Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
RMD Open ; 7(2)2021 05.
Article in English | MEDLINE | ID: mdl-34011674

ABSTRACT

OBJECTIVE: To investigate serum protein expression in participants with psoriatic arthritis (PsA) and changes after guselkumab treatment. METHODS: Participants with PsA were treated with guselkumab or placebo in the DISCOVER-1 and DISCOVER-2 studies. Serum levels of acute phase reactants C reactive protein (CRP) and serum amyloid A (SAA) and inflammatory cytokines/chemokines were measured at weeks 0, 4 and 24 in 300 study participants and 34 healthy controls (HCs). The PSUMMIT studies measured serum interleukin (IL)-17A, IL-17F and CRP after ustekinumab treatment and levels with ustekinumab versus guselkumab treatment were compared. RESULTS: Baseline serum levels of CRP, SAA, IL-6, IL-17A and IL-17F were elevated in participants with active PsA vs HCs (p<0.05, geometric mean (GM) ≥40% higher). Baseline T-helper cell 17 (Th17) effector cytokines were significantly associated with baseline psoriasis but not joint disease activity. Compared with placebo, guselkumab treatment resulted in decreases in serum CRP, SAA, IL-6, IL-17A, IL-17F and IL-22 as early as week 4 and continued to decrease through week 24 (p<0.05, GM decrease from baseline ≥33%). At week 24, IL-17A and IL-17F levels were not significantly different from HCs, suggesting normalisation of peripheral IL-23/Th17 axis effector cytokines postguselkumab treatment. Reductions in IL-17A/IL-17F levels were greater in guselkumab-treated versus ustekinumab-treated participants, whereas effects on CRP levels were similar. CONCLUSION: Guselkumab treatment reduced serum protein levels of acute phase and Th17 effector cytokines and achieved comparable levels to those in HCs. In participants with PsA, reductions of IL-17A and IL-17F were of greater magnitude after treatment with guselkumab than with ustekinumab.


Subject(s)
Arthritis, Psoriatic , Acute-Phase Proteins , Antibodies, Monoclonal, Humanized , Arthritis, Psoriatic/drug therapy , Cytokines , Double-Blind Method , Humans , Severity of Illness Index , Treatment Outcome
2.
Chembiochem ; 19(2): 121-125, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29120508

ABSTRACT

A fully synthetic MUC1-based cancer vaccine was designed and chemically synthesized containing an endogenous helper T-epitope (MHC class II epitope). The vaccine elicited robust IgG titers that could neutralize cancer cells by antibody-dependent cell-mediated cytotoxicity (ADCC). It also activated cytotoxic T-lymphocytes. Collectively, the immunological data demonstrate engagement of helper T-cells in immune activation. A synthetic methodology was developed for a penta-glycosylated MUC1 glycopeptide, and antisera of mice immunized by the new vaccine recognized such a structure. Previously reported fully synthetic MUC1-based cancer vaccines that elicited potent immune responses employed exogenous helper T-epitopes derived from microbes. It is the expectation that the use of the newly identified endogenous helper T-epitope will be more attractive, because it will activate cognate CD4+ T-cells that will provide critical tumor-specific help intratumorally during the effector stage of tumor rejection and will aid in the generation of sustained immunological memory.


Subject(s)
Cancer Vaccines/chemical synthesis , Cancer Vaccines/immunology , Glycopeptides/immunology , Mucin-1/immunology , Vaccines, Synthetic/immunology , Animals , Cancer Vaccines/chemistry , Glycopeptides/chemistry , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Conformation , Mucin-1/chemistry , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, Synthetic/chemistry
3.
Biomolecules ; 6(3)2016 06 29.
Article in English | MEDLINE | ID: mdl-27367740

ABSTRACT

Cancer vaccines have often failed to live up to their promise, although recent results with checkpoint inhibitors are reviving hopes that they will soon fulfill their promise. Although mutation-specific vaccines are under development, there is still high interest in an off-the-shelf vaccine to a ubiquitous antigen, such as MUC1, which is aberrantly expressed on most solid and many hematological tumors, including more than 90% of breast carcinomas. Clinical trials for MUC1 have shown variable success, likely because of immunological tolerance to a self-antigen and to poor immunogenicity of tandem repeat peptides. We hypothesized that MUC1 peptides could be optimized, relying on heteroclitic optimizations of potential anchor amino acids with and without tumor-specific glycosylation of the peptides. We have identified novel MUC1 class I peptides that bind to HLA-A*0201 molecules with significantly higher affinity and function than the native MUC1 peptides. These peptides elicited CTLs from normal donors, as well as breast cancer patients, which were highly effective in killing MUC1-expressing MCF-7 breast cancer cells. Each peptide elicited lytic responses in greater than 6/8 of normal individuals and 3/3 breast cancer patients. The CTLs generated against the glycosylated-anchor modified peptides cross reacted with the native MUC1 peptide, STAPPVHNV, suggesting these analog peptides may offer substantial improvement in the design of epitope-based vaccines.


Subject(s)
Cancer Vaccines , HLA-A2 Antigen , Mucin-1 , Peptides , T-Lymphocytes, Cytotoxic/immunology , Aged , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Female , Glycosylation , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Immune Tolerance , Interferon-gamma , MCF-7 Cells , Middle Aged , Mucin-1/immunology , Mucin-1/metabolism , Peptides/immunology , Peptides/metabolism
4.
PLoS One ; 11(1): e0145920, 2016.
Article in English | MEDLINE | ID: mdl-26788922

ABSTRACT

It remains challenging to produce decisive vaccines against MUC1, a tumor-associated antigen widely expressed by pancreas, breast and other tumors. Employing clinically relevant mouse models, we ruled out such causes as irreversible T-cell tolerance, inadequate avidity, and failure of T-cells to recognize aberrantly glycosylated tumor MUC1. Instead, every tested MUC1 preparation, even non-glycosylated synthetic 9mer peptides, induced interferon gamma-producing CD4(+) and CD8(+) T-cells that recognized glycosylated variants including tumor-associated MUC1. Vaccination with synthetic peptides conferred protection as long as vaccination was repeated post tumor challenge. Failure to revaccinate post challenge was associated with down-regulated tumor MUC1 and MHC molecules. Surprisingly, direct admixture of MUC1-expressing tumor with MUC1-hyperimmune T-cells could not prevent tumor outgrowth or MUC1 immunoediting, whereas ex vivo activation of the hyperimmune T-cells prior to tumor admixture rendered them curative. Therefore, surrogate T-cell preactivation outside the tumor bed, either in culture or by repetitive vaccination, can overcome tumor escape.


Subject(s)
Cancer Vaccines/therapeutic use , Mucin-1/genetics , Mucin-1/immunology , Neoplasms, Experimental/prevention & control , Peptides/therapeutic use , Animals , Antigens/chemistry , Antigens/immunology , Antigens/therapeutic use , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cell Line, Tumor , Glycosylation , Humans , Mice , Mice, Transgenic , Mucin-1/metabolism , Neoplasms, Experimental/immunology , Peptides/chemistry , Peptides/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape
5.
Chem Commun (Camb) ; 51(50): 10214-7, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26022217

ABSTRACT

A strategy for the linear synthesis of a sialylated glycolipopeptide cancer vaccine candidate has been developed using a strategically designed sialyl-Tn building block and microwave-assisted solid-phase peptide synthesis. The glycolipopeptide elicited potent humoral and cellular immune responses. T-cells primed by such a vaccine candidate could be restimulated by tumor-associated MUC1.


Subject(s)
Glycopeptides/chemistry , Mucin-1/chemistry , N-Acetylneuraminic Acid/chemistry , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Chemistry Techniques, Synthetic , Humans , Molecular Sequence Data
6.
Chembiochem ; 15(10): 1508-13, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24890740

ABSTRACT

The mucin MUC1 is overexpressed and aberrantly glycosylated by many epithelial cancer cells manifested by truncated O-linked saccharides. Although tumor-associated MUC1 has generated considerable attention because of its potential for the development of a therapeutic cancer vaccine, it has been difficult to design constructs that consistently induce cytotoxic T-lymphocytes (CTLs) and ADCC-mediating antibodies specific for the tumor form of MUC1. We have designed, chemically synthesized, and immunologically examined vaccine candidates each composed of a glycopeptide derived from MUC1, a promiscuous Thelper peptide, and a TLR2 (Pam3 CysSK4 ) or TLR9 (CpG-ODN 1826) agonist. It was found that the Pam3 CysSK4 -containing compound elicits more potent antigenic and cellular immune responses, resulting in a therapeutic effect in a mouse model of mammary cancer. It is thus shown, for the first time, that the nature of an inbuilt adjuvant of a tripartite vaccine can significantly impact the quality of immune responses elicited against a tumor-associated glycopeptide. The unique adjuvant properties of Pam3 CysSK4 , which can reduce the suppressive function of regulatory T cells and enhance the cytotoxicity of tumor-specific CTLs, are likely responsible for the superior properties of the vaccine candidate 1.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Breast Neoplasms/therapy , Cancer Vaccines/therapeutic use , Glycopeptides/therapeutic use , Mucin-1/therapeutic use , Toll-Like Receptor 2/agonists , Toll-Like Receptor 9/agonists , Adjuvants, Immunologic/chemistry , Amino Acid Sequence , Animals , Breast/immunology , Breast Neoplasms/immunology , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cell Line, Tumor , Female , Glycopeptides/chemistry , Glycopeptides/immunology , Glycosylation , Humans , Immunity, Cellular , Immunization , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mucin-1/chemistry , Mucin-1/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
7.
Proc Natl Acad Sci U S A ; 109(1): 261-6, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22171012

ABSTRACT

The mucin MUC1 is typically aberrantly glycosylated by epithelial cancer cells manifested by truncated O-linked saccharides. The resultant glycopeptide epitopes can bind cell surface major histocompatibility complex (MHC) molecules and are susceptible to recognition by cytotoxic T lymphocytes (CTLs), whereas aberrantly glycosylated MUC1 protein on the tumor cell surface can be bound by antibodies to mediate antibody-dependent cell-mediated cytotoxicity (ADCC). Efforts to elicit CTLs and IgG antibodies against cancer-expressed MUC1 have not been successful when nonglycosylated MUC1 sequences were used for vaccination, probably due to conformational dissimilarities. Immunizations with densely glycosylated MUC1 peptides have also been ineffective due to impaired susceptibility to antigen processing. Given the challenges to immuno-target tumor-associated MUC1, we have identified the minimum requirements to consistently induce CTLs and ADCC-mediating antibodies specific for the tumor form of MUC1 resulting in a therapeutic response in a mouse model of mammary cancer. The vaccine is composed of the immunoadjuvant Pam(3)CysSK(4), a peptide T(helper) epitope and an aberrantly glycosylated MUC1 peptide. Covalent linkage of the three components was essential for maximum efficacy. The vaccine produced CTLs, which recognized both glycosylated and nonglycosylated peptides, whereas a similar nonglycosylated vaccine gave CTLs which recognized only nonglycosylated peptide. Antibodies elicited by the glycosylated tripartite vaccine were significantly more lytic compared with the unglycosylated control. As a result, immunization with the glycosylated tripartite vaccine was superior in tumor prevention. Besides its own aptness as a clinical target, these studies of MUC1 are likely predictive of a covalent linking strategy applicable to many additional tumor-associated antigens.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Mucin-1/immunology , Neoplasms/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, Neoplasm/chemistry , Cancer Vaccines/chemistry , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Glycosylation , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Mammary Tumor Virus, Mouse/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/immunology , Vaccines, Synthetic/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...