3rd Annual Postdoctoral Science Symposium
August 1, 2013
Organized by the MD Anderson Postdoctoral Association
Sponsored by MD Anderson Cancer Center Alumni & Faculty
Associations and the Department of Trainee & Alumni Affairs
Symposium Agenda
August 1
Morning Sessions
9:00 – 9:15
Welcome Address
APSS Organizing Committee
9:15 – 10:15
Combinatorial mapping of the human
vasculature: towards ligand-directed therapies
and imaging agents
Renata Pasqualini , Ph.D., Professor,
Department of Genitourinary Medical
Oncology , MD Anderson Cancer Center
10:15 – 10:35
Personalized canine cancer therapy
Colleen O’Connor, Ph.D., Postdoctoral Fellow,
Department of Pediatrics Research, MD
Anderson Cancer Center
10:35 – 10:55
Mesenchymal stem cells promote skin invasion
and metastasis in inflammatory breast cancer
through an EGFR dependent mechanism
Lara C. Alvarez de Lacerda, Ph.D.,
Postdoctoral Fellow, Department of Radiation
Oncology, MD Anderson Cancer Center
10:55 – 11:15
Integrated analyses identify a master microRNA
regulatory network for the mesenchymal
subtype in serous ovarian cancer
Da Yang, Ph.D. Postdoctoral Fellow,
Department of Pathology, MD Anderson Center
11:15 – 12:30
Lunch/Poster Session
12:30 – 1:30
Breakout Sessions
1.
2.
3.
4.
5.
Reverse Phase Protein Microarrays (RPPA): Yiling Lu, M.D.
Bioinformatics: Nicholas Navin, Ph.D.
Cytometry by Time-Of-Flight (Cy-TOF): Jared Burks, Ph.D.
Non-coding RNA: Chang-Gong Liu, Ph.D.
Small Animal Imaging: James A. Bankson, Ph.D.
Classroom
S1.8331
Onstead
S3.8003
S5.8004
S5.8005
August 1
Afternoon Sessions
1:45 – 2:45
Oncogenome-selective vulnerabilities in lung
adenocarcinoma
Michael White, Ph.D., Professor, Department
of Cell Biology , UT Southwestern Medical
Center
2:45 – 3:05
Tobacco-related cancer disparities: the role
of health literacy
Diana W. Stewart, Ph.D., Instructor
Department of Health Disparities Research, MD
Anderson Cancer Center
3:05 – 3:25
Development of a force assay to monitor the
mobility of cancer cells
Varun Sreenivasan, Ph.D., Postdoctoral
Associate, Department of Otolaryngology,
Baylor College of Medicine
3:25 – 3:45
Dynamic changes in protein abundance and
cell cycle profiles revealed by profiling of human
embryonic stem cells during differentiation at
the single cell level by mass cytometry
Ryan McCarthy, Ph.D. Postdoctoral Fellow,
Department of Biochemistry, MD Anderson
Cancer Center
3:45 – 4:00
Coffee Break
4:00 – 5:00
Keynote
Normal and neoplastic stem cells
Irving L. Weissman, Professor , Department
of Pathology and Developmental Biology,
Stanford University
5:00 – 5:05
Closing Remarks
5:05 – 6:30
Reception
Abstracts
Renata Pasqualini, Ph.D.
Buchanan and Seeger Professor
of Medicine and Experimental Diagnostic Imaging
Department of Genitourinary Medical Oncology
MD Anderson Cancer Center
Education:
University of São Paulo, Brazil
BS
University of São Paulo, Brazil
PhD
The Children’s Hospital Harvard Medical School Postdoc
Dana Farber Cancer Institute
Postdoc
The Burnham Institute, La Jolla, CA
Sr. Fellow
1986
1990
1991
1994
1996
Notable Honors/Awards:
2006
Living Legend Faculty Achievement Award, UTMDACC
2010
Marcus Foundation Award, Marcus Foundation
2007
The Top 400 Inventors for 99% of patents filed since 1946
2009
Potu N. Rao Award for Excellence in Basic Science
2010
Distinguished Women in Science, Bio Houston
2011
The Lombroso Achievement Award, Weizmann Institute
of Science
2012
AACR – The Carcinoid Foundation Investigator Award
Research Interest:
• Human Vascular Mapping Project
• Ligand-directed Targeting, Nanotechnology
& Molecular-genetic Imaging
• Fingerprinting the Repertoire of
• Patient-derived Antibodies
• Obesity Reversal through Vascular
Targeting
• Hybridoma and hybridoma-free
Generation and Production of Antibodies
Email: rpasqual@mdanderson.org
Combinatorial mapping of the human vasculature:
towards ligand-directed therapies and imaging agents
As a matter of record, Drs. Arap and Pasqualini have a long-standing
collaboration and have led a joint laboratory since October 1999. Our
research group currently includes Ph.D. or M.D./Ph.D. students,
postdoctoral fellows, faculty (assistant and associate professors),
research technicians, and administrative staff members. Our central
working hypothesis is that differential protein expression in the
human vascular endothelium associated with normal or diseased
tissues offers the potential for developing novel diagnostic, imaging,
and therapeutic strategies. In essence, our research program uses
combinatorial library selection (peptide- and antibody-based) to
discover, validate, and exploit the vascular biochemical diversity of
endothelial cell surfaces that can be subsequently exploited towards
a new vascular-targeted pharmacology. Such targeting technologies
may lead to the development of ligand-directed, clinically applicable
agents for the treatment of cancer patients.
Translational
applications, such as first-in-man clinical trials, which have now
started within MDACC, will ultimately determine the value of this
strategy. Indeed, the Food and Drug Administration (FDA) granted
“safe-to-proceed” status for our first vascular-targeted IND in 2009.
A first-in-human study has already been completed with several
patients enrolled in this MDACC-sponsored clinical trial. A second
IND was filed mid-November 2010; subsequent questions from the
FDA were addressed though extensive additional experimentation,
and the second Phase I study is now active. Two other drugs are in
pre-IND stage, with several others in the pre-clinical laboratory
phase. Long-term, the broader vision of our research is to generate a
large-scale receptor map of the human vasculature
Colleen O’Connor, Ph.D.
Postdoctoral Fellow
Department of Pediatrics Research
MD Anderson Cancer Center
Education:
University of Kentucky, Lexington, Kentucky
PhD: Toxicology with Specialization in Immunology
University of Miami, Florida, Coral Gables, Florida
BS Double Major: Marine Science and Biology Minor: Chemistry
Notable Honors/Awards:
The Ben F. Love Fellowship in Innovative Cancer Therapies from the
Walter Frank Foundation
Trainee of the Quarter Winner (For Meritorious Work)
Janice Davis Singletary Lymphoma Fellowship
Trainee of the Quarter Nominee
University of Texas MD Anderson Cancer Center
Immunology Poster Contest Winner
University of Texas MD Anderson Cancer Center Immunology Retreat
Graduate Academic Year Fellowship
University of Kentucky
Henry King Stanford Academic Scholarship
University of Miami, Florida
TV Coverage of Scientific Reports Paper Results
ABC World News with Diane Sawyer
KPRC NBC Houston Evening News
Research Interest: Hematology, Oncology, Cell
Therapy, Gene Therapy, Comparative Oncology,
Immunology, T cells, Bone Marrow Transplants,
Biomarker Development, Nanostring,
Single Cell Analysis, MRI Imaging, Clinical Trials
(Human and Canine)
Email: coconnor@mdanderson.org
Personalized Canine cancer therapy
Personalized cancer therapy has come into focus in recent years with
newly developed immunotherapies complementing conventional
treatment modalities. However, the transition from bench to bedside
still faces significant headwinds. Mouse models often fall short in
predicting the efficacies and toxicities associated with T-cell therapy.
Thus, new models which more closely mimic the human condition are
urgently needed. The companion canine spontaneously develops
many of the same cancers found in humans, despite a functionally
intact immune system. Canine cancers mirror human malignancies
with similarities in mutagenesis, tumorigenic responses to
environmental toxins, documented oncogenic predispositions, and
intra-species genetic diversity. These parallels, coupled with their
relatively large body mass, afford a unique therapeutic model to
study human cancers. NHL is the most common malignancy of the
domestic dog, accounting for up to 25% of canine neoplasm and is
rarely curable. Similar to humans, the majority of NHL (60-80%) in
dogs arise from malignant B cells, most commonly diagnosed as Stage
III or IV at presentation. New approaches are needed to improve
patient survival beyond what combination chemotherapy (CHOP) can
currently achieve. Biological therapies, such as CAR+ or CARneg T-cell
therapy, can target disease by employing mechanisms independent of
chemo-radio-therapies. Clinical observations reveal that an
augmented pace of T-cell recovery after chemotherapy correlates
with improved tumor-free survival, suggesting the add-back of T cells
after chemotherapy may improve outcomes. To evaluate adoptive
immunotherapy treatment for B-lineage NHL, we expanded T cells
from client-owned canines diagnosed with NHL on artificial antigen
presenting cells (aAPC) in the presence of human interleukin (IL)-2
and IL-21. Graded doses of autologous T cells were infused after
CHOP chemotherapy and persisted for 49 days, homed to tumor sites,
and significantly improved survival. Serum thymidine kinase changes
predicted T-cell engraftment, while anti-tumor effects correlated with
neutrophil-to-lymphocyte ratios and granzyme B expression in
manufactured T cells. Our study supports the use of add-back T cells
after chemotherapy in NHL patients to improve prognosis and
survival.
Lara C. Alvarez de Lacerda, Ph.D.
Postdoctoral Fellow
Department of Radiation Oncology
MD Anderson Cancer Center
Education:
University of Coimbra (Portugal) - PharmD in Pharmaceutical Sciences
University of London (United Kingdom) – PhD in
Nanotechnology/Drug Delivery Systems
Notable Honors/Awards:
Postdoctoral Fellowship - Susan G. Komen for the Cure® Research
Program, USA
Postdoctoral Fellowship - Foundation for Science and Technology,
Ministry of Science and Education, Portugal
PhD Studentship - Foundation for Science and Technology, Ministry
of Science and Education, Portugal
AACR Scholar-In-Training Award - 2012 CTRC-AACR San Antonio
Breast Cancer Symposium, Susan G. Komen for the Cure®, USA
Trainee of the Quarter Award - Department of Trainee and Alumni
Affairs, University of Texas MD Anderson Cancer Center, USA
Research Interest:
My research project focuses on the role of the tumor
microenvironment in regulating the equilibrium among breast cancer
stem cells, progenitors, and differentiated cells. Additionally, I also
have interest in new approaches to better understand and treat
inflammatory breast cancer. My long-term
career goal is to develop targeted therapeutic
strategies that are specifically designed to
overcome disease recurrence by effectively
treating breast cancer stem cell populations.
Email: lcalvarez@mdanderson.org
Mesenchymal stem cells promote skin invasion and
metastasis in inflammatory breast cancer through an
EGFR dependent mechanism
Inflammatory breast cancer (IBC) is an aggressive variant of breast
cancer characterized by blocked breast lymphatics and clinically
apparent involvement of the skin resulting in erythema, swelling and
a high rate of resistance to therapy. Tumor-skin invasion in IBC
patients, despite being a rare event, is an event that develops very
rapidly, can spread throughout the upper body and ultimately results
in breast cancer death. Very little is known about this event and how
to prevent it. Mesenchymal stem cells (MSC) are multipotent
progenitor cells which are found in normal tissues, including the bone
marrow and adipose tissue. We hypothesized that MSC are critical
mediators of IBC clinical phenotype and represent targets for
treatment.
Our data shows a significant difference in the tumor-skin invasion,
between groups of mice injected with IBC cells SUM149 with or
without MSC. Furthermore, co-injection of MSC with IBC cells
significantly increased the spontaneous development of metastasis
after primary tumor resection. In vitro, MSC co-culture was
associated with increased EGFR signaling, and tumors in MSC coinjected animals had increased p-EGFR staining by IHC. Remarkably,
we found that Erlotinib inhibited the rapid development of
metastasis promoted by MSC co-injection, independently of primary
tumor growth rate.
In conclusion, MSC increased tumor-skin invasion and metastasis
development in an IBC pre-clinical model independent of growth
rate. This mechanism was due to increased EGFR signaling promoted
by MSC. Thus MSC-cancer cells interaction can be abolished by
Erlotinib. Further studies are needed to understand the role of MSC
on the IBC cell involvement in skin contributing to the aggressiveness
of this deadly disease.
Da Yang, Ph.D.
Postdoctoral Fellow
Department of Pathology
MD Anderson Cancer Center
Education:
Harbin Medical University
Harbin Medical University
Bachelor in Medicine 2005
Ph.D.
2009
Bioinformatics and Pharmacology
Notable Honors/Awards:
Odyssey Fellowship of The University of Texas MD Anderson Cancer
Center (2011)
Best Publication Award of The University of Texas MD Anderson
Cancer Center (2012)
The Diane Denson Tobola Fellowship in Ovarian Cancer Research
(2012)
The Harold C. and Mary L. Daily Endowment Fund Fellowship (2012)
The Diane Denson Tobola Fellowship in Ovarian Cancer Research
(2013)
Bristol-Myers Squibb Award in Clinical/Translational Research (2013)
Research Interest:
Cancer Genomics
Next generation sequencing analysis
miRNA network analysis
Personalized Medicine
Email: DYang3@mdanderson.org
Integrated analyses identify a master microRNA
regulatory network for the mesenchymal
subtype in serous ovarian cancer
Epithelial ovarian cancer (OvCa) is one of the most lethal
malignancies in women. Identification of molecular subtypes and
corresponding molecular driver events is critical for the development
of new therapies for OvCa patients. Here, we develop a
computational pipeline (Master mIRna Analysis for Cancer moLecular
subtype, MIRACLE) that aims to delineate the driver events,
especially driver miRNAs. Applying the MIRACLE pipeline to 459
serous OvCa cases in TCGA database revealed a core miRNAregulatory network for mesenchymal phenotype of OvCa. Using
genes in the regulatory network, we further characterized a robust
OvCa mesenchymal subtype significantly associated with poor
survival in 459 serous OvCa cases from TCGA and 560 cases from
three independent patient cohorts.
In the miRNA-regulatory network, eight key miRNAs were
predicted to regulate 89% of the targets. Among them are wellestablished EMT inhibitors, such as miR-200 family. In the follow-up
in vitro and in vivo experiments, we further demonstrated that miR506, whose function was previously uncharacterized, as a novel EMT
inhibitor by targeting SNAI2. Transfection of miR-506 augmented Ecadherin expression, inhibited cell migration and invasion, and
prevented TGFβ-induced EMT, while force expression of SNAI2 can
abolish miR-506’s effect. In human OvCa tissue, miR-506 expression
was correlated with decreased SNAI2, elevated E-cadherin, and
beneficial prognosis. To further explore the therapeutic efficacy of
miR-506 in OvCa, we treated OvCa orthotopic mouse models with
nanoparticle-incorporated miR-506. MiR-506 delivery can suppress
EMT and tumor growth in vivo, suggesting miR-506 may serve as a
potential therapeutic tool for OvCa patients.
Michael White, Ph.D.
Professor, Cell Biology
Associate Director, Basic Science, Simmons Cancer
Center
Education:
U. of North Carolina
Chapel Hill
CSHL
Ph.D.
Postdoctoral Fellow
1989-1992
1992-1995
Notable Honors/Awards:
Hortense and Morton Sanger Professorship in Oncology
The Sherry Wigley Crow Cancer Research Endowed Chair in Honor of
Robert Lewis Kirby, M.D.
Grant A. Dove Chair for Research in Oncology
Research Interest:
Cancer cell biology
Signal transduction
http://www.utsouthwestern.edu/labs/white-michael/
Email: michael.white@utsouthwestern.edu
Oncogenome-selective vulnerabilities in lung
adenocarcinoma
Diversity in the genetic lesions that drive cancer initiation and
progression is extreme. This diversity exists not only among tumors
from different patients, but also among cancer cells within the same
patient. This nefarious complexity is, in large measure, responsible
for the capacity of this disease to evade current best efforts for
effective therapy. “Personalized medicine” has been proposed in
response to this conundrum as a mechanism to tailor cancer
treatment to a specific tumor’s genetic and epigenetic characteristics.
However, selection of appropriate treatment is dramatically limited
by the paucity of appropriate drugs and by the difficulty of linking
treatment options to the appropriate patients. The challenge is to
identify authentic intervention targets for development of an
appropriately diverse cohort of therapies to contend with disease
heterogeneity. We are addressing this challenge by a focused
investigation of common vulnerabilities that arise as a consequence
of oncogene expression and tumor evolution. Here we will describe a
cancer intervention discovery pipeline using parallel genetic and
chemical perturbations within an extensive panel of cell lines
representative of the molecular lesions detected in lung cancer by
national and international cancer genome sequencing efforts. We
have found that current first line targeted therapies are discoverable
within this panel together with the enrollment biomarkers required
to stratify patient treatment regimens. Further, we have found that
new genetic and chemical vulnerabilities can be revealed that are
linked to recurrent mutations in lung cancer patients that are not
currently “actionable”. We are leveraging this approach to stratify
lung cancer subtypes and elaborate intervention targets that are
linked to those subtypes by robust molecular discriminators.
Diana W. Stewart, Ph.D.
Instructor
Department of Health Disparities Research
MD Anderson Cancer Center
Education:
2005
The University of Kansas, Lawrence, KS, B.A., Psychology,
English
2008
Louisiana State University, Baton Rouge, LA, M.A., Clinical
Psychology
2011
Louisiana State University, Baton Rouge, LA, Ph.D.,
Clinical Psychology, Minor in Behavioral Medicine
2010-2011 Clinical Internship, Behavioral Medicine Track, Alpert
Medical School at Brown University, Providence, RI
2011-2013 Cancer Prevention Research Postdoctoral Fellowship (NCI
R25T CA57730), Department of Health Disparities
Research, University of Texas MD Anderson Cancer
Center, Houston, TX
Notable Honors/Awards:
2011-present Platinum-level Trainee, Community Based
Participatory Research Trainee, Latinos Contra el Cancer Community
Networks Program Center, National Cancer Institute
2011-2013 Recipient, Competitive R25-T Postdoctoral Fellowship in
Cancer Prevention Research, NCI R25-T CA57730
2012-2014 Recipient, National Institutes of Health, Health
Disparities Research Loan Repayment Program
2013 Recipient, Mentored Faculty Fellowship, Duncan Family
Institute for Cancer Prevention and Risk Assessment
Research Interests:
My research program focuses on cancer prevention
and health promotion in underserved populations.
My specific interests and expertise are in tobacco
cessation for marginalized populations.
Email: dwstewart@mdanderson.org
Tobacco-related cancer disparities:
The role of health literacy
Compared to individuals in the general population, racial/ethnic
minorities and individuals with low socioeconomic status (SES) have
higher smoking rates and greater difficulty quitting smoking. Thus,
smoking has a striking impact on cancer disparities and mortality for
individuals from these populations. Low health literacy is one factor
that may be uniquely associated with poor cessation outcomes. This
presentation will describe three studies investigating relations
between health literacy and tobacco use and cessation, and potential
mechanisms of this association, among low-SES, racially/ethnically
diverse smokers. Implications and specific aims for future research
will be discussed.
Varun Sreenivasan, Ph.D.
Postdoctoral Associate
Department of Otolaryngology
Baylor College of Medicine
Education:
Macquarie University, Australia.
Ph.D. Physics
2012
Cochin University of
Science and Technology, India.
M.Sc. Photonics
2008
Notable Honors/Awards:
2012
2011
MQ BioFocus vacation scholarship, Sydney
Macquarie university post-graduate funding with Vice
Chancellor's commendation, Sydney
2011
Macquarie university post-graduate funding with Vice
Chancellor's commendation, Sydney
2008-2012 Macquarie postgraduate research scholarship, Sydney
2007
Summer research fellowship, Indian academy of sciences,
Chennai
Research Interests:
Molecular & cellular biophysics, cell signaling and motility, biological
& biomedical imaging, high resolution & single molecule optical
microscopy and nanoparticles.
Email: varun.sreenivasan@outlook.com
varun.sreenivasan@bcm.edu
Development of a force assay to monitor the mobility
of cancer cells
Cancer cells acquire motility by remodeling their cytoskeleton to form
migratory structures (e.g., filopodia). Such transformations are
dominated by F-actin re-assembly within the cytoplasm. These
processes produce protrusive forces (~1 pN) allowing cells to migrate
from their primary tumor site into surrounding stroma. Our goal is to
elucidate the motility of cancer cells in vitro, by developing an assay
to measure the protrusive forces and their time course.
An optical trap-based biosensor with a stiffness of 9.2×10-4 pN.nm1.mW-1 and time constant of 23 s-1.mW-1 is built. The trap levitates a
2-μm polystyrene bead 7 μm above the substrate. The bead
undergoes a displacement from its equilibrium position in proportion
to the force acting. The bead also serves as a handle to initiate the
formation of the filopodium-like structure. We use a metastatic
squamous cell carcinoma (HN31) of the pharynx, because they exhibit
constitutively active Rho-GTPases and can sustain actin remodeling.
The experiment involves bringing a cell in contact with the bead to
facilitate membrane adhesion and then pulling it apart to rupture
membrane-cytoskeletal bonds to initiate the formation of the
filopodium-like structure. Results show the maximum force required
for bond rupture is 70 pN, and decreases to 25 pN as the formed
structure reaches a stationary state. Riding upon the stationary value
are force fluctuations, which we associate with F-actin dynamics
occurring within the cytoplasm near the tip of the structure. We
suggest filopodium-like structures do form and we can monitor Factin dynamics at its leading edge.
Ryan McCarthy, Ph.D.
Postdoctoral Fellow
Department of Biochemistry
MD Anderson Cancer Center
Education:
Harvey Mudd College B.S. Mathematical Biology
University of Georgia Ph.D. Plant Biology
MD Anderson
Postdoctoral Fellow
2007
2012
Present
Notable Honors/Awards:
2013
Molecular Genetics of Cancer Training Program
2008/2010 Postdoctoral Fellowship, University of Georgia Plant
Biology Department Palfrey Grant, Harvey Mudd
2007
College Graduated with Distinction,
2007
Sigma Xi Inductee
2003-2007 National Merit Four Year Undergraduate Scholarship
Research Interest:
My research interests focus on the regulation of differentiation and
cell identity of human embryonic stem cells. I am particularly
interested in investigating the multiple roles of p53 as a regulator of
cell cycle, repressor of pluripotency factors and promoter of
differentiation. To better understand this process I utilize mass
cytometry for highly parametric profiling of cell populations
undergoing differentiation.
Email: rlmccarthy@mdanderson.org
Dynamic changes in protein abundance and cell cycle profiles
revealed by profiling of human embryonic stem cells during
differentiation at the single cell level by mass cytometry
Development hinges on the capability of pluripotent embryonic stem
cells (ESCs) to continuously self-renew and provide a source of
somatic cells through differentiation. ESCs are held in a pluripotent
state by core transcription factors including OCT4 and SOX2. During
early differentiation of human ESCs (hESCs) p53 actively promotes
differentiation by inducing cell cycle arrest, repressing pluripotency
factors and activating lineage-specific developmental genes. Our
understanding of the changes hESCs undergo during differentiation is
limited by the inherent heterogeneity of cellular states. To overcome
this we employed cytometry by time of flight mass spectrometry
(CyTOF) to quantify 30+ distinct protein abundances at the single cell
level, changing the heterogeneity of differentiation from an
impediment into an asset. Using CyTOF to quantify changes in cell
cycle distribution, histone modifications, pluripotency and lineagespecific marker proteins during differentiation we performed novel
computational analyses to define a continuum of cell states present
during differentiation and pinpointed key regulatory events. Upon
differentiation to either ectodermal or endodermal/mesodermal
lineages we observed p53 induction and accumulation of cells in G1
phase followed by the appearance of lineage-specific markers. p53
depletion increased proliferation and pluripotency factor abundance
in self-renewing hESCs and delayed but did not prevent
differentiation of hESCs. Additionally, G1 arrest was diminished but
not abolished during differentiation indicating compensation,
potentially by p53 homologs p63 and p73 or through p53
independent ERK-mediated cell cycle arrest pathways. Our findings
improve our understanding of p53’s non-apoptotic role in
differentiation and establish a unique system for further investigating
hESC differentiation.
Irving Weissman, M.D.
Keynote Speaker
Professor
Stanford University School of Medicine
Education:
Montana State College (now University)
Stanford University
B.S.
M.D.
1961
1965
Notable Honors/Awards:
Among others: The Robert Koch Award, Pasarow Award for
Outstanding Contribution to Cancer Biology, Election to the National
Academy of Sciences; California Scientist of the Year (2001); Jessie
Stevenson Kovalenko Medal, National Academy of Sciences Council;
Max Delbruck Award; Honorary Doctorates, from Montana State
University, Columbia University, Mt. Sinai School of Medicine.
Research Interest:
Dr. Weissman is an expert in the field of hematopoiesis (bloodformation), leukemia, and hematopoietic stem cells. The main focus
of his work for many years has been the purification, biology,
transplantation, and evolution of stem cells. His laboratory was first
to identify and isolate the blood-forming stem cell (hematopoietic
stem cell) in mice and humans, and to define many of the stages of
development between these stem cells and their differentiated
progeny. In recent years, Dr. Weissman has
studied the potential of CD47 as a
therapeutic target in multiple cancers and
identified cancer stem cells in various
hematologic and solid-tumor cancers.
Email: irv@stanford.edu
Normal and neoplastic stem cells
Following embryonic development, most of our tissues and organs
are continuously regenerated from tissue/organ specific stem cells.
The principal property that distinguishes such stem cells from their
daughter cells is self-renewal; when stem cells divide they give rise to
stem cells (by self-renewal) and progenitors (by differentiation). In
most tissues only the primitive stem cells self-renew. Stem cell
isolation and transplantation is the basis for regenerative medicine.
Self-renewal is dangerous, and therefore strictly regulated. Poorly
regulated self-renewal can lead to the genesis of cancer stem cells,
the only self-renewing cells in the cancerous tumor. The Weissman
lab has followed the progression from hematopoietic stem cells to
myelogenous leukemias. They have found that the developing cancer
clones progress at the stage of hematopoietic stem cells, until they
become fully malignant. At this point, the ‘leukemia’ stem cell moves
to a stage of a downstream oligolineage or multilineage progenitor
that has evaded programmed cell death and programmed cell
removal, while acquiring or keeping self-renewal. While there are
many ways to defeat programmed cell death and senescence, there
appears to be one dominant method to avoid programmed cell
removal—the expression of the cell surface ‘don’t eat me’ protein
CD47, the ligand for macrophage SIRP-alpha. All cancers tested
express CD47 to overcome expression of ‘eat me’ signals such as
calreticulin and asialogylycoproteins. Antibodies that block the CD47–
SIRP-alpha interaction enable phagocytosis and killing of the tumor
cells in vitro and in vivo. We expect to have the anti-CD47 therapies
in phase 1 trials in early 2014.
Breakout Sessions
Yiling Lu, M.D.
Associate Professor
Department of Systems Biology
MD Anderson Cancer Center
Education:
M.D. degree from Shanghai Jiao Tong University, College of Medicine
(Major in Medicine)
M. Med. from Shanghai Jiao Tong University, College of Medicine
(Major in Immunology and Microbiology)
Postdoctoral Training in Texas Tech University, HSC
(Immunology, Biochemistry and Molecular Biology)
Notable Honors/Awards:
The Susan Komen Breast Cancer Foundation: Rational design of
combinational targeted therapeutics in breast cancer: Towards
improving patient outcomes
NIH Roadmap Molecular Libraries Initiative: Reverse Phase Protein
Microarrays (RPPA) as a High Throughput Assay for Molecular
Screening
Breast SPORE Career Development Award: Identification of synthetic
lethal targets interacting with mTOR inhibition in breast cancer cells
expressing activated PI3K signaling network
Ovarian SPORE Development Research Award: Identification of
therapeutic targets interacting with autophagy in ovarian cancer
Research Interest:
My research focuses on identifying and characterizing novel cellular
targets for cancer therapy by Systems Biology
approach, which represents a paradigm shift from an
in-depth study of one molecule or a few molecules
at a time to a study of systemic integrated and
interacting molecules. Cancer systems biology
demonstrates that multiple genomic aberrations
in cancer cells integrate to alter cellular functions
at the level of protein.
Email: yilinglu@mdanderson.org
Functional Proteomics by Reverse Phase Protein Array
(RPPA) and its application in cancer biology
The Functional Proteomics Reverse Phase Protein Array (RPPA)
provides investigators with a powerful high throughput, quantitative,
cost-effective technology for functional proteomics studies. The RPPA
technology complements Mass Spectroscopy and tissue microarray
approaches to measure total and phospho- or cleaved- protein levels.
RPPA represents an antibody-based functional proteomics analysis
for both tumor tissue and cultured cells. Protein extracts denatured
by SDS followed by serial dilution (in order to define antigen-antibody
reaction in a linear range for accurate quantification) are arrayed on
nitrocellulose-coated slides and probed with validated antibodies that
recognize signaling molecules in their functional state. Signals are
captured by tyramide dye deposition and a DAB colorimetric
reaction. Data are collected and quantified using custom software
developed for this purpose. The values derived from the slope and
intercept are expressed relative to standard control cell lysates or
control peptides on the array. These values indicate the levels of
protein expression and modification. The sensitivity of RPPA
technology can detect signals at the concentration of a femtogram
from micrograms of starting materials. It characterizes, across large
numbers of tumor samples or cell lines, the basal protein expression
levels, growth factor- or ligand-induced effects, and time-resolved
responses appropriate for systems biology analysis. It provides
information to integrate the consequence of genetic aberrations in
cancer, to validate therapeutic targets, to demonstrate on- and offtarget activity of drugs, and to evaluate drug pharmacodynamics. We
have defined a set of standard lysates to be used in each array for
quality control of data generation and analysis.
Session 1. Reverse Phase Protein Array (RPPA)
Nicholas Navin, Ph.D.
Assistant Professor
Department of Genetics
MD Anderson Cancer Center
Education:
Skidmore College.
Stony Brook University
& Cold Spring Harbor Laboratory
Cold Spring Harbor Laboratory
B.S.
2002
Ph.D.
Postdoc
2010
2011
Notable Honors/Awards:
2005 Lindasy-Goldberg Fellowship
2008 King & Miller Fellowship
2009 NCI T32 Fellowship
2010 James Watson Suzhou Symposium Speaker
2012 AACR Princess Takamatsu Symposium Speaker
2012 Damon-Runyon Rachleff Innovation Award
Research Interest:
Single Cell Sequencing
Intratumor Heterogeneity
Genome Evolution
Cancer Genetics & Genomics
Email: nnavin@mdanderson.org
Bioinformatics for cancer biologists
The objective of this workshop is to provide a basic overview of the
tools and methods used by computational biologists to analyze largescale genomic datasets from human cancer patients. We will discuss
next-generation
sequencing
methods
including
genome
sequencing(DNA-seq) and transcriptome sequencing (RNA-seq). Next
we will cover tools used for detecting structural variants, copy
number changes and somatic mutations. We will then provide an
overview of the ongoing large-scale genomic sequencing projects
such as the Cancer Genome Atlas (TCGA) and the International
Cancer Genome Consortium (ICGC) and how to access data from
these projects using COSMIC and cBIO. We will also discuss how to
distinguish driver from passenger mutations using prediction
algorithms and mutation frequency. The topic of intratumor
heterogeneity will also be discussed and how clonal diversity can
affect the analysis of next-generation sequencing data. We will also
cover data visualization tools including the Integrated Genome
Viewer (IGV) and basic bioinformatics methods in multivariate
statistics. Common programming languages and operating systems
used by computational biologists will also be covered. Finally, we will
discuss advanced courses offered at different institution that can be
used to seek further training in computational biology, including the
Cold Spring Harbor Laboratory (CSHL), the European Molecular
Biology Laboratory (EMBL) and the Sanger Institute.
Session 2. Bioinformatics
Jared K. Burks, Ph.D.
Assistant Professor and Co-Director
Flow Cytometry & Cell Imaging Core Facility
MD Anderson Cancer Center
Education:
Degree-Granting Education
Florida Institute of Technology, Melbourne, FL, BS, 1995, Molecular
Biology
Florida Institute of Technology, Melbourne, FL, BS, 1995, PreProfessional Biology
Texas A&M University, College Station, TX, PHD, 2005, Biology
Postgraduate Training
Postdoctoral Associate, Baylor College of Medicine, Houston, TX,
1/2006-7/2007
Notable Honors/Awards:
2003
Entomology Graduate Student Forum Presentation
Award, Texas A&M University
2004
American Society of Virology 2004 Annual Meeting,
McGill University, Montreal, Canada
Research Interest:
As a member of the Flow Cytometry and Cellular Imaging Core Facility
I get to contribute to research across the spectrum here at MD
Anderson. I specifically enjoy applying imaging techniques to solve
problems.
Specifically, my interest is protein trafficking.
Email: jburks@mdanderson.org
Mass cytometry
Mass Cytometry is a burgeoning field where two very different
technologies meet, Flow Cytometry and ICP-Mass Spec. DVS Sciences
has developed an instrument based on an ICP-Mass Spec capable of
performing flow cytometry measurements in an environment
without spectral overlap and autofluorescence, thus removing the
need for compensation. As such this new system alleviates the
limitations of fluorescent based flow cytometry creating a paradigm
shifting new instrument capable of up to 100 parameters per cell.
Current limitations are ~35 parameters per cell. This instrument,
called a CyTOF, utilizes unique metal isotopes (non-radioactive) as
tags attached to antibodies to label your favorite proteins of interest.
This results in Massively High Parametric data per cell which is then
analyzed with SPADE Analysis. The limitations and benefits of the
instrumentation will be discussed with some example preliminary
datasets.
Session 3. Cytometry by Time-of-Flight (Cy-TOF)
Chang-Gong Liu, Ph.D.
Professor and Director
Sequencing & Non-coding RNA Program
Department of Experimental Therapeutics
MD Anderson Cancer Center
Education:
Ph.D. in Biochemistry-Molecular/Cellular Biology, 1995, University of
Vienna, Austria.
M. S. in Developmental Biology, 1989, Shandong University, Jinan,
P.R. China.
B. S. in Embryological Biology, 1982, Shandong University, Jinan, P.R.
China.
Notable Honors/Awards:
2000-Present Guest Professorship, Shandong University
1992-1995 Pre-Doctoral Fellowship, Austria National Science
Foundation
Research Interest:
Our research interest is to identify ncRNA biomarkers for cancer early
stage detection and diagnosis, later stage prognosis and therapeutic
targets collaboratively using ultra-throughput and cutting-edge
genomic technologies of microarray and next generation
sequencing. As a centralized sequencing and ncRNA Core services,
our primary responsibility is to provide NGS sequencing and ncRNA
services support to Investigators and facilitate cancer research. In
addition, we do collaboration with investigator for
in-house developments of new methodology and
applications using in-house existing technology and
expertise. Our previous collaborative efforts of
microRNA in cancer have resulted in over
hundreds of scientific research papers.
Email: cgliu@mdanderson.org
An integrated platform facilitate comprehensive
genomic and transcriptomic research in cancer
The technologies of microarray and next generation sequencing
enable us to analyze the changes of entire human genome and
transcriptome in one single experiment. Data analysis has indicated
more than 80% of human non-coding genome is transcribed into noncoding RNA (ncRNA). ncRNA includes large and small ncRNA that
transcribed from the genomic regions of intronic, intergenic,
antisense, interleaved, and overlapping with protein-coding genes.
Given evidences have revealed the crucial and critical rules of miRNA,
one class of ncRNA, expression in regulation of crucial processes of
normal development, cell proliferation, apoptosis, differentiation and
metabolism. Recent studies have identified different miRNA
expression signature patterns, by miRNA expression profiling,
associate with diagnosis and prognosis in human cancers and
revealed abnormal miRNA expression modulates a wide array of
growth and differentiation processes in human cancers. To explore
more about non-coding RNAs involved in cancer and expand our
concept in cancer genomic research, we have established a complete
and comprehensive analytical system to study ncRNA in cancers at
MD Anderson Cancer Center. This system includes 1) SOLiD™5500XL
ultra-throughput genome system for novel ncRNA discovery and
global profiling; 2) Ion Torrent PGM for targeted cancer related
genes; 3) in-house custom miRNA/ncRNA expression array for all
known targeted ncRNA expression profiling in cancer; 4)
identification of miRNA/ncRNA and target mRNA interaction
experimentally using NGS and Affymetrix mRNA profiling system
besides computing prediction and 5) the correlation studies of
differentially expressed ncRNA to it genomic copy number change by
using NGS and array CGH assays. These combined approaches will
allow us to gain full picture of cancer genomics and its differentially
expressed noncoding RNA as biomarkers.
Session 4. Non-coding RNA
James A. Bankson, Ph.D.
Associate Professor
Department of Imaging Physics,
MD Anderson Cancer Center
Education:
1994 Texas A&M University
2000 Texas A&M University
2002 UT MDACC
BS, Electrical Engineering
PhD, Electrical Engineering
Postdoctoral Fellowship,
Imaging Physics
Notable Honors/Awards:
2003-2011 Assistant Professor
2006-Present
Deputy Director, Small Animal Imaging Facility
2007-Present
Ad hoc member, various NIH grant review
panels
2011-Present
Associate Professor, with award of Tenure
2011-2013
Associate Editor, IEEE Transactions on
Biomedical Engineering, Medical Physics
2013
Graduate, UT MDACC Faculty Leadership
Academy
Research Interests:
• Imaging science
• Advanced biomedical imaging
• Magnetic resonance imaging and
spectroscopy
• Signal and image processing
• RF and systems engineering
• Cancer imaging
• Metabolic imaging with
hyperpolarized tracers
Email: jbankson@mdanderson.org
The small animal imaging facility
The Small Animal Imaging Facility (SAIF) is an core resource at The
University of Texas MD Anderson Cancer Center. It was established
to ensure that cancer researchers, regardless of their academic
training, have access to cutting-edge imaging technologies and to the
expertise that is necessary for their successful integration into
routine cancer research. SAIF consists of instrumentation, specially
trained staff, faculty affiliates with academic focus in imaging
sciences, and infrastructure that facilitates data management and
scheduling of services. Services include magnetic resonance imaging
and spectroscopy, nuclear imaging, x-ray computed tomography,
image-guided radiation therapy, ultrasound, photoacoustic, and
optical (fluorescence/bioluminescence) imaging. Faculty and staff are
available for consultation through all phases of research, from
preliminary estimation of feasibility, through experiment design, data
acquisition, analysis, and interpretation, and summary of results in
grants, manuscripts, progress reports, and other media. SAIF is a feefor-service facility with fees that compare favorably with similar
preclinical/translational imaging cores at other institutions.
Dedicated staff can carry out all aspects of imaging experiments, or
users can be trained to operate SAIF instrumentation for self-directed
imaging services. Recent advances in imaging technologies inform on
the structure, function, and composition of tissue and disease with
unprecedented specificity and resolution, and the Small Animal
Imaging Facility is an efficient conduit for the integration of emerging
imaging sciences into cancer research.
http://www.mdanderson.org/saif
Session 5. Small Animal Imaging
Poster Presentations
Poster Presentations
No
Name
Institution
Title
1 Alexander, Angela
MDACC Targeting Deregulated Cyclin E in
Inflammatory Breast Cancer via
Sequential Combination Therapy
With CDK Inhibitors
2 Appikonda, Srikanth
MDACC Histone acetylation recruits
TRIM24 to wrestle SUMO
3 Au Yeung, Chi Lam
MDACC Leptin up-regulates matrix
metalloproteinase-1 in
metastatic ovarian cancer cells in
the omental tumor
microenvironment
4 Bhadane, Minal
UTHSC
Evaluating severity of poststroke spasticity through resting
joint angle measurement
5 Bird, Kathleen N.
UTHSC
Real-time Sonographic
Measurements at the Bedside
can Evaluate Sarcopenia in the
Elderly Traumatized Patient
6 Brugmann, William
MDACC NK cells exhibit cytotoxic activity
against medulloblastoma cells in
vitro and in vivo
7 Wang, Chao
MDACC The role of Numb expression and
nuclear translocation in
endometrial cancer
8 Chen, Jixin
Rice
Multi-Loop DNA Hairpin
Folding/Unfolding
Poster Presentations
No
Name
Institution
Title
9 Cobanoglu, Zehra
MDACC SNDX-275 IS A POTENTIAL
IMMUNOMODULATOR FOR NK
CELL IMMUNOTHERAPY OF
MEDULLOBLASTOMA
10 Correa-Fernández,
Virmarie
MDACC Simultaneous Treatment of AtRisk Drinking and Tobacco
Dependence among Puerto
Ricans
11 Cox, Aaron
Baylor
12 Datar, Vrushali
MDACC Study of Histone Methyl
Transferase G9a Inhibition in
ATRT and Medulloblastoma
13 Fan, Xiaozhou
MDACC Engagement of the ICOS
pathway markedly enhances
efficacy of CTLA-4 blockade in
cancer immunotherapy
14 Forget, MarieAndree
MDACC Genetically modified artificial
antigen-presenting cells (aAPC)
to improve expansion,
phenotype and function of
tumor infiltrating cells for
adoptive cell therapy in
melanoma
15 Gehlot, Prashasnika
MDACC Bio-mathematical modeling of
Yucatan pigs – a pilot report
Extreme obesity induces massive
β-cell expansion through selfrenewal and does not alter cell
fate
Poster Presentations
No
Name
Institution
Title
16 Sim, Geok Choo
MDACC Immunotherapy with High Dose
IL-2 in Metastatic Melanoma
Patients Induces Dramatic
Changes in an Activated ICOS+ Tregulatory Cell Subset that is
Associated with Clinical
Response
17 Haymaker, Cara
MDACC Enhanced functional and survival
properties of a CD8+BTLA+ T-cell
subset from human metastatic
melanoma
18 Hossain,
Mohammad
MDACC The tyrosine kinase receptor
Tie2 plays a significant role in the
radioresistance of malignant
gliomas and DNA repair
19 Hu, Zheng
MDACC GENOME-WIDE ANALYSIS OF
REPLICATIVE LIFE SPAN
EXTENSION MEDIATED VIA
ALTERING HISTONE LEVELS IN
YEAST S. CEREVISIAE
20 Hylin, Michael
UTHSC
21 Irwin, Mary
MDACC The Antioxidant Heme
Oxygenase 1 Promotes
Proliferation and Survival of Flt3ITD-positive AML
Characterization of the severity
of neurocognitive and
histopathological deficits
following a mild fluid percussion
injury
Poster Presentations
No
Name
22 Carey, Jason
Institution
Title
MDACC Dinaciclib a potent CDK inhibitor,
abrogates cell cycle response in
TNBC cells and induces
susceptibility to PARP inhibition
23 Taube, Joseph
MDACC
Epigenetic silencing of
microRNA-203 is required for
EMT and cancer stem cell
properties
24 Jung, Su Yon
MDACC
Associations Between Sedentary
Behavior And Cancer-Related
Biomarkers
25 Kota, Daniel
UTHSC
Mesenchymal Stem/Progenitor
cell pre-conditioning with Poly
I:C Increases monocytic leukemia
cell line binding through
Hyaluronic acid
26 Krukowski, Karen
MDACC
Contribution of Peripheral
Immune Cells to ChemotherapyInduced Neuropathy
Rice
Monocyte/ Macrophage
Response to Nanofibrous
Multidomain Peptide Hydrogels
28 Lee, Hyun
UTHSC
YAP is activated by shear stress
to promote metastatic behavior
in prostate cancer cells
29 Lee, Youngbok
MDACC
Real Time In Vivo Metabolic
Profiling of Cancer Using
Hyperpolarized Imaging Agents
27 Kumar, Vivek
Poster Presentations
No
Name
Institution
Title
30 Liu, Chen
UTHSC
Pathogenic tissue
transglutaminase stabilizes
placental AT1 receptor by
ubiquitination-preventing
isopeptide modification in
preeclampsia
31 Liu, Kevin
Rice
Pleiotropy and Polygenes in
Adaptive Hybridized Gene
Clusters in Mice
32 Li, Xu
MDACC Targeting Myc-overexpressed
difficult-to-treat cancers
33 Low, Kwang Huei
MDACC LMWE induced genomic
instability via mis-regulated DNA
replication licensing machinery
34 McCullough,
Christopher
MDACC Metabolic Biomarker for Ovarian
Cancer Determined by High Field
Nuclear Magnetic Resonance
Spectroscopy
35 Morcos, Faruck
Rice
Conformational changes leave an
evolutionary footprint across
protein lineages
36 Morelli, M. Pia
MDACC Heterogeneity of acquired KRAS
and EGFR mutations in colorectal
cancer patients treated with
anti-EGFR monoclonal antibodies
37 Moss, Tyler
MDACC miR365-1 upregulates ERα and
HER2 and is associated with poor
outcome in patients
Poster Presentations
No
Name
38 Ornelas, Argentina
Institution
Title
MDACC Imaging Cell Death Through
Minimalist Molecular Probes
39 Pradeep, Sunila
MDACC Hematogenous metastasis of
ovarian cancer: rethinking mode
of spread
40 Purwaha, Preeti
MDACC Targeted LC-MS/MS
metabolomics for quantitation of
amino acids with application to
studies of L-asparaginase
treatment
41 Riquelme, Erick
MDACC VEGF/VEGFR-2 upregulates EZH2
expression and EZH2 depletion
reduce promotion of the
malignant phenotype in lung
adenocarcinoma cells, enhancing
the response to platinum-based
and VEGFR-2–targeted therapy
42 Say, Amanda
MDACC Generation of LMW-E knock-in
human mammary epithelial cells
and mice
43 Sehgal, Lalit
MDACC EZH2 and RBM5 cooperate to
mediate resistance to FAS
mediated apoptosis by
regulating the function of FAS
Natural Antisense Transcript in B
cell Lymphoma
44 Sehgal, Vasudha
MDACC Identification of candidate
miRNA Associated with Survival
for Breast Cancer Patients
Poster Presentations
No
Name
45 Seviour, Elena
Institution
MDACC
Title
46 Shao, Long Jiang
Baylor
Highly Specific Targeting of the
TMPRSS2/ERG Fusion Gene in
Prostate Cancer Using Liposomal
Nanotechnology
47 Shirazi, Fazal
MDACC Diet modification and metformin
(MET) have a beneficial effect in
a fly model of obesity and
disseminated mucormycosis
48 Singhmar, Pooja
MDACC GRK2-kinase activity inhibits
Epac-Rap1 signaling to prevent
transition from acute to chronic
pain
49 Sorokin, Alexey
MDACC Aberrant expression of
proPTPRN2 confers resistance to
apoptosis in breast tumors
50 Sreenivasan, Varun
Baylor
Silent membrane potential and
voltage at maximum
amplification: Are they
coincident in outer hair cells of
the mammalian
cochlea?
51 Srivastava, Priyanka
UTHSC
Simulating membrane binding of
K-Ras monomers and dimers
52 Sun, Hongqiang
Baylor
Epidemiological trends and the
advances of Treatments of
Amphetamine-type Stimulants
(ATS) in China
Poster Presentations
No
Name
53 Tang, Hongwei
Institution
Title
MDACC Gene-environment interactions
in smoking-associated pancreatic
cancer: A GWAS data analysis
54 Vichaya, Elisabeth
MDACC Evaluating the role of
neuroinflammation in head and
neck cancer-related symptoms
55 von Maltzan,
Kristine
MDACC Melanocyte Activation by UVRinduced Calprotectin
56 Wadhwa, Roopma
MDACC Incidence of brain metastases
following trimodality therapy
(TMT) in patients with
esophageal and
gastroesophageal cancer
57 Wang, Wenqi
MDACC Defining New Regulators for
Hippo Pathway
58 Watson, Ian
MDACC Comprehensive Molecular
Characterization of Regional
Metastatic Melanoma
59 Whiting, Nicholas
MDACC Towards the Implementation of
Hyperpolarized, Functionalized
Silicon Nanoparticles as In Vivo
Molecular Imaging Agents for
the Early Detection of Cancer by
MRI
MDACC Indoleamine 2,3 dioxygenase
(IDO1) is a Key Regulator of
Depressive-like Behavior but not
of Mechanical Hyperalgesia in
the Spared Nerve Injury Model
of Neuropathic Pain
60 Zhou, Wenjun
MDACC
Postdoctoral Association
All postdoctoral fellows in MD Anderson Cancer Center are
automatically members of the Postdoctoral Association (PDA). The
PDA Executive Committee (PDAEC) strives to improve, enhance, and
enrich the postdoctoral fellowship experience at MD Anderson by
planning monthly events and fostering interactions between
members of the Texas Medical Center (TMC).
Ways to contact the PDA and find information:
•
•
•
•
http://www.mdanderson.org/education-andresearch/education-and-training/schools-andprograms/research-training/postdoctoral-association/index.html
postdoctoralassociation@mdanderson.org
http://www.linkedin.com/groups?gid=4420426
http://www.facbook.com/#!/groups/83955564561/
MDACC
Postdoctoral Association
Executive Committee (PDAEC)
PDAEC Co-Chair:
Lara Alvarez de Lacerda, Ph.D
Lara is a Susan G. Komen Postdoctoral Fellow,
training with Dr. Wendy A. Woodward in the
Department of Radiation Oncology. Her
research focuses on the role of the tumor
microenvironment in regulating the
equilibrium among breast cancer stem cells,
progenitor, and differentiated cells.
PDAEC Co-Chair
Melissa Singh, Ph.D.
Melissa is a postdoctoral fellow in the
laboratory of Dr. Joya Chandra in the
Department of Pediatrics Research. Her
current research focuses on targeting histone
deacetylases and demethylases as a rational
combination therapy for the treatment of
glioblastoma.
Secretary and Publicity Co-Chair:
Shadia Zaman, Ph.D.
Shadia is a postdoctoral fellow in the
department of Experimental Therapeutics.
She is currently training with Dr. Varsha
Gandhi to identify and validate novel
therapeutics for the treatment of
hematological malignancies such as multiple
myeloma.
Policy Co-Chair: Haley Peters, Ph.D.
MDACC
Postdoctoral Association
Executive Committee (PDAEC)
Speakers Chair
Prashasnika Gehlot, Ph.D.
Prashasnika is a postdoctoral fellow in the
Department of Surgical Oncology under the
mentoring of Dr. Steven Curley. She is
currently working on hepatocellular cancer
and liver cancer stem cells with animal
models.
International Co-Chairs:
Amir Hamdi, M.D.
Dong Yang, Ph.D.
Events Co-Chair:
Becket Hui
RCR Seminars Co-Chair:
Enrique Fuentes-Mattei, Ph.D.
Enrique is currently training in the
Department of Molecular and Cellular
Oncology under the mentorship of Dr. MongHong Lee and Dr. Sai-Ching Yeung. His main
areas of research include toxicology,
molecular biology, receptor mechanisms,
obesity, diabetes and carcinogenesis
South Campus Chair:
Diane Scaduto, Ph.D.
MDACC
Postdoctoral Association
Executive Committee (PDAEC)
Science Park Chair:
Brent Ferguson, Ph.D.
Brent is a postdoctoral fellow in the
Department of Molecular Carcinogenesis at
Science Park in Smithville, TX working under
the direction of Dr. Marcelo Aldaz. His
research is focused on understanding the
function of the putative tumor suppressor
gene WWOX in various types of cancer.
Postdoc Liaison Chair:
Figen Beceren-Braun, Ph.D.
Figen is a postdoctoral fellow in the
laboratory of Dr. Tomasz Zal in the
Department of Immunology. Her research
focuses on restoring the anti-tumor immunity
of lymphocytes in tumor microenvironment.
APSS Working Group:
Becket Hui
Melissa Singh, Ph.D.
Fabiola Gomez, Ph.D.
Fabiola is a postdoctoral fellow in the
laboratory of Dr. Varsha Gandhi in the
Department of Experimental Therapeutics.
She is working on developing strategies to
combine Pim kinase inhibitors with other
agents in chronic lymphocytic leukemia.
Associate Director, Research Trainee
Programs:
Martha Skender, MPH
MDACC
Postdoctoral Association
Executive Committee (PDAEC)
Mentor Award Co-Chairs:
Rob Cowan, Ph.D.
Argentina Ornelas , Ph.D.
Qingshan (Carly) Yang, Ph.D.
Program Manager, Office of Postdoctoral
Affairs:
Tracy Jennifer Costello, Ph.D.
Tracy is charged with assisting Martha
Skender with the planning, organization,
direction setting and evaluation of the
academic and career development
programming for the Institution's research
postdoctoral fellow population, including
strategic oversight of the MD Anderson
Postdoctoral Association.
Acknowledgements
T
he MD Anderson PDAEC would like to extend formal gratitude to
Dr. Ronald DePhino, MD Anderson President, for his continued
support and encouragement of all MD Anderson trainees.
Furthermore , we would like to thank Dr. Oliver Bogler, and the MD
Anderson Department of Trainee and Alumni Affairs (TAA), as well as
the Alumni and Faculty Association (AFA) for their generous
sponsorship and assistance with this event. We appreciate the effort
it took to secure the funding necessary to help establish and execute
this symposium. PDEAC would also like to thank MD Anderson Faculty
Advisory Committee for providing endless assistance and mentorship.
We would like to especially recognize Martha Skender and Kristina
Brown from Office of Trainee and Alumni Affairs , Tracy Costello from
Office of Postdoctoral Affairs, and Sandra Craft from Medical Graphics
and Photography for their help putting together the symposium.
A very special thanks to all the members of PDAEC and former
member Chris Vellano, this symposium was made possible by all your
hard work, organization, and support.
2013 Working Group
• Melissa Singh
• Fabiola Gomez
• Becket Hui
Notes
The Mission of APSS
The Annual Postdoctoral Science Symposium (APSS) was initiated on
August 4th 2011, by the MD Anderson Postdoctoral Association to
provide a platform for talented postdoctoral fellows to present their
work to a wider audience.
Our ultimate vision is to develop this event into a symposium that
incorporates all Texas Medical Center (TMC)-affiliated institutions
providing postdocs from a unique opportunity to learn, interact, and
foster new collaborations. With your support and participation, we
can make this happen.