The University of Toledo Cancer Research Symposium

Cancer Symposium 2018 Presentations

Photo from 2018 symposium

TOLEDO CANCER RESEARCH SYMPOSIUM

FRIDAY, DECEMBER 7, 2018

2018 Presentations

01. Addressing the Proteogenomic Platform in Precision Therapeutics
Alexzander Asea, Ph.D., Department of Medicine, UTCOMLS

02. Genomic Sequencing Platform Using Foundation
Thomas Blomquist. M.D., Ph.D., Department of Pathology, UTCOMLS

03. Inhibiting Bromodomain Protein 9 (BRD9) in Melanoma
Ivana de la Serna, Ph.D., Department of Cancer Biology, UTCOMLS

04. Targeting the Cytoskeleton and Formin Machinery to Halt Tumor Cell Invasion
Kathryn Eisenmann, Ph.D., Department of Cancer Biology, UTCOMLS

05. The CD3's Compound Library and Its Direction Toward Anticancer Research
Paul Erhardt, Ph.D., Department of Medicinal & Biological Chemistry, College of Pharmacy, UT

06. Restoration of Nitric Oxide Synthase Function as a Novel lmmunotherapy for Breast Cancer
Saari Furuta, Ph.D., Department of Cancer Biology, UTCOMLS

07. Identification of Novel Regulators of lnvadopodia Formation
Rafael Garcia-Mata, Ph.D., Department of Biological Sciences, College of Natural Sciences and Mathematics, UT

08. Shared Resources Available in the UToledo Integrated Core Facilities to Aid in Cancer Research
Andrea L. Kalinoski, Ph.D., Department of Surgery and Integrated Core Labs, UTCOMLS

09. Repurposing of FDA Approved Drugs for Precision BIRC5 Targeted Therapy for Pancreatic Ductal Adenocarcinoma
Shi-He Liu, M.D., Juehua Yu, Robert Damoiseaux, Robbi Sanchez, F. Charles Brunicardi, Department of Surgery, UTCOMLS
Molecular Screening Shared Resource UCLA, Los Angeles, CA

10. Mitochondrial Fission Regulator 2 (MTFR2) Contributes to Faithful Chromosomal Segregation
Yibo Luo, Ph.D., Song-Tao Liu, Department of Biological Sciences, College of Natural Sciences and Mathematics, UT

11. Kinome Array Profiling of Complex Biological Samples
Robert Smith, M.D., Ph.D., Department of Neurosciences, UTCOMLS

12. Unveiling a New Collaborative Precision Cancer Clinical Trials Program, Born of the Academic Affiliation between the University of Toledo, College of Medicine and Life Sciences and ProMedica Health System: Overall Vision and Initial Steps
John Nemunaitis, M.D., Department of Medicine, UTCOMLS
Adam Walters, M.D., M.S., Gynecologic Oncology, Promedica Physicians

13. Repurposing Drugs and the FDA Approved Drug Library
Zhixing Kevin Pan, M.D., Ph.D., Department of Medical Microbiology and Immunology, UTCOMLS

14. The Role of CXCR4-LASP1 Axis in Triple-Negative Breast Cancer
Dayanidhi Raman, Ph.D., Department of Cancer Biology, UTCOMLS

15. A Nuclear Complex of AR and TM4SF3 is a Novel Target for Prostate Cancer Therapy
Lirim Shemshedini, Ph.D., Department of Biological Sciences, College of Natural Sciences and Mathematics, UT

16. Targeting NAD and NADP-Dependent Cell Signaling for New Anti-Cancer Therapy
James Slama, Ph.D., Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, UT

17. Novel Technologies to Overcome Drug Resistance in Cancer
Amit K. Tiwari, Ph.D., Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, UT

18. Targeting the Expression of Tumor Suppressor Gene RKIP for Cancer Therapy
Kam Yeung, Ph.D., Department of Cancer Biology, UTCOMLS

19. Targeted Drug Discovery Program, a Small Business Start-up Model and Perspectives at UT
Jian-Ting Zhang, Ph.D., Department of Pharmacology and Toxicology, Indiana University School of Medicine


01. Addressing the Proteogenomic Platform in Precision Therapeutics

Alexzander Asea, Ph.D.
Department of Medicine
UTCOMLS

The Precision Therapeutics Proteogenomics Diagnostics Center is the diagnostic arm of the Precision Therapy Program at the University of Toledo College of Medicine & Life Sciences and ProMedica. The rationale for our existence is that disease progression and drug responses vary significantly from patient to patient. Precision medicine, also called personalized medicine, is an approach to patient care that allows doctors to select specific treatments that are most likely to help specific patients based on a proteogenomic understanding of their disease. Specifically, genomic data and transcriptomic data are used to generate customized protein sequence databases to help interpret proteomic data. In turn, the proteomic data provide protein-level validation of the gene expression data and help refine gene models. The enhanced gene models can help improve protein sequence databases for traditional proteomic analysis. To illustrate how excited the research community is about proteogenomics, about two years ago, the National Cancer Institute (NCI) established the Office of Cancer Clinical Proteomic Research with the aim to improve prevention, early detection, diagnosis, and treatment of cancer by enhancing the understanding of the molecular_mechanisms of cancer, advancing proteome/proteogenome science and technology development through community resources (data and reagents), and accelerating the translation of molecular findings into the clinic. This office set up the Applied Proteogenomics Organizational Learning and Outcomes (APOLLO) network which is a collaboration between the NCI, The Department of Defense (DoD) and the Veterans Administration (VA) to incorporate proteogenomics into patient care.


02. Genomic Sequencing Platform Using Foundation 1

Thomas Blomquist, M.D., Ph.D.
Department of Pathology
UTCOMLS

Management of advanced stage cancer is steadily incorporating next generation sequencing (NGS) assays such as Foundation One CDx (F1 CDx; 324 genes) as part of standard of care. Supporting this practice is accruing evidence that targeted treatments directed toward each tumor's genetic profile results in improved progression free survival with reduced side-effects. For many advanced cancer patients there is a trend toward minimally invasive procedures to obtain tissue for diagnosis, staging and the myriad of molecular tests to achieve precision care.

However, an unexpected outcome is that large panel NGS assays require more tissue than is routinely obtained from minimally invasive procedures. Tissue inadequacy from this consequence, may lead to rebiopsy, alternative molecular tests that may be suboptimal, or lack of suitable treatment options for advanced stage cancer patients. In this session, I will discuss efforts at UTMC to optimize tissue adequacy for Foundation One CDx testing through provider education and intraoperative consultation, with the goal of providing a one-and-done biopsy experience for our patients.


03. Inhibiting Bromodomain Protein 9 (BRD9) in Melanoma

Ivana de la Serna, Ph.D.
Department of Cancer Biology
UTCOMLS

Epigenetic regulators are emerging as promising targets in cancer therapeutics. We have identified the bromodomain protein, BRD9 as a novel epigenetic target in melanoma. Bromodomain proteins act as readers of the histone code by binding acetylated histones and regulating gene expression through recruitment of regulatory proteins and modulation of chromatin structure. Our data show that BRD9 expression is high in patient derived melanoma samples and correlates with poorer survival. Chemical inhibition of BRD9 enhances the antitumor effect of BRAF inhibitors in cultured melanoma cells and results in changes in the expression of genes associated with resistance to BRAF inhibitors. Depletion of BRD9 by siRNA has a similar effect on gene expression as do chemical inhibitors, thus confirming the on-target effects of the drugs. Our current studies are focused on testing the effects of BRD9 inhibitors in vivo and on elucidating the mechanisms by which BRD9 inhibitors exert their anticancer effects.


04. Targeting the Cytoskeleton and Formin Machinery to Halt Tumor Cell Invasion

Kathryn Eisenmann, Ph.D.
Department of Cancer Biology
UTCOMLS

Glioblastoma multiforme (GBM) is the most common cancer originating in adult brains. What makes GBM so deadly is its capacity to spread, or invade, throughout the brain. Failure of current GBM therapies to target invasive cells partly explains why these treatments confer only minimal survival advantages: invasive tumors lack easily-defined margins, making complete surgical removal impossible, and invasive GBM cells are inherently more resistant to chemotherapy and radiation. GBM cell invasion mechanisms are understudied and represent potential targets for drug development. Proteins controlling assembly/disassembly of the cell skeleton, or cytoskeleton, underlie cell movement and when targeted may halt tumor progression. Diaphanous (mDia)-related formins are proteins with critical roles in cancer cell invasion. mDias are cellular "nanomachines" that build filaments that generate force to drive cell movement. Drugs that turn this nanomachine on (agonists) or off (antagonist) target cytoskeleton assembly. We published that mDia agonists called intramimics (IMMs) halts invasion in established GBM lab cells. We now test the antiinvasion efficacy of IMMs in cells grown directly from GBM patient tumors. Patient cells spontaneously formed invasive three-dimensional (30) structures called neurospheres. In all GBM patient samples, IMMs significantly and effectively halted neurosphere invasion and warrants further development.


05. The CD3's Compound Library and Its Direction Toward Anticancer Research

Paul Erhardt, Ph.D.
Department of Medicinal & Biological Chemistry
College of Pharmacy, UT

The Center for Drug Design and Development (CD3) is a UToledo core resource for translational research related to the pursuit of small molecule biomarkers, diagnostics, therapeutic or disease preventative agents. It can provide directly or through a closely-knit network, all of the interdisciplinary expertise and activities needed to move a therapeutic concept conducive to small molecule approaches, all the way to an IND submission. One of its resources is a proprietary compound library that can be used in a directed manner to optimize hit compounds emanating from frontline screening. This library has been used repeatedly to advance several UToledo research programs aimed at developing novel anticancer therapies. Ongoing collaborative programs in this area include the following mechanistic strategies: "Methuosis" for glioblastoma (W. Maltese); "Methuophagy"for colon cancer (A. Tiwari); "C3" repurposed mix for BIRC5-dependent pancreatic cancer (C. Brunicardi, S-H. Liu, J. Nemunaitis); "PAM lnhibitors"for lung cancer (P. Erhardt); ''AR-TM4SF3"for prostate cancer (L. Shemshedini); and, "MAD2"for breast cancer (S-T. Liu).


06. Restoration of Nitric Oxide Synthase Function as a Novel lmmunotherapy for Breast Cancer

Saori Furuta, Ph.D.
Department of Cancer Biology
UTCOMLS

Breast cancer is the second leading cause of cancer-associated death among women in the US. Despite a number of cancer-targeted drugs, breast cancer frequently becomes resistant to them. Thus, as an adjuvant therapy, cancer immunotherapy, which induces immune cells to attack cancer cells, has been explored intensively. However, clinical studies report only modest responses of breast cancer to immunotherapy, and there is no FDA-approved immunotherapy for this disease unlike other cancer types. Such failures are largely attributed to the immuno-suppressive microenvironment of breast cancer. Thus, there is an urgent need  to develop a method to correct the unsuitable microenvironment of breast cancer. We hypothesize that supplement of sepiapterin, the naturally-produced cofactor of nitric oxide synthase, could correct the immuno-suppressive microenvironment of breast cancer, facilitating cancer cell-killing by immune cells. Sepiapterin has been safely utilized in humans to treat certain metabolic disorders. We showed that sepiapterin effectively converted immunosuppressive (M2, tumor-promoting) to immuno-stimulatory (M1, tumor-killing) tumor-associated macrophages (TAMs). Sepiapterin also dramatically inhibited proliferation of a panel of breast cancer cells by downmodulating tumorigenic HER2 and TGFl3. These results demonstrate the utility of sepiapterin as a novel immunotherapy agent that simultaneously targets TAMs and cancer cells to collaboratively inhibit breast cancer growth.


07. Identification of Novel Regulators of lnvadopodia Formation

Rafael Garica-Mata, Ph.D.
Department of Biological Sciences
College of Natural Sciences and Mathematics, UT

The migration of cancer cells away from the primary tumor and their subsequent metastasis to distant organs is the leading cause of mortality in cancer patients. Metastatic cells escape the primary tumor and enter the bloodstream by developing specialized cellular structures called invadopodia that degrade the extracellular matrix surrounding the tumor to allow invasion. lnvadopodia formation is regulated by a family of proteins called Rho GTPases, which function in virtually all stages of cancer progression. In contrast to other cancer-associated genes that are frequently mutated in cancer, mutations in Rho genes are extremely rare in tumors. In contrast, RhoGEFs and RhoGAPs, two protein families that function upstream of the Rho GTPases to regulate their activity, are often misregulated or mutated in cancer. Our goal is to identify novel regulators of invadopodia formation within the RhoGEF and RhoGAP families. It is our expectation that the identified RhoGEFs and RhoGAPs can be used to develop specific inhibitors to slow down or prevent invadopodia formation and thus, invasion and metastasis in different cancer types. RhoGEFs and RhoGAPs inhibitors are emerging as a promising approach to cancer therapy, and studies suggest that such inhibitors appear to block many functions associated with tumorigenesis.


08. Shared Resources Available in the UToledo Integrated Core Facilities to Aid in Cancer Research

Andrea L. Kalinoski, Ph.D.
Department of Surgery
UTCOMLS

The University of Toledo Integrated Core Facility (ICF) is a centralized facility comprised of shared services, cutting edge scientific equipment, and scientific expertise which is available to both internal and external users. The ICF is composed of three major laboratory facilities which include the Advanced Microscopy & Imaging Center, Flow Cytometry Core, and the Histology Core for your experimental needs. The Advanced Microscopy & Imaging Center offers access to confocal and conventional fluorescence microscopes with image analysis resources for live cells, fixed cells or whole model organisms. Also available are high throughput protein characterization and kinome profiling arrays to determine protein activity. The Flow Cytometry Core Facility provides access to instrumentation, assistance and guidance to perform the measurement and purification of cell populations or particles of interest. The Histology Core provides services associated with the microscopic study of cells and tissues from plant or animal origin offering histological processing, sectioning, staining and digital imaging of frozen or formalin-fixed, paraffin-embedded tissues. The ICF maintains and supports the state-of-the-art equipment that enables cutting-edge research to be performed by helping researchers economically and efficiently take advantage of innovative technology and collaborate with experts in the field. The goal of the ICF is to aid in the production of high quality scientific data for use in publications, grant preparation, or other scientific endeavors at the University of Toledo.


09. Repurposing of FDA Approved Drugs for Precision B1RC5 Targeted Therapy for Pancreatic Ductal Adenocarcinoma

Shi-He Liu, Juehua Yu, Robert Damoiseaux, Robbi Sanchez, F. Charles Brunicardi
Department of Surgery, UTCOMLS
Molecular Screening Shared Resource UCLA, Los Angeles CA

Precision cancer therapy requires matching of the tumor's genomic profile to targeted therapy. In this study, we have tested the hypothesis that genomic profiling using RNA-Seq and high throughput screening (HTS) of an FDA approved drug library using a target gene superpromoter (SP) can provide precision cancer therapy for pancreatic adenocarcinoma (PDAC) using repurposed FDA approved drugs. B1RC5 was identified a target for PDAC and selected for HTS of FDA approved drugs using B1RC5-SP driven luciferase assay. Based on extensive in vitro testing, and a list of criteria to enhance translational potential, simvastatin, metformin and digoxin, (C3), were determined to be the optimal combination and then tested in three in vivo studies using PDAC xenograft mouse models. Following therapy, PDAC tumor growth was significantly suppressed as a result from complete suppression of B1RC5 expression and increased apoptosis, without toxicity in any mice. RNA Seq of PDAC tumors revealed decreased cell proliferation gene expression, decreased ATP/energy gene expression and markedly increase cell death gene expression after C3 therapy. Our study demonstrates a feasible cost-effective strategy of repurposing FDA-approved drugs for targeted precision PDAC therapy using a super-promoter of the target gene in a clinically relevant timeframe.


10. Mitochondrial Fission Regulator 2 (MTFR2) Contributes to Faithful Chromosomal Segregation

Yibo Luo, Ph.D., Song-Tao Liu, Ph.D.
Department of Biological Sciences
College of Natural Sciences and Mathematics, UT

The human genome in a cell consists of two parts: the chromosomes in the nucleus and the mitochondrial DNA (mtDNA). During cell division (mitosis), duplicated chromosomes are separated and equally distributed into two daughter cells. Meanwhile, cytoplasmic mitochondria carrying mtDNA are also fragmented and distributed into daughter cells. Whether and how chromosomal segregation and mitochondrial fission is coordinated during mitosis is an important and interesting question. Here we addressed the functions of a poorly characterized protein, MTFR2, in both mitochondrial fission and chromosome segregation. We showed that MTFR2 is a mitochondrial protein and localized on the mitochondrial outer membrane. MTFR2 overexpression induced premature mitochondrial fission during interphase.

Conversely, when MTFR2 was depleted, the mitochondria stayed as tubular network during mitosis. Interestingly, when MTFR2 was depleted from cells, the duration of mitosis was prolonged and the chromosomal mis-segregation rate was significantly increased. Moreover, the MTFR2 depleted cells became more resistant to Taxol induced apoptosis. Taken together, our results demonstrate that MTFR2 contributes to both mitochondrial fission and chromosomal segregation and suggest MTFR2 loss of function could be involved in tumorigenesis.

Keywords: MTFR2, Mitochondrial fission, Mitosis, Tumorigenesis


11. Kinome Array Profiling of Complex Biological Samples

Robert Smith, M.D., Ph.D.
Department of Neurosciences
UTCOMLS

Studies of mRNA expression using RNA sequencing technologies have provided important new leads and insights for scientific discovery of disease processes. However, approaches that focus on mRNA or protein levels fail to capture the functional aspects of many genes. For example, there may be no changes in mRNA expression, but a large change in the same gene's level of enzyme activity. This caveat is of particular concern for protein networks that rely on enzymatic activity to effect changes in biological function in disease states. To address this challenge in the field, we have adapted the Pamgene12 kinome array platform for use in postmortem brain and other difficult to obtain biological substrates to assess broad-based networks of protein kinase enzyme activity in complex biological samples. This novel approach will identify novel disease pathways, provide new drug leads in model systems, and permit sophisticated personalized medicine assessments for the afflicted.


12. Unveiling a New Collaborative Precision Cancer Clinical Trials Program, Born of the Academic Affiliation between the University of Toledo, College of Medicine and Life Sciences and ProMedica Health System: Overall Vision and Initial Steps 

John Nemunaitis, M.D.
Department of Medicine
UTCOMLS

Adam Walters, M.D., M.S.
Gynecologic Oncology
ProMedica Physicians

The process of providing new precision therapy technologies to our cancer community involves basic science, early clinical testing to define optimal dose and schedule (translational research) and efficacy testing to prove the new therapy provides a safe approach and increases patient benefit over what we currently use as standard care (registration trial).

UTCOM and ProMedica have combined resources to establish an affiliated cancer research program which will pursue preclinical, translational and registration trial development of new precision therapies. Precision therapy involves the selection of a specific target therapy for management of disease based on a molecular understanding of that disease. Our combined mission is to extend treatment options for cancer patients though precision therapies following molecular profile analysis. This unique approach combines "beside to bench ... and back ... "processes that integrate standard of care and clinical trials with basic science. It is also an innovative opportunity for registration trial development and education as a NEW standard of care. Furthermore, this will focus diversity in care, advocacy, and work force onto the individual patient. FDA supported benefits over standard of care have now been demonstrated with nearly 100 precision therapies, predominately available at UTCOM and ProMedica involving multiple molecular targets. Initial assessment will be reviewed of the first cancer patients in clinical trial as a result of the affiliated program already demonstrate evidence of major cancer benefit. Multiple clinical trial opportunities are now open for accrual!


13. Repurposing Drugs and the FDA Approved Drug Library

Zhixing Kevin Pan, M.D., Ph.D.
Department of Medical Microbiology and Immunology
UTCOMLS

The process of developing new medications can cost billions of dollars and take decades worth of research. To bypass this extensive and expensive process, we use a plan of attack called drug repositioning strategy (drug repurposing). We take an existing drug, perform experiments to determine if the drug can be used for other diseases. In these cases, because much of the work has already been done in approving the drugs, it is much easy for regulatory agencies to grant the use of an already approved drug for new purposes.

We have implemented this strategy by screening of the library of 1,200 FDA approved drugs. We have discovered that an already approved drug called rolipram, which is currently being used for COPD, can increase the mouse survival rate in our bacteria-induced septic shock animal model. These studies should provide preclinical information that will allow us to initiate clinical studies for rolipram in victims with sepsis and septic shock induced by bacteria. The FDA-approved drug library is already being used by various faculty including Dr. Taylor, Dr. Chattopadhyay, Dr. Pan from the Department of MMI, Dr. Brunicardi from the Department of Surgery, and Dr. Yeung from the Department Cancer Biology. This library is greatly expanding what questions that our researchers may have in the College and providing important and pre-clinical data for new grant applications.


14. THE ROLE OF CXCR4-LASP1 AXIS IN TRIPLE-NEGATIVE BREAST CANCER

Dayanidhi Raman, Ph.D.
Department of Cancer Biology
UTCOMLS

Breast cancer frequently spreads (metastasizes) to distant sites like lungs and cause a havoc by disrupting its function. Most breast cancer patients who die or experience serious pain do so because of these distant tumors. The tumors that have spread are difficult to remove and furthermore resistant to chemotherapies. Therefore, drugs that specifically and potently targets the metastatic tumor cells are needed. In order to develop such new drugs, we propose to study the role of the CXCR4-LASP1 pathway in metastatic breast cancer cells. Many cancer types including breast cancer have "hijacked" the CXCR4 receptor and use it to move throughout the body. We have identified a novel mechanism of LASP1 that may play a role in tumor cell survival and metastasis. The sole aim is to increase patient longevity and survival.


15. A Nuclear Complex of AR and TM4SF3 is a Novel Target for Prostate Cancer Therapy 

Lirim Shemshedini, Ph.D.
Department of Biological Sciences
College of Natural Sciences and Mathematics, UT

Androgen Receptor (AR) is key in the development and progression of prostate cancer to the usually lethal Castration Resistant Prostate Cancer (CRPC). While the most effective drug therapies target AR (androgen ablation or anti-androgens), these therapies have limited efficacy clinically and prostate tumors often develop resistance, suggesting a strong need for alternative ways to regulate and disrupt AR. Our lab has recently identified a novel mechanism of AR regulation via the transmembrane protein TM4SF3 (Transmembrane 4 superfamily 3).

We have discovered that a fraction of TM4SF3 is in the cytosol and is able to physically interact with AR in a ligand-dependent manner, resulting in the mutual stabilization and nuclear co-localization of the two proteins. Importantly, TM4SF3 also interacts with and stabilizes AR(V7), a variant AR protein that is lacking the ligand-binding domain and is responsible for a common form of CRPC. We have recently mapped interaction regions of AR and TM4SF3 and will use this information to design peptides targeting the AR/TM4SF3 complex. This study, together with future ChiP-Seq and RNA-Seq studies, will allow us to study the importance of TM4FS3 in AR biology in prostate cancer, providing information that can used to possibly make a more effective prostate cancer therapy.


16. Targeting NAD and NADP-Dependent Cell Signaling for New Ant- Cancer Therapy

James Slama, Ph.D.
Department of Medicinal and Biological Chemistry
College of Pharmacy and Pharmaceutical Sciences, UT

NAO+ and NADP+-the pyridine dinucleotides-are derived from vitamin 83 (niacin and niacinamide) and are well known to medicine and biochemistry as essential co-substrates in metabolism What is less appreciated is that NAO+ and NADP+ participate in specialized roles which are essential to cell regulation. For example, NAO+ is polymerized in the cell nucleus following DNA damage, producing ADP-ribose polymers. ADP-ribose polymer turnover enables dividing cells to survive DNA damage. Disrupting this process with polymerase inhibitors yielded anti-cancer PARP inhibitors-olaparib, rucaparib and niraparib. We have made inhibitors of ADP-ribose polymer degradation, which are predicted to act similarly. As a second example, a small change in the structure of NADP yields nicotinic acid adenine dinucleotide phosphate or NAADP. NAADP is an intracellular second messenger which controls ion-channels and causes intracellular Ca2+ ion release. Cell Ca2+ in turn, regulates cell division, cell motility and secretory processes. My laboratory is committed to understanding NAADP mediated signaling through the isolation and identification of the intracellular NAADP receptor protein. We employ a chemical biology approach utilizing photoactive NAADP affinity tags. Identification of the NAADP receptor will enable us to develop antagonists that we predict will be an important new class of antitumor drugs.


17. Novel Technologies to Overcome Drug Resistance in Cancer

Amit K. Tiwari, Ph.D.
Department of Pharmacology and Experimental Therapeutics
College of Pharmacy and Pharmaceutical Sciences, UT

Multidrug resistance (MOR) is a major impediment to cancer chemotherapy. MOR to current therapies, typically develop due to overexpression of ATP-binding efflux transporters, metastasis and reduced sensitivity to apoptosis, resulting in early relapse and shorter survival. We hypothesize that new opportunities to treat MOR in cancer patients could arise from the identification of compounds that are not a substrate of efflux transporters and can trigger a cell death mechanism that does not depend upon classical apoptotic pathways. In this regard, we have recently developed 2 novel technologies. First deals with the discovery of a unique, structurally constrained pyridinyl probes which induce a form of non-apoptotic cell death characterized by simultaneous induction of dysregulated macropinocytosis (self-drinking) and macroautophagy (self-eating). We have defined this unique type of caspase-independent cell death 'methuophagy' (the neologism arises from the Greek 'methuo', to drink to intoxication and 'phagy', self-eating). Our new class of small molecular probes (patented and out-licensed) that do not involve classical apoptotic pathways, cause methuophagy by inducing the formation and accumulation of fluid-filled, heterogeneous vacuoles and autophagosomes, resulting in metabolic failure, loss of membrane integrity, and detachment of cells from the substratum. Methuophagy produces hypervacuolization and extensive autophagy without cell shrinkage, chromatin condensation, mitochondrial membrane potential alteration, or nuclear membrane damage. The second technology deals with discovery of a novel class of pyrimidinhydrazinylidene compounds that selectively induces a unique NANI (non-apoptotic-necroptotic inducing i.e. caspase-independent programmed cell injury) cell death in specific cancer cells. The identified novel compound will be particularly beneficial for therapy of drug resistant and aggressive cancers. A contribution to adjuvant therapy is envisioned for these new class of drugs that kill cancer cells by inducing a non-apoptotic mechanism distinct from ONAalkylating, apoptosis-inducing agents and receptor-targeted therapeutics. Finally, the novel probes identified will allow us to understand the biology of unique non-apoptosis-induced cell death.


18. Targeting the Expression of Tumor Suppressor Gene RKIP for Cancer Therapy 

Kam Yeung, Ph.D.
Department of Cancer Biology,
UTCOMLS

Raf Kinase Inhibitory Protein (RKIP) is a well-established tumor suppressor whose expression is often diminished in a plethora of solid and hematological malignancies. Loss or diminution of RKIP expression was significantly associated with shorter disease-free survival in cancer patients. Importantly restoration of RKIP expression in low-RKIP-expressing cancer cells decreases total tumor burden in experimental mouse cancer models. As such, restoring RKIP expression represents a promising strategy for the development of anti-cancer therapy. Recent studies showed that a temporary halt of the RKIP gene expression in cancer cells was the cause of reduced RKIP expression in cancer. To identify small-molecule activators that will increase RKIP gene activity, we developed a novel cell-based screening assay. Using CRISPR/Cas9 genome editing technology, we generated a knock-in nanoluciferase reporter cancer cell line that uses bioluminescent nanoluciferase as a proxy for endogenous RKIP gene activity. The described reporter assay will enable the high-throughput screening of drug library to identify potential novel drugs for cancer therapy.


19. Targeted Drug Discovery Program, A Small Business Startup Model and Perspectives at UT

Jian- Ting Zhang, Ph.D.
Department of Pharmacology and Toxicology
Indiana University School of Medicine

In this presentation, I will reflect on the history regarding how I started a drug discovery program at Indiana University School of Medicine, going through different cycles of fund raising to support this program. I will then elaborate on one of our successful stories on targeting survivin as an example. Finally, I will close the presentation on perspectives for possibly \ establishing such a program at UT/Promedica and hope to initiate a discussion on this possibility.

Survivin is a member of the Inhibitor of apoptosis (IAP) gene family, existing as a homo-dimer. It is over-expressed essentially in all cancers, but not expressed in most adult normal tissues. Ectopic survivin over-expression causes inhibition of cell death induced by intrinsic and extrinsic stimuli in cell lines and in animals. Survivin has also been shown to contribute to radiotherapy and chemotherapy resistance, and inhibition of survivin sensitizes cancer cells to these treatments. Treatments with molecular probes such as antisense oligonucleotide, ribozyme, siRNA, and dominant negative mutant all resulted in caspase-dependent cell death and increased apoptosis induced by radiation and anticancer drugs. These findings clearly established survivin as an ideal target for discovery of anticancer therapeutics. Unfortunately, survivin belongs to a group of proteins that are considered undruggable due to lack of enzymatic activities.

Despite the fact that different approaches have been attempted and tested in targeting survivin expression, none of these studies has resulted in an FDA-approved drug due to a variety of issues. We took a different approach by targeting the dimerization domain with a hypothesis that inhibiting survivin dimerization would lead to survivin degradation via proteasome due to cellular quality control system. Through a combination of computational analysis of the dimerization interface, in-silica screening, biochemical, medicinal chemistry, cell-based and animal studies, we have identified effective and specific survivin inhibitors for further development.

Last Updated: 6/27/22