Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Cancer Immunol Immunother ; 69(4): 663-675, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31980914

ABSTRACT

Cholesteryl pullulan (CHP) is a novel antigen delivery system. CHP and New York esophageal squamous cell carcinoma 1 (NY-ESO-1) antigen complexes (CHP-NY-ESO-1) present multiple epitope peptides to the MHC class I and II pathways. Adjuvants are essential for cancer vaccines. MIS416 is a non-toxic microparticle that activates immunity via the nucleotide-binding oligomerization domain 2 (NOD2) and TLR9 pathways. However, no reports have explored MIS416 as a cancer vaccine adjuvant. We conducted a first-in-human clinical trial of CHP-NY-ESO-1 with MIS416 in patients with NY-ESO-1-expressing refractory solid tumors. CHP-NY-ESO-1/MIS416 (µg/µg) was administered at 100/200, 200/200, 200/400 or 200/600 (cohorts 1, 2, 3 and 4, respectively) every 2 weeks for a total of 6 doses (treatment phase) followed by one vaccination every 4 weeks until disease progression or unacceptable toxicity (maintenance phase). The primary endpoints were safety and tolerability, and the secondary endpoint was the immune response. In total, 26 patients were enrolled. Seven patients (38%) continued vaccination in the maintenance phase. Grade 3 drug-related adverse events (AEs) were observed in six patients (23%): anorexia and hypertension were observed in one and five patients, respectively. No grade 4-5 drug-related AEs were observed. Eight patients (31%) had stable disease (SD). Neither augmentation of the NY-ESO-1-specific IFN-γ-secreting CD8+ T cell response nor an increase in the level of anti-NY-ESO-1 IgG1 was observed as the dose of MIS416 was increased. In a preclinical study, adding anti-PD-1 monoclonal antibody to CHP-NY-ESO-1 and MIS416 induced significant tumor suppression. This combination therapy is a promising next step.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Membrane Proteins/immunology , Neoplasms/immunology , Nod2 Signaling Adaptor Protein/immunology , Toll-Like Receptor 9/immunology , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Neoplasm/blood , Antibodies, Neoplasm/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Female , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Male , Mice, Inbred BALB C , Middle Aged , Neoplasms/pathology , Neoplasms/therapy , Nod2 Signaling Adaptor Protein/metabolism , Toll-Like Receptor 9/metabolism , Vaccination/methods
2.
Acta Neuropathol Commun ; 8(1): 2, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31915070

ABSTRACT

There is great interest in understanding how the central nervous system (CNS) communicates with the immune system for recruitment of protective responses. Infiltrating phagocytic monocytes and granulocytes are implicated in neuroinflammation in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). To investigate how CNS endogenous signals can be harnessed to promote anti-inflammatory programs, we have used a particulate Toll-like receptor 9 and nucleotide-oligomerization domain 2 bispecific innate ligand (MIS416), to address whether its phagocytosis within the CNS recruits protective myeloid cells. We find that MIS416 injected intrathecally into the cerebrospinal fluid via the cisterna magna induced a local chemokine response that recruited blood-derived monocytes and neutrophils to the CNS. These cells phagocytosed MIS416. The increase in EAE severity normally seen from time of onset did not occur in mice receiving MIS416. This suppression of disease symptoms was dependent on expression of the type I interferon receptor (IFNAR). Transfer of intrathecal MIS416-induced neutrophils suppressed EAE in recipient mice, while monocytes did not transfer protection. MIS416-induced neutrophils showed increased IL-10 expression that was IFNAR1-driven. In contrast to intrathecal administration, intravenous administration of MIS416 led to monocyte but not neutrophil infiltration to the CNS. We thus identify a CNS-intrinsic and -specific phagocytosis-induced recruitment of anti-inflammatory neutrophils that contribute to CNS homeostasis and may have therapeutic potential.


