Network


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

Hotspot


Dive into the research topics where H. Charles Manning is active.

Publication


Featured researches published by H. Charles Manning.


Cancer Research | 2011

A Kinome-Wide Screen Identifies the Insulin/IGF-I Receptor Pathway as a Mechanism of Escape from Hormone Dependence in Breast Cancer

Emily M. Fox; Todd W. Miller; Justin M. Balko; Maria G. Kuba; Violeta Sanchez; R. Adam Smith; Shuying Liu; Ana M. Gonzalez-Angulo; Gordon B. Mills; Fei Ye; Yu Shyr; H. Charles Manning; Elizabeth Buck; Carlos L. Arteaga

Estrogen receptor α (ER)-positive breast cancers adapt to hormone deprivation and become resistant to antiestrogens. In this study, we sought to identify kinases essential for growth of ER(+) breast cancer cells resistant to long-term estrogen deprivation (LTED). A kinome-wide siRNA screen showed that the insulin receptor (InsR) is required for growth of MCF-7/LTED cells. Knockdown of InsR and/or insulin-like growth factor-I receptor (IGF-IR) inhibited growth of 3 of 4 LTED cell lines. Inhibition of InsR and IGF-IR with the dual tyrosine kinase inhibitor OSI-906 prevented the emergence of hormone-independent cells and tumors in vivo, inhibited parental and LTED cell growth and PI3K/AKT signaling, and suppressed growth of established MCF-7 xenografts in ovariectomized mice, whereas treatment with the neutralizing IGF-IR monoclonal antibody MAB391 was ineffective. Combined treatment with OSI-906 and the ER downregulator fulvestrant more effectively suppressed hormone-independent tumor growth than either drug alone. Finally, an insulin/IGF-I gene expression signature predicted recurrence-free survival in patients with ER(+) breast cancer treated with the antiestrogen tamoxifen. We conclude that therapeutic targeting of both InsR and IGF-IR should be more effective than targeting IGF-IR alone in abrogating resistance to endocrine therapy in breast cancer.


Clinical Cancer Research | 2009

Inhibition of Mammalian Target of Rapamycin Is Required for Optimal Antitumor Effect of HER2 Inhibitors against HER2-Overexpressing Cancer Cells

Todd W. Miller; James T. Forbes; Chirayu Shah; Shelby K. Wyatt; H. Charles Manning; Maria Graciela Olivares; Violeta Sanchez; Teresa C. Dugger; Nara de Matos Granja; Archana Narasanna; Rebecca S. Cook; J. Phillip Kennedy; Craig W. Lindsley; Carlos L. Arteaga

Purpose: A significant fraction of HER2-overexpressing breast cancers exhibit resistance to the HER2 antibody trastuzumab. Hyperactivity of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway confers trastuzumab resistance, and mammalian target of rapamycin (mTOR) is a major downstream effector of PI3K/AKT. Therefore, we examined whether mTOR inhibitors synergize with trastuzumab. Experimental Design: Immunocompetent mice bearing HER2+ mammary tumors were treated with trastuzumab, the mTOR inhibitor rapamycin, or the combination. Mice were imaged for tumor cell death using an optical Annexin-V probe and with [18F]FDG positron emission tomography. The signaling and growth effects of the mTOR inhibitor RAD001 on HER2+ cells treated with trastuzumab or lapatinib were evaluated. Results: Treatment of mice with trastuzumab plus rapamycin was more effective than single-agent treatments, inducing complete regression of 26 of 26 tumors. The combination induced tumor cell death (Annexin-V binding) and inhibited FDG uptake. Rapamycin inhibited mTOR and tumor cell proliferation as determined by phosphorylated S6 and Ki-67 immunohistochemistry, respectively. In culture, the combination of RAD001 plus trastuzumab inhibited cell growth more effectively than either drug alone. Trastuzumab partially decreased PI3K but not mTOR activity. Knockdown of TSC2 resulted in HER2-independent activation of mTOR and dampened the response to trastuzumab and lapatinib. Treatment with the HER2 inhibitor lapatinib decreased phosphorylated S6 and growth in TSC2-expressing cells but not in TSC2-knockdown cells. Conclusions: Inhibition of PI3K and mTOR are required for the growth-inhibitory effect of HER2 antagonists. These findings collectively support the combined use of trastuzumab and mTOR inhibitors for the treatment of HER2+ breast cancer. (Clin Cancer Res 2009;15(23):7266–76)


