Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 11(3)2023 03.
Article in English | MEDLINE | ID: mdl-36948505

ABSTRACT

BACKGROUND: This phase 1 study evaluated PF-06753512, a vaccine-based immunotherapy regimen (PrCa VBIR), in two clinical states of prostate cancer (PC), metastatic castration-resistant PC (mCRPC) and biochemical recurrence (BCR). METHODS: For dose escalation, patients with mCRPC received intramuscular PrCa VBIR (adenovirus vector and plasmid DNA expressing prostate-specific membrane antigen (PSMA), prostate-specific antigen (PSA), and prostate stem cell antigen (PSCA)) with or without immune checkpoint inhibitors (ICIs, tremelimumab 40 or 80 mg with or without sasanlimab 130 or 300 mg, both subcutaneous). For dose expansion, patients with mCRPC received recommended phase 2 dose (RP2D) of PrCa VBIR plus tremelimumab 80 mg and sasanlimab 300 mg; patients with BCR received PrCa VBIR plus tremelimumab 80 mg (Cohort 1B-BCR) or tremelimumab 80 mg plus sasanlimab 130 mg (Cohort 5B-BCR) without androgen deprivation therapy (ADT). The primary endpoint was safety. RESULTS: Ninety-one patients were treated in dose escalation (mCRPC=38) and expansion (BCR=35, mCRPC=18). Overall, treatment-related and immune-related adverse events occurred in 64 (70.3%) and 39 (42.9%) patients, with fatigue (40.7%), influenza-like illness (30.8%), diarrhea (23.1%), and immune-related thyroid dysfunction (19.8%) and rash (15.4%), as the most common. In patients with mCRPC, the objective response rate (ORR, 95% CI) was 5.6% (1.2% to 15.4%) and the median radiographic progression-free survival (rPFS) was 5.6 (3.5 to not estimable) months for all; the ORR was 16.7% (3.6% to 41.4%) and 6-month rPFS rate was 45.5% (24.9% to 64.1%) for those who received RP2D with measurable disease (n=18). 7.4% of patients with mCRPC achieved a ≥50% decline in baseline PSA (PSA-50), with a median duration of 4.6 (1.2-45.2) months. In patients with BCR, 9 (25.7%) achieved PSA-50; the median duration of PSA response was 3.9 (1.9-4.2) and 10.1 (6.9-28.8) months for Cohorts 5B-BCR and 1B-BCR. Overall, antigen specific T-cell response was 88.0% to PSMA, 84.0% to PSA, and 80.0% to PSCA. CONCLUSIONS: PrCa VBIR overall demonstrated safety signals similar to other ICI combination trials; significant side effects were seen in some patients with BCR. It stimulated antigen-specific immunity across all cohorts and resulted in modest antitumor activity in patients with BCR without using ADT. TRIAL REGISTRATION NUMBER: NCT02616185.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Vaccines , Male , Humans , Prostatic Neoplasms, Castration-Resistant/drug therapy , Docetaxel/therapeutic use , Prostate-Specific Antigen , Androgen Antagonists/therapeutic use , Immunotherapy , Hormones/therapeutic use
2.
Nat Commun ; 13(1): 1768, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365657

ABSTRACT

Myeloid cells play key roles in cancer immune suppression and tumor progression. In response to tumor derived factors, circulating monocytes and granulocytes extravasate into the tumor parenchyma where they stimulate angiogenesis, immune suppression and tumor progression. Chemokines, cytokines and interleukins stimulate PI3Kγ-mediated Rap1 activation, leading to conformational changes in integrin α4ß1 that promote myeloid cell extravasation and tumor inflammation Here we show that PI3Kγ activates a high molecular weight form of myosin light chain kinase, MLCK210, that promotes myosin-dependent Rap1 GTP loading, leading to integrin α4ß1 activation. Genetic or pharmacological inhibition of MLCK210 suppresses integrin α4ß1 activation, as well as tumor inflammation and progression. These results demonstrate a critical role for myeloid cell MLCK210 in tumor inflammation and serve as basis for the development of alternative approaches to develop immune oncology therapeutics.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Myosin-Light-Chain Kinase , Neoplasms , Cell Adhesion/physiology , Humans , Inflammation , Molecular Weight , Myeloid Cells/metabolism , Myosin-Light-Chain Kinase/metabolism , Neoplasms/genetics
3.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article in English | MEDLINE | ID: mdl-33846242

