Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Language
Publication year range
1.
Bioconjug Chem ; 26(11): 2249-60, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26332743

ABSTRACT

Antibody-drug conjugates (ADC) have emerged as potent antitumor drugs that provide increased efficacy, specificity, and tolerability over chemotherapy for the treatment of cancer. ADCs generated by targeting cysteines and lysines on the antibody have shown efficacy, but these products are heterogeneous, and instability may limit their dosing. Here, a novel technology is described that enables site-specific conjugation of toxins to antibodies using chemistry to produce homogeneous, potent, and highly stable conjugates. We have developed a cell-based mammalian expression system capable of site-specific integration of a non-natural amino acid containing an azide moiety. The azide group enables click cycloaddition chemistry that generates a stable heterocyclic triazole linkage. Antibodies to Her2/neu were expressed to contain N6-((2-azidoethoxy)carbonyl)-l-lysine at four different positions. Each site allowed over 95% conjugation efficacy with the toxins auristatin F or a pyrrolobenzodiazepine (PBD) dimer to generate ADCs with a drug to antibody ratio of >1.9. The ADCs were potent and specific in in vitro cytotoxicity assays. An anti Her2/neu conjugate demonstrated stability in vivo and a PBD containing ADC showed potent efficacy in a mouse tumor xenograph model. This technology was extended to generate fully functional ADCs with four toxins per antibody. The high stability of the azide-alkyne linkage, combined with the site-specific nature of the expression system, provides a means for the generation of ADCs with optimized pharmacokinetic, biological, and biophysical properties.


Subject(s)
Amino Acids/chemistry , Azides/chemistry , Click Chemistry , Cycloaddition Reaction , Immunoconjugates/chemistry , Amino Acids/genetics , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Azides/metabolism , Humans , Immunoconjugates/genetics , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Male , Mice , Neoplasms/drug therapy , Protein Engineering , Rats, Sprague-Dawley , Receptor, ErbB-2/immunology
2.
Protein Eng Des Sel ; 28(10): 467-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26271488

ABSTRACT

Targeting more than one molecule in multifactorial diseases involving several disease mediators may provide improved therapeutic efficacy. Psoriasis is a multifactorial disease in which interleukin (IL)-6 and IL-23 are important disease mediators because they facilitate development of Th17 cells; widely accepted to be associated with psoriasis. To meet the need for new therapeutics, we aimed to create a clinically relevant bispecific drug, by combining the inhibitory properties of anti-IL-6 and anti-IL-23 antibodies, exhibiting high affinity, high stability and the ability to be produced in high yield. The bispecific molecule AZ17 was created by combining high affinity binding domains originating from monoclonal antibodies targeting human IL-6 and IL-23. To allow for high and efficient production, AZ17 was assembled by site-specific bioconjugation from two individual single chain fragment variables that were synthesized separately in Escherichia coli. To improve stability and extend pharmacokinetics, a flexible poly-ethylene glycol molecule was used as linker. In preclinical psoriasis models, AZ17 reduced IL-23-induced ear inflammation and improved psoriasis in a xenograft transplantation model where psoriasis skin is transplanted onto immune-deficient mice. The data presented here suggest AZ17 to be a promising drug candidate in psoriasis and other inflammatory diseases associated with Th17 cell development.


Subject(s)
Antibodies, Bispecific/immunology , Interleukin-23/immunology , Interleukin-6/immunology , Molecular Targeted Therapy , Psoriasis/drug therapy , Psoriasis/immunology , Transplantation, Heterologous , Animals , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/therapeutic use , Antibody Specificity , Disease Models, Animal , Female , Humans , Mice , Rats
3.
J Exp Med ; 202(5): 673-85, 2005 Sep 05.
Article in English | MEDLINE | ID: mdl-16147978

ABSTRACT

Human cytomegalovirus (HCMV) infections of immunocompetent hosts are characterized by a dynamic, life-long interaction in which host immune responses, particularly of T cells, restrain viral replication and prevent disease but do not eliminate the virus or preclude transmission. Because HCMV is among the largest and most complex of known viruses, the T cell resources committed to maintaining this balance have never been characterized completely. Here, using cytokine flow cytometry and 13,687 overlapping 15mer peptides comprising 213 HCMV open reading frames (ORFs), we found that 151 HCMV ORFs were immunogenic for CD4(+) and/or CD8(+) T cells, and that ORF immunogenicity was influenced only modestly by ORF expression kinetics and function. We further documented that total HCMV-specific T cell responses in seropositive subjects were enormous, comprising on average approximately 10% of both the CD4(+) and CD8(+) memory compartments in blood, whereas cross-reactive recognition of HCMV proteins in seronegative individuals was limited to CD8(+) T cells and was rare. These data provide the first glimpse of the total human T cell response to a complex infectious agent and will provide insight into the rules governing immunodominance and cross-reactivity in complex viral infections of humans.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/genetics , Immunologic Memory/immunology , Adult , Cytomegalovirus Infections/genetics , Female , Flow Cytometry , Humans , Immunogenetics , Male , Middle Aged , Open Reading Frames/genetics , Peptides/immunology , Serologic Tests
4.
Infect Immun ; 72(12): 7231-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15557648

