Melissa M - Indiana University School of Medicine



Curriculum Vitae

Melissa (Limp-Foster) Fishel, Ph.D. (DOB : 06/04/1974, Indianapolis, IN USA)

Associate Research Professor

Department of Pediatrics, Wells Center for Pediatric Research

Indiana University School of Medicine

1044 W. Walnut St. R4 321

Indianapolis, IN 46202

Office: (317) 274-8810

Lab: (317) 278-0579

Fax (317) 274-8679

mfishel@iu.edu

Current Position

Associate Professor, Research

Academic Appointment

2001-2004 Post-Doctoral Fellow, Hematology/Oncology, Laboratory of M. Eileen Dolan, The University of Chicago, Chicago, IL

2004-2008 Assistant Research Professor, Pediatrics, Hematology/Oncology, Herman B Wells Center for Pediatric Research, Laboratory of Mark R. Kelley, Indiana University School of Medicine, Indianapolis, IN

2008-2015 Assistant Research Professor, Pediatrics, Hematology/Oncology, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN

2015-present Associate Research Professor, Pediatrics, Hematology/Oncology, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN

Education

1992-1996 B.S., Chemistry (cum laude), University of Dayton, Dayton, OH

1996-2001 Ph.D., Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN

2001-2005 Post doctoral Fellowship, The University of Chicago, Laboratory of Eileen Dolan

Awards / Fellowships

1996-2000 Graduate Assistance in Areas of National Need (GAANN) Fellowship

1999 Environmental Mutagen Society Travel Award

1999-2000 Cancer Biology Training Fellowship, Indiana University Cancer Center, Indianapolis, IN

2000-2001 BC991226 CDMRP. Predoctoral training grant, DOD. DNA base excision repair (BER) and cancer gene therapy: Use of the human N-Methylpurine DNA Glycosylase (MPG) to sensitize breast cancer cells to low dose chemotherapy. $22,000 per year for 3 years. (Mentor; Kelley)

2003-2004 Cancer Biology Training Grant, The University of Chicago, Chicago, IL

2006-2007 Marsha Rivkin Scientific Scholar Award Recipient

2015 Nominated for Associate Scientific Advisorship for Early-Career Translational Scientists with Science Translational Medicine

Professional Society Memberships

American Association for Cancer Research

Women in Cancer Research

Research-focus Groups

Member of IU Simon Cancer Center; Tumor Microenvironment and Metastases (TMM) program (2013 – present)

Member of IU Simon Cancer Center; Experimental and Developmental Therapeutics (EDT) program (2005 – 2013)

Member of Pancreatic Cancer Signature Center Initiative (2011 – present)

Member of IU Simon Cancer Center Pancreatic Cancer Working Group (2008 -– present)

Member of IU Simon Cancer Center Brain Tumor Working Group (2010 – present)

Member of IU Simon Cancer Center Chemotherapy Induced Peripheral Neuropathy Working Group (2013 - present)

Member of Pediatric Hematology Oncology (PHO) Translational Research working group (2007 – 2015)

Member of Pediatric Molecular Oncology & Experimental Therapeutics group (2015 – present)

Teaching Responsibilities

General Chemistry Laboratory, CHM123L and CHM124L. (1995-1996) Responsible for lectures, general lab supervision, grading students (~22 students).

Fundamental Molecular Biology, G865 “DNA Repair and Its Translational Uses,” Lecturer (2000), 1 Lecture/Year.

Cancer Biology IV: Frontiers in Cancer Research, “Exploiting Molecular Targets in Cancer Therapy,” Lecturer (2003-2004), 1 Lecture/year.

Teaching Assistant, BioSci25108 Cancer Biology. (Winter 2004) Responsible for 1-2 lectures, homework sets, leading discussion groups, grading students’ work and examinations.

Mammalian DNA Repair & Disease, G837. (Spring 2007, 2008) “Base Excision Repair,” 3 Lectures/year.

Chemical Carcinogenesis, F819. (Spring 2008) “DNA damage in Carcinogenesis,” 1 lecture/year.

Neoplastic Determinants, J842. (Fall 2013, 2014) “DNA Repair,” 1 lecture/year.

Advanced Concepts in Nuclear and Cytosolic Signaling, G751. (Spring 2014 – 2017) “Jak/STATs,” 1 lecture/year.

Graduate Thesis and Qualifying Exam Committees

2016 PhD Qualification Exam Committee for Antja-Voy Hartley, Department of Pharmacology and Toxicology

Laboratory Mentoring

Undergraduate Students

• Nona Hollenbaugh (2004 & 2005)

• Amanda Meyer (2009)

• Amy Dreischerf (2009 & 2010)

• Lily Wang (2010)

• John Zhang (2011)

Medical Students

• Jeff Florek (2013, Winner of Poster Presentation Award)

• Devika Chakrabarti (2016)

Graduate Students

• David McIlwain (2013-present)

• Derek Logsdon (2013-present)

• Jennifer Beebe (2015-present)

Post-doctoral Fellows

• Yanlin Jiang (2007 – 2013)

• Huiwen Cheng (2013 – 2015)

• Fenil Shah (2015-present)

Fellows

• Safi Shahda, M.D. (July 2011 – 2013)

Internal and Community Service

Indiana University School of Medicine Tissue Procurement and Distribution Core Advisory Committee October 2014 – present

Indiana University School of Medicine Laboratory Animal Resource Center Advisory Committee July 2013 – present

Indiana University School of Medicine SRPAM Poster Reviewer September 2013 – 2016

Wells Center for Pediatric Research Summer Intern Judge August 2017

High School Research Mentoring Programs

Molecular Medicine in Action (MMIA) siRNA module (March 2009 – 2012, 2014 – 2016)

Molecular Medicine in Action (MMIA) Tumor Microenvironment supermodule (2017)

Molecular Medicine in Action (MMIA) for Teaching Professionals siRNA module (Oct. 2009 – 2012)

Advisory Board for Project Lead the Way Biomedical Program– Greenfield Central High School and Warren Central High School (2010-present)

Community Service

2009 - present Advisory Board Member “Project Lead the Way” Biomedical section

June 2010 American Cancer Society Relay for Life, Volunteer for decorating luminaries to honor survivors and victims of cancer

October 2010 Panel Discussion Cancer projects for Crawfordsville High School students

Aug 2012, 2013 Judge for Student Research Program in Academic Medicine (SRPAM) for Indiana University School of Medicine medical students

April 2013 WishTV8 interview with pancreatic cancer patient, highlighting Fishel and Kelley labs’ pancreatic cancer projects to raise awareness for disease and need for research (April 8, 2013)

May 2013 Advisory Board Member, Biomedical program in “Project Lead the Way” receives

national certification

March 2015 Lab Tour for the students that organize the Purdue University Dance Marathon

April 2015 Lab Tour for the students that organize the Floyd Central High School Dance Marathon

February 2016 Indianapolis Public Schools Center for Inquiry Career Day guest speaker

March 2016 Lab Tour for the Galvin family donors of Pancreatic Cancer Research

April 2016 Lab tour for the Chuck Strong donors

April 2016 Carmel Dance Marathon presentation for Riley Children’s Foundation

August 2016 ChuckStrong Leadership Luncheon for IU Cancer Research

August 2016 IU Dance Marathon Parents’ Luncheon guest speaker

Sept 2016 St. Baldrick’s Foundation Lab Tour

Oct 2016 Career Panel for Riley Children’s Hospital visiting Peru High School students

Oct 2016 Riley Children’s Foundation demonstrations

Oct 2016 WTTV4 interview with Debby Knox, highlighting Fishel and Kelley labs’ pancreatic cancer project in bringing Ref-1 redox inhibitor to clinic (Airs Nov, 2016)

Sept 2017 Team Leader for Torch Relay, Children’s Miracle Network Hospitals, Indianapolis, IN

Internal Service

Cancer Biology Research Club, Director (2011 - present)

Peer Review and Mentoring Committee (PRMC) July 2016 - present

Journal Reviewer

Cancer Chemotherapy and Pharmacology (2004-present)

Molecular and Cellular Biochemistry (2008-present)

Hepatology (2011-present)

Anti-oxidant & Redox Signaling (2011-present)

Chemical Research in Toxicology (2010)

Bioorganic & Medicinal Chemistry (2011)

PLoS ONE (2011-present)

Molecular Carcinogenesis (2011-present)

Molecular Biology of the Cell (2013)

Cancer Medicine (2013)

Mutation Research (2014)

Editorial Boards

The Scientific World Journal (2011 – 2012)

Journal of Cellular and Molecular Medicine, Assoc Editor (2017- present)

Grant Reviews

Italian Ministry of Health, Cancer Program for Young Investigators, 7 proposals evaluated (August 2009)

Ad hoc Reviewer, Associazione Italiana per la Ricerca sul Cancro  (AIRC), Milano, Italy: “Temozolomide-based immuno-chemotherapy of haematological malignancies: preclinical and clinical studies,” Italian Ministry of Health (September 2009)

Indiana University Melvin and Bren Simon Cancer Center Translational Research Acceleration Collaboration (ITRAC), Preclinical Project Grants (2011, 2012)

Simmons Clinical Studies Fund, Wells Center for Pediatric Research (Nov. 2012)

Merilyn Hester Scholarship Fund, Indiana University Simon Cancer Center (June 2013)

Italian Ministry of Health, Health and Biomedical Research (June 2013)

Indiana Clinical and Translational Sciences Institute (CTSI), Pilot Funding for Research Use of Core Facilities (June 2014)

Indiana Clinical and Translational Sciences Institute (CTSI) Biorepository Grants (January 2015)

IUSCC American Cancer Society, Pilot Project Grants (May 2015)

Indiana Clinical and Translational Sciences Institute (CTSI), Pilot Funding for Research Use of Core Facilities (July 2015)

DOD Peer Review Cancer Research Program (Special Focus: Pancreatic Cancer) (Nov 2015)

IUSCC American Cancer Society, Pilot Project Grants (May 2016)

DOD Peer Review Cancer Research Program (Special Focus: Pancreatic Cancer) (Nov 2016)

NIH Study Section, Basic Mechanisms of Cancer Therapeutics, ad hoc Reviewer (Feb 2017)

National Cancer Institute reviewer for  “Consortium for Pancreatic Ductal Adenocarcinoma Translational studies on the Tumor Microenvironment” (June 2017)

Formal Research Seminars Given

February 2004 Chicago State University, Chicago, IL

April 2007 Pennington Biomedical Research Center, Baton Rouge, LA. A Workshop on Cellular

Trafficking, Signals, and Switches

February 2010 Roche Applied Science, Indianapolis, IN. “Targeting the two major functions of

APE1/Ref-1 in the fight against cancer”

October 2010 Roche Applied Science, Boston, MA. xCELLigence Symposium “Targeting the Redox Function of APE1/Ref-1 in Pancreatic Cancer”

Nov. 2012 Grand Rounds, IU Simon Cancer Center, Indianapolis, IN. “Dual Targeting of APE1/Ref-1 and STAT3 in Pancreatic Cancer”

May 2013 Acea Biosciences, Inc. Webinar Series, “Identifying Novel Combination Therapeutic Targets for Pancreatic Cancer:  Real-Time Label-Free Monitoring Cancer Cell Survival & Migration.”  

October 2013 Colloquium Series - University of Toronto Mississauga, Mississauga, ON Canada. “Probing the interaction between pancreatic tumor and its microenvironment”

February 2015 Pharmacology & Toxicology Seminar Series, Indianapolis, IN “Probing the interaction between pancreatic tumor and its microenvironment via modulation of the STAT3 pathway”

Informal Seminars

March 2007 Ovar’coming Together – Research Tours, Indiana University Simon Cancer Center, Indianapolis, IN

Dec. 2007 Pediatrics Hematology/Oncology Translational Research Group, “APE1 as a target in cancer”

March 2009 Pediatrics Hematology/Oncology Translational Research Group, “Molecular targeting in cancer with a focus on Ref-1”

Dec. 2009 Cancer Biology Research Club, “Two targets in one protein: APE1 and Ref-1 in cancer”

June 2010 Pediatrics Hematology/Oncology Clinical Research Group, “Targeting the redox function of APE1/Ref-1in the fight against cancer”

Nov. 2010 Cancer Biology Research Club, “APE1/Ref-1 as a target in pancreatic cancer”

Oct. 2011 Cancer Biology Research Club, “Targeting the redox function of APE1/Ref-1in the fight against pancreatic cancer”

August 2012 Pediatrics Hematology/Oncology Translational Research Group Dual Targeting of APE1/Ref-1 and STAT3 in Cancer”

May 2013 IUSCC Pancreatic Program Meeting; Update on Fishel Lab findings in PDAC

August 2013 Pediatric Faculty Research Seminar, “3-Dimensional Modeling in Cancer: Novel Targets and Combination Therapy”

March 2014 First Annual Hypoxic Cell Culture Research Talk and Roundtable Discussion, Indianapolis, IN. “Hypoxic Cell Culture Innovations in Cancer and Stem Cell Research”

Nov. 2014 Jordan-Rieger Fund for Pancreatic Cancer Fundraising Event

April 2017 Midwest Tumor Microenvironment Meeting, St. Louis, MO “Single-cell RNA sequencing and Reverse phase protein arrays identify novel roles and interacting partners for APE1 in Pancreatic Ductal Adenocarcinoma Microenvironment”, Post Doc Short talk

Current Grant Support

W81XWH-14-1-0525 (Jerde,PI); Co-I Fishel 09/30/14-09/29/17 0.6 calendar months

Dept. Of Defense Overcoming Drug Resistant Prostate Cancer with APE1/Ref-1 Blockade

Goals: Investigate the role of Ref-1 in regulating critical transcription factors and the role in Taxane drug resistance

R01 CA167291-01A1 Kelley/Fishel (co-PIs) 01/01/13-12/31/17 4.80 Calendar Months

NIH $444,567

Novel Role of Ref-1 in Pancreatic Etiology and Progression

Goals: Study the role and interaction of Ref-1 in tumor and stroma of PDAC.Goals: To determine the outcome of inhibiting the function of Ref-1 in PDAC as well as in the tumor microenvironment using both orthotopic and genetically engineered mouse models.

R01 (Konieczny/Flick/Fishel)

NIH 1.2 calendar moths in Years 3 and 4

Thrombin-dependent mechanisms of pancreatic ductal adenocarcinoma disease

Goals: Define the mechanisms by which Fibrin and PAR-1, the central effector molecules downstream of thrombin generation, accelerate PDAC pathogenesis

Role: Co-Investigator

DHART SPORE Development Research Program (Fishel, PI) 09/1/16 – 8/30/17

Targeting STAT3 with Potent, Salicylic Acid-based Inhibitors in Malignant Peripheral Nerve Sheath Tumors

Goal: Evaluate STAT3 as a target in MPNST

Pending

R01 (Fishel/Kelley/Han/Koneiczny, MPI) 09/01/17-08/31/21 2.4 calendar months

“Mechanistic identification of novel drug combinations using PDAC avatar for bench-to-clinic acceleration”

Goals: Establish an innovative in vitro pipeline of drug evaluation capable of studying the mechanism behind synergistic combinations of chemotherapy.

R01 (Kelley/Fehrenbacher, MPI) 09/01/17-08/31/21 0.6 calendar months

NIH $2,108,922

Targeting oxidative DNA damage to prevent and treat chemotherapy-induced peripheral neuropathy (CIPN) using a first-in-class APE1 modifier

Goals: Explore the DNA repair function of APE1 in protection again chemotherapy-induced neuropathy

Role: Co-Investigator

Previous Grant Support

1R43CA171344-01A1 Kelley (PI), Co-I; Fishel and Cardoso 07/01/13-06/30/15 (NCE)   0.5 Calendar Months (10% Effort) SBIR           $188,069                         

Novel Therapeutic Strategy for Refractory and Relapse Childhood Acute Leukemia

Goals: To develop a novel therapeutic strategy for childhood acute lymphoblastic leukemia (ALL), with focus on relapsed T-cell leukemia.

Fishel (PI) 8/1/14 – 7/31/15 No Salary Support

Eli Lilly PSI Pilot Funds in Oncology / IU Simon Cancer Center

Using the Transgenic and Knock-Out Mouse Core to understand the role of Ref-1 redox activity

Goals: Use CRISPR technology to knockout APE1/Ref-1 and then replace it with a redox deficient APE1/Ref-1

IUPUI Signature Center Initiative for the Cure of Glioblastoma (Fishel/Rickus) No Salary Support

4/1/14 – 3/31/16 $13,000

Multi-cellular, Multi-niche 3D Culture Model of Glioblastoma

Pilot Funding (Use of Core Facilities) (Fishel) 03/01/14 – 02/28/16 No Salary Support

CTSI/IU $9,060

Development of Novel Potent Salicylic Acid-Based STAT3 Inhibitors for Cancer Therapy

Pilot Funding (IU Health Strategic Research Initiative in Oncology) (Fishel) 06/01/14 – 05/30/15

No Salary Support $19,588

Pharmacokinetic / Pharmacodynamic studies for Newly Identified STAT3 Inhibitors. 

Indiana Simon Cancer Center Translational Research Acceleration Collaboration (ITRAC) (Ivan/Fishel/Radovich) 7/1/13- 6/30/14 No Salary Support $40,000

Roles of Hypoxia-Regulated Linc-RNAs in Pancreatic Cancer

Goals: Elucidate the role of Linc-RNAs in the resistance of Pancreatic Cancer

Fishel (PI) 1/1/14 – 6/31/14 No Salary Support

ACEA Biosciences Innovative Grant

Using the iCELLigence system to evaluate the impact of the tumor microenvironment upon tumor cell proliferation and migration after modulation of APE1

Goals: Testing the role of APE1 redox function in the interaction of dorsal root ganglia, pancreatic cancer cells, and cancer-associated fibroblasts as a mimic of the tumor microenvironment

Kelley (PI)   Fishel (co-PI)                           7/1/13-6/30/14       20% Effort

ApeX Therapeutics Contract                                                                                                                      

Testing ApeX compounds for efficacy in leukemia models

Goals:  Testing new Ref-1 redox inhibitors in leukemia models for efficacy

Indiana Simon Cancer Center Translational Research Acceleration Collaboration (ITRAC) (Fishel/Cardoso/Kelley) 6/1/11- 6/30/13 No Salary Support $50,000

“Ref-1/APE1 redox function as a novel molecular target in ALL”

Pilot Funding (Use of Core Facilities) (Fishel) 01/01/12-12/31/13 No Salary Support

CTSI/IU $10,000

Preclinical Studies of a Novel Dual Targeting Strategy for Pancreatic Cancer

Biomedical Research Grant (Fishel) 07/01/12-06/30/13 No Salary Support

$40,000

Role of APE1 in Cancer-Associated Fibroblasts and Stellate Cell Biology in Pancreatic Cancer

Program Project Planning P3 (Kelley/Howard) 10/01/11-04/01/13              0 calendar months

CTSI $100,000                  No salary support

Model for Transformative Science using a Multi-Investigative Team Approach  

Fishel/Cardoso Project #5 “Targeting two key survival pathways in pancreatic cancer” $20,000

Simmons Clinical Fund “Improved response to chemotherapy through inhibition of APE1 in Pediatric Brain Tumors” 01/01/10 – 12/31/10. PI: M.L. Fishel, Year 1 $25,000

Showalter Research Trust Fund Fishel (PI) 07/01/10 – 06/30/11

“Investigation of a novel target in pancreatic cancer: redox signaling protein APE1/Ref-1 and its effects on proliferation and metastatic potential”

NIH/NCI Pancreatic Pilot R21 CA122298-01A1 “Chemosensitization of Pancreatic Tumors via Inhibition of a DNA BER Enzyme, Ape1.” 05/01/07-04/30/09, PI: M.L. Fishel, 40% Effort, TDC – Year 1 $100,000; Year 2 $120,000.

Marsha Rivkin Center for Ovarian Cancer Research “Enhancement of ovarian cancer to chemotherapeutic agents, cisplatin and TMZ, using small molecules, BG and MX” 05/01/06 – 04/30/07, PI: M.L. Fishel, 30% Effort, TDC – $30,000

Ovar’coming Ovarian Cancer support grant; PI: Fishel, M.L.11/1/2006 – 10/30/2007. $25,000 direct costs for supplies only.

Patents

1. Combination Therapy for Treating Cancer, Mark Kelley and Melissa Fishel, Provisional Application Number 61/547,149

2. New Salicylic Acids Derivatives, Pharmaceutically Acceptable Salt thereof, Composition thereof, and Method of Use thereof, Patrick Gunning, Sina Haftchenary, Brent Page, Artee Luchman, Samuel Weiss, and Melissa Fishel, Patent Number 9650399, Issued 05/16/2017

3. Compounds and Methods for Treating Leukemia, Mark Kelley, Angelo Cardoso and Melissa Fishel, Patent Number 9315481, Issued 04/19/2016

4. Therapeutic Agent for Tuberous Sclerosis Complex (TSC), Mark R. Kelley and Melissa L. Fishel, Provisional Application Number 62/520,733

5. Methods of Targeting APE1/Ref-1 to Inhibit Hypoxia Signaling Genes, Mark R. Kelley and Melissa L. Fishel, Provisional Application Number 62/307,000

6. Use of APE/REF-1 Redox Specific Inhibitors for Treating Metastatic Prostate Cancer, Mark R Kelley, Travis Jerde, Melissa L Fishel, Provisional Application Number 62/450,125

7. Use of APE1/REF-1 Inhibitors in Combination Therapies for Treatment of Cancer, Mark R Kelley and Melissa L Fishel, Provisional Application Number 62/521,082

Publications

1. Xu, Y., Hansen, W.K., Rosenquist, T.A., Williams, D.A., Limp-Foster, M., and Kelley, M.R. (2001) Protection of mammalian cells against chemotherapeutic agents thiotepa, BCNU and mafosfamide using the DNA base excision repair genes Fpg and Ogg1; Implications for protective gene therapy applications. Journal of Pharmacology and Experimental Therapeutics, 296: 825-31.

2. Kreklau, E.K., Limp-Foster, M., Liu, N., Xu, Y., Kelley, M.R., and Erickson, L.C. (2001) A Novel Fluorometric Oligonucleotide Assay to Measure O6-Methylguanine DNA Methyltransferase, Methylpurine DNA Glycosylase, 8-Oxoguanine DNA Glycosylase, and Abasic Endonuclease Activities: DNA Repair Status in Human Breast Carcinoma Cells Overexpressing Methylpurine DNA Glycosylase. Nucleic Acids Research, 29: 2558-66.

3. Seo Y.R., Limp-Foster, M., Kelley M.R., and Smith, M.L. (2002). Role for p53 in base excision DNA repair: In vivo evidence. Oncogene 21: 731-7.

4. Fishel, M.L., Seo, Y.R., Smith, M.L., and Kelley, M.R. (2003) Imbalancing the DNA base excision repair pathway in the mitochondria; Targeting and overexpressing N-methylpurine DNA glycosylase in mitochondria leads to enhanced cell killing. Cancer Research, 63: 608-15.

5. Fishel, M.L., Delaney, S.M., Durtan, L. J., Hansen, R. J., Zuhowski, E.G., Moschel, R.C., Egorin, M.J., and Dolan, M.E. (2003) Enhancement of platinum-induced cytotoxicity by O6-benzylguanine, Molecular Cancer Therapeutics, 2:633-40.

6. Fishel, M.L., Newell, D.R., Griffin, R.J., Davison, R., Wang, L.-Z., Curtin, N., Zuhowski, E.G., Kasza, K., Egorin, M.J., Moschel, R.C., and Dolan, M.E. (2004) Effect of cell cycle inhibition on cisplatin cytotoxicity, J Pharmacol. Exp. Ther, 312(1): 206-13.

7. Fishel, M.L., Gamcsik, M.P., Zuhowski, E.G., Maher, V.M., Karrison, T., Moschel, R.C., Egorin, M.J., and Dolan, M.E. (2005) Role of GSH and NER in modulation of cisplatin activity with O6-benzylguanine, Cancer Chemother. Pharmacol., 55(4): 333-42.

8. Fishel, M.L., Rabik, C.A., Bleibel, W.B., Xinmin, L., Moschel, R.C. and Dolan, M.E. (2006) Role of GADD34 in modulation of cisplatin cytotoxicity. Biochem Pharm. 71: 239-47.

9. Fishel, M.L., He, Y., Smith, M.L. and Kelley, M.K. (2007) Manipulation of Base Excision Repair (BER) to Sensitize Ovarian Cancer Cells to Alkylating Agent Temozolomide, Clin. Can. Res. 13:74-81.

10. Fishel, M.L., He, Y., Reed, A.M., Chin-Sinex, H., Hutchins, G.D., Mendonca, M.S., and Kelley, M.R. (2008) Knockdown of the DNA repair and redox signaling protein Ape1/Ref-1 blocks ovarian cancer cell and tumor growth. DNA Repair. 7: 177-86.

11. Luo, M., Delaplane, S., Jiang, A., Reed, A., He, Y., Fishel, M., Nyland II, R.., Borch, RF., Qiao, X., Georgiadis, MM. and Kelley, MR. (2008) Role of the multifunctional DNA repair and redox signaling protein Ape1/Ref-1 in cancer and endothelial cells: Small molecule inhibition of Ape1’s redox function. Antioxidant and Redox Signaling, Nov; 10(11): 1853-1867. PMCID: PMC2587278.

12. Rabik, CA., Fishel, ML., Holleran, JL, Kasza, K., Kelley, MR., Egorin, MJ. and Dolan, ME. (2008) Enhancement of cisplatin cytotoxicity by O6-benzylguanine involves endoplasmic reticulum stress. J Pharmacol Exp Ther. Nov;327(2):442-52 PMID: 18664592

13. Jiang Y, Guo C, Fishel ML, Wang Z-Y, Vasko MR and Kelley MR. (2009) Role of APE1 in differentiated neuroblastoma SH-SY5Y cells in response to oxidative stress; Use of APE1 small molecule inhibitors to delineate APE1 functions. DNA Repair Nov 2, 8(11): 1273-82 PMCID: PMC2765866

14. Tate, C.M., Fishel, M.L., Holleran, J.L., Egorin,M.J., and Skalnik, D.G. (2009) Embryonic Stem Cells Lacking the Epigenetic Regulator Cfp1 are Hypersensitive to DNA-Damaging Agents and Exhibit Decreased Ape1/Ref-1 Protein Expression and Endonuclease Activity. DNA Repair (Amst), 8(12): 1411-23. PMCID: PMC2787775.

15. Bapat A, Glass, LS, Luo M, Fishel ML, Long EC, Georgiadis MM, Kelley MR. (2010) Novel small molecule inhibitor of Ape1 endonuclease blocks proliferation and reduces viability of glioblastoma cells. Journal of Pharm and Exp Therapeutics, September 334(3): 988-998. PMCID: PMC2939666.

16. Jiang Y, Zhou S, Sandusky GE, Kelley MR, and Fishel ML. (2010) Reduced expression of DNA repair and redox signaling protein APE1/Ref-1 impairs human pancreatic cancer cell survival, proliferation, and cell cycle progression. Cancer Investigation, 28 (9): 885-895. PMCID: PMC2966714.

17. Fishel ML, Colvin ES, Luo M, Kelley MR, Robertson KA (2010) Inhibition of the Redox Function of APE1/Ref-1 in Myeloid Leukemia Cell Lines Results in a Hypersensitive Response to Retinoic Acid-induced Differentiation and Apoptosis. Experimental Hematology. Dec; 38(12):1178-88. PMCID: PMC2998713

18. Fishel ML, Jiang Y, Rajeshkumar NV, Scandura G, Sinn AL, He Y, Shen C, Jones DR, Pollok KE, Ivan M, Maitra A, Kelley MR. (2011). Impact of APE1/Ref-1 Redox Inhibition on Pancreatic Tumor Growth. Molecular Cancer Therapeutics. Sep;10(9):1698-708.PMCID: PMC3170439.

19. Cardoso AA, Jiang Y, Luo M, Reed AR, He Y, Kelley MR, Fishel ML. (2012) APE1/Ref-1 Regulates STAT3 Transcriptional Activity and APE1/Ref-1-STAT3 Dual-Targeting Effectively Inhibits Pancreatic Cancer Cell Survival. PLos One, (10): e47462 PMCID: PMC3477158

20. Parajuli B, Georgiadis TM, Fishel ML, Hurley TD. (2014) Development of Selective Inhibitors for Human Aldehyde Dehydrogenase 3A1 (ALDH3A1) for the Enhancement of Cyclophosphamide Cytotoxicity. ChemBioChem. 2014 Mar 21;15(5):701-12. PMCID: PMC4043872

21. Parajuli B, Fishel ML, Hurley TD. (2014) Selective ALDH3A1 inhibition by potent benzimidazole analogs increase mafosfamide sensitivity in cancer cells. Journal of Medicinal Chemistry. Jan 23;57(2):449-61. PMCID: PMC3988914

22. Shannon HE*, Fishel ML*, Xie J, Gu D, McCarthy BP, Riley AA, Sinn AL, Silver JM, Peterman K, Kelley MR, Hanenberg H, Korc M, Pollok KE, Territo PR. (2015) Longitudinal Bioluminescence Imaging of Primary versus Abdominal Metastatic Tumor Growth in Orthotopic Pancreatic Tumor Models in NOD/SCIDy(-/-) Mice. Pancreas. Jan; 44(1): 64-75. PMCID: PMC4262664 *Corresponding authors

23. Fishel ML, Wu X, Devlin CM, Logsdon DP, Jiang Y, Luo M, He Y, Yu Z, Tong Y, Lipking KP, Maitra A, Rajeshkumar NV, Scandura G, Kelley MR, Ivan M. (2015) Apurinic/Apyrimidinic Endonuclease /Redox Factor-1 (APE1/Ref-1) redox function negatively regulates NRF2. J Biol Chem. Jan 30;290(5):3057-68. PMCID: PMC4317024.

24. Arpin CC, Mac S, Jiang Y, Cheng H, Page BDG, Kamocka MM, Haftchenary S, Su H, Ball D, Rosa DA, Lai P-S, Gomez-Biagi RF, Ali AM, Rana R, Hanenberg H, Kerman K, McElyea KC, Sandusky GE, Gunning PT*, Fishel ML*. (2016) Applying small molecule STAT3 protein inhibitors as pancreatic cancer therapeutics. Mol Cancer Ther. 15(5), 794-805 PMID: 26873728 * Co-last authorship

25. Logsdon DP, Grimard M, Luo M, Shahda S, Jiang Y, Tong Y, Yu Z, Zyromski N, Schipani E, Carta F, Supuran CT, Korc M, Ivan M, Kelley MR, Fishel ML*. Regulation of HIF1α under hypoxia by APE1/Ref-1 impacts CA9 expression: Dual-targeting in patient-derived 3D pancreatic cancer models. Mol Cancer Ther 2016 Nov;15(11):2722-32 PMID: 27535970. * Corresponding author.

26. Kelley MR, Wikel JH, Guo C, Pollok KE, Bailey BJ, Wireman R, Fishel ML, MR Vasko. Identification of new chemical entities targeting APE1 for the prevention of chemotherapy-induced peripheral neuropathy (CIPN). J Pharmacol Exp Ther 2016 Nov;359(2):300-09 PMID: 27608656.

27. Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE, Hill R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2016 Sept, doi: 10.1038/onc.2016.353, PMID: 27669441.

28. Lee J, Yakubov B, Ivan C, Jones D, Caperell-Grant A, Fishel ML, Cardenas H, and Matei D. Tissue Transglutaminase Activates Cancer-Associated Fibroblasts and Induces Gemcitabine Resistance in Pancreatic Cancer. Neoplasia 2016 Nov, 18:689-98, .

29. Zeleniak AE, Huang W, Brinkman MK, Fishel ML, and Hill R. Loss of MTSS1 Results in Increased Metastatic Potential in Pancreatic Cancer. (2017) Oncotarget Mar 7;8(10):16473-87. doi: 10.18632/oncotarget.14869. PMID:28146435.

30. Huang W, Zeleniak AE, Fishel ML, and Hill R. (2017) PTEN-Dependent Stabilization of MTSS1 (Metastasis Suppressor 1) Promotes Primary Cilia Formation in Pancreatic Cancer-Associated Fibroblasts; Submitted to Journal of Biological Chemistry.

31. Ding, J, Fishel ML, Reed AM, Ketchem E, Czader M, Cardoso AA, and Kelley MR. (2017) Ref-1/APE1 as Transcriptional Regulator and Novel Therapeutic Target in Pediatric T-cell Leukemia; Mol Cancer Ther. 2017 Apr 26. pii: molcanther.0099.2017. doi: 10.1158/1535-7163.MCT-17-0099. [Epub ahead of print], PMID:28446640

32. Prabhu L, Wei H, Chen L, Demir O, Sandusky G, Sun E, Wang J, Mo J, Zeng L, Fishel ML, Safa A, Amaro R, Korc M, Zhang Z-Y, Lu T. (2017) Adapting AlphaLISA high throughput screen to discover a novel small-molecule inhibitor targeting protein arginine methyltransferase 5 in pancreatic and colorectal cancers. Oncotarget, doi:  10.18632/oncotarget.18102. PMCID: PMC5522311.

33. Shah F*, Goossens E*, Atallah NM, Grimard M, Kelley MR and Fishel ML. (2017) Characterizing gene expression changes and identifying novel pathways following APE1/Ref-1 knockdown in Pancreatic Ductal Adenocarcinoma using Single-cell RNA Sequencing. Mol Oncol. 2017 Sep 18. doi: 10.1002/1878-0261.12138. [Epub ahead of print]. PMID: 28922540 *Co-First authors.

34. McIlwain DW, Fishel ML, Kelley MR, Jerde TJ. (2017) APE1/Ref-1 Redox-Specific Inhibition Decreases Survivin Protein Levels and Induces Cell Cycle Arrest in Prostate Cancer Cells. Oncotarget, In Press.

35. Herrera-Perez M, Voytik-Harbin SL, Sarkaria JN, Pollok KP, Fishel ML, Rickus J. (2017) Presence of stromal cells in a bioengineered tumor microenvironment alters glioblastoma migration and response to STAT3 inhibition. Submitted to Cancer Letters.

36. Shouksmith AE, Grimard M, Geletu M, Luchman HA, Heaton WL, de Araujo ED, Berger-Becvar A, Gawel JM, Bakhshinyan D,Adile AA,_VenugopalC, Johns AE, Al-Qaysi O, Lewis AM, O’Hare T, Deininger MW, Singh SK, Weiss S, Fishel ML, and Gunning PT. (2017) Identification and characterization of AES-135, a potent HDAC inhibitor with biological effects in 3D patient-derived tumour spheroids of pancreatic cancer. Submitted to Journal of Medicinal Chemistry.

Reviews

1. Evans, A., Limp-Foster, M., and Kelley, M.R. (2000) Going APE over Ref-1. Mutation Research, 40043:1-26.

2. Limp-Foster, M. and Kelley, M.R. (2000) DNA repair and gene therapy : Implications for translational uses. Environmental and Molecular Mutagenesis, 35:71-81.

3. Fishel, M.L., Vasko, M.R. and Kelley, M.R. (2006) DNA Repair in Neurons: So if they don’t divide, what’s to repair? Mutation Research, 2006 Jul 29.

4. Kelley, M.R. and Fishel, M.L. (2007) DNA repair proteins as molecular targets for cancer therapeutics. Anticancer Agents in Medicinal Chemistry, 8(4): 417–25.

5. Fishel, M.L. and Kelley, M.R. (2008) The DNA base excision repair protein Ape1/Ref-1 as a therapeutic and chemopreventive target. Molecular Aspects of Medicine 28:375-95.

6. Bapat, A., Fishel, M.L., and Kelley, M.R. (2009) Going Ape as an Approach to Cancer Therapeutics. Antioxid RedoxSignaling, 11: 651-68.

7. Reed AM, Fishel ML, Kelley MR. (2009) Small molecule inhibitors of proteins involved in base excision repair potentiate the anti-tumorigenic effect of existing chemotherapeutics and irradiation. Future Oncol. June 5(5): 713-26. PMCID: PMC2756177

8. Kelley MR, Georgiadis MM, Fishel ML. (2012) APE1/Ref-1 Role in Redox Signaling: Translational Applications of Targeting the Redox Function of the DNA Repair/Redox Protein APE1/Ref-1. Current Molecular Pharmacology 5 (1): 36-53. PMCID: PMC3319314.

9. Kelley MR, Logsdon D, Fishel ML. (2014) Targeting DNA repair pathways for cancer treatment: what’s new? Future Oncology 10(7): 1215-37. PMCID: PMC4125008

10. Zimmers TA, Fishel ML, Bonetto, A. (2016) STAT3 in the systemic inflammation of cancer cachexia. Seminars in Cell and Developmental Biology, 54:28-41. PMID: 26860754

11. Shah F, Logsdon DP, Fehrenbacher J, Kelley MR, and Fishel ML. (2017) Exploiting the APE1-Ref-1 node in cancer signaling and other diseases: from bench to clinic. Npj Precis Oncol, 1(19); doi:10.1038/s41698-017-0023-0. PMID: 28825044

Book Chapters

1. Kelley, MR, Georgiadis, MM, Fishel, ML. (2010) DNA Repair and Redox Signaling. In: The Tumor Microenvironment (Bagley, R., ed.) Springer (part of Springer Science+Business Media), New York, NY. pp 133-168.

2. Vascotto, C. & Fishel, M.L. (2012) Blockade of Base Excision Repair:  Inhibition of small lesions results in big consequences to cancer cells, in DNA Repair in Cancer Therapy: Molecular Targets and Clinical Implications, Kelley, M., Ed. Academic Press/Elsevier: 29-54

3. Fishel ML, Vascotto C, Kelley MR. (2013) DNA Base Excision Repair Therapeutics: Summary of BER targets with a focus on APE1. (233-287)  In: Madhusudan S, Wilson III D, editors. DNA Repair and Cancer:  Bench to Clinic. Enfield, NH Science Publishers, an imprint of Edenbridge Ltd, British Channel Islands. 

8. Kelley, MR and Fishel, ML. (2013) DNA Repair and Cancer Therapeutics. In Principles of Molecular Diagnostics and Personalized Cancer Therapy. Lippincott Williams & Wilkins. p.566-579 

9. Fishel, ML and Kelley, MR. (2016) Introduction and Overview of DNA Repair Targets: From Bench to Clinic. In DNA Repair in Cancer Therapy: Molecular Targets and Clinical Applications (2nd Edition). Fishel, M.L. & Kelley, M., Ed. Academic Press/Elsevier.

Abstracts

1. Xu, Y., Hansen, W. K., Clapp, W., New, S., Deutsch, W. A., Limp-Foster, M., Roberts, A., Williams, D. A., and Kelley, M. R. Retroviral mediated protection against chemotherapeutic agents using the DNA base excision repair genes fpg and Ogg1. AACR annual meeting, San Diego, CA; 1998.

2. Limp-Foster, M., Xu,Y., Williams, D.A., and Kelley, M.R. Gene transfer of the human DNA base excision repair (BER) N-methylpurine-DNA glycosylase to sensitize tumor cells to chemotherapy. International Conference of Gene Therapy, San Diego, CA; Nov. 1998.

3. Limp-Foster, M., Xu, Y., Williams, D. A., and Kelley, M. R. DNA base excision repair (BER) and gene transfer: Use of the human N-Methylpurine DNA glycosylase (MPG) to sensitize tumor cells to chemotherapy. Environmental Mutagen Society annual meeting, Washington, DC; April 1999.

4. Moore, D.H., Michael, H., Tritt, R. Parsons, S.H., Limp-Foster, M., and Kelley, M.R. Altered expression of the multifunctional DNA repair/redox Ape/ref-1 enzyme in ovarian cancer. AACR annual meeting, San Francisco, CA; April 2000.

5. Limp-Foster, M., Xu,Y., Hansen, W.K., Parsons, S.H., Tritt, R., Kobune, M., Williams, D.A., and Kelley, M.R. DNA base excision repair (BER) genes in gene therapy: Translational applications. DNA Repair Defects Meeting, AACR Special Conference, San Diego, CA; Jan. 2000.

6. Limp-Foster, M., Rinne, M., Xu, Y., and Kelley, M.R. Imbalancing DNA Base Excision Repair (BER): Use of Nuclear and Mitochondrial-Targeted Human N-Methylpurine DNA Glycosylase (MPG) to Sensitize Breast Cancer Cells to Low Dose Chemotherapy. AACR Annual Meeting, New Orleans, LA; March 2001.

7. Fishel, M.L., Connell, P., Siddiqui, N., and Dolan, M.E. Effect of O6-benzylguanine on nitrogen mustard- and cisplatin-induced toxicity in head and neck cancer cells. AACR Annual Meeting, San Francisco, CA April 2002.

8. Seo, Y-R., Fishel, M. L., Kelley, M.R. and Smith, M.L. Implication of p53 in base excision DNA repair: in vivo evidence. AACR Annual Meeting, San Francisco, CA, April 6-10, 2002

9. Fishel, M.L., Zuhowski, E.G., Friesen, L.D., Egorin, M., and Dolan, M.E. Enhancement of platinum-induced toxicity and DNA damage with O6-Benzylguanine (O6-BG). AACR Annual Meeting, Washington DC July 2003.

10. Fishel, M.L., Li, X., and Dolan, M.E. Genes important in modulation of cisplatin cytotoxicity. Annual Meeting of the American Association for Cancer Research, Orlando, FL March, 2004.

11. Reed, A., Fishel, M., Jiang, Y., Pollok, K. and Kelley, M.R. Enhancement of temozolomide (TMZ) brain tumor cell killing using a small molecule inhibitor of the human apurinic/apyrimidinic endonuclease DNA base excision repair enzyme (Ape1/Ref-1) function. American Society of Pediatric Hematology/Oncology (ASPHO) and Society of Pediatric Research (SPR) annual meeting; May, 2005.

12. Fishel, M.L., He, Y., Dolan, M.E. and Kelley, M.R. Small molecule enhancement of cisplatin and temozolomide chemotherapeutic agents for the treatment of ovarian cancer. EORTC Meeting, Philadelphia, PA November 2005.

13. Rabik, C.A., Fishel, M.L. Dolan, M.E. The role of the ER stress pathway in modulation of platinating agents. Annual Meeting of the American Association for Cancer Research, Anaheim, CA 2005.

14. Fishel, M.L., He, Y., Cai, S., Pollok, K.E., and Kelley, M.R. Enhancement of ovarian cancer treatment via manipulation of the base excision repair pathway after temozolomide treatment. Annual Meeting of the American Association for Cancer Research, Washington DC April 2006.

15. Rabik CA, Fishel ML, Delaney, S.M., Ottley, C.J., Pearson, D.G., Tilby, M.J., Dolan, M.E. O6-Benzylguanine enhances DNA damage caused by cisplatin in head and neck cancer cell lines. Annual Meeting of the American Association for Cancer Research, Washington DC April 2006.

16. Fishel M, He Y, Dolan ME, and Kelley MR. Enhancement of chemotherapeutic agents for the treatment of ovarian cancer. Molecular Targets and Cancer Therapeutics AACR meeting. April, 2006.

17. Bapat A, Fishel ML, Georgiadis MM, and Kelley MR. High Throughput Screen for Inhibitors of APE1. Indiana University Cancer Research Day, Indianapolis, IN April 2007.

18. Fishel ML, Jiang Y, Reed AM, He Y, and Kelley MR. Sensitization of pancreatic cancer cells via inhibition of Ape1, a DNA base excision repair enzyme and redox signaling protein. Annual Meeting of the American Association for Cancer Research, Denver, CO April 2009.

19. Fishel ML, Reed AM, Luo M, and Kelley MR. Inhibition of APE1/Ref-1 Redox Function as an Anti-Angiogenic Molecular Target. Annual Meeting of the American Association for Cancer Research, Denver, CO April 2009.

20. Fishel ML, Reed AM, Luo M, and Kelley MR. Inhibition of APE1/Ref-1 Redox Function as an Anti-Angiogenic Molecular Target. American Association for Cancer Research Annual Meeting, Denver, CO 2009.

21. Fishel ML & Kelley MR. Sensitization of Pancreatic Cancer Cells via Inhibition of APE1/Ref-1, a DNA Base Excision Repair and Redox Signaling Protein: Novel target and small molecule development. NCI Translational Science Meeting, Vienna, VA 2009.

22. Bapat A, Glass LS, Luo M, Fishel ML, Reed A, Long EC, Georgiadis MM, Kelley MR. Novel small molecule inhibitor of the endonuclease function of the APE1 DNA repair and redox signaling enzyme blocks proliferation and reduces viability of glioblastoma cells. AACR 101st Annual Meeting, Washington DC, April 17 – 21, 2010.

23. Fishel ML, Jiang Y, Reed AM, Kelley MR, and Cardoso AA. Dual Targeting of APE1/Ref-1 and STAT3 in pancreatic cancer cells: dramatic effect on survival. Indiana University Cancer Research Day, Indianapolis, IN May 2011.

24. Fishel ML, Jiang Y, Luo M, Reed AM, He Y, Cardoso AM, Kelley, MR.C173/C173 - APE1/Ref-1-STAT3 dual-targeting synergize to effectively inhibit pancreatic cancer cell survival. Molecular Targets and Cancer Therapeutics, San Francisco, CA, November 12 – 16, 2011.

25. Shahda S, Kelley MR, and Fishel ML. Inhibition of APE1 in PDAC cells results in reduction in hypoxia signaling gene expression in vitro, Indiana University Cancer Research Day, Indianapolis, IN May 2012. Honorable mention in Fellow Poster Presentations.

26. Ding J, Luo M, Czader M, Reese R, Batra S, Carlesso N, Fishel ML, Kelley MR, Cardoso AA. Ref-1 is master regulator of leukemia T-cell survival, and a new therapeutic target for relapsed childhood acute lymphoblastic leukemia. Oral Presentation at the 2012 Midwest Blood Club, Indianapolis, IN, March 15 – 16, 2012.

27. Miller SJ, Wenning MJ, Bills RG, Sliva P, Labarrere CA, Kelley MR, Fishel ML, Unthank JL.  Impact of redox signaling inhibition on collateral growth in young, healthy rats. Experimental Biology 2013, Boston, MA April 20-24, 2013. Published in FASEB J. 2013, 27:685.2

28. McIlwain D, Fishel ML, Wang L, Snider B, Zhang JT, Kelley MK, Jerde TJ. APE1/Ref-1 regulates survivin-mediated drug resistance in prostate cancer cells. Journal of Urology, 189 (4): Sup e991, 2013.

29. McIlwain DM, Fishel ML, Wang L, Snider BM, Zhang JT, Kelley MR, Jerde TJ. APE1/REF-1 regulates survivin-mediated drug resistance in prostate cancer cells. Presented at the Annual meeting of the American Urological Association, San Diego, CA, May 6, 2013.

30. Unthank JL, Kelley MR, Fishel ML, Barrere C, Miller S. Redox Signaling Inhibition and Collateral Growth/ MCS President’s Symposium II: Rapid Fire Discussion of Novel Trends at Experimental Biology Boston, MA, April 20, 2013

31. McIlwain DM, Fishel ML, Wang L, Snider BM, Zhang JT, Kelley MR, Jerde TJ. APE1/REF-1 regulates survivin-mediated drug resistance in prostate cancer cells. Presented at the Annual meeting of the Society for Basic Urological Research (SBUR) “10th World Congress on Urological Research”, Nashville, TN, November 21-24, 2013

32. McIlwain DM, Fishel ML, Wang L, Zhang JT, Kelley MR, Jerde TJ. Redox Factor 1 regulates drug resistance in prostate cancer cells via survival protein induction. Presented at the American Association for Cancer Research-Prostate Cancer Foundation Special Conference on Advances in Prostate Cancer Research, San Diego, CA, January 18-21, 2014

33. McIlwain DM, Fishel ML, Wang L, Snider BM, Zhang JT, Kelley MR, Jerde TJ. APE1/REF-1 regulates survivin-mediated drug resistance in prostate cancer cells. 2014 John P. Donohue Visiting Professor Series & First Annual Indiana Basic Urological Research Symposium, Indianapolis, IN, February 28, 2014

34. Shannon HE, Fishel ML,, Xie J, Gu D, McCarthy BP, Riley AA, Sinn AL, Silver JM, Kelley MR, Hanenberg H, Korc M, Pollok KE and Territo PR. Longitudinal bioluminescence imaging of primary versus abdominal metastatic tumor growth in orthotopic pancreatic tumor models in NOD/SCID((-/-) mice. American Association for Cancer Research, 2014.

35. Fishel ML, Cheng H, Kamocka MM, Hanenberg H, Zyromski N, Korc M, Kelley MR. Redox factor 1 (Ref-1) signaling in the interaction between pancreatic tumor cells and cancer-associated fibroblasts. Pancreatic Cancer: Innovations in Research and Treatment, AACR special conference, May 2014.

36. Fishel ML, Cheng H, Kamocka MM, Hanenberg H, Zyromski N, Korc M, Kelley MR. Redox factor 1 (Ref-1) signaling in the interaction between pancreatic tumor cells and cancer-associated fibroblasts. Indiana University Simon Cancer Center, Cancer Research Day, May 2014.

37. McIlwain DM, Fishel ML, Wang L, Snider BM, Zhang JT, Kelley MR, Jerde TJ. APE1/REF-1 regulates survival proteins in prostate cancer, CTSI National Conference, April 2015.

38. Fishel ML, Logsdon DP, Grimard ML, Supuran CT, Zyromski N, Ivan M, Kelley MR, Shah F. Targeting Ref1/APE1 pathway inhibition in pancreatic cancer using APX3330 for clinical trials, AACR Annual Meeting, New Orleans, LA, April 2016.

39. McElyea KC, Jacobsen MH, Schmidt M, Cheng H, Kelley MR, Sandusky GE, Fishel ML. Efficacy study of APX3330, a Ref-1 redox inhibitor, and Gemcitabine in a mouse pancreatic ductal adenocarcinoma model. AACR Annual Meeting, New Orleans, LA, April 2016.

40. Fishel ML, Grimard ML, Kelley MR, Rosa DA, Shouksmith A, Tin G, Park J, Gunning PT. Development of STAT3 dual-targeting strategies for the treatment of pancreatic cancer. AACR Annual meeting, New Orleans, LA, April 2016.

................
................

In order to avoid copyright disputes, this page is only a partial summary.

Google Online Preview   Download