Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Theranostics ; 10(17): 7510-7526, 2020.
Article in English | MEDLINE | ID: mdl-32685002

ABSTRACT

Tumor-associated macrophages (TAMs) enhance tumor growth in mice and are correlated with a worse prognosis for breast cancer patients. While early therapies sought to deplete all macrophages, current therapeutics aim to reprogram pro-tumor macrophages (M2) and preserve those necessary for anti-tumor immune responses (M1). Recent studies have shown that c-MYC (MYC) is induced in M2 macrophages in vitro and in vivo where it regulates the expression of tumor-promoting genes. In a myeloid lineage MYC KO mouse model, MYC had important roles in macrophage maturation and function leading to reduced tumor growth. We therefore hypothesized that targeted delivery of a MYC inhibitor to established M2 TAMs could reduce polarization toward an M2 phenotype in breast cancer models. Methods: In this study, we developed a MYC inhibitor prodrug (MI3-PD) for encapsulation within perfluorocarbon nanoparticles, which can deliver drugs directly to the cytosol of the target cell through a phagocytosis independent mechanism. We have previously shown that M2-like TAMs express significant levels of the vitronectin receptor, integrin ß3, and in vivo targeting and therapeutic potential was evaluated using αvß3 integrin targeted rhodamine-labeled nanoparticles (NP) or integrin αvß3-MI3-PD nanoparticles. Results: We observed that rhodamine, delivered by αvß3-rhodamine NP, was incorporated into M2 tumor promoting macrophages through both phagocytosis-independent and dependent mechanisms, while NP uptake in tumor suppressing M1 macrophages was almost exclusively through phagocytosis. In a mouse model of breast cancer (4T1-GFP-FL), M2-like TAMs were significantly reduced with αvß3-MI3-PD NP treatment. To validate this effect was independent of drug delivery to tumor cells and was specific to the MYC inhibitor, mice with integrin ß3 knock out tumors (PyMT-Bo1 ß3KO) were treated with αvß3-NP or αvß3-MI3-PD NP. M2 macrophages were significantly reduced with αvß3-MI3-PD nanoparticle therapy but not αvß3-NP treatment. Conclusion: These data suggest αvß3-NP-mediated drug delivery of a c-MYC inhibitor can reduce protumor M2-like macrophages while preserving antitumor M1-like macrophages in breast cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Macrophages/drug effects , Nanoparticles/administration & dosage , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/pathology , Carcinogenesis/drug effects , Carcinogenesis/immunology , Cell Line, Tumor/transplantation , Drug Evaluation, Preclinical , Female , Fluorocarbons/administration & dosage , Fluorocarbons/chemistry , Gene Knockout Techniques , Humans , Integrin alphaVbeta3 , Integrin beta3 , Macrophages/immunology , Macrophages/metabolism , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Nanoparticles/chemistry , Phagocytosis , Primary Cell Culture , Prodrugs/administration & dosage , Proto-Oncogene Proteins c-myc/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
2.
Nat Commun ; 9(1): 275, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348537

ABSTRACT

Most cancer patients succumb to disseminated disease because conventional systemic therapies lack spatiotemporal control of their toxic effects in vivo, particularly in a complicated milieu such as bone marrow where progenitor stem cells reside. Here, we demonstrate the treatment of disseminated cancer by photoactivatable drugs using radiopharmaceuticals. An orthogonal-targeting strategy and a contact-facilitated nanomicelle technology enabled highly selective delivery and co-localization of titanocene and radiolabelled fluorodeoxyglucose in disseminated multiple myeloma cells. Selective ablation of the cancer cells was achieved without significant off-target toxicity to the resident stem cells. Genomic, proteomic and multimodal imaging analyses revealed that the downregulation of CD49d, one of the dimeric protein targets of the nanomicelles, caused therapy resistance in small clusters of cancer cells. Similar treatment of a highly metastatic breast cancer model using human serum albumin-titanocene formulation significantly inhibited cancer growth. This strategy expands the use of phototherapy for treating previously inaccessible metastatic disease.


