Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Nano ; 7(3): 2132-44, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23373542

ABSTRACT

Tumor-associated inflammation mediates the development of a systemic immunosuppressive milieu that is a major obstacle to effective treatment of cancer. Inflammation has been shown to promote the systemic expansion of immature myeloid cells which have been shown to exert immunosuppressive activity in laboratory models of cancer as well as cancer patients. Consequentially, significant effort is underway toward the development of therapies that decrease tumor-associated inflammation and immunosuppressive cells. The current study demonstrated that a previously described deep tissue imaging modality, which utilized indocyanine green-loaded calcium phosphosilicate nanoparticles (ICG-CPSNPs), could be utilized as an immunoregulatory agent. The theranostic application of ICG-CPSNPs as photosensitizers for photodynamic therapy was shown to block tumor growth in murine models of breast cancer, pancreatic cancer, and metastatic osteosarcoma by decreasing inflammation-expanded immature myeloid cells. Therefore, this therapeutic modality was termed PhotoImmunoNanoTherapy. As phosphorylated sphingolipid metabolites have been shown to have immunomodulatory roles, it was hypothesized that the reduction of immature myeloid cells by PhotoImmunoNanoTherapy was dependent upon bioactive sphingolipids. Mechanistically, PhotoImmunoNanoTherapy induced a sphingosine kinase 2-dependent increase in sphingosine-1-phosphate and dihydrosphingosine-1-phosphate. Furthermore, dihydrosphingosine-1-phosphate was shown to selectively abrogate myeloid lineage cells while concomitantly allowing the expansion of lymphocytes that exerted an antitumor effect. Collectively, these findings revealed that PhotoImmunoNanoTherapy, utilizing the novel nontoxic theranostic agent ICG-CPSNP, can decrease tumor-associated inflammation and immature myeloid cells in a sphingosine kinase 2-dependent manner. These findings further defined a novel myeloid regulatory role for dihydrosphingosine-1-phosphate. PhotoImmunoNanoTherapy holds the potential to be a revolutionary treatment for cancers with inflammatory and immunosuppressive phenotypes.


Subject(s)
Immunotherapy/methods , Nanoparticles/therapeutic use , Neoplasms, Experimental/therapy , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Photochemotherapy/methods , Sphingosine/analogs & derivatives , Animals , Cell Line, Tumor , Combined Modality Therapy , Female , Humans , Indocyanine Green/administration & dosage , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Nude , Myeloid Cells/immunology , Myeloid Cells/metabolism , Nanoparticles/chemistry , Nanotechnology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Silicates/chemistry , Sphingosine/metabolism , Xenograft Model Antitumor Assays
2.
ACS Nano ; 4(9): 4967-70, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20873786

ABSTRACT

The ability to apply nanomaterials as targeted delivery agents for drugs and other therapeutics holds promise for a wide variety of diseases, including many types of cancer. A nanodelivery vehicle must demonstrate in vivo efficacy, diminished or no toxicity, stability, improved pharmacokinetics, and controlled-release kinetics. In this issue, Lee et al. construct polymer nanobins that fulfill these requirements and demonstrate effective delivery of doxorubicin in vivo to breast cancer cells. This Perspective explores the outlook for these nanobins as well as other technologies in this field and the challenges that lie ahead.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/metabolism , Coated Materials, Biocompatible/pharmacokinetics , Coated Materials, Biocompatible/toxicity , Delayed-Action Preparations , Drug Carriers/metabolism , Drug Carriers/pharmacokinetics , Drug Carriers/toxicity , Humans , Nanocapsules/chemistry , Nanocapsules/toxicity , Nanomedicine , Nanoparticles/toxicity
3.
Article in English | MEDLINE | ID: mdl-20135691

ABSTRACT

Near infrared imaging has presented itself as a powerful diagnostic technique with potential to serve as a minimally invasive, nonionizing method for sensitive, deep tissue diagnostic imaging. This potential is further realized with the use of nanoparticle (NP)-based near infrared (NIR) contrast agents that are not prone to the rapid photobleaching and instability of their organic counterparts. This review discusses applications that have successfully demonstrated the utility of nanoparticles for NIR imaging, including NIR-emitting semiconductor quantum dots (QDs), resonant gold nanoshells, and dye-encapsulating nanoparticles. NIR QDs demonstrate superior optical performance with exceptional fluorescence brightness stability. However, the heavy metal composition and high propensity for toxicity hinder future application in clinical environments. NIR resonant gold nanoshells also exhibit brilliant signal intensities and likewise have none of the photo- or chemical-instabilities characteristic of organic contrast agents. However, concerns regarding ineffectual clearance and long-term accumulation in nontarget organs are a major issue for this technology. Finally, NIR dye-encapsulating nanoparticles synthesized from calcium phosphate (CP) also demonstrate improved optical performances by shielding the component dye from undesirable environmental influences, thereby enhancing quantum yields, emission brightness, and fluorescent lifetime. Calcium phosphate nanoparticle (CPNP) contrast agents are neither toxic, nor have issues with long-term sequestering, as they are readily dissolved in low pH environments and ultimately absorbed into the system. Though perhaps not as optically superior as QDs or nanoshells, these are a completely nontoxic, bioresorbable option for NP-based NIR imaging that still effectively improves the optical performance of conventional organic agents.


Subject(s)
Nanoparticles , Nanotechnology/methods , Spectroscopy, Near-Infrared/methods , Animals , Contrast Media/chemistry , Humans , Neoplasms/diagnosis , Neoplasms/drug therapy , Swine
4.
ACS Nano ; 4(3): 1279-87, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20180585

ABSTRACT

The early diagnosis of cancer is the critical element in successful treatment and long-term favorable patient prognoses. The high rate of mortality is mainly attributed to the tendency for late diagnoses as symptoms may not occur until the disease has metastasized, as well as the lack of effective systemic therapies. Late diagnosis is often associated with the lack of timely sensitive imaging modalities. The promise of nanotechnology is presently limited by the inability to simultaneously seek, treat, and image cancerous lesions. This study describes the design and synthesis of fluorescent calcium phosphosilicate nanocomposite particles (CPNPs) that can be systemically targeted to breast and pancreatic cancer lesions. The CPNPs are a approximately 20 nm diameter composite composed of an amorphous calcium phosphate matrix doped with silicate in which a near-infrared imaging agent, indocyanine green (ICG), is embedded. In the present studies, we describe and validate CPNP bioconjugation of human holotransferrin, anti-CD71 antibody, and short gastrin peptides via an avidin-biotin or a novel PEG-maleimide coupling strategy. The conjugation of biotinylated human holotransferrin (diferric transferrin) and biotinylated anti-CD71 antibody (anti-transferrin receptor antibody) to avidin-conjugated CPNPs (Avidin-CPNPs) permits targeting of transferrin receptors, which are highly expressed on breast cancer cells. Similarly, the conjugation of biotinylated pentagastrin to Avidin-CPNPs and decagastrin (gastrin-10) to PEG-CPNPs via PEG-maleimide coupling permits targeting of gastrin receptors, which are overexpressed in pancreatic cancer lesions. These bioconjugated CPNPs have the potential to perform as a theranostic modality, simultaneously enhancing drug delivery, targeting, and imaging of breast and pancreatic cancer tumors.


Subject(s)
Breast Neoplasms/metabolism , Nanoparticles/chemistry , Pancreatic Neoplasms/metabolism , Silicates/chemistry , Animals , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Drug Design , Humans , Mice , Organ Specificity , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism
6.
Nano Lett ; 8(12): 4108-15, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19367837

ABSTRACT

Encapsulation of imaging agents and drugs in calcium phosphate nanoparticles (CPNPs) has potential as a nontoxic, bioresorbable vehicle for drug delivery to cells and tumors. The objectives of this study were to develop a calcium phosphate nanoparticle encapsulation system for organic dyes and therapeutic drugs so that advanced fluoresence methods could be used to assess the efficiency of drug delivery and possible mechanisms of nanoparticle bioabsorption. Highly concentrated CPNPs encapsulating a variety of organic fluorophores were successfully synthesized. Well-dispersed CPNPs encapsulating Cy3 amidite exhibited nearly a 5-fold increase in fluorescence quantum yield when compared to the free dye in PBS. FCS diffusion data and cell staining were used to show pH-dependent dissolution of the particles and cellular uptake, respectively. Furthermore, an experimental hydrophobic cell growth inhibitor, ceramide, was successfully delivered in vitro to human vascular smooth muscle cells via encapsulation in CPNPs. These studies demonstrate that CPNPs are effective carriers of dyes and drugs for bioimaging and, potentially, for therapeutic intervention.


Subject(s)
Calcium Phosphates/chemistry , Drug Carriers , Nanocomposites , Organic Chemicals/chemistry , Animals , Cattle , Microscopy, Electron, Transmission
7.
Nano Lett ; 8(12): 4116-21, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19367878

ABSTRACT

Paradigm-shifting modalities to more efficiently deliver drugs to cancerous lesions require the following attributes: nanoscale-size, targetability, and stability under physiological conditions. Often, these nanoscale drug delivery vehicles are limited due to agglomeration, poor solubility, or cytotoxicity. Thus, we have designed a methodology to encapsulate hydrophobic antineoplastic chemotherapeutics within a 20-30 nm diameter, pH-responsive, nonagglomerating, nontoxic calcium phosphate nanoparticle matrix. In the present study, we report on calcium phosphate nanocomposite particles (CPNPs) that encapsulate both fluorophores and chemotherapeutics, are colloidally stable in physiological solution for an extended time at 37 degrees C and can efficaciously deliver hydrophobic antineoplastic agents, such as ceramide, in several cell model systems.


Subject(s)
Antineoplastic Agents/therapeutic use , Calcium Phosphates/chemistry , Drug Carriers , Nanocomposites , Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage
8.
ACS Nano ; 2(10): 2075-84, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-19206454

ABSTRACT

Early detection is a crucial element for the timely diagnosis and successful treatment of all human cancers but is limited by the sensitivity of current imaging methodologies. We have synthesized and studied bioresorbable calcium phosphate nanoparticles (CPNPs) in which molecules of the near-infrared (NIR) emitting fluorophore, indocyanine green (ICG), are embedded. The ICG-CPNPs demonstrate exceptional colloidal and optical characteristics. Suspensions consisting of 16 nm average diameter particles are colloidally stable in physiological solutions (phosphate buffered 0.15 M saline (PBS), pH 7.4) with carboxylate or polyethylene glycol (PEG) surface functionality. ICG-doped CPNPs exhibit significantly greater intensity at the maximum emission wavelength relative to the free constituent fluorophore, consistent with the multiple molecules encapsulated per particle. The quantum efficiency per molecule of the ICG-CPNPs is 200% greater at 0.049 +/- 0.003 over the free fluorophore in PBS. Photostability based on fluorescence half-life of encapsulated ICG in PBS is 500% longer under typical clinical imaging conditions relative to the free dye. PEGylated ICG-CPNPs accumulate in solid, 5 mm diameter xenograft breast adenocarcinoma tumors via enhanced retention and permeability (EPR) within 24 h after systemic tail vein injection in a nude mouse model. Ex situ tissue imaging further verifies the facility of the ICG-CPNPs for deep-tissue imaging with NIR signals detectable from depths up to 3 cm in porcine muscle tissue. Our ex vivo and in vivo experiments verify the promise of the NIR CPNPs for diagnostic imaging in the early detection of solid tumors.


Subject(s)
Breast Neoplasms/pathology , Calcium Phosphates , Drug Carriers/chemistry , Indocyanine Green , Microscopy, Fluorescence/methods , Nanoparticles/chemistry , Animals , Cell Line, Tumor , Humans , Image Enhancement/methods , Infrared Rays , Mice , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...