Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Control Release ; 343: 303-313, 2022 03.
Article in English | MEDLINE | ID: mdl-35104570

ABSTRACT

Interactions between different cell types in the tumor microenvironment (TME) affect tumor growth. Tumor-associated fibroblasts produce C-X-C motif chemokine ligand 13 (CXCL13) which recruits B cells to the TME. B-cells in the TME differentiate into regulatory B cells (Bregs) (IL-10+CD1d+CD5+CD138+CD19+). We highlight these Breg cells as a new important factor in the modulation of the immunosuppressive TME in different desmoplastic murine tumor models. In addition, CXCL13 also stimulates epithelial-mesenchymal transition (EMT) of the tumor cells. The tumorigenic roles of CXCL13 led us to explore an innovative anti-cancer strategy based on delivering plasmid DNA encoding a CXCL13 trap to reduce Bregs differentiation and normalize EMT, thereby suppressing tumor growth. CXCL13 trap suppressed tumor growth in pancreatic cancer, BRAF-mutant melanoma, and triple-negative breast cancer. In this study, following treatment, the affected tumor remained dormant resulting in prolonged progression-free survival of the host.


Subject(s)
B-Lymphocytes, Regulatory , Cancer-Associated Fibroblasts , Pancreatic Neoplasms , Triple Negative Breast Neoplasms , Animals , B-Lymphocytes, Regulatory/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Humans , Mice , Pancreatic Neoplasms/metabolism , Triple Negative Breast Neoplasms/therapy , Tumor Microenvironment
2.
Adv Mater ; 30(52): e1805007, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30387230

ABSTRACT

The development and progression of colorectal cancer (CRC) is closely related to gut microbiome. Here, the impact of lipopolysaccharide (LPS), one of the most prevalent products in the gut microbiome, on CRC immunotherapy is investigated. It is found that LPS is abundant in orthotopic CRC tissue and is associated with low responses to anti-PD-L1 mAb therapy, and clearance of Gram-negative bacteria from the gut using polymyxin B (PmB) or blockade of Toll-like receptor 4 using TAK-242 will both relieve the immunosuppressive microenvironment and boost T-cell infiltration into the CRC tumor. Further, an engineered LPS-targeting fusion protein is designed and its coding sequence is loaded into a lipid-protamine-DNA (LPD) nanoparticle system for selective expression of LPS trap protein and blocking LPS inside the tumor, and this nanotrapping system significantly relieves the immunosuppressive microenvironment and boosts anti-PD-L1 mAb therapy against CRC tumors. This LPS trap system even attenuates CRC liver metastasis when applied, suggesting the importance of blocking LPS in the gut-liver axis. The strategy applied here may provide a useful new way for treating CRC as well as other epithelial cancers that interact with mucosa microbiome.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Immunotherapy , Lipopolysaccharides/metabolism , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Animals , Cell Line, Tumor , Colorectal Neoplasms/immunology , Gastrointestinal Microbiome/immunology , Gram-Negative Bacteria/metabolism , Liver Neoplasms/immunology , Mice , Nanoparticles/administration & dosage , Neoplasm Transplantation
3.
ACS Nano ; 11(9): 8690-8706, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28809532

ABSTRACT

Pancreatic tumors are known to be resistant to immunotherapy due to the extensive immune suppressive tumor microenvironment (TME). We hypothesized that CXCL12 and PD-L1 are two key molecules controlling the immunosuppressive TME. Fusion proteins, called traps, designed to bind with these two molecules with high affinity (Kd = 4.1 and 0.22 nM, respectively) were manufactured and tested for specific binding with the targets. Plasmid DNA encoding for each trap was formulated in nanoparticles and intravenously injected to mice bearing orthotopic pancreatic cancer. Expression of traps was mainly seen in the tumor, and secondarily, accumulations were primarily in the liver. Combination trap therapy shrunk the tumor and significantly prolonged the host survival. Either trap alone only brought in a partial therapeutic effect. We also found that CXCL12 trap allowed T-cell penetration into the tumor, and PD-L1 trap allowed the infiltrated T-cells to kill the tumor cells. Combo trap therapy also significantly reduced metastasis of the tumor cells to other organs. We conclude that the trap therapy significantly modified the immunosuppressive TME to allow the host immune system to kill the tumor cells. This can be an effective therapy in clinical settings.