Subject(s)
Mammary Neoplasms, Experimental/therapy , Multiple Myeloma/therapy , Organometallic Compounds/administration & dosage , Photochemotherapy , Radiopharmaceuticals/administration & dosage , Animals , Cell Line , Drug Resistance, Neoplasm , Female , Integrin alpha4beta1 , Mice, Inbred C57BL , Mice, SCID , Micelles , Molecular Targeted Therapy , Nanoparticles , Rats , Serum Albumin, Human , Xenograft Model Antitumor Assays
3.
Nanomedicine ; 11(3): 601-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25652900

ABSTRACT

High-relaxivity T1-weighted (T1w) MR molecular imaging nanoparticles typically present high surface gadolinium payloads that can elicit significant acute complement activation (CA). The objective of this research was to develop a high T1w contrast nanoparticle with improved safety. We report the development, optimization, and characterization of a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC; 138±10 (Dav)/nm; PDI: 0.06; zeta: -27±2 mV). High r1 particulate relaxivity with minute additions of Gd-DOTA-lipid conjugate to the MnOL nanocolloid surface achieved an unexpected paramagnetic synergism. This hybrid MnOL-Gd NC provided optimal MR TSE signal intensity at 5 nM/voxel and lower levels consistent with the level expression anticipated for sparse biomarkers, such as neovascular integrins. MnOL NC produced optimal MR TSE signal intensity at 10 nM/voxel concentrations and above. Importantly, MnOL-Gd NC avoided acute CA in vitro and in vivo while retaining minimal transmetallation risk. From the clinical editor: The authors developed a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC) in this study. These were used as a high-relaxivity paramagnetic MR molecular imaging agent in experimental models. It was shown that MnOL-Gd NC could provide high T1w MR contrast for targeted imaging. As the level of gadolinium used was reduced, there was also reduced risk of systemic side effects from complement activation.


Subject(s)
Complement Activation/drug effects , Contrast Media , Gadolinium , Magnetic Resonance Imaging , Manganese , Nanoparticles , Animals , Biomarkers/blood , Colloids , Contrast Media/adverse effects , Contrast Media/chemistry , Contrast Media/pharmacology , Drug Evaluation, Preclinical , Gadolinium/adverse effects , Gadolinium/chemistry , Gadolinium/pharmacology , Manganese/adverse effects , Manganese/chemistry , Manganese/pharmacology , Mice , Nanoparticles/adverse effects , Nanoparticles/chemistry
4.
Theranostics ; 5(2): 124-33, 2015.
Article in English | MEDLINE | ID: mdl-25553103

ABSTRACT

Photoacoustic (PA) tomography enables multiscale, multicontrast and high-resolution imaging of biological structures. In particular, contrast-enhanced PA imaging offers high-sensitivity noninvasive imaging of neovessel sprout formation and nascent tubules, which are important biomarkers of malignant tumors and progressive atherosclerotic disease. While gold nanoparticles or nanorods have been used as PA contrast agents, we utilized high-density copper oleate small molecules encapsulated within a phospholipid surfactant (CuNPs) to generate a soft nanoparticle with PA contrast comparable to that from gold. Within the NIR window, the copper nanoparticles provided a 4-fold higher signal than that of blood. ανß3-integrin targeting of CuNPs in a Matrigel(TM) angiogenesis mouse model demonstrated prominent (p<0.05) PA contrast enhancement of the neovasculature compared with mice given nontargeted or competitively inhibited CuNPs. Furthermore, incorporation of a Sn 2 lipase-labile fumagillin prodrug into the CuNP outer lipid membrane produced marked antiangiogenesis in the same model when targeted to the ανß3-integrin, providing proof of concept in vivo for the first targeted PA - drug delivery agent.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Copper/metabolism , Cyclohexanes/metabolism , Fatty Acids, Unsaturated/metabolism , Integrin alphaVbeta3/metabolism , Neovascularization, Pathologic/diagnosis , Neovascularization, Pathologic/therapy , Animals , Disease Models, Animal , Lipase/metabolism , Mice, Nude , Nanoparticles/metabolism , Photoacoustic Techniques/methods , Prodrugs/metabolism , Sesquiterpenes/metabolism
5.
ACS Nano ; 8(7): 7305-17, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-24941020