Subject(s)
Brain/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Monocytes/metabolism , Neutrophils/metabolism , Signal Transduction , Spinal Cord/metabolism , Animals , Brain/immunology , Disease Models, Animal , Female , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Phagocytosis , Spinal Cord/immunology
3.
Nucleic Acid Ther ; 28(4): 225-232, 2018 08.
Article in English | MEDLINE | ID: mdl-29893623

ABSTRACT

MIS416 is a microparticulate formulation derived from propionibacterium acnes cell wall skeletons with intrinsic adjuvant activity. Conjugates of MIS416-SS-peptide containing a disulfide linkage facilitate the cytoplasmic delivery and release of peptides in antigen-presenting cells (APCs). We hypothesized that MIS416-siRNA (small interfering RNA) conjugates, containing a disulfide linkage between MIS416 and the siRNA, would allow cytoplasmic release of siRNA in APCs. MIS416-SS-siStat3 conjugates added to cell culture medium of monolayers of DCs in culture flasks successfully targeted Stat3 mRNA in DCs in vitro without transfection, downregulating Stat3 mRNA and protein levels. These results suggest that MIS416-SS-siRNA conjugates can be used as a novel siRNA delivery system for the knockdown of mRNA levels in APCs.


Subject(s)
Cancer Vaccines/genetics , Genetic Therapy , RNA, Small Interfering/genetics , STAT3 Transcription Factor/genetics , Antigen-Presenting Cells/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Gene Transfer Techniques , Humans , Immunoconjugates/therapeutic use , RNA, Small Interfering/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/immunology
4.
Front Immunol ; 9: 1078, 2018.
Article in English | MEDLINE | ID: mdl-29892282

ABSTRACT

Human adult stem cells, including umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs), have recently been considered a promising alternative treatment for inflammatory bowel disease (IBD) due to their unique immunomodulatory properties and ability to promote tissue regeneration. However, despite many years of research and pre-clinical studies, results from clinical trials using these cells have been diverse and conflicting. This discrepancy is caused by several factors, such as poor engraftment, low survival rate, and donor-dependent variation of the cells. Enhancement of consistency and efficacy of MSCs remains a challenge for the feasibility of cell-based therapy. In this study, we investigated whether administration of MIS416, a novel microparticle that activates NOD2 and TLR9 signaling, could enhance the therapeutic efficacy of hUCB-MSCs against Crohn's disease, using dextran sulfate sodium (DSS)-induced colitis model. Colitis was experimentally induced in mice by using 3% DSS, and mice were administered a retro-orbital injection of MIS416 and subsequent intraperitoneal injection of hUCB-MSCs. Mice were examined grossly, and blood, spleen, and colon tissues were subsequently collected for further ex vivo analyses. To explore the effects of MIS416 on the therapeutic process, hUCB-MSCs and primary isolated immune cells were cultured with MIS416, and in vitro assays were performed. Compared to the single administration of hUCB-MSCs, co-administration with MIS416 improved the therapeutic efficiency of the stem cells by significantly alleviating the symptoms of IBD. Interestingly, MIS416 did not exert any direct effect on the immunomodulatory capacity of hUCB-MSCs. Instead, systemically injected MIS416 altered the immune milieu in the colon which caused hUCB-MSCs to be more readily recruited toward the lesion site and to suppress inflammation more efficiently. In addition, considerable numbers of regulatory immune cells were stimulated as a result of the cooperation of MIS416 and hUCB-MSCs. These findings indicate that co-administration with MIS416 enhances the therapeutic potential of hUCB-MSCs by systemically regulating the immune response, which might be an effective strategy for overcoming the current obstacles to stem cell therapy in clinical practice.


Subject(s)
Cancer Vaccines/immunology , Colitis/etiology , Fetal Blood/cytology , Immunomodulation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Cell Cycle , Cell Movement/immunology , Cellular Microenvironment/immunology , Chemokine CCL2/biosynthesis , Colitis/pathology , Colitis/therapy , Cytokines/biosynthesis , Dextran Sulfate/adverse effects , Disease Models, Animal , Humans , Immunity, Innate , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Milieu Therapy , Regeneration , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
5.
Article in English | MEDLINE | ID: mdl-29177070

ABSTRACT

BACKGROUND: While disease progression can be readily monitored in early stage relapsing multiple sclerosis (MS), it is more challenging for secondary progressive multiple sclerosis (SPMS). This advanced stage of disease has distinct pathophysiology due to compartmentalization of neuroinflammatory activity within the central nervous system, resulting in increased incidence and severity of cognitive dysfunction. The shift in the dominant disease pathways is underscored by the failure of relapsing therapies to benefit SPMS patients, highlighting the need for novel treatment strategies and clinical trial endpoints that are well-aligned with potential benefits. The Expanded Disability Status Scale (EDSS) is widely used but is weighted towards ambulatory ability, lacking sensitivity to other aspects of neurological impairment experienced in more severely disabled SPMS patients, so may not effectively capture their clinical status.To investigate the feasibility of an alternative clinical trial endpoint model for a phase 2B trial of an immune modulator for SPMS, the potential for treatment efficacy-based patient-centered outcomes was assessed within the context of a before and after, 12-week clinical trial of safety and tolerability. METHODS: Patients treated with MIS416 for 12 weeks were evaluated for clinical status at baseline and end of dosing, using the established Multiple Sclerosis Functional Composite, Short Form Health Survey, and Expanded Disability Status Scale. Responder status was determined for eight outcome measures based on minimally important change, defined using published studies. To evaluate the patients' immune response to MIS416, blood plasma samples collected at baseline and pre- and 24-h post doses 1-4 were analyzed using multiplex cytokine quantification assays. RESULTS: Using a combination of patient-centered outcomes, MIS416 treatment was associated with improved clinical status for 10/11 patients: eight patients showed improvement on two to five outcome measures, five of which also showed improvement by EDSS. Multi-dimensional scaling analysis of MIS416-induced factors quantified in individual patients, revealed immune response patterns which had a strong concordance with the extent of the patients' clinical response. CONCLUSIONS: The data support the feasibility of using patient-centered outcomes as additional clinical trial endpoints, for determining the efficacy of disease-modifying therapies, in secondary progressive multiple sclerosis patients. TRIAL REGISTRATION: ClinicalTrial.gov, NCT01191996.

6.
PLoS One ; 10(12): e0145403, 2015.
Article in English | MEDLINE | ID: mdl-26695183

ABSTRACT

MIS416 is an intact minimal cell wall skeleton derived from Proprionibacterium acnes that is phagocytosed by antigen presenting cells, including dendritic cells (DCs). This property allows MIS416 to be exploited as a vehicle for the delivery of peptide antigens or other molecules (for example, nucleic acids) to DCs. We previously showed that covalent (non-cleavable) conjugation of OVA, a model antigen derived from ovalbumin, to MIS416 enhanced immune responses in DCs in vivo, compared to unconjugated MIS416 and OVA. Intracellular trafficking promotes the lysosomal degradation of MIS416, leading to the destruction of MIS416 plus the associated cargos conjugated to MIS416. However, lysosomal degradation of cargo may not be desired for some MIS416 conjugates. Here we have investigated whether a cleavable linkage could facilitate release of the cargo in the cytoplasm of DCs to avoid lysosomal degradation. DCs were treated in vitro with disulfide-containing conjugates, and as hypothesised faster release of SIINFEKL peptide in the cytoplasm of DCs was observed with the inclusion of a disulfide bond between MIS416 and cargo. The inclusion of a cleavable disulfide bond in the conjugates did not significantly alter the amount of SIINFEKL antigens presented on MHC I molecules on DCs as compared with conjugates without a disulfide bond. However, the conjugates containing disulfide-linkages performed either slightly better (p<0.05) than, or the same as conjugates without a disulfide bond with respect to in vitro OT-1 T-cell proliferation induced by the presentation of SIINFEKL antigens on DCs, or DC activation studies, respectively. However, disulfide-containing conjugates were less effective than conjugates without a disulfide bond in in vivo cytotoxicity assays. In conclusion, inclusion of a disulfide bond in MIS416-peptide conjugates was associated with efficient release of peptides in the cytoplasm of DCs, an important consideration for MIS416-mediated delivery of degradation-sensitive cargoes. However, treatment of DCs with disulfide-containing conjugates did not significantly alter the presentation of peptide antigens on MHC class I molecules to T-cells, or greatly enhance antigen-associated T-cell proliferation in vitro.


Subject(s)
Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Disulfides/chemistry , Vaccines, Conjugate/chemistry , Animals , Antigen Presentation/drug effects , Cancer Vaccines/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Drug Carriers/chemistry , Drug Carriers/pharmacology , Mice , Ovalbumin/chemistry , Ovalbumin/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Spleen/cytology , Vaccines, Conjugate/pharmacology
7.
Oncotarget ; 6(13): 11310-26, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25888637

ABSTRACT

Epithelial ovarian cancer (EOC) is typically diagnosed at advanced stages, and is associated with a high relapse rate. Patients in remission are ideal candidates for immunotherapy aimed at cure or prolonging disease-free periods. However, immunosuppressive pathways in the tumor microenvironment are obstacles to durable anti-tumor immunity. In a metastatic syngeneic mouse model of EOC, immunosuppressive macrophages and myeloid-derived suppressor cells (MDSCs) accumulate in the local tumor environment. In addition, resident peritoneal macrophages from non-tumor-bearing mice were highly immunosuppressive, abrogating stimulated T cell proliferation in a cell contact-dependent manner. Immunization with microparticles containing TLR9 and NOD-2 ligands (MIS416) significantly prolonged survival in tumor-bearing mice. The strategy of MIS416 immunization followed by anti-CD11b administration further delayed tumor progression, thereby establishing the proof of principle that myeloid depletion can enhance vaccine efficacy. In patients with advanced EOC, ascites analysis showed substantial heterogeneity in the relative proportions of myeloid subsets and their immunosuppressive properties. Together, these findings point to immunosuppressive myeloid cells in the EOC microenvironment as targets to enhance vaccination. Further studies of myeloid cell accumulation and functional phenotypes in the EOC microenvironment may identify patients who are likely to benefit from vaccination combined with approaches that deplete tumor-associated myeloid cells.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Cancer Vaccines/administration & dosage , Myeloid Cells/immunology , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/therapy , Tumor Escape , Tumor Microenvironment , Vaccination , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Ascites/immunology , CD11b Antigen/immunology , Cancer Vaccines/immunology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Disease Progression , Female , Humans , Ligands , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Nod2 Signaling Adaptor Protein/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , T-Lymphocytes/immunology , Time Factors , Toll-Like Receptor 9/immunology
8.
Vaccine ; 29(3): 545-57, 2011 Jan 10.
Article in English | MEDLINE | ID: mdl-21034827

ABSTRACT

Propionibacterium acnes was modified using biochemical extraction methods generating a suspension of microparticles (MIS416) comprising a minimal cell wall skeleton rich in immunostimulatory crosslinked muramyl dipeptide repeats and native bacterial DNA fragments, each which have known adjuvant activity. In vitro studies demonstrated that MIS416 was readily internalized by human myeloid and plasmacytoid DC inducing cytokine secretion and cell activation/maturation. Vaccination studies in mice using OVA as a model antigen demonstrated that MIS416 acts as a Th1 adjuvant, promoting cross-priming of cytotoxic CD8(+) T cell responses and enhanced anti-tumour immunity. Covalent attachment of OVA to MIS416 enabling simultaneous delivery of antigen and adjuvant to the antigen presentation system resulted in a dose-sparing vaccine formulation. Preclinical GLP toxicology studies demonstrated that MIS416 has a favorable safety profile in mouse and rabbit supporting its use in human vaccine formulations.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/administration & dosage , Adjuvants, Immunologic/administration & dosage , Cross-Priming , DNA, Bacterial/administration & dosage , Ovalbumin/immunology , Propionibacterium acnes/chemistry , Th1 Cells/immunology , Acetylmuramyl-Alanyl-Isoglutamine/isolation & purification , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/isolation & purification , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Cytokines/metabolism , DNA, Bacterial/isolation & purification , Dendritic Cells/immunology , Endocytosis , Female , Humans , Male , Mice , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Vaccination/methods
9.
Biochem J ; 366(Pt 2): 459-69, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12027803

ABSTRACT

Members of both Myc and nuclear factor kappaB (NF-kappaB) families of transcription factors are found overexpressed or inappropriately activated in many forms of human cancer. Furthermore, NF-kappaB can induce c-Myc gene expression, suggesting that the activities of these factors are functionally linked. We have discovered that both c-Myc and v-Myc can induce a previously undescribed, truncated form of the RelA(p65) NF-kappaB subunit, RelA(p37). RelA(p37) encodes the N-terminal DNA binding and dimerization domain of RelA(p65) and would be expected to function as a trans-dominant negative inhibitor of NF-kappaB. Surprisingly, we found that RelA(p37) no longer binds to kappaB elements. This result is explained, however, by the observation that RelA(p37), but not RelA(p65), forms a high-molecular-mass complex with c-Myc. These results demonstrate a previously unknown functional and physical interaction between RelA and c-Myc with many significant implications for our understanding of the role that both proteins play in the molecular events underlying tumourigenesis.


Subject(s)
Genes, myc , NF-kappa B/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Amino Acid Sequence , Binding Sites , Cell Line , DNA-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Neoplasms/genetics , Proto-Oncogene Mas , Recombinant Proteins/metabolism , Sequence Deletion , Transcription Factor RelA , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL