Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Bioconjug Chem ; 29(4): 1111-1118, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29412642

ABSTRACT

The limited delivery of chemotherapy agents to cancer cells and the nonspecific action of these agents are significant challenges in oncology. We have previously developed a customizable drug delivery and activation system in which a nucleic acid functionalized gold nanoparticle (Au-NP) delivers a drug that is selectively activated within a cancer cell by the presence of an mRNA unique to the cancer cell. The amount of drug released from sequestration to the Au-NP is determined by both the presence and the abundance of the cancer cell specific mRNA in a cell. We have now developed this technology for the potent, but difficult to deliver, topoisomerase I inhibitor SN-38. Herein, we demonstrate both the efficient delivery and selective release of SN-38 from gold nanoparticles in Ewing sarcoma cells with resulting efficacy in vitro and in vivo. These results provide further preclinical validation for this novel cancer therapy and may be extendable to other cancers that exhibit sensitivity to topoisomerase I inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Gold/chemistry , Irinotecan/pharmacology , Metal Nanoparticles/chemistry , RNA, Messenger/metabolism , Sarcoma, Ewing/genetics , Topoisomerase I Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , In Vitro Techniques , Irinotecan/chemistry , Irinotecan/pharmacokinetics , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacokinetics
2.
Bone ; 47(1): 12-22, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20233612

ABSTRACT

Despite palliative treatments, tumor-induced bone disease (TIBD) remains highly debilitating for many cancer patients and progression typically results in death within two years. Therefore, more effective therapies with enhanced anti-resorptive and cytotoxic characteristics are needed. We developed bisphosphonate-chemotherapeutic conjugates designed to bind bone and hydrolyze, releasing both compounds, thereby targeting both osteoclasts and tumor cells. This study examined the effects of our lead compound, MBC-11 (the anhydride formed between arabinocytidine (AraC)-5'-phosphate and etidronate), on bone tumor burden, bone volume, femur bone mineral density (BMD), and overall survival using two distinct mouse models of TIBD, the 4T1/luc breast cancer and the KAS-6/1-MIP1alpha multiple myeloma models. In mice orthotopically inoculated with 4T1/luc mouse mammary cells, MBC-11 (0.04 microg/day; s.c.) reduced the incidence of bone metastases to 40% (4/10), compared to 90% (9/10; p=0.057) and 100% (5/5; p=0.04) of PBS- or similarly-dosed, zoledronate-treated mice, respectively. MBC-11 also significantly decreased bone tumor burden compared to PBS- or zoledronate-treated mice (p=0.021, p=0.017, respectively). MBC-11 and zoledronate (0.04 microg/day) significantly increased bone volume by two- and four-fold, respectively, compared to PBS-treated mice (p=0.005, p<0.001, respectively). In mice systemically injected with human multiple myeloma KAS-6/1-MIP1alpha cells, 0.04 and 4.0 microg/day MBC-11 improved femur BMD by 13% and 16%, respectively, compared to PBS (p=0.025, p=0.017, respectively) at 10 weeks post-tumor cell injection and increased mean survival to 95 days compared to 77 days in mice treated with PBS (p=0.047). Similar doses of zoledronate also improved femur BMD (p< or =0.01 vs PBS) and increased mean survival to 86 days, but this was not significantly different than in PBS-treated mice (p=0.53). These results demonstrate that MBC-11 decreases bone tumor burden, maintains bone structure, and may increase overall survival, warranting further investigation as a treatment for TIBD.


Subject(s)
Antimetabolites/therapeutic use , Bone Diseases/drug therapy , Bone Diseases/etiology , Diphosphonates/therapeutic use , Neoplasms/complications , Nucleosides/therapeutic use , Animals , Antimetabolites/pharmacology , Bone Density/drug effects , Bone Diseases/physiopathology , Bone and Bones/drug effects , Bone and Bones/pathology , Bone and Bones/physiopathology , Cell Line, Tumor , Cell Proliferation/drug effects , Diphosphonates/chemistry , Diphosphonates/pharmacology , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, SCID , Multiple Myeloma/pathology , Neoplasm Transplantation , Nucleosides/pharmacology , Organ Size/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...