ABSTRACT

Chlamydia trachomatis is an obligate intracellular gram-negative bacterium responsible for a wide spectrum of diseases in humans. Both genital and ocular C. trachomatis infections are associated with tissue inflammation and pathology. Dendritic cells (DC) play an important role in both innate and adaptive immune responses to microbial pathogens and are a source of inflammatory cytokines. To determine the potential contribution of DC to the inflammatory process, human DC were infected with C. trachomatis serovar E or L2. Both C. trachomatis serovars were found to infect and replicate in DC. Upon infection, DC up-regulated the expression of costimulatory (B7-1) and cell adhesion (ICAM-1) molecules. Furthermore, chlamydial infection induced the secretion of interleukin-1beta (IL-1beta), IL-6, IL-8, IL-12p70, IL-18, and tumor necrosis factor alpha (TNF-alpha). The mechanisms involved in Chlamydia-induced IL-1beta and IL-18 secretion differed from those of the other cytokines. Chlamydia-induced IL-1beta and IL-18 secretion required infection with viable bacteria and was associated with the Chlamydia-induced activation of caspase-1 in infected host cells. In contrast, TNF-alpha and IL-6 secretion did not require that the Chlamydia be viable, suggesting that there are at least two mechanisms involved in the Chlamydia-induced cytokine secretion in DC. Interestingly, an antibody to Toll-like receptor 4 inhibited Chlamydia-induced IL-1beta, IL-6, and TNF-alpha secretion. The data herein demonstrate that DC can be infected by human C. trachomatis serovars and that chlamydial components regulate the secretion of various cytokines in DC. Collectively, these data suggest that DC play a role in the inflammatory processes caused by chlamydial infections.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/pathogenicity , Cytokines/biosynthesis , Dendritic Cells/physiology , Caspase 1/metabolism , Cell Adhesion Molecules/biosynthesis , Dendritic Cells/microbiology , Enzyme Activation , Humans , Interleukin-1/biosynthesis , Interleukin-18/biosynthesis , Membrane Glycoproteins , Receptors, Cell Surface , Toll-Like Receptor 4 , Toll-Like Receptors
5.
J Immunol ; 173(11): 6905-13, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15557186

ABSTRACT

The intracellular bacterial pathogen Chlamydia is sequestered from the host cell cytoplasm by remaining within an inclusion body during its replication cycle. Nevertheless, CD8(+) T cells recognizing Chlamydia Ags in the context of MHC class I molecules are primed during infection. We have recently described derivation of Chlamydia-specific human CD8(+) T cells by using infected dendritic cells as a surrogate system to reflect Chlamydia-specific CD8(+) T cell responses in vivo. These CD8(+) T cell clones recognize chlamydial Ags processed via the conventional class Ia processing pathway, as assessed by treatment of infected APC with lactacystin and brefeldin A, suggesting that the Ags are translocated from the chlamydial inclusion into the host cell cytosol. In this study, outer membrane protein 2 (OmcB) was identified as the Ag recognized by one of these Chlamydia-specific human CD8(+) T cells, and we defined the HLA*A0101-restricted epitope from this Ag. CD8(+) T cell responses to this epitope were present at high frequencies in the peripheral blood of both of two HLA*A0101 donors tested. In vitro chlamydial growth was completely inhibited by the OmcB-specific CD8(+) T cell clone independently of lytic mechanisms. OmcB is a 60-kDa protein that has been postulated to be associated with the Chlamydia outer membrane complex. The subcellular localization of OmcB to the cytosol of infected cells, as determined by conventional MHC class I Ag processing and presentation, suggests the possibility of an additional, cytosolic-associated function for this protein.