ABSTRACT

Antiangiogenesis has been extensively explored for the treatment of a variety of cancers and certain inflammatory processes. Fumagillin, a mycotoxin produced by Aspergillus fumigatus that binds methionine aminopeptidase 2 (MetAP-2), is a potent antiangiogenic agent. Native fumagillin, however, is poorly soluble and extremely unstable. We have developed a lipase-labile fumagillin prodrug (Fum-PD) that eliminated the photoinstability of the compound. Using αvß3-integrin-targeted perfluorocarbon nanocarriers to deliver Fum-PD specifically to angiogenic vessels, we effectively suppressed clinical disease in an experimental model of rheumatoid arthritis (RA). The exact mechanism by which Fum-PD-loaded targeted nanoparticles suppressed inflammation in experimental RA, however, remained unexplained. We herein present evidence that Fum-PD nanotherapy indirectly suppresses inflammation in experimental RA through the local production of endothelial nitric oxide (NO). Fum-PD-induced NO activates AMP-activated protein kinase (AMPK), which subsequently modulates macrophage inflammatory response. In vivo, NO-induced AMPK activation inhibits mammalian target of rapamycin (mTOR) activity and enhances autophagic flux, as evidenced by p62 depletion and increased autolysosome formation. Autophagy in turn mediates the degradation of IkappaB kinase (IKK), suppressing the NF-κB p65 signaling pathway and inflammatory cytokine release. Inhibition of NO production by N(G)-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, reverses the suppression of NF-κB-mediated inflammatory response induced by Fum-PD nanotherapy. These unexpected results uncover an activity of Fum-PD nanotherapy that may be further explored in the treatment of angiogenesis-dependent diseases.


Subject(s)
Cyclohexanes/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fatty Acids, Unsaturated/metabolism , Macrophages/drug effects , Nanomedicine , Nitric Oxide/metabolism , Prodrugs/pharmacology , AMP-Activated Protein Kinases/metabolism , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/metabolism , Animals , Arthritis/drug therapy , Arthritis/immunology , Arthritis/metabolism , Arthritis/pathology , Cyclohexanes/chemistry , Cytokines/metabolism , Enzyme Activation/drug effects , Fatty Acids, Unsaturated/chemistry , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lipase/metabolism , Macrophages/cytology , Male , Mice , Nanoparticles , Prodrugs/metabolism , Prodrugs/therapeutic use , Sesquiterpenes/chemistry , Sesquiterpenes/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism
6.
Biomaterials ; 33(33): 8632-40, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22922023

ABSTRACT

Nanoparticle-based therapeutics are emerging technologies that have the potential to greatly impact the treatment of many human diseases. However, drug instability and premature release from the nanoparticles during circulation currently preclude clinical translation. Herein, we use a lipase-labile (Sn 2) fumagillin prodrug platform coupled with a unique lipid surface-to-surface targeted delivery mechanism, termed contact-facilitated drug delivery, to counter the premature drug release and overcome the inherent photo-instability of fumagillin, an established anti-angiogenic agent. We show that α(v)ß(3)-integrin targeted fumagillin prodrug nanoparticles, administered at 0.3 mg of fumagillin prodrug/kg of body weight suppress the clinical disease indices of KRN serum-mediated arthritis in a dose-dependent manner when compared to treatment with the control nanoparticles with no drug. This study demonstrates the effectiveness of this lipase-labile prodrug nanocarrier in a relevant preclinical model that approximates human rheumatoid arthritis. The lipase-labile prodrug paradigm offers a translatable approach that is broadly applicable to many targeted nanosystems and increases the translational potential of this platform for many diseases.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Cyclohexanes/metabolism , Cyclohexanes/therapeutic use , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/therapeutic use , Lipase/metabolism , Nanoparticles/chemistry , Animals , Cyclohexanes/chemistry , Electrophoresis , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Unsaturated/chemistry , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred C57BL , Sesquiterpenes/chemistry , Sesquiterpenes/metabolism , Sesquiterpenes/therapeutic use
7.
Nanomedicine (Lond) ; 7(10): 1507-19, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22709347

ABSTRACT

BACKGROUND: The chemical instability of antiangiogenic fumagillin, combined with its poor retention during intravascular transit, requires an innovative solution for clinical translation. We hypothesized that an Sn-2 lipase-labile fumagillin prodrug, in combination with a contact-facilitated drug delivery mechanism, could be used to address these problems. METHODS: α(v)ß(3)-targeted and nontargeted nanoparticles with and without fumagillin in the prodrug or native forms were evaluated in vitro and in vivo in the Matrigel™ (BD Biosciences, CA, USA) plug model of angiogenesis in mice. RESULTS: In vitro experiments demonstrated that the new fumagillin prodrug decreased viability at least as efficacious as the parent compound, on an equimolar basis. In the Matrigel mouse angiogenesis model, α(v)ß(3)-fumagillin prodrug decreased angiogenesis as measured by MRI (3T), while the neovasculature was unaffected with the control nanoparticles. CONCLUSION: The present approach resolved the previously intractable problems of drug instability and premature release in transit to target sites.


