9 research outputs found

    Distribution and efficacy of chemotherapeutics in the treatment of preclinical brain metastases of breast cancer

    Get PDF
    Brain metastases are a critical, life-threatening problem for women with advanced metastatic breast cancer. Approximately 80% of women with disseminated central lesions are unable to survive the first year after diagnosis. Despite the breakdown of the blood-brain barrier, chemotherapeutics have limited penetration and distribution into brain metastases and are unable to induce cytotoxicity in the tumor. Limiting the development of new treatments for brain metastases of breast cancer, there are no commercially available in vitro models available that accurately model, and mimic the functionality of, the in vivo blood-tumor barrier (BTB). In an attempt to address the aforementioned problem, the following connected, but independent aims were proposed and completed in a novel microfluidic device: (1) Determine the permeability of three passive markers and one subject to efflux, in blood-brain barrier (BBB) and BTB models (2) Determine if trastuzumab crosses the BBB and BTB barrier in both in vivo and in vitro models (3) Evaluate if the microfluidic BBB and BTB models are relevant and comparable to current in vivo models. Further, based on the data presented herein, additional questions and trials have evolved into an evolution of the current microfluidic chip, discussed in the final chapter. This dissertation incorporates multiple innovative and complex experiments, which suggest that the current microfluidic chip accurately portrays the BBB and BTB when compared to the in vivo barriers, and is a readily available and rapid throughput model for all cancer, as well as BBB, researchers

    NKTR-102 Efficacy versus irinotecan in a mouse model of brain metastases of breast cancer

    Get PDF
    Background: Brain metastases are an increasing problem in women with invasive breast cancer. Strategies designed to treat brain metastases of breast cancer, particularly chemotherapeutics such as irinotecan, demonstrate limited efficacy. Conventional irinotecan distributes poorly to brain metastases; therefore, NKTR-102, a PEGylated irinotecan conjugate should enhance irinotecan and its active metabolite SN38 exposure in brain metastases leading to brain tumor cytotoxicity. Methods: Female nude mice were intracranially or intracardially implanted with human brain seeking breast cancer cells (MDA-MB-231Br) and dosed with irinotecan or NKTR-102 to determine plasma and tumor pharmacokinetics of irinotecan and SN38. Tumor burden and survival were evaluated in mice treated with vehicle, irinotecan (50 mg/kg), or NKTR-102 low and high doses (10 mg/kg, 50 mg/kg respectively). Results: NKTR-102 penetrates the blood-tumor barrier and distributes to brain metastases. NKTR-102 increased and prolonged SN38 exposure (\u3e20 ng/g for 168 h) versus conventional irinotecan (\u3e1 ng/g for 4 h). Treatment with NKTR-102 extended survival time (from 35 days to 74 days) and increased overall survival for NKTR-102 low dose (30 % mice) and NKTR-102 high dose (50 % mice). Tumor burden decreased (37 % with 10 mg/kg NKTR-102 and 96 % with 50 mg/kg) and lesion sizes decreased (33 % with 10 mg/kg NKTR-102 and 83 % with 50 mg/kg NKTR-102) compared to conventional irinotecan treated animals. Conclusions: Elevated and prolonged tumor SN38 exposure after NKTR-102 administration appears responsible for increased survival in this model of breast cancer brain metastasis. Further, SN38 concentrations observed in this study are clinically achieved with 145 mg/m2 NKTR-102, such as those used in the BEACON trial, underlining translational relevance of these results

    Liposomal Irinotecan Accumulates in Metastatic Lesions, Crosses the Blood-Tumor Barrier (BTB), and Prolongs Survival in an Experimental Model of Brain Metastases of Triple Negative Breast Cancer

    Get PDF
    Purpose—The blood-tumor barrier (BTB) limits irinotecan distribution in tumors of the central nervous system. However, given that the BTB has increased passive permeability we hypothesize that liposomal irinotecan would improve local exposure of irinotecan and its active metabolite SN-38 in brain metastases relative to conventional irinotecan due to enhanced-permeation and retention (EPR) effect. Methods—Female nude mice were intracardially or intracranially implanted with human brain seeking breast cancer cells (brain metastases of breast cancer model). Mice were administered vehicle, non-liposomal irinotecan (50 mg/kg), liposomal irinotecan (10 mg/kg and 50 mg/kg) intravenously starting on day 21. Drug accumulation, tumor burden, and survival were evaluated. Results—Liposomal irinotecan showed prolonged plasma drug exposure with mean residence time (MRT) of 17.7 ± 3.8 h for SN-38, whereas MRT was 3.67 ± 1.2 for non-liposomal irinotecan. Further, liposomal irinotecan accumulated in metastatic lesions and demonstrated prolonged exposure of SN-38 compared to non-liposomal irinotecan. Liposomal irinotecan achieved AUC values of 6883 ± 4149 ng-h/g for SN-38, whereas non-liposomal irinotecan showed significantly lower AUC values of 982 ± 256 ng-h/g for SN-38. Median survival for liposomal irinotecan was 50 days, increased from 37 days (
    corecore