Subject(s)
B7-H1 Antigen/immunology , Carcinoma, Pancreatic Ductal/therapy , Chemokine CXCL12/immunology , DNA/therapeutic use , Immunotherapy/methods , Animals , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Chemokine CXCL12/antagonists & inhibitors , Genetic Therapy/methods , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Escape , Tumor Microenvironment
4.
Biomaterials ; 141: 260-271, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28700955

ABSTRACT

The ability to generate potent immunotherapies locally and transiently for the treatment of cancers is a promising strategy to improve efficacy and decrease off-target toxicities. Here, we explored an alternative approach for the delivery of immunotherapeutic agents, in which we deliver the pDNA of an engineered PD-L1 trap and/or CXCL12 trap to the nucleus of liver hepatocytes via a lipid calcium phosphate nanoparticle. This strategy greatly increased the concentrations of immunotherapeutic agents in the local tissue, allowing the therapy to inhibit the accumulation of immune suppressive cells and liver metastasis. Furthermore, we find that the lipid calcium phosphate nanoparticles containing the pCXCL12 trap resolved the formation of immune suppressive ectopic lymphoid structures, while the pPD-L1 trap promoted T-cell survival and migration into the liver following vaccination against tumor antigens (>180% increase in survival). This approach showed superior efficacy in the treatment of the liver metastasis compared to free protein immunotherapies. This strategy should be considered as an approach to support liver metastasis therapies as well as for future research interested in manipulating the chemokine/cytokine immune factors within the liver. SIGNIFICANCE: Our approach results in transient liver specific expression of gene immunotherapies with improved efficacy and reduced off-target toxicities over traditional systemically administered immunotherapies. This approach would allow clinicians to manipulate the liver and immune microenvironment to resist cancer invasion, improve organ health, and prolong patient survival.


Subject(s)
B7-H1 Antigen/genetics , Cancer Vaccines/therapeutic use , Chemokine CXCL12/genetics , Colorectal Neoplasms/pathology , DNA/therapeutic use , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/pathology , Calcium Phosphates/chemistry , DNA/administration & dosage , DNA/genetics , Female , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Immunotherapy/methods , Lipids/chemistry , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/therapeutic use
5.
J Lipid Res ; 57(4): 650-62, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26921357

ABSTRACT

Sec14-like phosphatidylinositol transfer proteins (PITPs) play important biological functions in integrating multiple aspects of intracellular lipid metabolism with phosphatidylinositol-4-phosphate signaling. As such, these proteins offer new opportunities for highly selective chemical interference with specific phosphoinositide pathways in cells. The first and best characterized small molecule inhibitors of the yeast PITP, Sec14, are nitrophenyl(4-(2-methoxyphenyl)piperazin-1-yl)methanones (NPPMs), and a hallmark feature of NPPMs is their exquisite targeting specificities for Sec14 relative to other closely related Sec14-like PITPs. Our present understanding of Sec14::NPPM binding interactions is based on computational docking and rational loss-of-function approaches. While those approaches have been informative, we still lack an adequate understanding of the basis for the high selectivity of NPPMs among closely related Sec14-like PITPs. Herein, we describe a Sec14 motif, which we term the VV signature, that contributes significantly to the NPPM sensitivity/resistance of Sec14-like phosphatidylinositol (PtdIns)/phosphatidylcholine (PtdCho) transfer proteins. The data not only reveal previously unappreciated determinants that govern Sec14-like PITP sensitivities to NPPMs, but enable predictions of which Sec14-like PtdIns/PtdCho transfer proteins are likely to be NPPM resistant or sensitive based on primary sequence considerations. Finally, the data provide independent evidence in support of previous studies highlighting the importance of Sec14 residue Ser173 in the mechanism by which NPPMs engage and inhibit Sec14-like PITPs.


Subject(s)
Phospholipid Transfer Proteins/antagonists & inhibitors , Phospholipid Transfer Proteins/chemistry , Piperazines/pharmacology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/chemistry , Amino Acid Motifs , Drug Design , Drug Resistance/genetics , Lipid Metabolism/drug effects , Molecular Docking Simulation , Mutation, Missense , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Piperazines/chemistry , Piperazines/metabolism , Protein Conformation , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...