Clinical Cancer Research | 2008

Molecular Imaging of Therapeutic Response to Epidermal Growth Factor Receptor Blockade in Colorectal Cancer

H. Charles Manning; Nipun B. Merchant; A. Coe Foutch; John Virostko; Shelby K. Wyatt; Chirayu Shah; Eliot T. McKinley; Jingping Xie; Nathan Mutic; M. Kay Washington; Bonnie LaFleur; M. N. Tantawy; Todd E. Peterson; M. Sib Ansari; Ronald M. Baldwin; Mace L. Rothenberg; Darryl J. Bornhop; John C. Gore; Robert J. Coffey

Purpose: To evaluate noninvasive molecular imaging methods as correlative biomarkers of therapeutic efficacy of cetuximab in human colorectal cancer cell line xenografts grown in athymic nude mice. The correlation between molecular imaging and immunohistochemical analysis to quantify epidermal growth factor (EGF) binding, apoptosis, and proliferation was evaluated in treated and untreated tumor-bearing cohorts. Experimental Design: Optical imaging probes targeting EGF receptor (EGFR) expression (NIR800-EGF) and apoptosis (NIR700-Annexin V) were synthesized and evaluated in vitro and in vivo. Proliferation was assessed by 3′-[18F]fluoro-3′-deoxythymidine ([18F]FLT) positron emission tomography. Assessment of inhibition of EGFR signaling by cetuximab was accomplished by concomitant imaging of NIR800-EGF, NIR700-Annexin V, and [18F]FLT in cetuximab-sensitive (DiFi) and insensitive (HCT-116) human colorectal cancer cell line xenografts. Imaging results were validated by measurement of tumor size and immunohistochemical analysis of total and phosphorylated EGFR, caspase-3, and Ki-67 immediately following in vivo imaging. Results: NIR800-EGF accumulation in tumors reflected relative EGFR expression and EGFR occupancy by cetuximab. NIR700-Annexin V accumulation correlated with cetuximab-induced apoptosis as assessed by immunohistochemical staining of caspase-3. No significant difference in tumor proliferation was noted between treated and untreated animals by [18F]FLT positron emission tomography or Ki-67 immunohistochemistry. Conclusions: Molecular imaging can accurately assess EGF binding, proliferation, and apoptosis in human colorectal cancer xenografts. These imaging approaches may prove useful for serial, noninvasive monitoring of the biological effects of EGFR inhibition in preclinical studies. It is anticipated that these assays can be adapted for clinical use.


Magnetic Resonance Imaging | 2011

Magnetic resonance in the era of molecular imaging of cancer.

John C. Gore; H. Charles Manning; C. Chad Quarles; Kevin W. Waddell; Thomas E. Yankeelov

Magnetic resonance imaging (MRI) has played an important role in the diagnosis and management of cancer since it was first developed, but other modalities also continue to advance and provide complementary information on the status of tumors. In the future, there will be a major continuing role for noninvasive imaging in order to obtain information on the location and extent of cancer, as well as assessments of tissue characteristics that can monitor and predict treatment response and guide patient management. Developments are currently being undertaken that aim to provide improved imaging methods for the detection and evaluation of tumors, for identifying important characteristics of tumors such as the expression levels of cell surface receptors that may dictate what types of therapy will be effective and for evaluating their response to treatments. Molecular imaging techniques based mainly on radionuclide imaging can depict numerous, specific, cellular and molecular markers of disease and have unique potential to address important clinical and research challenges. In this review, we consider what continuing and evolving roles will be played by MRI in this era of molecular imaging. We discuss some of the challenges for MRI of detecting imaging agents that report on molecular events, but highlight also the ability of MRI to assess other features such as cell density, blood flow and metabolism which are not specific hallmarks of cancer but which reflect molecular changes. We discuss the future role of MRI in cancer and describe the use of selected quantitative imaging techniques for characterizing tumors that can be translated to clinical applications, particularly in the context of evaluating novel treatments.


