ/cm2 absorbed (corresponding to a carrier density of 2.2 x 10(21) cm(-3)). The diffraction pattern was captured over a range of delay times with a time resolution of 350 fs. All of the detected Bragg peaks exhibited intensity loss with a time constant of less than 2 ps. Beyond the initial decay, there was no further change in the diffracted intensity up to 700 ps. We find that the loss of intensity in the diffracted orders is accounted for by the Debye-Waller effect on a time scale indicative of a thermally driven process as opposed to an electronically driven one. Furthermore, the relaxation time constant is consistent with the excitation regime where the phonon emission rate is reduced due to carrier screening.


Subject(s)
Microscopy, Electron/methods , Silicon/chemistry , Electrons , Sensitivity and Specificity , Silicon/radiation effects , Time Factors , Ultraviolet Rays
16.
Oncoimmunology ; 5(5): e1122861, 2016 May.
Article in English | MEDLINE | ID: mdl-27467920

ABSTRACT

Immune suppression is recognized as a hallmark of cancer and this notion is largely based on studies on cellular immunity. Our recent studies have demonstrated a potential new mechanism of cancer suppression of immunity by impairment of antibody effector function mediated by proteolytic enzymes in the tumor microenvironment.

17.
Immunol Lett ; 172: 29-39, 2016 04.
Article in English | MEDLINE | ID: mdl-26905931

ABSTRACT

Pathogens that induce acute and chronic infections, as well as certain cancers, employ numerous strategies to thwart host cellular and humoral immune defenses. One proposed evasion mechanism against humoral immunity is a localized expression of extracellular proteases that cleave the IgG hinge and disable host IgG functions. Host immunity appears to be prepared to counter such a proteolytic tactic by providing a group of autoantibodies, denoted anti-hinge antibodies that specifically bind to cleaved IgGs and provide compensating functional restoration in vitro. These respective counter-measures highlight the complex interrelationships among pathogens and host immunity and suggested to us a possible means for therapeutic intervention. In this study, we combined an investigation of pathogen-mediated proteolysis of host IgGs with an immunization strategy to boost host anti-hinge antibodies. In a Staphylococcus aureus infection model using an artificial tissue cage (wiffle ball) implanted into rabbits, cleaved rabbit IgGs were detected in abundance in the abscesses of untreated animals early after infection. However, in animals previously immunized with peptide analogs of the cleaved IgG hinge to generate substantial anti-hinge antibody titers, S. aureus colony formation was markedly reduced compared to control animals or those similarly immunized with a scrambled peptide sequence. The results of this study demonstrate that extensive local proteolysis of IgGs occurs in a test abscess setting and that immunization to increase host anti-hinge antibodies provided substantial acute protection against bacterial growth.


Subject(s)
Abscess/immunology , Immunoglobulin G/immunology , Peptide Fragments/immunology , Recombinant Fusion Proteins/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Bacterial Load , Disease Models, Animal , Drug Combinations , Freund's Adjuvant/immunology , Hemocyanins/genetics , Humans , Immune Evasion , Immunity, Humoral , Immunization , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Plant Extracts , Proteolysis , Rabbits
18.
J Am Coll Cardiol ; 40(2): 360-6, 2002 Jul 17.
Article in English | MEDLINE | ID: mdl-12106945

ABSTRACT

OBJECTIVES: This study was designed to assess the effect of abciximab on platelet and leukocyte deposition 60 min after stent insertion in nonhuman primates. BACKGROUND: Although it is well established that abciximab improves both short- and long-term clinical outcomes after stent placement, there have been no studies assessing its effect on early platelet and leukocyte deposition. METHODS: Cynomolgus monkeys were pretreated with aspirin and either saline or a 0.4 mg/kg bolus of abciximab, and then subjected to angioplasty and Palmaz-Schatz stent placement in the common iliac artery or abdominal aorta. After 60 min, animals were euthanized and the stented artery was evaluated by immunohistochemistry and morphometry. RESULTS: Complete occlusion of the stented vessel with a thin fibrin(ogen) meshwork and trapped blood occurred in two saline-treated and two abciximab-treated animals. In the four remaining saline-treated animals, a layer of erythrocytes trapped in a network of fibrin(ogen) was noted close to the vessel wall, and this was covered by a layer of large, irregular platelet thrombi. Leukocytes formed a monolayer on top of the platelets and near stent struts. In the four remaining abciximab-treated animals, the mean erythrocyte area was 65% smaller (p = 0.070), the platelet aggregate area was 89% smaller (p = 0.049) and the luminal area was 59% larger (p = 0.004). A monolayer of leukocytes also formed on top of the platelets and near stent struts. CONCLUSIONS: In control stented blood vessels in this study, platelet thrombi formed not at the vessel wall, but on top of an erythrocyte-rich layer, and platelets recruited leukocytes. Abciximab decreased the size of platelet thrombi, but did not prevent leukocyte recruitment.


Subject(s)
Antibodies, Monoclonal/pharmacology , Anticoagulants/pharmacology , Arteries/pathology , Blood Platelets/drug effects , Immunoglobulin Fab Fragments/pharmacology , Leukocytes/drug effects , Platelet Aggregation Inhibitors/pharmacology , Stents/adverse effects , Thrombosis/prevention & control , Abciximab , Animals , Aorta, Abdominal/pathology , Arteries/drug effects , Arteries/injuries , Iliac Artery/pathology , Immunohistochemistry , Macaca fascicularis , Microscopy, Electron , Thrombosis/etiology , Time Factors
19.
MAbs ; 7(3): 494-504, 2015.
Article in English | MEDLINE | ID: mdl-25933349

ABSTRACT

Cytotoxic therapeutic monoclonal antibodies (mAbs) often mediate target cell-killing by eliciting immune effector functions via Fc region interactions with cellular and humoral components of the immune system. Key functions include antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC). However, there has been increased appreciation that along with cell-killing functions, the induction of antibody-dependent cytokine release (ADCR) can also influence disease microenvironments and therapeutic outcomes. Historically, most Fc engineering approaches have been aimed toward modulating ADCC, ADCP, or CDC. In the present study, we describe an Fc engineering approach that, while not resulting in impaired ADCC or ADCP, profoundly affects ADCR. As such, when peripheral blood mononuclear cells are used as effector cells against mAb-opsonized tumor cells, the described mAb variants elicit a similar profile and quantity of cytokines as IgG1. In contrast, although the variants elicit similar levels of tumor cell-killing as IgG1 with macrophage effector cells, the variants do not elicit macrophage-mediated ADCR against mAb-opsonized tumor cells. This study demonstrates that Fc engineering approaches can be employed to uncouple macrophage-mediated phagocytic and subsequent cell-killing functions from cytokine release.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neoplasm , Antibody-Dependent Cell Cytotoxicity , Cytokines/immunology , Immunoglobulin Fc Fragments , Macrophages/immunology , Neoplasms/drug therapy , Protein Engineering , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/genetics , Cell Line, Tumor , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/pharmacology , Neoplasms/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
20.
Mol Cancer Ther ; 14(3): 681-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25552368

ABSTRACT

Primary and acquired resistance to anticancer antibody immunotherapies presents significant clinical challenges. Here, we demonstrate that proteolytic inactivation of cancer-targeting antibodies is an unappreciated contributor to cancer immune evasion, and the finding presents novel opportunities for therapeutic intervention. A single peptide bond cleavage in the IgG1 hinge impairs cancer cell killing due to structural derangement of the Fc region. Hinge-cleaved trastuzumab gradually accumulated on the surfaces of HER2-expressing cancer cell lines in vitro, and was greatly accelerated when the cells were engineered to express the potent bacterial IgG-degrading proteinase (IdeS). Similar to cancer-related matrix metalloproteinases (MMP), IdeS exposes a hinge neoepitope that we have developed an antibody, mAb2095-2, to specifically target the epitope. In in vitro studies, mAb2095-2 restored the lost antibody-dependent cell-mediated cytotoxicity functionality of cell-bound single-cleaved trastuzumab (scIgG-T). In vivo, mAb2095-2 rescued the impaired Fc-dependent tumor-suppressive activity of scIgG-T in a xenograft tumor model and restored the recruitment of immune effector cells into the tumor microenvironment. More importantly, an Fc-engineered proteinase-resistant version of mAb2095-2 rescued trastuzumab antitumor efficacy in a mouse tumor model with human cancer cells secreting IdeS, whereas trastuzumab alone showed significantly reduced antitumor activity in the same model. Consistently, an Fc-engineered proteinase-resistant version of trastuzumab also greatly improved antitumor efficacy in the xenograft tumor model. Taken together, these findings point to a novel cancer therapeutic strategy to rescue proteolytic damage of antibody effector function by an Fc-engineered mAb against the hinge neoepitope and to overcome cancer evasion of antibody immunity.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Neoplasms/drug therapy , Neoplasms/immunology , Proteolysis/drug effects , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Immunoglobulin G/immunology , MCF-7 Cells , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL