Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 4 de 4
1.
MAbs ; 11(6): 1101-1112, 2019.
Article En | MEDLINE | ID: mdl-31161859

Forced degradation experiments of monoclonal antibodies (mAbs) aid in the identification of critical quality attributes (CQAs) by studying the impact of post-translational modifications (PTMs), such as oxidation, deamidation, glycation, and isomerization, on biological functions. Structure-function characterization of mAbs can be used to identify the PTM CQAs and develop appropriate analytical and process controls. However, the interpretation of forced degradation results can be complicated because samples may contain mixtures of asymmetrically and symmetrically modified mAbs with one or two modified chains. We present a process to selectively create symmetrically and asymmetrically modified antibodies for structure-function characterization using the bispecific DuoBody® platform. Parental molecules mAb1 and mAb2 were first stressed with peracetic acid to induce methionine oxidation. Bispecific antibodies were then prepared from a mixture of oxidized or unoxidized parental mAbs by a controlled Fab-arm exchange process. This process was used to systematically prepare four bispecific mAb products: symmetrically unoxidized, symmetrically oxidized, and both combinations of asymmetrically oxidized bispecific mAbs. Results of this study demonstrated chain-independent, 1:2 stoichiometric binding of the mAb Fc region to both FcRn receptor and to Protein A. The approach was also applied to create asymmetrically deamidated mAbs at the asparagine 330 residue. Results of this study support the proposed 1:1 stoichiometric binding relationship between the FcγRIIIa receptor and the mAb Fc. This approach should be generally applicable to study the potential impact of any modification on biological function.


Antibodies, Bispecific/chemistry , Antibodies, Monoclonal/chemistry , Immunoglobulin G/chemistry , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Asparagine/chemistry , Asparagine/immunology , Humans , Immunoglobulin G/immunology , Mass Spectrometry , Methionine/chemistry , Methionine/immunology , Oxidation-Reduction , Receptors, IgG/chemistry , Receptors, IgG/immunology , Structure-Activity Relationship
2.
Vaccine ; 25(17): 3338-46, 2007 Apr 30.
Article En | MEDLINE | ID: mdl-17292519

MUC1 (mucin 1) is a transmembrane glycoprotein normally expressed on epithelia of the pancreas, breast, prostate, colon, and lung. However, this self-antigen is over-expressed and aberrantly glycosylated in adenocarcinomas, thereby making it a potential target for immunotherapy. Toward this goal, DNA plasmids encoding human MUC1 (pMUC1) and mouse interleukin-18 (pmuIL-18) were developed, and previous work demonstrated pMUC1/pmuIL18 vaccination protected MUC1 transgenic mice (MUC1.Tg) from subcutaneous tumor challenge. This report shows that pMUC1/pmuIL-18 is effective in preventing and treating pulmonary metastases in MUC1.Tg mice. Vaccination with pMUC1 or pmuIL-18 alone was insufficient to elicit measurable anti-tumor effects. However, co-administration of pMUC1 with pmuIL-18 reduced the incidence of lung tumors and prolonged survival. Furthermore, pMUC1/pmuIL-18 immunization protected mice from challenge with MUC1+ tumors, but not from MUC1- tumors, indicating that the anti-tumor effect is antigen-specific. More importantly, pMUC1/pmuIL-18 was effective in treating established tumors. Finally, in vivo antibody-mediated lymphocyte depletion and neutralization of interferon gamma (IFNgamma) revealed that CD8+ T cells and IFNgamma mediate the anti-tumor immunity. Collectively, these results demonstrate that pMUC1/pmuIL-18 breaks tolerance to MUC1, and induces antigen-specific immunity with protective and therapeutic benefit. This suggests that pMUC1/pmuIL-18 DNA vaccination may provide clinical benefit for patients with MUC1+ tumors.


Cancer Vaccines/immunology , Interleukin-18/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mucin-1/immunology , Vaccines, DNA/immunology , Administration, Cutaneous , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Interferon-gamma/physiology , Interleukin-18/genetics , Mice , Mice, Transgenic , Mucin-1/genetics , Vaccination
3.
Nanotechnology ; 18(26): 265301, 2007 Jul 04.
Article En | MEDLINE | ID: mdl-21730395

In this work, we described the processes of synthesizing free-standing indium nanoclusters using inverse micelles and microemulsions as well as synthesizing organic-encapsulated indium nanoclusters using alkanethiols as the organic encapsulants. The synthesized organic-encapsulated indium nanoclusters have demonstrated the feasibilities to be used as plastic compatible soft metal contacts for emerging organic devices. The homogeneously distributed indium nanoclusters with sizes of 10-30 nm have been fabricated on a few different plastic substrates. By changing the alkanethiol carbon chain length and the sizes of the indium nanoclusters, the annealing temperature required to form low-resistance indium thin film conductors has been reduced to 80-100 °C, which is acceptable for a variety of organic thin films.

4.
Vaccine ; 24(16): 3340-52, 2006 Apr 12.
Article En | MEDLINE | ID: mdl-16472547

MUC1 (mucin 1) is a tumor-associated antigen that is overexpressed in many adenocarcinomas. Active immunotherapy targeting tumors expressing MUC1 could have great treatment value. MUC1 DNA vaccines were evaluated in MUC1 transgenic (MUC1.Tg) mice challenged with MC38/MUC1+ tumor cells. Vaccination with MUC1 plasmid DNA (pMUC1) alone was insufficient to induce tumor protection. However, co-administration of pMUC1 with a plasmid encoding murine interleukin-18 (pmuIL-18) resulted in significant tumor protection and survival after tumor challenge. Protection was durable in the absence of additional vaccination, as demonstrated by continued protection of vaccinated mice following tumor rechallenge. Mice surviving challenges with MC38/MUC1+ cells showed significant protection after challenge with MUC1(-) MC38 tumor cells, suggesting that these mice had developed immune responses to epitopes shared between the tumor cell lines. Antibody-mediated depletion of lymphocyte subsets demonstrated that protection was due largely to CD4+ T cells. This work demonstrates that a naked DNA vaccine can break tolerance to MUC1 and induce an immune response capable of mediating both significant protection from tumor challenge and increased survival.


Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Interleukin-18/immunology , Mucins/immunology , Neoplasms, Experimental/prevention & control , Vaccines, DNA/immunology , Adjuvants, Immunologic , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Base Sequence , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Female , Genetic Vectors , Interleukin-18/genetics , Lymphocyte Depletion , Lymphocyte Subsets/immunology , Mice , Mice, Transgenic , Molecular Sequence Data , Mucin-1 , Mucins/genetics , Neoplasms, Experimental/immunology , Plasmids , Survival Analysis , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
...