Dr. Emmanuel Katsanis Biography

Emmanuel Katsanis Biography

Dr. Emmanuel Katsanis, is a Professor of Pediatrics, Medicine, Immunobiology and Pathology at the University of Arizona College of Medicine, Tucson.  He holds the Louise Thomas Endowed Chair in pediatric cancer research and the Peter and Paula Fasseas Chair in cancer research.

Dr. Katsanis was born in Quebec City and grew up in Montreal, Canada. He received his M.D. degree (1980) from the National and Kapodistrian University of Athens Medical School in Greece.  He then returned to Canada for postdoctoral research (1981–1982) followed by a rotating internship (1982–1983) at McGill University in Montreal, and a pediatric residency (1983–1987) at the Children’s Hospital of Eastern Ontario/University of Ottawa.  He then moved to the University of Minnesota for his fellowship training in pediatric hematology, oncology and bone marrow transplantation (1987–1990) followed by additional postdoctoral work in tumor and transplant immunology (1990–1991) leading to an Assistant Professor position in the Department of Pediatrics at the University of Minnesota (1991-1997).  In 1997, he relocated to the University of Arizona and was promoted to Associate Professor (tenured) in 1998 and Professor in 2004. He became the Division Chief of Pediatric Hematology-Oncology in 2005 and the Director of the University of Arizona Cancer Center’s Hematopoietic Cell Therapy and Transplant Program in 2012.

Dr. Katsanis has published over 130 peer-reviewed articles and 11 book chapters in the areas of cancer immunology and hematopoietic stem cell transplantation. He has received extramural funding from Medical Research Council of Canada, the National Institutes of Health, the Department of Defense, the Canadian and American Cancer Societies, and the Leukemia and Lymphoma Society. He has served on numerous NIH grant study sections and other national and international granting organizations and several journal editorial boards. Dr. Katsanis has received numerous awards for his research, some of which include the Gordon E. Richard Award from the Canadian Cancer Society, the Young Investigator Award from American Society of Pediatric Hematology/Oncology, the Irvine McQuarrie Research Scholar Award, the Clinical Oncology Career Development Award from the American Cancer Society and the Leading-Edge Researcher Award from the University of Arizona.

As a post-doctor fellow and junior faculty member at the University of Minnesota, his research focused on encapsulation of interleukin-2 (IL-2) into liposomes for improved biodistribution and enhanced in vivo immuno-stimulation1. Combination of liposomal IL-2 with anti-CD3 and IL-2 activated and propagated T cells exhibited superior in vivo anti-tumor activity in murine tumor models2. Moreover, in the human system he demonstrated that anti-CD3 plus IL-2 can generate effector cells with high cytolytic function in heavily treated patients with leukemia/lymphoma or following hematopoietic cell transplantation (HCT)3. Additionally, he investigated the in vivo effects of cyclophosphamide on IL-2-induced cytolytic function demonstrating that CY may be used more effectively in IL-2-based immunotherapy if consideration is given to timing4. Subsequent work focused on elucidating anti-neuroblastoma immune responses to various genetically modified tumor cell vaccines in an orthotopic retroperitoneal murine neuroblastoma model that his laboratory developed5. More translational work evaluated novel cytokine approaches in HCT to enhance anti-lymphoma/leukemia responses. Findings included accelerated hematopoiesis observed in a phase I/II clinical trial with IL-1a and effective generation of anti-tumor activity post-HCT with IL-15 and with less toxicity than that seen with IL-26,7.

After moving to the University of Arizona, his laboratory discovered a novel technique for enriching multiple chaperones from tumor lysate using solution-isoelectric focusing8. They demonstrated that tumor-derived chaperone-rich cell lysates (CRCL) carry antigenic peptides, which can be taken up by dendritic cells (DCs) and cross-presented to T cells9. Tumor-derived CRCL vaccines induced protective immune responses against a diverse range of murine tumor types in different genetic backgrounds. When compared to purified heat shock protein 70 (HSP70), single antigenic peptide or unfractionated lysate, CRCL had superior ability to activate/mature DCs and was able to induce potent, long lasting and tumor specific T-cell-mediated immunity10. Moreover, CRCL based vaccines were effectively combined with chemotherapeutic agents11. CRCL vaccine moved to phase I clinical trials. His research program expanded into investigating the regulatory networks involved in tumor-induced immunosuppression in different settings and designing strategies that overcome cancer escape. His laboratory first observed that imatinib mesylate a tyrosine kinase inhibitor can target T regulatory cells (Treg) and be used as an immunomodulatory agent12. Additionally, his research team provided important insights on the cross-talk between myeloid derived suppressor cells (MDSCs) and Treg13.

More recent work in Dr. Katsanis’ laboratory has focused on pre- and post-hematopoietic cell transplant (HCT) immune modulation by bendamustine and its effects on immune reconstitution, graft-versus-host disease and graft-versus-tumor effects. Promising findings in preclinical models14 with post-transplant bendamustine led to a Phase Ia/Ib investigator initiated trial evaluating clinical endpoints and effects on immune reconstitution15. Additional ongoing preclinical research in his laboratory is in the cancer vaccine arena studying an implantable device platform.

 

1.            Anderson PM, et al. Cancer Res. 1990;50(6):1853-1856.

2.            Loeffler CM, et al. Cancer Res. 1991;51(8):2127-2132.

3.            Katsanis E, et al. Blood. 1991;78(5):1286-1291.

4.            Katsanis E, et al. Cancer Immunol Immunother. 1991;34(2):74-78.

5.            Katsanis E, et al. Cancer Gene Ther. 1996;3(2):75-82.

6.            Katsanis E, et al. Transplantation. 1996;62(6):872-875.

7.            Katsanis E, et al. J Clin Immunol. 1994;14(3):205-211.

8.            Graner MW, et al. Cancer Immunol Immunother. 2003;52(4):226-234.

9.            Zeng Y, et al. Blood. 2003;101(11):4485-4491.

10.          Feng H, et al. Blood. 2003;101(1):245-252.

11.          Zeng Y, et al. Int J Cancer. 2004;110(2):251-259.

12.          Larmonier N, et al. J Immunol. 2008;181(10):6955-6963.

13.          Centuori SM, et al. J Leukoc Biol. 2012;92(5):987-997.

14.          Stokes J, et al. Br J Haematol. 2016;174(1):102-116.

15.          Katsanis E, et al. eJHaem 2020;1(May 26):1-7.