The objective of this research proposal is to maximize the cure rates of relapsed or refractory (R/R) non-Hodgkin lymphomas (NHL) by enhancing the capacity for targeted radionuclide therapies to comprehensively eliminate residual disease. NHL is the most commonly diagnosed hematologic malignancy in the United States and several subtypes portend poor outcomes, including so called ?double hit? and ?triple hit? disease. In addition, indolent lymphoma remains generally incurable. While CAR T (chimeric antigen receptor T cell therapy) offers promise for R/R diffuse large B cell lymphoma, durable complete response rates have been reported in only 30-40% of patients. NHL relapse is believed to result from the persistence of small tumor cell reserves that elude treatment. The notable heterogeneity of NHL contributes to malignant cell escape and complicates a uniform approach to therapy. Independent of responsiveness to other interventions however, virtually all NHL is exquisitely radiation sensitive, giving radionuclide based approaches a unique advantage. Radiolabeled anti-CD20 antibodies can achieve remissions in up to 95% of lymphoma patients failing conventional combination chemotherapy when myeloablative doses of the radioimmunotherapy (RIT) are administered with stem cell support. While these responses are encouraging, low tumor-to non-tumor ratios of absorbed radiation, toxicity, and unacceptable rates of relapse form the rationale for the ongoing work of this grant proposal. The proposed projects leverage novel RIT delivery methods developed during prior funding periods and shown to dramatically improve tumor-to- normal organ ratios of absorbed radiation. Clinical experience dictates that multi-agent combinations are generally most effective in the treatment of NHL. This proposal explores three RIT combination strategies with a goal of generating unvarying efficacy across NHL subtypes through targeting the presumptive cause of relapse, occult residual disease. To achieve this goal, Aim 1 will employ the rational pairing of two targeted radionuclides, first using the longer path length and modest linear energy transfer (LET) of ?-emitter therapy to safely debulk NHL tumors, followed by an extremely high LET and short path length ?-emitter to eliminate residual disease.
Aim 2 will explore combining the ability of RIT to trigger tumor cell death with the pro-apoptotic efficacy of emergent BH3 mimetics.
Aim 3 will examine RIT combined with rationally selected immune checkpoint inhibitors, in a novel tumor vaccine approach to eradicating residual disease. Collectively these proposed studies test the hypothesis that RIT combination approaches will reduce the total radiation dose required to effectively treat NHL, in a manner that facilitates rapid translation through our non-Hodgkin lymphoma clinical RIT program.

Public Health Relevance

Approximately 77,200 Americans will be diagnosed with non-Hodgkin?s lymphoma in 2020 and fewer than half will be cured with conventional treatments. Lymphomas are exquisitely sensitive to radiation and radiolabeled monoclonal antibodies can be very effective at inducing remission in most lymphoma patients, but the disease frequently recurs. This grant proposal describes the development of several innovations that combine radiolabeled antibodies with each other, or with other drugs, to prevent relapse by comprehensively eliminating every lymphoma cell.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
2R01CA076287-20
Application #
10210966
Study Section
Radiation Therapeutics and Biology Study Section (RTB)
Program Officer
Capala, Jacek
Project Start
1998-01-01
Project End
2026-01-31
Budget Start
2021-02-01
Budget End
2022-01-31
Support Year
20
Fiscal Year
2021
Total Cost
Indirect Cost
Name
Fred Hutchinson Cancer Research Center
Department
Type
DUNS #
078200995
City
Seattle
State
WA
Country
United States
Zip Code
98109
Green, Damian J; O'Steen, Shyril; Lin, Yukang et al. (2018) CD38-bispecific antibody pretargeted radioimmunotherapy for multiple myeloma and other B-cell malignancies. Blood 131:611-620
Greenbaum, Adam M; Green, Damian J; Holmberg, Leona A et al. (2018) Bendamustine, etoposide, and dexamethasone to mobilize peripheral blood hematopoietic stem cells for autologous transplantation in non-Hodgkin lymphoma. Blood Res 53:223-226
Green, Damian J; Press, Oliver W (2017) Whither Radioimmunotherapy: To Be or Not To Be? Cancer Res 77:2191-2196
Kern, Hanna B; Srinivasan, Selvi; Convertine, Anthony J et al. (2017) Enzyme-Cleavable Polymeric Micelles for the Intracellular Delivery of Proapoptotic Peptides. Mol Pharm 14:1450-1459
O'Steen, Shyril; Green, Damian J; Gopal, Ajay K et al. (2017) Venetoclax Synergizes with Radiotherapy for Treatment of B-cell Lymphomas. Cancer Res 77:3885-3893
Orozco, Johnnie J; Kenoyer, Aimee; Balkin, Ethan R et al. (2016) Anti-CD45 radioimmunotherapy without TBI before transplantation facilitates persistent haploidentical donor engraftment. Blood 127:352-9
Green, Damian J; Frayo, Shani L; Lin, Yukang et al. (2016) Comparative Analysis of Bispecific Antibody and Streptavidin-Targeted Radioimmunotherapy for B-cell Cancers. Cancer Res 76:6669-6679
Green, D J; Bensinger, W I; Holmberg, L A et al. (2016) Bendamustine, etoposide and dexamethasone to mobilize peripheral blood hematopoietic stem cells for autologous transplantation in patients with multiple myeloma. Bone Marrow Transplant 51:1330-1336
Cassaday, Ryan D; Stevenson, Philip A; Gooley, Theodore A et al. (2015) High-dose CD20-targeted radioimmunotherapy-based autologous transplantation improves outcomes for persistent mantle cell lymphoma. Br J Haematol 171:788-97
Frost, Sofia H L; Frayo, Shani L; Miller, Brian W et al. (2015) Comparative efficacy of 177Lu and 90Y for anti-CD20 pretargeted radioimmunotherapy in murine lymphoma xenograft models. PLoS One 10:e0120561

Showing the most recent 10 out of 61 publications