Subject(s)
Cyclohexanes/therapeutic use , Fatty Acids, Unsaturated/therapeutic use , Lipase/therapeutic use , Nanomedicine , Neovascularization, Pathologic/therapy , Prodrugs/therapeutic use , Animals , Biological Availability , Cells, Cultured , Cyclohexanes/pharmacokinetics , Fatty Acids, Unsaturated/pharmacokinetics , Humans , Mice , Rats , Sesquiterpenes/pharmacokinetics , Sesquiterpenes/therapeutic use
8.
FASEB J ; 23(9): 2978-85, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19376816

ABSTRACT

The purpose of this study was to assess whether an alternative treatment approach that targets angiogenesis, delivered through ligand-targeted nanotherapy, would ameliorate inflammatory arthritis. Arthritis was induced using the K/BxN mouse model of inflammatory arthritis. After arthritis was clearly established, mice received three consecutive daily doses of alpha(v)beta(3)-targeted fumagillin nanoparticles. Control groups received no treatment or alpha(v)beta(3)-targeted nanoparticles without drugs. Disease score and paw thickness were measured daily. Mice that received alpha(v)beta(3)-targeted fumagillin nanoparticles showed a significantly lower disease activity score (mean score of 1.4+/-0.4; P<0.001) and change in ankle thickness (mean increase of 0.17+/-0.05 mm; P<0.001) 7 d after arthritis induction, whereas the group that received alpha(v)beta(3)-targeted nanoparticles without drugs exhibited a mean arthritic score of 9.0 +/- 0.3 and mean change in ankle thickness of 1.01 +/- 0.09 mm. Meanwhile, the group that received no treatment showed a mean arthritic score of 9.8 +/- 0.5 and mean change in ankle thickness of 1.05 +/- 0.10 mm. Synovial tissues from animals treated with targeted fumagillin nanoparticles also showed significant decrease in inflammation and angiogenesis and preserved proteoglycan integrity. Ligand-targeted nanotherapy to deliver antiangiogenic agents may represent an effective way to treat inflammatory arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Cyclohexanes/therapeutic use , Fatty Acids, Unsaturated/therapeutic use , Integrin alphaVbeta3/drug effects , Animals , Arthritis, Experimental/pathology , Cyclohexanes/pharmacology , Drug Delivery Systems/methods , Fatty Acids, Unsaturated/pharmacology , Inflammation/drug therapy , Mice , Nanoparticles/therapeutic use , Neovascularization, Pathologic/drug therapy , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Treatment Outcome
9.
J Am Chem Soc ; 130(29): 9186-7, 2008 Jul 23.
Article in English | MEDLINE | ID: mdl-18572935

ABSTRACT

Although gadolinium has been the dominant paramagnetic metal for MR paramagnetic contrast agents, the recent association of this lanthanide with nephrogenic systemic fibrosis, an untreatable disease, has spawned renewed interest in alternative metals for MR molecular imaging. We have developed a self-assembled, manganese(III)-labeled nanobialys (1), a toroidal-shaped MR theranostic nanoparticle. In this report, Mn(III) nanobialys are characterized as MR molecular imaging agents for targeted detection of fibrin, a major biochemical feature of thrombus. A complementary ability of nanobialys to incorporate chemotherapeutic compounds with greater than 98% efficiency and to retain more than 80% of these drugs after infinite sink dissolution, point to the theranostic potential of this platform technology.


Subject(s)
Drug Delivery Systems/methods , Magnetic Resonance Angiography/methods , Manganese/chemistry , Nanoparticles/chemistry , Biotin/chemistry , Camptothecin/administration & dosage , Camptothecin/chemistry , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Fibrin/analysis , Fibrin/chemistry , Humans , Micelles , Microscopy, Atomic Force , Organometallic Compounds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL