Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Carol Lee is active.

Publication


Featured researches published by Carol Lee.


Biomaterials | 2011

MRI monitoring of intratumoral drug delivery and prediction of the therapeutic effect with a multifunctional thermosensitive liposome

Tatsuaki Tagami; Warren D. Foltz; Mark J. Ernsting; Carol Lee; Ian F. Tannock; Jonathan P. May; Shyh-Dar Li

Non-invasive in vivo imaging of drug distribution enables real-time monitoring and prediction of therapeutic responses to treatment. We have developed a thermosensitive liposomal formulation (HaT: Hyperthermia-activated-cytoToxic) consisting of DPPC and Brij78, a formulation that enhanced drug delivery compared to the lyso-lipid temperature sensitive liposomes (LTSL). Here we report the development of a multifunctional HaT liposome co-encapsulating Gd-DTPA (an MRI probe) and doxorubicin (DOX), which simultaneously releases and reports on drug delivery in a locally heated tumor. The temperature-dependent release profiles of DOX from HaT were closely related to the change in the MR T(1) relaxation time, in which DOX was 100% released at 40-42 °C in 3 min, accompanied by a 60% reduction in T(1). By T(1) relaxometry analysis, no Gd-DTPA leakage was detected in 30 min at 30-37 °C. In the in vivo study, DOX uptake in the tumor was quantitatively correlated with T(1) response (R(2) = 0.98) and the patterns of the T(1) image and the intratumoral DOX uptake were matched, in which both signals were predominantly detected in the highly perfused tumor periphery. Finally, the extent of T(1) relaxation enhancement in the heated tumor successfully predicted the antitumor efficacy in a standard pharmacological response model (R(2) = 0.98).


BMC Cancer | 2010

The distribution of the therapeutic monoclonal antibodies cetuximab and trastuzumab within solid tumors

Carol Lee; Ian F. Tannock

BackgroundPoor distribution of some anticancer drugs in solid tumors may limit their anti-tumor activity.MethodsHere we used immunohistochemistry to quantify the distribution of the therapeutic monoclonal antibodies cetuximab and trastuzumab in relation to blood vessels and to regions of hypoxia in human tumor xenografts. The antibodies were injected into mice implanted with human epidermoid carcinoma A431 or human breast carcinoma MDA-MB-231 transfected with ERBB2 (231-H2N) that express high levels of ErbB1 and ErbB2 respectively, or wild-type MDA-MB-231, which expresses intermediate levels of ErbB1 and low levels of ErbB2.ResultsThe distribution of cetuximab in A431 xenografts and trastuzumab in 231-H2N xenografts was time and dose dependent. At early intervals after injection of 1 mg cetuximab into A431 xenografts, the concentration of cetuximab decreased with increasing distance from blood vessels, but became more uniformly distributed at later times; there remained however limited distribution and binding in hypoxic regions of tumors. Injection of lower doses of cetuximab led to heterogeneous distributions. Similar results were observed with trastuzumab in 231-H2N xenografts. In MDA-MB-231 xenografts, which express lower levels of ErbB1, homogeneity of distribution of cetuximab was achieved more rapidly.ConclusionsCetuximab and trastuzumab distribute slowly, but at higher doses achieve a relatively uniform distribution after about 24 hours, most likely due to their long half-lives in the circulation. There remains poor distribution within hypoxic regions of tumors.


Clinical Cancer Research | 2005

Reduction of Intracellular pH as a Strategy to Enhance the pH-Dependent Cytotoxic Effects of Melphalan for Human Breast Cancer Cells

Philip Wong; Carol Lee; Ian F. Tannock

The microenvironment within solid tumors is slightly acidic, and manipulation of this extracellular acidity to cause intracellular acidification might be used to increase selective antitumor effects of some anticancer drugs. Potential mechanisms include inhibition of repair of DNA damage and inhibition of repopulation of tumor cells between successive courses of chemotherapy. Here, we evaluate the influence of extracellular pH (pHe) and of two agents that lead to intracellular acidification (cariporide and S3705) on toxicity of melphalan for two human breast cancer cell lines (MDA-MB231 and MCF7). Both the total number and number of colony-forming cells were evaluated during and after three sequential weekly drug treatments. Our results indicate the following: (a) Slow or absent repopulation after the first course of treatment that is influenced minimally by pHe. (b) Rapid repopulation after the second course of treatment that may be inhibited at low pHe. (c) Effects of low pHe following treatment with melphalan to increase cell kill. (d) Small effects of incubation in cariporide and S3705 at low pHe to increase the net cell kill after treatment with melphalan. Although these results add to evidence that manipulation of intracellular pH within the acidic environment of solid tumors can influence the effects of chemotherapy, they are too small and inconsistent to warrant clinical evaluation.


Clinical Cancer Research | 2013

Use of the Proton Pump Inhibitor Pantoprazole to Modify the Distribution and Activity of Doxorubicin: A Potential Strategy to Improve the Therapy of Solid Tumors

Krupa J. Patel; Carol Lee; Qian Tan; Ian F. Tannock

Purpose: Limited drug distribution within solid tumors is an important cause of drug resistance. Basic drugs (e.g., doxorubicin) may be sequestered in acidic organelles, thereby limiting drug distribution to distal cells and diverting drugs from their target DNA. Here we investigate the effects of pantoprazole, a proton pump inhibitor, on doxorubicin uptake, and doxorubicin distribution and activity using in vitro and murine models. Experimental Design: Murine EMT-6 and human MCF-7 cells were treated with pantoprazole to evaluate changes in endosomal pH using fluorescence spectroscopy, and uptake of doxorubicin using flow cytometry. Effects of pantoprazole on tissue penetration of doxorubicin were evaluated in multilayered cell cultures (MCC), and in solid tumors using immunohistochemistry. Effects of pantoprazole to influence tumor growth delay and toxicity because of doxorubicin were evaluated in mice. Results: Pantoprazole (>200 μmol/L) increased endosomal pH in cells, and also increased nuclear uptake of doxorubicin. Pretreatment with pantoprazole increased tissue penetration of doxorubicin in MCCs. Pantoprazole improved doxorubicin distribution from blood vessels in solid tumors. Pantoprazole given before doxorubicin led to increased growth delay when given as single or multiple doses to mice bearing MCF7 xenografts. Conclusions: Use of pantoprazole to enhance the distribution and cytotoxicity of anticancer drugs in solid tumors might be a novel treatment strategy to improve their therapeutic index. Clin Cancer Res; 19(24); 6766–76. ©2013 AACR.


Journal of Pediatric Surgery | 2017

Breast milk-derived exosomes promote intestinal epithelial cell growth

Alison Hock; Hiromu Miyake; Bo Li; Carol Lee; Leonardo Ermini; Yuhki Koike; Yong Chen; Pekka Määttänen; Augusto Zani; Agostino Pierro

BACKGROUND Breast milk administration prevents necrotizing enterocolitis (NEC). However, the mechanism remains unclear. Exosomes are cell-derived vesicles highly present in human milk and regulate intercellular signaling, inflammation, and immune response. We hypothesized that milk-derived exosomes beneficially affect intestinal epithelial cells. METHODS Rat milk was collected, and exosomes were isolated using ExoQuick reagent and visualized by Nanoparticle Tracking Analysis. Protein was extracted from encapsulating exosomes, and concentration was measured. 2×104 intestinal epithelial cells (IEC-18) were treated for five hours with 0.5-μg/μl exosomes, an equal volume of exosome-free milk, or control solution (PBS). IEC-18 viability was measured using a colorimetric assay (MTT), and gene expression was analyzed by qRT-PCR. Data were compared using one-way ANOVA with Bonferroni post-test. RESULTS Rat milk was collected, and exosome isolation was confirmed. Compared to control, treatment with exosomes significantly increased IEC viability, proliferation, and stem cell activity (all p<0.05). However, administration of exosome-free milk had less significant effects. CONCLUSIONS Rat milk-derived exosomes promote IEC viability, enhance proliferation, and stimulate intestinal stem cell activity. These findings provide insight into the mechanism of action of breast milk in the intestines. Exosome administration is a promising prevention method for infants at risk of developing NEC when breastfeeding is not tolerated.


Oncogene | 2011

hTID-1 defines a novel regulator of c-Met Receptor signaling in renal cell carcinomas

Emma Copeland; Steve Balgobin; Carol Lee; Maria Rozakis-Adcock

The c-Met receptor tyrosine kinase (MetR) is frequently overexpressed and constitutively phosphorylated in a number of human malignancies. Activation of the receptor by its ligand, hepatocyte growth factor (HGF), leads to increased cell proliferation, motility, survival and disruption of adherens junctions. In this study, we show that hTid-1, a DNAJ/Hsp40 chaperone, represents a novel modulator of the MetR signaling pathway. hTid-1 is a co-chaperone of the Hsp70 family of proteins, and has been shown to regulate a number of cellular signaling proteins including several involved in tumorigenic and apoptotic pathways. In this study we demonstrate that hTid-1 binds to unphosphorylated MetR and becomes dissociated from the receptor upon HGF stimulation. Overexpression of the short form of hTid-1 (hTid-1S) in 786-0 renal clear cell carcinomas (RCCs) enhances MetR kinase activity leading to an increase in HGF-mediated cell migration with no discernible effect on cell proliferation. By contrast, knockdown of hTid-1 markedly impairs both the onset and amplitude of MetR phosphorylation in response to HGF without altering receptor protein levels. hTid-1-depleted cells display defective migratory properties, coincident with inhibition of ERK/MAP kinase and STAT3 pathways. Taken together, our findings denote hTid-1S as an essential regulatory component of MetR signaling. We propose that the binding of hTid-1S to MetR may stabilize the receptor in a ligand-competent state and this stabilizing function may influence conformational changes that take place during the catalytic cycle that promote kinase activation. Given the prevalence of HGF/MetR pathway activation in human cancers, targeted inhibition of hTid-1 may be a useful therapeutic in the management of MetR-dependent malignancies.


Journal of Pediatric Surgery | 2016

Intestinal epithelial cell injury is rescued by hydrogen sulfide

Bo Li; Augusto Zani; Zechariah Martin; Carol Lee; Elke Zani-Ruttenstock; Simon Eaton; Agostino Pierro

BACKGROUND/PURPOSE Oxidative stress is implicated in the pathogenesis of necrotizing enterocolitis (NEC). Hydrogen sulfide (H2S) has been reported to have a protective function against oxidative stress in the gut. We hypothesize that administration of H2S can help decrease intestinal epithelial cell injury in vitro. METHODS Intestinal epithelial cells (IEC-18) were treated with 200μM hydrogen peroxide (H2O2) for 21h. At 21h sodium hydrosulfide (NaHS), an H2S donor, was administered as a rescue treatment at two different concentrations: 0.1mM and 0.2mM. At 24h, cell viability was measured using a colorimetric assay (MTT). Oxidative stress was studied by glutathione peroxidase (GPx) activity and thiobarbituric acid reactive substances (TBARS). IL-6 and TNFα levels were tested to study inflammation. Data were presented as mean±SD and compared using one-way ANOVA with Bonferroni post-test. RESULTS Compared to control, H2O2-treated IEC-18 had reduced viability (p<0.01), lower GPx activity (p<0.01), higher TBARS levels (p<0.01), and increased IL6 and TNFα (p<0.001). Compared to H2O2-treated IEC-18, treatment with 0.2mM NaHS rescued viability (p<0.01), increased GPx activity (p<0.05), and reduced TBARS (p<0.01), IL6 and TNFα (p<0.001). CONCLUSIONS H2S successfully rescues epithelial cell damage induced by oxidative stress in vitro. This indicates that H2S could be a potential pharmacological intervention in conditions like NEC.


Journal of Pediatric Surgery | 2017

Intestinal epithelial injury induced by maternal separation is protected by hydrogen sulfide

Bo Li; Carol Lee; Zechariah Martin; Xinpei Li; Yuhki Koike; Alison Hock; Elke Zani-Ruttenstock; Augusto Zani; Agostino Pierro

PURPOSE Oxidative stress has been implicated in the pathogenesis of various neonatal diseases involving the intestine. Hydrogen sulfide (H2S) has been shown to protect against oxidative stress. We hypothesized that administration of sodium hydrosulfide (NaHS), an H2S donor, to neonatal mice can decrease the intestinal epithelial injury associated with maternal separation (MS). METHODS C57BL/6 mice received either intraperitoneal phosphate buffered saline (PBS; n=10) or NaHS (1mg/kg/day; n=10), followed by MS for 3h daily between postnatal day P5 and P9. Control neonatal mice were untreated and were not exposed to MS (n=10). Proximal colon was harvested and analyzed for crypt length, goblet cell number per crypt, oxidative stress and inflammation. Groups were compared using one-way ANOVA with Bonferroni post-test. RESULTS Compared to controls, MS+PBS mice had shorter crypt lengths, fewer goblet cells per crypt, reduced glutathione peroxidase activity, increased expression of thiobarbituric acid reactive substances and inducible nitric oxide synthase mRNA, as well as increased IL-6, TNFα and myeloperoxidase. Administration of NaHS significantly counteracted these negative effects of MS. CONCLUSIONS H2S protects the colon from the epithelial damage, oxidative stress and inflammation caused by maternal separation. This study provides insights on the pathogenesis of neonatal bowel diseases and indicates the potential for a pharmacological intervention to rescue the colonic epithelium. LEVEL OF EVIDENCE n/a - animal and laboratory study.


Journal of Pediatric Surgery | 2016

Endoplasmic reticulum stress is involved in the colonic epithelium damage induced by maternal separation

Bo Li; Augusto Zani; Carol Lee; Elke Zani-Ruttenstock; Ziyi Zhang; Xinpei Li; Wan Ip; Tanja Gonska; Agostino Pierro

BACKGROUND Maternal separation (MS) leads to intestinal barrier dysfunction in neonatal mice. Endoplasmic reticulum (ER) stress is associated with apoptosis and pro-inflammatory response induction. We hypothesized that MS induced gut damage is associated with ER stress and that administration of an ER stress inhibitor protects gut damage. METHODS C57BL/6 mice received intraperitoneal PBS (n=10) or Salubrinal (1mg/kg/day, n=10). MS was performed soon after treatment for 3h daily between P5 and P9. Ten untreated neonatal mice served as control. The colon was harvested on P9 and analyzed for ER stress markers (BiP, CHOP), apoptosis (CC3), goblet cell number per crypt and crypt length (Alcian blue, hematoxylin/eosin), and transcellular permeability (Ussing chamber). Groups were compared using one-way ANOVA with Bonferroni post-test. RESULTS Compared to controls, MS mice had higher relative protein expression of ER stress and apoptosis markers (p<0.05) and reduced goblet cell number per crypt and crypt length (p<0.001). In comparison to PBS mice, Salubrinal treated mice had higher goblet cell number (p<0.05), crypt length (p<0.001), and lower transcellular permeability (p<0.05). CONCLUSIONS Maternal separation induces ER stress and causes colon damage, but ER stress inhibitor protects morphology and permeability. This provides insights on bowel pathogenesis and potential novel treatments for diseases such as necrotizing enterocolitis.


Scientific Reports | 2017

Inhibition of corticotropin-releasing hormone receptor 1 and activation of receptor 2 protect against colonic injury and promote epithelium repair

Bo Li; Carol Lee; Tali Filler; Alison Hock; Richard Y. Wu; Qi Li; Shigang Chen; Yuhki Koike; Wan Ip; Lijun Chi; Elke Zani-Ruttenstock; Pekka Määttänen; Tanja Gonska; Paul Delgado-Olguin; Augusto Zani; Philip M. Sherman; Agostino Pierro

Maternal separation (MS) in neonates can lead to intestinal injury. MS in neonatal mice disrupts mucosal morphology, induces colonic inflammation and increases trans-cellular permeability. Several studies indicate that intestinal epithelial stem cells are capable of initiating gut repair in a variety of injury models but have not been reported in MS. The pathophysiology of MS-induced gut injury and subsequent repair remains unclear, but communication between the brain and gut contribute to MS-induced colonic injury. Corticotropin-releasing hormone (CRH) is one of the mediators involved in the brain-gut axis response to MS-induced damage. We investigated the roles of the CRH receptors, CRHR1 and CRHR2, in MS-induced intestinal injury and subsequent repair. To distinguish their specific roles in mucosal injury, we selectively blocked CRHR1 and CRHR2 with pharmacological antagonists. Our results show that in response to MS, CRHR1 mediates gut injury by promoting intestinal inflammation, increasing gut permeability, altering intestinal morphology, and modulating the intestinal microbiota. In contrast, CRHR2 activates intestinal stem cells and is important for gut repair. Thus, selectively blocking CRHR1 and promoting CRHR2 activity could prevent the development of intestinal injuries and enhance repair in the neonatal period when there is increased risk of intestinal injury such as necrotizing enterocolitis.

Collaboration


Dive into the Carol Lee's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bo Li

University of Toronto

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiromu Miyake

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Bo Li

University of Toronto

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yong Chen

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge