16 research outputs found

    A clinical evaluation of an ex vivo organ culture system to predict patient response to cancer therapy

    Get PDF
    IntroductionEx vivo organ cultures (EVOC) were recently optimized to sustain cancer tissue for 5 days with its complete microenvironment. We examined the ability of an EVOC platform to predict patient response to cancer therapy.MethodsA multicenter, prospective, single-arm observational trial. Samples were obtained from patients with newly diagnosed bladder cancer who underwent transurethral resection of bladder tumor and from core needle biopsies of patients with metastatic cancer. The tumors were cut into 250 μM slices and cultured within 24 h, then incubated for 96 h with vehicle or intended to treat drug. The cultures were then fixed and stained to analyze their morphology and cell viability. Each EVOC was given a score based on cell viability, level of damage, and Ki67 proliferation, and the scores were correlated with the patients’ clinical response assessed by pathology or Response Evaluation Criteria in Solid Tumors (RECIST).ResultsThe cancer tissue and microenvironment, including endothelial and immune cells, were preserved at high viability with continued cell division for 5 days, demonstrating active cell signaling dynamics. A total of 34 cancer samples were tested by the platform and were correlated with clinical results. A higher EVOC score was correlated with better clinical response. The EVOC system showed a predictive specificity of 77.7% (7/9, 95% CI 0.4–0.97) and a sensitivity of 96% (24/25, 95% CI 0.80–0.99).ConclusionEVOC cultured for 5 days showed high sensitivity and specificity for predicting clinical response to therapy among patients with muscle-invasive bladder cancer and other solid tumors

    TL-118 and Gemcitabine Drug Combination Display Therapeutic Efficacy in a MYCN Amplified Orthotopic Neuroblastoma Murine Model – Evaluation by MRI

    No full text
    <div><p>Neuroblastoma (NB) is the most common extra-cranial pediatric solid tumor with up to 50% of NB patients classified as having high-risk disease with poor long-term survival rates. The poor clinical outcome and aggressiveness of high-risk NB strongly correlates with enhanced angiogenesis, suggesting anti-angiogenic agents as attractive additions to the currently insufficient therapeutics. TL-118, a novel drug combination has been recently developed to inhibit tumor angiogenesis. In the current study, we used the SK-N-BE (2) cell line to generate orthotopic NB tumors in order to study the combinational therapeutic potential of TL-118 with either Gemcitabine (40 mg/kg; IP) or Retinoic acid (40 mg/kg; IP). We show that TL-118 treatment (n = 9) significantly inhibited tumor growth, increased cell apoptosis, reduced proliferation and extended mouse survival. Moreover, the reciprocal effect of TL-118 and Gemcitabine treatment (n = 10) demonstrated improved anti-tumor activity. The synergistic effect of these drugs in combination was more effective than either TL or Gemcitabine alone (n = 9), via significantly reduced cell proliferation (p<0.005), increased apoptosis (p<0.05) and significantly prolonged survival (2-fold; p<0.00001). To conclude, we demonstrate that the novel drug combination TL-118 has the ability to suppress the growth of an aggressive NB tumor. The promising results with TL-118 in this aggressive animal model may imply that this drug combination has therapeutic potential in the clinical setting.</p></div

    Treatment effect on tumor growth and mouse survival.

    No full text
    <p>A. Tumor volume (mm<sup>3</sup>) for each individual mouse, as measured from T<sub>2</sub>W MRI images as a function of days post cell inoculation in control (n = 19), Gemcitabine (Gem; n = 6), TL-118<sup>1/4</sup> (n = 9) and TL<sup>1/4</sup>+ Gem combination (n = 10) treated mice. The dashed line indicates the maximal survival day of the control-treated mice. The b-values represent the average exponential coefficients of each treatment group. The b-values of all the treated groups (Gem, TL<sup>1/4</sup> and TL<sup>1/4</sup>+ Gem) were significantly lower compared to control (p<0.0001). B. Representative T<sub>2</sub>W anatomical axial images of Control, Gem, TL-118<sup>1/4</sup> and TL<sup>1/4</sup>+ Gem treated tumors that were acquired on the indicated days (Bar = 1 cm) C. Kaplan-Meier survival analysis for each of the treated groups (*p<0.05; **P<0.0001; ***p<0.00001 compared to control). D. Box and Whisker plots of mean calculated b-values for each treated group (black square – median; * p<0.0001).</p

    Effects of the different therapies on NB tumor proliferation and apoptosis.

    No full text
    <p>Representative histological sections of control (1<sup>st</sup> column), TL-118<sup>1/4</sup> (2<sup>nd</sup> column), Gem (3<sup>rd</sup> column) and TL<sup>1/4</sup> + Gem (4<sup>th</sup> column) treated mice. Slides were immuno-stained for proliferation (BrdU) (A) and apoptosis (TUNEL) (B); Quantification of BrdU positive (C) and TUNEL positive (D) cells/HPF analyzed from 10 randomly selected HPF/tumor ; n = 3–6 mice/group. Original magnification is indicated on the right image of each row. * p = 0.01, ** p<0.005. The histological sections were taken at the end of the experiments when the tumor load/mouse reached ethical limits.</p

    Effects of TL therapy combinations on NB tumor vascularization and perfusion.

    No full text
    <p>A. Representative T<sub>2</sub>W fast SE images (top) and the corresponding ΔSco<sub>2</sub> (middle row) and ΔSo<sub>2</sub> (bottom) maps of control (left), TL-118<sup>1/4</sup> (middle column) and TL<sup>1/4</sup> + Gem (right) treated tumors (Bar = 1 cm). B. Mean ΔSo<sub>2</sub> values of the tumor and liver-ROI's calculated for control (blue), TL-118<sup>1/4</sup> (green) and TL<sup>1/4</sup> + Gem (purple) treated mice. C. Representative histological slides immuno-stained with the smooth muscle marker α-SMA (upper-rows) and with the endothelial cell marker CD31 (lower-rows), of control (top), TL-118<sup>1/4</sup> (middle) and TL<sup>1/4</sup> + Gem (bottom) treated tumors. Photographs were taken from the peripheral (left column) and central (right column) regions (original magnification ×20) demonstrating the higher vascularity in tumor periphery. D. Quantification of α-SMA positive vessels/HPF was determined from the tumor center (dark) and peripheral (light) areas for each of the treatment groups. (Mean ± SE; n = 5–7 mice/group).</p

    RNA-seq

    No full text

    Table_1_A clinical evaluation of an ex vivo organ culture system to predict patient response to cancer therapy.docx

    No full text
    IntroductionEx vivo organ cultures (EVOC) were recently optimized to sustain cancer tissue for 5 days with its complete microenvironment. We examined the ability of an EVOC platform to predict patient response to cancer therapy.MethodsA multicenter, prospective, single-arm observational trial. Samples were obtained from patients with newly diagnosed bladder cancer who underwent transurethral resection of bladder tumor and from core needle biopsies of patients with metastatic cancer. The tumors were cut into 250 μM slices and cultured within 24 h, then incubated for 96 h with vehicle or intended to treat drug. The cultures were then fixed and stained to analyze their morphology and cell viability. Each EVOC was given a score based on cell viability, level of damage, and Ki67 proliferation, and the scores were correlated with the patients’ clinical response assessed by pathology or Response Evaluation Criteria in Solid Tumors (RECIST).ResultsThe cancer tissue and microenvironment, including endothelial and immune cells, were preserved at high viability with continued cell division for 5 days, demonstrating active cell signaling dynamics. A total of 34 cancer samples were tested by the platform and were correlated with clinical results. A higher EVOC score was correlated with better clinical response. The EVOC system showed a predictive specificity of 77.7% (7/9, 95% CI 0.4–0.97) and a sensitivity of 96% (24/25, 95% CI 0.80–0.99).ConclusionEVOC cultured for 5 days showed high sensitivity and specificity for predicting clinical response to therapy among patients with muscle-invasive bladder cancer and other solid tumors.</p

    Table_3_A clinical evaluation of an ex vivo organ culture system to predict patient response to cancer therapy.docx

    No full text
    IntroductionEx vivo organ cultures (EVOC) were recently optimized to sustain cancer tissue for 5 days with its complete microenvironment. We examined the ability of an EVOC platform to predict patient response to cancer therapy.MethodsA multicenter, prospective, single-arm observational trial. Samples were obtained from patients with newly diagnosed bladder cancer who underwent transurethral resection of bladder tumor and from core needle biopsies of patients with metastatic cancer. The tumors were cut into 250 μM slices and cultured within 24 h, then incubated for 96 h with vehicle or intended to treat drug. The cultures were then fixed and stained to analyze their morphology and cell viability. Each EVOC was given a score based on cell viability, level of damage, and Ki67 proliferation, and the scores were correlated with the patients’ clinical response assessed by pathology or Response Evaluation Criteria in Solid Tumors (RECIST).ResultsThe cancer tissue and microenvironment, including endothelial and immune cells, were preserved at high viability with continued cell division for 5 days, demonstrating active cell signaling dynamics. A total of 34 cancer samples were tested by the platform and were correlated with clinical results. A higher EVOC score was correlated with better clinical response. The EVOC system showed a predictive specificity of 77.7% (7/9, 95% CI 0.4–0.97) and a sensitivity of 96% (24/25, 95% CI 0.80–0.99).ConclusionEVOC cultured for 5 days showed high sensitivity and specificity for predicting clinical response to therapy among patients with muscle-invasive bladder cancer and other solid tumors.</p
    corecore