ABSTRACT

Precision medicine in oncology leverages clinical observations of exceptional response. Toward an understanding of the molecular features that define this response, we applied an integrated, multiplatform analysis of RNA profiles derived from clinically annotated glioblastoma samples. This analysis suggested that specimens from exceptional responders are characterized by decreased accumulation of microglia/macrophages in the glioblastoma microenvironment. Glioblastoma-associated microglia/macrophages secreted interleukin 11 (IL11) to activate STAT3-MYC signaling in glioblastoma cells. This signaling induced stem cell states that confer enhanced tumorigenicity and resistance to the standard-of-care chemotherapy, temozolomide (TMZ). Targeting a myeloid cell restricted an isoform of phosphoinositide-3-kinase, phosphoinositide-3-kinase gamma isoform (PI3Kγ), by pharmacologic inhibition or genetic inactivation disrupted this signaling axis by reducing microglia/macrophage-associated IL11 secretion in the tumor microenvironment. Mirroring the clinical outcomes of exceptional responders, PI3Kγ inhibition synergistically enhanced the anti-neoplastic effects of TMZ in orthotopic murine glioblastoma models. Moreover, inhibition or genetic inactivation of PI3Kγ in murine glioblastoma models recapitulated expression profiles observed in clinical specimens isolated from exceptional responders. Our results suggest key contributions from tumor-associated microglia/macrophages in exceptional responses and highlight the translational potential for PI3Kγ inhibition as a glioblastoma therapy.


Subject(s)
Glioblastoma/metabolism , Microglia/metabolism , Temozolomide/pharmacology , Adult , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Drug Resistance, Neoplasm/physiology , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Interleukin-11/immunology , Interleukin-11/metabolism , Male , Mice, Inbred C57BL , Mice, Nude , Microglia/physiology , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Temozolomide/metabolism , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/physiology
4.
Nat Commun ; 9(1): 5379, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30568188

ABSTRACT

Myeloid cells are recruited to damaged tissues where they can resolve infections and tumor growth or stimulate wound healing and tumor progression. Recruitment of these cells is regulated by integrins, a family of adhesion receptors that includes integrin CD11b. Here we report that, unexpectedly, integrin CD11b does not regulate myeloid cell recruitment to tumors but instead controls myeloid cell polarization and tumor growth. CD11b activation promotes pro-inflammatory macrophage polarization by stimulating expression of microRNA Let7a. In contrast, inhibition of CD11b prevents Let7a expression and induces cMyc expression, leading to immune suppressive macrophage polarization, vascular maturation, and accelerated tumor growth. Pharmacological activation of CD11b with a small molecule agonist, Leukadherin 1 (LA1), promotes pro-inflammatory macrophage polarization and suppresses tumor growth in animal models of murine and human cancer. These studies identify CD11b as negative regulator of immune suppression and a target for cancer immune therapy.


Subject(s)
Benzoates/therapeutic use , CD11b Antigen/metabolism , Macrophages/metabolism , Melanoma, Experimental/immunology , MicroRNAs/metabolism , Thiohydantoins/therapeutic use , Animals , Benzoates/pharmacology , CD11b Antigen/agonists , Macrophages/drug effects , Melanoma, Experimental/drug therapy , Mice, Transgenic , Neovascularization, Pathologic , Proto-Oncogene Proteins c-myc/metabolism , Thiohydantoins/pharmacology
5.
Cancer Immunol Res ; 5(11): 957-968, 2017 11.
Article in English | MEDLINE | ID: mdl-28963139

ABSTRACT

Immunosuppressive myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) accumulate in tumors where they inhibit T cell-mediated antitumor immune responses and promote tumor progression. Myeloid cell PI3Kγ plays a role in regulating tumor immune suppression by promoting integrin α4-dependent MDSC recruitment to tumors and by stimulating the immunosuppressive polarization of MDSCs and TAMs. Here, we show that integrin α4 promotes immunosuppressive polarization of MDSCs and TAMs downstream of PI3Kγ, thereby inhibiting antitumor immunity. Genetic or pharmacological suppression of either PI3Kγ or integrin α4 blocked MDSC recruitment to tumors and also inhibited immune suppressive myeloid cell polarization, thereby reducing expression of IL10 and increasing expression of IL12 and IFNγ within tumors. Inhibition of PI3Kγ or integrin α4 within tumors stimulated dendritic cell and CD8+ T-cell recruitment and maturation, as well as tumor cell cytotoxicity in vivo, thereby inhibiting tumor growth. As blockade of PI3Kγ or integrin α4 prevents accumulation of MDSC and reduces myeloid cell expression of immunosuppressive factors that stimulate tumor immune escape, these results highlight PI3Kγ and integrin α4 as targets for the design of cancer therapeutics. Cancer Immunol Res; 5(11); 957-68. ©2017 AACR.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/immunology , Integrin alpha4beta1/immunology , Neoplasms/immunology , Animals , Antibodies/pharmacology , Cell Line, Tumor , Cell Polarity/drug effects , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/genetics , Cytokines/immunology , Female , Immune Tolerance , Integrin alpha4beta1/antagonists & inhibitors , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/immunology , Neoplasms/drug therapy , Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors
6.
JCI Insight ; 2(18)2017 09 21.
Article in English | MEDLINE | ID: mdl-28931759

ABSTRACT

Checkpoint inhibitors have demonstrated efficacy in patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). However, the majority of patients do not benefit from these agents. To improve the efficacy of checkpoint inhibitors, intratumoral (i.t.) injection with innate immune activators, TLR7 and TLR9 agonists, were tested along with programmed death-1 receptor (PD-1) blockade. The combination therapy suppressed tumor growth at the primary injected and distant sites in human papillomavirus-negative (HPV-negative) SCC7 and MOC1, and HPV-positive MEER syngeneic mouse models. Abscopal effects and suppression of secondary challenged tumor suggest that local treatment with TLR agonists in combination with anti-PD-1 provided systemic adaptive immunity. I.t. treatment with a TLR7 agonist increased the ratio of M1 to M2 tumor-associated macrophages (TAMs) and promoted the infiltration of tumor-specific IFNγ-producing CD8+ T cells. Anti-PD-1 treatment increased T cell receptor (TCR) clonality of CD8+ T cells in tumors and spleens of treated mice. Collectively, these experiments demonstrate that combination therapy with i.t. delivery of TLR agonists and PD-1 blockade activates TAMs and induces tumor-specific adaptive immune responses, leading to suppression of primary tumor growth and prevention of metastasis in HNSCC models.


Subject(s)
Head and Neck Neoplasms/therapy , Immunotherapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Heterografts , Humans , Killer Cells, Natural/immunology , Macrophages/immunology , Mice , Mice, Inbred C3H , Toll-Like Receptor 7/agonists , Toll-Like Receptor 9/agonists , Tumor Microenvironment
8.
Nature ; 539(7629): 437-442, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27642729

ABSTRACT

Macrophages play critical, but opposite, roles in acute and chronic inflammation and cancer. In response to pathogens or injury, inflammatory macrophages express cytokines that stimulate cytotoxic T cells, whereas macrophages in neoplastic and parasitic diseases express anti-inflammatory cytokines that induce immune suppression and may promote resistance to T cell checkpoint inhibitors. Here we show that macrophage PI 3-kinase γ controls a critical switch between immune stimulation and suppression during inflammation and cancer. PI3Kγ signalling through Akt and mTor inhibits NFκB activation while stimulating C/EBPß activation, thereby inducing a transcriptional program that promotes immune suppression during inflammation and tumour growth. By contrast, selective inactivation of macrophage PI3Kγ stimulates and prolongs NFκB activation and inhibits C/EBPß activation, thus promoting an immunostimulatory transcriptional program that restores CD8+ T cell activation and cytotoxicity. PI3Kγ synergizes with checkpoint inhibitor therapy to promote tumour regression and increased survival in mouse models of cancer. In addition, PI3Kγ-directed, anti-inflammatory gene expression can predict survival probability in cancer patients. Our work thus demonstrates that therapeutic targeting of intracellular signalling pathways that regulate the switch between macrophage polarization states can control immune suppression in cancer and other disorders.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Immune Tolerance/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Class Ib Phosphatidylinositol 3-Kinase/genetics , Female , Humans , Inflammation/immunology , Lymphocyte Activation , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neoplasms/immunology , Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/metabolism , Tumor Escape/immunology
9.
Cancer Discov ; 6(8): 870-85, 2016 08.
Article in English | MEDLINE | ID: mdl-27179037

ABSTRACT

UNLABELLED: Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a low 5-year survival rate, yet new immunotherapeutic modalities may offer hope for this and other intractable cancers. Here, we report that inhibitory targeting of PI3Kγ, a key macrophage lipid kinase, stimulates antitumor immune responses, leading to improved survival and responsiveness to standard-of-care chemotherapy in animal models of PDAC. PI3Kγ selectively drives immunosuppressive transcriptional programming in macrophages that inhibits adaptive immune responses and promotes tumor cell invasion and desmoplasia in PDAC. Blockade of PI3Kγ in PDAC-bearing mice reprograms tumor-associated macrophages to stimulate CD8(+) T-cell-mediated tumor suppression and to inhibit tumor cell invasion, metastasis, and desmoplasia. These data indicate the central role that macrophage PI3Kγ plays in PDAC progression and demonstrate that pharmacologic inhibition of PI3Kγ represents a new therapeutic modality for this devastating tumor type. SIGNIFICANCE: We report here that PI3Kγ regulates macrophage transcriptional programming, leading to T-cell suppression, desmoplasia, and metastasis in pancreas adenocarcinoma. Genetic or pharmacologic inhibition of PI3Kγ restores antitumor immune responses and improves responsiveness to standard-of-care chemotherapy. PI3Kγ represents a new therapeutic immune target for pancreas cancer. Cancer Discov; 6(8); 870-85. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 803.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Macrophages/metabolism , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Biomarkers , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/genetics , Class Ib Phosphatidylinositol 3-Kinase/genetics , Disease Models, Animal , Disease Progression , Gene Expression , Gene Knockout Techniques , Heterografts , Humans , Immunomodulation , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Male , Mice , Mice, Knockout , Mice, Transgenic , Mortality , Neoplasm Metastasis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenols/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Pteridines/pharmacology , Xenograft Model Antitumor Assays
10.
Cancer Discov ; 6(3): 270-85, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26715645

ABSTRACT

UNLABELLED: Pancreas ductal adenocarcinoma (PDAC) has one of the worst 5-year survival rates of all solid tumors, and thus new treatment strategies are urgently needed. Here, we report that targeting Bruton tyrosine kinase (BTK), a key B-cell and macrophage kinase, restores T cell-dependent antitumor immune responses, thereby inhibiting PDAC growth and improving responsiveness to standard-of-care chemotherapy. We report that PDAC tumor growth depends on cross-talk between B cells and FcRγ(+) tumor-associated macrophages, resulting in T(H)2-type macrophage programming via BTK activation in a PI3Kγ-dependent manner. Treatment of PDAC-bearing mice with the BTK inhibitor PCI32765 (ibrutinib) or by PI3Kγ inhibition reprogrammed macrophages toward a T(H)1 phenotype that fostered CD8(+) T-cell cytotoxicity, and suppressed PDAC growth, indicating that BTK signaling mediates PDAC immunosuppression. These data indicate that pharmacologic inhibition of BTK in PDAC can reactivate adaptive immune responses, presenting a new therapeutic modality for this devastating tumor type. SIGNIFICANCE: We report that BTK regulates B-cell and macrophage-mediated T-cell suppression in pancreas adenocarcinomas. Inhibition of BTK with the FDA-approved inhibitor ibrutinib restores T cell-dependent antitumor immune responses to inhibit PDAC growth and improves responsiveness to chemotherapy, presenting a new therapeutic modality for pancreas cancer.


Subject(s)
Cell Communication/immunology , Immune System/cytology , Immune System/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Disease Models, Animal , Disease Progression , Humans , Leukocytes/immunology , Leukocytes/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pancreatic Neoplasms/genetics , Receptors, IgG/metabolism , Signal Transduction
11.
Proc Natl Acad Sci U S A ; 110(31): 12750-5, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23852726

ABSTRACT

Studies of DNA methylation from fungi, plants, and animals indicate that gene body methylation is ancient and highly conserved in eukaryotic genomes, but its role has not been clearly defined. It has been postulated that regulation of alternative splicing of transcripts was an original function of DNA methylation, but a direct experimental test of the effect of methylation on alternative slicing at the whole genome level has never been performed. To do this, we developed a unique method to administer RNA interference (RNAi) in a high-throughput and noninvasive manner and then used it to knock down the expression of DNA methyl-transferase 3 (dnmt3), which is required for de novo DNA methylation. We chose the honey bee (Apis mellifera) for this test because it has recently emerged as an important model organism for studying the effects of DNA methylation on development and social behavior, and DNA methylation in honey bees is predominantly on gene bodies. Here we show that dnmt3 RNAi decreased global genomic methylation level as expected and in addition caused widespread and diverse changes in alternative splicing in fat tissue. Four different types of splicing events were affected by dnmt3 gene knockdown, and change in two types, exon skipping and intron retention, was directly related to decreased methylation. These results demonstrate that one function of gene body DNA methylation is to regulate alternative splicing.


Subject(s)
Alternative Splicing/physiology , Bees/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Insect Proteins/metabolism , RNA Interference , Animals , Bees/genetics , Behavior, Animal , DNA (Cytosine-5-)-Methyltransferases/genetics , Gene Knockdown Techniques , Insect Proteins/genetics , Social Behavior
12.
Biomaterials ; 31(11): 3079-86, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20092889

ABSTRACT

RNA interference (RNAi) is a useful in vitro research tool, but its application as a safe and effective therapeutic agent may benefit from improved understanding of mechanisms of exogenous siRNA delivery, including cell trafficking and sorting patterns. We report the development of a transfection reagent for siRNA delivery which employs a distinctive non-digestive mode of particle-cell membrane interaction through the formation of a hemifusion complex resulting in lipid raft transport of cargo to the cytosol, bypassing the usual endosomal nanoparticle uptake pathway. We further demonstrate markedly enhanced efficacy over conventional transfection agents for suppressing endothelial cell expression of upregulated vascular adhesion molecules.


Subject(s)
Emulsions/chemistry , Endothelial Cells/metabolism , Fluorocarbons/chemistry , Nanoparticles/chemistry , Nucleotides/metabolism , RNA, Small Interfering , Animals , Cations/chemistry , Cell Line , Cell Membrane Structures/metabolism , Drug Carriers/chemistry , Drug Carriers/metabolism , Indicators and Reagents/metabolism , Lipids/chemistry , Mice , Nucleotides/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection/methods
13.
Nanomedicine ; 5(3): 359-67, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19523428

ABSTRACT

We have designed multifunctional nanoparticulate reporter bioprobes capable of targeting vascular cell adhesion molecule 1 (VCAM-1), which is up-regulated in numerous inflammatory processes. These perfluorocarbon-cored nanoparticles emit a unique (19)F magnetic resonance (MR) signature, providing the potential to localize and quantify VCAM-1 expression in early atherosclerosis. Nanoparticle-VCAM-1 targeting specificity was confirmed by in vitro binding and competition studies. ApoE-null and control C57-BL6 mice (n = 6/group), fed a Western diet for 35 weeks, were injected i.v. with targeted or non-targeted nanoparticles. After two hours, kidneys were excised and prepared for analysis. ApoE-null kidneys exhibited increased VCAM-1-targeted nanoparticle content over healthy controls by (19)F MR spectroscopy (36.5+8.8 vs. 9.3+2.2 x 10(8)/g, P < .05), which correlated with increased VCAM-1 staining (2.5 +/- 1.3% vs. 0.9 +/- 0.3%, P < .05); their relative biodistributions were confirmed by fluorescence microscopy and MR imaging. These molecular imaging agents offer new approaches for detection, quantification, and longitudinal evaluation of early inflammation utilising (19)F MR spectroscopy and imaging. FROM THE CLINICAL EDITOR: Multifunctional nanoparticulate reporter bioprobes capable of targeting vascular cell adhesion molecule 1 (VCAM-1) are reported in this paper. These perfluorocarbon-cored nanoparticles offer new approaches for detection, quantification, and longitudinal evaluation of early inflammation utilising 19F MR spectroscopy and imaging.


Subject(s)
Apolipoproteins E/deficiency , Diet , Inflammation/pathology , Kidney/blood supply , Molecular Probe Techniques , Nanoparticles/chemistry , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Immunohistochemistry , Kidney/pathology , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Mice , Microscopy, Fluorescence , Tissue Distribution
14.
Ann Biomed Eng ; 37(10): 1922-33, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19184435

ABSTRACT

A broad array of nanomaterials is available for use as contrast agents for molecular imaging and drug delivery. Due to the lack of endogenous background signal in vivo and the high NMR sensitivity of the (19)F atom, liquid perfluorocarbon nanoemulsions make ideal agents for cellular and magnetic resonance molecular imaging. The perfluorocarbon core material is surrounded by a lipid monolayer which can be functionalized with a variety of agents including targeting ligands, imaging agents and drugs either individually or in combination. Multiple copies of targeting ligands (approximately 20-40 monoclonal antibodies or 200-400 small molecule ligands) serve to enhance avidity through multivalent interactions while the composition of the particle's perfluorocarbon core results in high local concentrations of (19)F. Additionally, lipophilic drugs contained within molecularly targeted nanoemulsions can result in contact facilitated drug delivery to target cells. Ultimately, the dual use of perfluorocarbon nanoparticles for both site targeted drug delivery and molecular imaging may provide both imaging of disease states as well as conclusive evidence that drug delivery is localized to the area of interest. This review will focus on liquid perfluorocarbon nanoparticles as (19)F molecular imaging agents and for targeted drug delivery in cancer and cardiovascular disease.


Subject(s)
Diagnostic Imaging/trends , Drug Delivery Systems/trends , Fluorocarbons/therapeutic use , Molecular Probe Techniques/trends , Nanoparticles/therapeutic use , Contrast Media , Emulsions/chemistry , Fluorocarbons/chemistry , Image Enhancement/methods , Nanoparticles/chemistry
15.
J Biomed Mater Res A ; 88(1): 205-12, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18286622

ABSTRACT

Sphingosine 1-phosphate (S1P) promotes endothelial cell migration in vitro and may potentially impact the endothelialization of implanted biomaterials. However, the effects of S1P on endothelial cells (EC) in flowing blood could be negligible due to preactivation of signaling cascades. We previously developed biomaterials that release S1P and wished to determine through in vitro experiments the extent to which EC respond to S1P added to human platelet poor plasma. We found that addition of 200 nM S1P to platelet poor plasma significantly increased cell migration in two migration models. A lower concentration of S1P added to plasma (100 nM) did not increase endothelial cell migration rates, while the cell migration response was saturated above 200 nM S1P. Expression of the main S1P receptor in EC, S1P(1), was elevated in plasma compared to low serum medium, but addition of VEGF or fluid flow elicited a further increase in S1P(1) mRNA, consistent with the synergistic effects observed between S1P, VEGF, and fluid flow. Thus, sustained delivery of S1P from biomaterials might only enhance endothelial cell migration if the concentration of S1P at the surface of the material stimulated adjacent EC to the same extent as approximately 200 nM S1P added to plasma.


Subject(s)
Blood Circulation , Cell Movement/drug effects , Endothelial Cells/physiology , Lysophospholipids/pharmacology , Sphingosine/analogs & derivatives , Biocompatible Materials/therapeutic use , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Humans , Models, Biological , Receptors, Lysosphingolipid/biosynthesis , Sphingosine/pharmacology , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/pharmacology
16.
Biomacromolecules ; 7(4): 1335-43, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16602758

ABSTRACT

While protein growth factors promote therapeutic angiogenesis, delivery of lipid factors such as sphingosine 1-phosphate (S1P) may provide better stabilization of newly formed vessels. We developed a biomaterial for the controlled delivery of S1P, a bioactive lipid released from activated platelets. Multiarm poly(ethylene glycol)-vinyl sulfone was cross-linked with albumin, a lipid-transporting protein, to form hydrogels. The rate of S1P release from the materials followed Fickian kinetics and was dependent upon the presence of lipid carriers in the release solution. Delivery of S1P from RGD-modified hydrogels increased the cell migration speed of endothelial cells growing on the materials. The materials also induced angiogenesis in the chorioallantoic membrane assay. Our data demonstrate that the storage and release of lipid factors provides a new route for the induction of angiogenesis by artificial materials.


Subject(s)
Hydrogels/chemistry , Lysophospholipids/chemistry , Polyethylene Glycols/chemistry , Sphingosine/analogs & derivatives , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Drug Delivery Systems , Endothelial Cells/drug effects , Endothelial Cells/physiology , Gels/chemistry , Humans , Hydrogels/chemical synthesis , Hydrogels/pharmacology , Molecular Structure , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/pharmacology , Sphingosine/chemistry , Time Factors
17.
Ann Biomed Eng ; 33(8): 1003-14, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16133909

ABSTRACT

The rational design of drug delivery systems requires the ability to predict the environment-specific responses of target cells to the delivered drug. Here we describe the in vitro effects of fluid shear stress, vascular endothelial growth factor (VEGF), and sphingosine 1-phosphate (S1P) on the migration of human umbilical vein endothelial cells (HUVEC). Endothelial cell migration into a scrape wound was enhanced in S1P- or VEGF-stimulated HUVEC by the addition of fluid shear stress. In both cases, scrape wound closure rates were near a maximal value that was not exceeded when cells were exposed to all three factors. We also found that cell migration into a scrape wound due to S1P stimulation was correlated with the S1P1 mRNA concentration, in systems where cell migration was not already near maximal. The present work represents our initial steps toward predicting cell migration based upon the activation state of the receptors and enzymes involved in the chemokinetic response. These results also illustrate the importance of context-dependent analysis of cell signaling cascades.


Subject(s)
Cell Movement/drug effects , Endothelial Cells/physiology , Lysophospholipids/pharmacology , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Umbilical Veins/physiology , Vascular Endothelial Growth Factors/pharmacology , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/cytology , Humans , Lysophospholipids/metabolism , Receptors, Lysosphingolipid/biosynthesis , Signal Transduction/physiology , Sphingosine/metabolism , Sphingosine/pharmacology , Stress, Mechanical , Umbilical Veins/cytology , Vascular Endothelial Growth Factors/metabolism , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...