Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1331480, 2024.
Article in English | MEDLINE | ID: mdl-38545103

ABSTRACT

Macrophages are critical regulators of the tumor microenvironment and often present an immuno-suppressive phenotype, supporting tumor growth and immune evasion. Promoting a robust pro-inflammatory macrophage phenotype has emerged as a therapeutic modality that supports tumor clearance, including through synergy with immune checkpoint therapies. Polyglucose nanoparticles (macrins), which possess high macrophage affinity, are useful vehicles for delivering drugs to macrophages, potentially altering their phenotype. Here, we examine the potential of functionalized macrins, synthesized by crosslinking carboxymethyl dextran with L-lysine, as effective carriers of immuno-stimulatory drugs to tumor-associated macrophages (TAMs). Azide groups incorporated during particle synthesis provided a handle for click-coupling of propargyl-modified ß-cyclodextrin to macrins under mild conditions. Fluorescence-based competitive binding assays revealed the ability of ß-cyclodextrin to non-covalently bind to hydrophobic immuno-stimulatory drug candidates (Keq ~ 103 M-1), enabling drug loading within nanoparticles. Furthermore, transcriptional profiles of macrophages indicated robust pro-inflammatory reprogramming (elevated Nos2 and Il12; suppressed Arg1 and Mrc1 expression levels) for a subset of these immuno-stimulatory agents (UNC2025 and R848). Loading of R848 into the modified macrins improved the drug's effect on primary murine macrophages by three-fold in vitro. Intravital microscopy in IL-12-eYFP reporter mice (24 h post-injection) revealed a two-fold enhancement in mean YFP fluorescence intensity in macrophages targeted with R848-loaded macrins, relative to vehicle controls, validating the desired pro-inflammatory reprogramming of TAMs in vivo by cell-targeted drug delivery. Finally, in an intradermal MC38 tumor model, cyclodextrin-modified macrin NPs loaded with immunostimulatory drugs significantly reduced tumor growth. Therefore, efficient and effective repolarization of tumor-associated macrophages to an M1-like phenotype-via drug-loaded macrins-inhibits tumor growth and may be useful as an adjuvant to existing immune checkpoint therapies.


Subject(s)
Nanoparticles , Neoplasms , beta-Cyclodextrins , Animals , Mice , Pharmaceutical Preparations , Tumor-Associated Macrophages , Nanoparticles/chemistry , Phenotype , Tumor Microenvironment
2.
Cancer Immunol Res ; 10(1): 40-55, 2022 01.
Article in English | MEDLINE | ID: mdl-34795032

ABSTRACT

Macrophages often abound within tumors, express colony-stimulating factor 1 receptor (CSF1R), and are linked to adverse patient survival. Drugs blocking CSF1R signaling have been used to suppress tumor-promoting macrophage responses; however, their mechanisms of action remain incompletely understood. Here, we assessed the lung tumor immune microenvironment in mice treated with BLZ945, a prototypical small-molecule CSF1R inhibitor, using single-cell RNA sequencing and mechanistic validation approaches. We showed that tumor control was not caused by CSF1R+ cell depletion; instead, CSF1R targeting reshaped the CSF1R+ cell landscape, which unlocked cross-talk between antitumoral CSF1R- cells. These cells included IFNγ-producing natural killer and T cells, and an IL12-producing dendritic cell subset, denoted as DC3, which were all necessary for CSF1R inhibitor-mediated lung tumor control. These data indicate that CSF1R targeting can activate a cardinal cross-talk between cells that are not macrophages and that are essential to mediate the effects of T cell-targeted immunotherapies and promote antitumor immunity.See related Spotlight by Burrello and de Visser, p. 4.


Subject(s)
Dendritic Cells/immunology , Immunotherapy/methods , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lung Neoplasms/therapy , Animals , Benzothiazoles/pharmacology , Cell Line, Tumor , Female , Lung Neoplasms/immunology , Mice , Mice, Inbred C57BL , Picolinic Acids/pharmacology , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism , Xenograft Model Antitumor Assays
3.
Nat Biomed Eng ; 2(8): 578-588, 2018 08.
Article in English | MEDLINE | ID: mdl-31015631

ABSTRACT

Tumour-associated macrophages are abundant in many cancers, and often display an immune-suppressive M2-like phenotype that fosters tumour growth and promotes resistance to therapy. Yet, macrophages are highly plastic and can also acquire an anti-tumorigenic M1-like phenotype. Here, we show that R848, an agonist of the toll-like receptors TLR7 and TLR8 identified in a morphometric-based screen, is a potent driver of the M1 phenotype in vitro and that R848-loaded ß-cyclodextrin nanoparticles (CDNP-R848) lead to efficient drug delivery to tumour-associated macrophages in vivo. As a monotherapy, the administration of CDNP-R848 in multiple tumour models in mice altered the functional orientation of the tumour immune microenvironment towards an M1 phenotype, leading to controlled tumour growth and protecting the animals against tumour rechallenge. When used in combination with the immune checkpoint inhibitor anti-PD-1, we observed improved immunotherapy response rates, including in a tumour model resistant to anti-PD-1 therapy alone. Our findings demonstrate the ability of rationally engineered drug-nanoparticle combinations to efficiently modulate tumour-associated macrophages for cancer immunotherapy.


Subject(s)
Immunotherapy/methods , Macrophages/drug effects , Nanoparticles/chemistry , Neoplasms/therapy , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Macrophages/immunology , Mice , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
4.
Chem Commun (Camb) ; 54(1): 42-45, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29185561

ABSTRACT

The receptor tyrosine kinase Mer (MERTK) is a promising drug target in cancer, where it can influence the metastasis-promoting signaling of both tumor cells and immune cells alike; however, no small molecule probes currently exist to selectively image Mer. In this work, we design and synthesize a selective near-infrared fluorescent molecular probe of Mer (MERi-SiR). Confocal microscopy of metastases in mice reveals predominant probe accumulation in Mer-expressing tumor-associated macrophages.


Subject(s)
Macrophages/pathology , Spectroscopy, Near-Infrared , c-Mer Tyrosine Kinase/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Macrophages/cytology , Macrophages/metabolism , Mice , Microscopy, Confocal , Molecular Docking Simulation , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Protein Structure, Tertiary , Pyrimidines/chemistry , Pyrroles/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Structure-Activity Relationship , c-Mer Tyrosine Kinase/antagonists & inhibitors , c-Mer Tyrosine Kinase/genetics
5.
Sci Transl Med ; 9(392)2017 05 31.
Article in English | MEDLINE | ID: mdl-28566423

ABSTRACT

Efficient delivery of therapeutic nanoparticles (TNPs) to tumors is critical in improving efficacy, yet strategies that universally maximize tumoral targeting by TNP modification have been difficult to achieve in the clinic. Instead of focusing on TNP optimization, we show that the tumor microenvironment itself can be therapeutically primed to facilitate accumulation of multiple clinically relevant TNPs. Building on the recent finding that tumor-associated macrophages (TAM) can serve as nanoparticle drug depots, we demonstrate that local tumor irradiation substantially increases TAM relative to tumor cells and, thus, TNP delivery. High-resolution intravital imaging reveals that after radiation, TAM primarily accumulate adjacent to microvasculature, elicit dynamic bursts of extravasation, and subsequently enhance drug uptake in neighboring tumor cells. TAM depletion eliminates otherwise beneficial radiation effects on TNP accumulation and efficacy, and controls with unencapsulated drug show that radiation effects are more pronounced with TNPs. Priming with combined radiation and cyclophosphamide enhances vascular bursting and tumoral TNP concentration, in some cases leading to a sixfold increase of TNP accumulation in the tumor, reaching 6% of the injected dose per gram of tissue. Radiation therapy alters tumors for enhanced TNP delivery in a TAM-dependent fashion, and these observations have implications for the design of next-generation tumor-targeted nanomaterials and clinical trials for adjuvant strategies.


Subject(s)
Drug Delivery Systems , Macrophages/pathology , Nanoparticles/chemistry , Neoplasms/blood supply , Neoplasms/radiotherapy , Animals , Cell Count , Cell Line, Tumor , Chemistry, Pharmaceutical , Combined Modality Therapy , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Extravasation of Diagnostic and Therapeutic Materials/pathology , Humans , Intravital Microscopy , Macrophages/drug effects , Macrophages/radiation effects , Mice, Nude , Neoplasms/drug therapy , Permeability , Phagocytes/drug effects , Phagocytes/pathology , Phagocytes/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Xenograft Model Antitumor Assays
6.
Sci Transl Med ; 9(389)2017 05 10.
Article in English | MEDLINE | ID: mdl-28490665

ABSTRACT

Monoclonal antibodies (mAbs) targeting the immune checkpoint anti-programmed cell death protein 1 (aPD-1) have demonstrated impressive benefits for the treatment of some cancers; however, these drugs are not always effective, and we still have a limited understanding of the mechanisms that contribute to their efficacy or lack thereof. We used in vivo imaging to uncover the fate and activity of aPD-1 mAbs in real time and at subcellular resolution in mice. We show that aPD-1 mAbs effectively bind PD-1+ tumor-infiltrating CD8+ T cells at early time points after administration. However, this engagement is transient, and aPD-1 mAbs are captured within minutes from the T cell surface by PD-1- tumor-associated macrophages. We further show that macrophage accrual of aPD-1 mAbs depends both on the drug's Fc domain glycan and on Fcγ receptors (FcγRs) expressed by host myeloid cells and extend these findings to the human setting. Finally, we demonstrate that in vivo blockade of FcγRs before aPD-1 mAb administration substantially prolongs aPD-1 mAb binding to tumor-infiltrating CD8+ T cells and enhances immunotherapy-induced tumor regression in mice. These investigations yield insight into aPD-1 target engagement in vivo and identify specific Fc/FcγR interactions that can be modulated to improve checkpoint blockade therapy.


Subject(s)
Antibodies, Monoclonal/immunology , Programmed Cell Death 1 Receptor/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Receptors, IgG/metabolism
7.
Nat Commun ; 8: 14293, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176769

ABSTRACT

Involvement of the immune system in tumour progression is at the forefront of cancer research. Analysis of the tumour immune microenvironment has yielded a wealth of information on tumour biology, and alterations in some immune subtypes, such as tumour-associated macrophages (TAM), can be strong prognostic indicators. Here, we use optical tissue clearing and a TAM-targeting injectable fluorescent nanoparticle (NP) to examine three-dimensional TAM composition, tumour-to-tumour heterogeneity, response to colony-stimulating factor 1 receptor (CSF-1R) blockade and nanoparticle-based drug delivery in murine pulmonary carcinoma. The method allows for rapid tumour volume assessment and spatial information on TAM infiltration at the cellular level in entire lungs. This method reveals that TAM density was heterogeneous across tumours in the same animal, overall TAM density is different among separate pulmonary tumour models, nanotherapeutic drug delivery correlated with TAM heterogeneity, and successful response to CSF-1R blockade is characterized by enhanced TAM penetration throughout and within tumours.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Lewis Lung/diagnostic imaging , Imaging, Three-Dimensional/methods , Lung Neoplasms/diagnostic imaging , Macrophages/immunology , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Female , Humans , Lung/diagnostic imaging , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophage Colony-Stimulating Factor/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Perfusion/methods , Pyrroles/pharmacology , Pyrroles/therapeutic use , RAW 264.7 Cells , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Tomography, X-Ray Computed , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
8.
Nat Chem Biol ; 13(2): 168-173, 2017 02.
Article in English | MEDLINE | ID: mdl-27918558

ABSTRACT

Quantitation of drug target engagement in single cells has proven to be difficult, often leaving unanswered questions in the drug development process. We found that intracellular target engagement of unlabeled new therapeutics can be quantitated using polarized microscopy combined with competitive binding of matched fluorescent companion imaging probes. We quantitated the dynamics of target engagement of covalent BTK inhibitors, as well as reversible PARP inhibitors, in populations of single cells using a single companion imaging probe for each target. We then determined average in vivo tumor concentrations and found marked population heterogeneity following systemic delivery, revealing single cells with low target occupancy at high average target engagement in vivo.


Subject(s)
Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Single-Cell Analysis , Dose-Response Relationship, Drug , Humans , Molecular Structure , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Substrate Specificity , Tumor Cells, Cultured
9.
ACS Med Chem Lett ; 5(5): 522-6, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24900873

ABSTRACT

The synthetic cryptocaryols A and B and a series of their analogues have been evaluated for their cytotoxicity and their ability to stabilize the tumor suppressor PDCD4. Cytotoxicities in the 3 to 30 µM range were found. Both the cytotoxicity and PDCD4 stabilizing ability were tolerant of large stereochemical changes to the molecule. Co-dosing studies with cryptocaryols A and B and several known cancer drugs showed no measuable enhancement in cancer drug cytotoxicity.

10.
ACS Chem Biol ; 8(12): 2771-7, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24093441

ABSTRACT

Aminoglycosides are broad-spectrum antibiotics that are used for the treatment of severe Gram-negative and Gram-positive bacterial infections. While bactericidal effects of aminoglycosides are due to binding to the 30S subunit of the bacterial ribosome, aminoglycosides can affect protein synthesis, intracellular calcium levels, and levels of reactive oxygen species (ROS) in eukaryotic cells. While aminoglycosides can be cytotoxic at high concentrations, our results show that at much lower doses, gentamicin can be implemented as a sensitizing agent for the NSCLC cell line NCI-H460, increasing the efficacy of camptothecin, digitoxin, and vinblastine in vitro. We have also established that this sensitization is reliant on the ROS response generated by gentamicin.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Digitoxin/pharmacology , Gentamicins/pharmacology , Vinblastine/pharmacology , Antibiotics, Antineoplastic/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Camptothecin/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Digitoxin/chemistry , Drug Synergism , Gentamicins/chemistry , High-Throughput Screening Assays , Humans , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism , Vinblastine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...