Subject(s)
Antigen Presentation/immunology , Bacterial Outer Membrane Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Chlamydia trachomatis/immunology , Chlamydia trachomatis/metabolism , Adolescent , Adult , Amino Acid Sequence , Bacterial Outer Membrane Proteins/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Transformed , Cells, Cultured , Chlamydia trachomatis/growth & development , Clone Cells , Cytotoxicity Tests, Immunologic , Epitope Mapping/methods , Epitopes, T-Lymphocyte/immunology , Female , HLA-A Antigens/immunology , HLA-A Antigens/metabolism , HeLa Cells , Humans , Male , Molecular Sequence Data , Molecular Weight , Peptide Fragments/immunology , Peptide Fragments/metabolism
6.
J Immunol ; 171(8): 4278-86, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14530352

ABSTRACT

CD8(+) T cells are a key immune component for the eradication of many intracellular pathogens. This study aims to characterize the human CD8(+) T cell response to naturally processed chlamydial Ags in individuals exposed to the intracellular pathogen Chlamydia trachomatis. By using C. trachomatis-infected autologous dendritic cells (DCs) as stimulators, Chlamydia-reactive CD8(+) T cell responses were detected in all 10 individuals tested. The majority of the Chlamydia-reactive CD8(+) T cells were non-MHC class Ia restricted in all three of the individuals tested. From one donor, three non-class Ia-restricted and two class Ia-restricted Chlamydia-specific CD8(+) T cells were cloned and characterized further. All five T cell clones secreted IFN-gamma in response to autologous DCs infected with viable Chlamydia, but not with DCs pulsed with inactivated chlamydial elementary bodies. MHC class Ia-restricted and non-class Ia-restricted responses were inhibited by DC treatment with a proteasomal inhibitor and an endoplasmic reticulum-Golgi transport inhibitor, suggesting that these T cells recognize a peptide Ag translocated to the host cell cytosol during infection that is processed via the classical class Ia Ag-processing pathway. Even though both restricted and nonrestricted CD8(+) T cells produced IFN-gamma in response to Chlamydia-infected fibroblasts, only the non-class Ia-restricted cells were lytic for these targets. The class Ia-restricted CTLs, however, were capable of cytolysis as measured by redirected killing. Collectively, these data demonstrate that both class Ia-restricted and non-classically restricted CD8(+) T cells are elicited in C. trachomatis-exposed individuals. Their role in host immunity remains to be elucidated.


Subject(s)
Antigens, CD1/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Chlamydia trachomatis/immunology , HLA Antigens/immunology , Histocompatibility Antigens Class I/immunology , Antigen Presentation/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Transformed , Cells, Cultured , Clone Cells , Cytotoxicity Tests, Immunologic , Dendritic Cells/immunology , Dendritic Cells/microbiology , Epitopes, T-Lymphocyte/immunology , Female , HeLa Cells , Humans , Interferon-gamma/biosynthesis , Lymphocyte Activation/immunology , Lymphocyte Count , Male
7.
J Immunol ; 170(3): 1498-503, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12538713

ABSTRACT

Linear peptides (SynB vectors) with specific sequence motifs have been identified that are capable of enhancing the transport of a wide range of molecules into cells. These peptide vectors have been used to deliver exogenous peptides and protein Ags across the cell membrane and into the cytoplasm of cells. Specifically, in vitro analysis indicated that these SynB peptides enhanced the uptake of two 9-mer peptide Ags, NP(147-155) and Mtb(250-258) (T cell epitopes of influenza nucleoprotein and Mycobacterium tuberculosis, respectively) and the M. tuberculosis Ag Mtb8.4 protein, into K562 cells when covalently linked to the respective Ags. Furthermore, selected SynB vectors, when conjugated to these same Ags and used as immunogens, resulted in considerably enhanced Ag-specific CTL responses. Several SynB vectors were tested and resulted in varying levels of cellular uptake. The efficiency of uptake correlated with the ability of the SynB construct to deliver each epitope in vivo and induce specific CTL responses in mice. These data suggest that peptide vectors, such as SynB that transport target Ags across the cell membrane in a highly efficient manner, have significant potential for vaccine delivery.