Clinical Cancer Research | 2009

Imaging Biomarkers Predict Response to Anti-HER2 (ErbB2) Therapy in Preclinical Models of Breast Cancer

Chirayu Shah; Todd W. Miller; Shelby K. Wyatt; Eliot T. McKinley; Maria Graciela Olivares; Violeta Sanchez; Donald D. Nolting; Jason R. Buck; Ping Zhao; M. Sib Ansari; Ronald M. Baldwin; John C. Gore; Rachel Schiff; Carlos L. Arteaga; H. Charles Manning

Purpose: To evaluate noninvasive imaging methods as predictive biomarkers of response to trastuzumab in mouse models of HER2-overexpressing breast cancer. The correlation between tumor regression and molecular imaging of apoptosis, glucose metabolism, and cellular proliferation was evaluated longitudinally in responding and nonresponding tumor-bearing cohorts. Experimental Design: Mammary tumors from MMTV/HER2 transgenic female mice were transplanted into syngeneic female mice. BT474 human breast carcinoma cell line xenografts were grown in athymic nude mice. Tumor cell apoptosis (NIR700-Annexin V accumulation), glucose metabolism [2-deoxy-2-[18F]fluoro-d-glucose positron emission tomography ([18F]FDG-PET)], and proliferation [3′-[18F]fluoro-3′-deoxythymidine-PET ([18F]FLT-PET)] were evaluated throughout a biweekly trastuzumab regimen. Imaging metrics were validated by direct measurement of tumor size and immunohistochemical analysis of cleaved caspase-3, phosphorylated AKT, and Ki67. Results: NIR700-Annexin V accumulated significantly in trastuzumab-treated MMTV/HER2 and BT474 tumors that ultimately regressed but not in nonresponding or vehicle-treated tumors. Uptake of [18F]FDG was not affected by trastuzumab treatment in MMTV/HER2 or BT474 tumors. [18F]FLT-PET imaging predicted trastuzumab response in BT474 tumors but not in MMTV/HER2 tumors, which exhibited modest uptake of [18F]FLT. Close agreement was observed between imaging metrics and immunohistochemical analysis. Conclusions: Molecular imaging of apoptosis accurately predicts trastuzumab-induced regression of HER2+ tumors and may warrant clinical exploration to predict early response to neoadjuvant trastuzumab. Trastuzumab does not seem to alter glucose metabolism substantially enough to afford [18F]FDG-PET significant predictive value in this setting. Although promising in one preclinical model, further studies are required to determine the overall value of [18F]FLT-PET as a biomarker of response to trastuzumab in HER2+ breast cancer.


Magnetic Resonance Imaging | 2012

Simultaneous PET-MRI in Oncology: A Solution Looking for a Problem?

Thomas E. Yankeelov; Todd E. Peterson; Richard G. Abramson; David Garcia-Izquierdo; Lori R. Arlinghaus; Xia Li; Nkiruka C. Atuegwu; Ciprian Catana; H. Charles Manning; Zahi A. Fayad; John C. Gore

With the recent development of integrated positron emission tomography-magnetic resonance imaging (PET-MRI) scanners, new possibilities for quantitative molecular imaging of cancer are realized. However, the practical advantages and potential clinical benefits of the ability to record PET and MRI data simultaneously must be balanced against the substantial costs and other requirements of such devices. In this review, we highlight several of the key areas where integrated PET-MRI measurements, obtained simultaneously, are anticipated to have a significant impact on clinical and/or research studies. These areas include the use of MR-based motion corrections and/or a priori anatomical information for improved reconstruction of PET data, improved arterial input function characterization for PET kinetic modeling, the use of dual-modality contrast agents, and patient comfort and practical convenience. For widespread acceptance, a compelling case could be made if the combination of quantitative MRI and specific PET biomarkers significantly improves our ability to assess tumor status and response to therapy, and some likely candidates are now emerging. We consider the relative advantages and disadvantages afforded by PET-MRI and summarize current opinions and evidence as to the likely value of PET-MRI in the management of cancer.