Subject(s)
Antigens, Bacterial/administration & dosage , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/administration & dosage , Lymphocyte Activation , Nucleoproteins/administration & dosage , Peptide Fragments/administration & dosage , RNA-Binding Proteins , T-Lymphocytes, Cytotoxic/immunology , Viral Core Proteins/administration & dosage , Amino Acid Sequence , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , K562 Cells , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Mycobacterium tuberculosis/immunology , Nucleocapsid Proteins , Nucleoproteins/immunology , Nucleoproteins/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Tumor Cells, Cultured , Viral Core Proteins/immunology , Viral Core Proteins/metabolism
8.
Clin Cancer Res ; 8(11): 3407-18, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429628

ABSTRACT

To determine the toxicity and immunogenicity of the HER-2/neu, HLA-A2-restricted peptide E75 in patients with metastatic breast and ovarian cancer, 14 patients were vaccinated with escalating amounts of E75 (100, 500, and 1000 microg) mixed with 250 microg granulocyte macrophage colony-stimulating factor as adjuvant. Each vaccine dose was administered in a total volume of 1.5 ml divided into four intradermal injections and administered weekly for 4 weeks, followed by monthly boosts for a total of 10 injections. Vaccinations were well tolerated without significant toxicity. Blood was drawn before, at 8 weeks, and up to 13-16 months after vaccination for measurement of cellular immunity. Seven of 8 patients tested had significant delayed type hypersensitivity to E75 defined as >5 mm induration. Peripheral blood mononuclear cells from 5 of 9 patients tested proliferated to E75 with a stimulation index of > or = 2.0. Of 8 vaccinated patients tested for induction of a CTL response, 4 responded to stimulation by autologous dendritic cells plus cytokines by eliciting E75-specific lytic activity consistent with the presence of activated/memory cells, 2 others after in vitro stimulation with E75 + interleukin-12 +/- anti-CD152(33KD), whereas 2 others did not respond. Four patients with E75-specific CTLs present specifically recognized E75 on indicator tumors as demonstrated by cold-target inhibition of tumor lysis. These 4 patients showed E75-specific IFN-gamma production. peripheral blood mononuclear cell from 3 of these patients proliferated to E75, but stimulation indices were higher in the prevaccine samples. All 4 of the patients showed DTH responses to E75. These results demonstrate that vaccination with E75+ granulocyte macrophage colony-stimulating factor can induce both peptide-specific IFN-gamma and epitope specific CTLs, which lyse HER-2/neu+ tumors in stage IV patients.


Subject(s)
Antigens, Neoplasm/therapeutic use , Breast Neoplasms/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunoconjugates , Ovarian Neoplasms/therapy , Peptide Fragments/pharmacology , Receptor, ErbB-2/therapeutic use , Abatacept , Adult , Aged , Antigens/metabolism , Antigens, CD , Antigens, Differentiation/metabolism , CTLA-4 Antigen , Cancer Vaccines , Cell Division , Epitopes , Female , HLA-A2 Antigen/biosynthesis , Humans , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lymphocytes/drug effects , Middle Aged , Peptide Fragments/chemistry , Peptides/chemistry , T-Lymphocytes, Cytotoxic/metabolism , Time Factors
9.
Journal of Immunology ; 161(11): 6171-6179, 1998.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP, SESSP-IBACERVO | ID: biblio-1064258

ABSTRACT

We have evaluated the ability of the Leishmania protein LeIF to influence the Th1/Th2 cytokine responses and the generation of LeIF-specific T cell clones in the absence of adjuvant. We characterized LeIF-specific T cell responses in Leishmania major. Infected and uninfected BALB/c mice. These mice develop a strong Th2 response during infection with L. major. When lymph node cells from infected BALB/c mice were stimulated in vitro with LeIF, only IFN-ã (and no detectable IL-4) was found in the culture supernatant. In addition, LeIF down-regulated Leishmania Ag-specific IL-4 production by lymph node cells from infected BALB/c mice. Subsequently, Th responses were evaluated in naive BALB/c mice following immunization with LeIF. T cell clones derived from mice immunized with LeIF preferentially secreted IFN-ã. Finally, to understand the basis for the preferential Th1 cytokine bias observed with LeIF, the ability of LeIF to influence the early cytokine profile was evaluated in splenocytes of SCID mice. We found that LeIF stimulated fresh spleen cells from naive SCID mice to secrete IFN-ã by IL-12/IL-18-dependent mechanisms. The N-terminal half of the molecule (amino acid residues 1-226) maintained the ability to stimulate IFN-ã from splenocytes of SCID mice. Finally, we also demonstrated that LeIF was able to provide partial protection of BALB/c mice against L. major. Thus, our results suggest the potential of LeIF as a Th1-type adjuvant and as a therapeutic and prophylactic vaccine Ag for leishmaniasis when used with other leishmanial Ags


Subject(s)
Animals , Leishmania , Cytokines
SELECTION OF CITATIONS
SEARCH DETAIL
...