Oncology Reports | 2012

Evaluation of CD44 and CD133 as cancer stem cell markers for colorectal cancer

Chunxia Wang; Jingping Xie; Jiasong Guo; H. Charles Manning; John C. Gore; Ning Guo

Colorectal cancer (CRC) is a major cause of cancer-related mortality in the world. Recently, a number of studies have demonstrated that cancer stem cells (CSCs) present in colorectal cancer tissues, are responsible for resistance to conventional therapies. Therefore, effective recognition of CSCs is of great importance. In the present study, to explore the potential characterizations of CSCs by the expression of specific cell surface markers such as CD133 and CD44, we screened six CRC cell lines using western blotting, immunofluorescence and flow cytometry. SW620, one of the six cell lines analyzed, was sorted into four subpopulations by fluorescence activated cell sorting (FACS). The capability of colony formation, proliferation rate, apoptosis, drug resistance, as well as their migratory and invasion potential were detected. The results revealed that the combination of CD44 and CD133 correlates with the features of CSCs in SW620 cells. CD44 positive cells showed more robust colony formation, higher proliferation, less spontaneous apoptosis, a higher resistance to drug-induced cell death, and were enriched after drug treatment. Among CD44 positive SW620 cells, the CD133 negative subpopulation was more migratory and invasive, which means that CD44+CD133− correlates with most of features proposed for CSCs. Overall, the data presented herein showed that CRCs have a wide range of expression for CD44 and CD133; it is unlikely the CSCs can be characterized by any single marker or the same set of markers for all colon cancer cells. For SW620 cells, the CSCs are likely represented by the CD44+CD133− surface marker. This finding of CSC markers represented by one positive and one negative is in line with CSCs in other tumors, such as CD34+CD38− for acute myeloid leukemia; CD44+CD24− for breast and pancreatic tumors. The absence of surface molecule(s) on CSCs will make it even more difficult to track and target this group of minority cells.


PLOS ONE | 2013

Limits of [18F]-FLT PET as a Biomarker of Proliferation in Oncology

Eliot T. McKinley; Gregory D. Ayers; R. Adam Smith; Samir Saleh; Ping Zhao; Mary Kay Washington; Robert J. Coffey; H. Charles Manning

Background Non-invasive imaging biomarkers of cellular proliferation hold great promise for quantifying response to personalized medicine in oncology. An emerging approach to assess tumor proliferation utilizes the positron emission tomography (PET) tracer 3’-deoxy-3’[18F]-fluorothymidine, [18F]-FLT. Though several studies have associated serial changes in [18F]-FLT-PET with elements of therapeutic response, the degree to which [18F]-FLT-PET quantitatively reflects proliferative index has been continuously debated for more that a decade. The goal of this study was to elucidate quantitative relationships between [18F]-FLT-PET and cellular metrics of proliferation in treatment naïve human cell line xenografts commonly employed in cancer research. Methods and Findings [18F]-FLT-PET was conducted in human cancer xenograft-bearing mice. Quantitative relationships between PET, thymidine kinase 1 (TK1) protein levels and immunostaining for proliferation markers (Ki67, TK1, PCNA) were evaluated using imaging-matched tumor specimens. Overall, we determined that [18F]-FLT-PET reflects TK1 protein levels, yet the cell cycle specificity of TK1 expression and the extent to which tumors utilize thymidine salvage for DNA synthesis decouple [18F]-FLT-PET data from standard estimates of proliferative index. Conclusions Our findings illustrate that [18F]-FLT-PET reflects tumor proliferation as a function of thymidine salvage pathway utilization. Unlike more general proliferation markers, such as Ki67, [18F]-FLT PET reflects proliferative indices to variable and potentially unreliable extents. [18F]-FLT-PET cannot discriminate moderately proliferative, thymidine salvage-driven tumors from those of high proliferative index that rely primarily upon de novo thymidine synthesis. Accordingly, the magnitude of [18F]-FLT uptake should not be considered a surrogate of proliferative index. These data rationalize the diversity of [18F]-FLT-PET correlative results previously reported and suggest future best-practices when [18F]-FLT-PET is employed in oncology.


Journal of Ultrasound in Medicine | 2010

Volume of preclinical xenograft tumors is more accurately assessed by ultrasound imaging than manual caliper measurements

Gregory D. Ayers; Eliot T. McKinley; Ping Zhao; Jordan M. Fritz; Rebecca E. Metry; Brenton C. Deal; Katrina M. Adlerz; Robert J. Coffey; H. Charles Manning

Objective. The volume of subcutaneous xenograft tumors is an important metric of disease progression and response to therapy in preclinical drug development. Noninvasive imaging technologies suitable for measuring xenograft volume are increasingly available, yet manual calipers, which are susceptible to inaccuracy and bias, are routinely used. The goal of this study was to quantify and compare the accuracy, precision, and inter‐rater variability of xenograft tumor volume assessment by caliper measurements and ultrasound imaging. Methods. Subcutaneous xenograft tumors derived from human colorectal cancer cell lines (DLD1 and SW620) were generated in athymic nude mice. Experienced independent reviewers segmented 3‐dimensional ultrasound data sets and collected manual caliper measurements resulting in tumor volumes. Imaging‐ and caliper‐derived volumes were compared with the tumor mass, the reference standard, determined after resection. Bias, precision, and inter‐rater differences were estimated for each mouse among reviewers. Bootstrapping was used to estimate mean and confidence intervals of variance components, intraclass correlation coefficients (ICCs), and confidence intervals for each source of variation. Results. The average deviation from the true volume and inter‐rater differences were significantly lower for ultrasound volumes compared with caliper volumes (P = .0005 and .001, respectively). Reviewer ICCs for ultrasound and caliper measurements were similarly low (1%), yet caliper volume variance was 1.3‐fold higher than for ultrasound. Conclusions. Ultrasound imaging more accurately, precisely, and reproducibly reflects xenograft tumor volume than caliper measurements. These data suggest that preclinical studies using the xenograft burden as a surrogate end point measured by ultrasound imaging require up to 30% fewer animals to reach statistical significance compared with analogous studies using caliper measurements.


Gastroenterology | 2013

Aurora kinase A promotes inflammation and tumorigenesis in mice and human gastric neoplasia.

Ahmed M. Katsha; Mohammed Soutto; Vikas Sehdev; DunFa Peng; M. Kay Washington; M. Blanca Piazuelo; M. N. Tantawy; H. Charles Manning; Pengcheng Lu; Yu Shyr; Jeffrey Ecsedy; Abbes Belkhiri; Wael El–Rifai

BACKGROUND & AIMS Chronic inflammation contributes to the pathogenesis of gastric tumorigenesis. The aurora kinase A (AURKA) gene is frequently amplified and overexpressed in gastrointestinal cancers. We investigated the roles of AURKA in inflammation and gastric tumorigenesis. METHODS We used quantitative real-time reverse transcription polymerase chain reaction, immunofluorescence, immunohistochemistry, luciferase reporter, immunoblot, co-immunoprecipitation, and in vitro kinase assays to analyze AGS and MKN28 gastric cancer cells. We also analyzed Tff1(-/-) mice, growth of tumor xenografts, and human tissues. RESULTS We correlated increased expression of AURKA with increased levels of tumor necrosis factor-α and inflammation in the gastric mucosa of Tff1(-/-) mice (r = 0.62; P = .0001). MLN8237, an investigational small-molecule selective inhibitor of AURKA, reduced nuclear staining of nuclear factor-κB (NF-κB) p65 in human gastric cancer samples and mouse epithelial cells, suppressed NF-κB reporter activity, and reduced expression of NF-κB target genes that regulate inflammation and cell survival. Inhibition of AURKA also reduced growth of xenograft tumors from human gastric cancer cells in mice and reversed the development of gastric tumors in Tff1(-/-) mice. AURKA was found to regulate NF-κB activity by binding directly and phosphorylating IκBα in cells. Premalignant and malignant lesions from the gastric mucosa of patients had increased levels of AURKA protein and nuclear NF-κB, compared with healthy gastric tissue. CONCLUSIONS In analyses of gastric cancer cell lines, human tissue samples, and mouse models, we found AURKA to be up-regulated during chronic inflammation to promote activation of NF-κB and tumorigenesis. AURKA inhibitors might be developed as therapeutic agents for gastric cancer.

Collaboration


Dive into the H. Charles Manning's collaboration.

Top Co-Authors

Avatar

Robert J. Coffey

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eliot T. McKinley

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael L. Nickels

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ping Zhao

Vanderbilt University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M. Kay Washington

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge