4.8 Article

Low-dose targeted radionuclide therapy renders immunologically cold tumors responsive to immune checkpoint blockade

Journal

SCIENCE TRANSLATIONAL MEDICINE
Volume 13, Issue 602, Pages -

Publisher

AMER ASSOC ADVANCEMENT SCIENCE
DOI: 10.1126/scitranslmed.abb3631

Keywords

-

Funding

  1. NIH [1K08CA241319-01, 1DP5OD024576-01, U01CA233102, R35CA197078, U54CA232568, TL1TR002375, F30CA250263, T32CA009206, P01CA250972]
  2. University of Wisconsin Carbone Cancer Center from UWCCC [P30CA014520]
  3. UW Graduate School, the Waisman Center Core Grant from the National Institute of Child Health and Human Development [U54 HD090256]
  4. NIH MSTP Training Grant [T32 GM008692]
  5. Radiological Society of North America Research Fellow Grant [RF1716]
  6. American Society of Clinical Oncology Hayden Family Foundation Young Investigator Award [12805]
  7. Midwest Athletes Against Childhood Cancer
  8. Hillman Cancer Center Early Career Fellowship for Innovative Cancer Research
  9. Bentson Translational Research Fellowship
  10. Archeus Technologies [NM600]

Ask authors/readers for more resources

The study demonstrates that delivering radiation semiselectively to tumors through targeted radionuclide therapy (TRT) can enhance response to immune checkpoint inhibitors (ICIs), leading to therapeutic efficacy and tumor-specific T cell memory in murine models. Combination of TRT and ICI activates production of proinflammatory cytokines, promotes tumor infiltration by CD8(+) T cells, and reduces metastases, suggesting a promising approach to improve response to ICIs for various tumor types.
Molecular and cellular effects of radiotherapy on tumor microenvironment (TME) can help prime and propagate antitumor immunity. We hypothesized that delivering radiation to all tumor sites could augment response to immunotherapies. We tested an approach to enhance response to immune checkpoint inhibitors (ICIs) by using targeted radionuclide therapy (TRT) to deliver radiation semiselectively to tumors. NM600, an alkylphosphocholine analog that preferentially accumulates in most tumor types, chelates a radioisotope and semiselectively delivers it to the TME for therapeutic or diagnostic applications. Using serial Y-86-NM600 positron emission tomography (PET) imaging, we estimated the dosimetry of Y-90-NM600 in immunologically cold syngeneic murine models that do not respond to ICIs alone. We observed strong therapeutic efficacy and reported optimal dose (2.5 to 5 gray) and sequence for Y-90-NM600 in combination with ICIs. After combined treatment, 45 to 66% of mice exhibited complete response and tumor-specific T cell memory, compared to 0% with Y-90-NM600 or ICI alone. This required expression of STING in tumor cells. Combined TRT and ICI activated production of proinflammatory cytokines in the TME, promoted tumor infiltration by and clonal expansion of CD8(+) T cells, and reduced metastases. In mice bearing multiple tumors, combining TRT with moderate-dose (12 gray) external beam radiotherapy (EBRT) targeting a single tumor augmented response to ICIs compared to combination of ICIs with either TRT or EBRT alone. The safety of TRT was confirmed in a companion canine study. Low-dose TRT represents a translatable approach to promote response to ICIs for many tumor types, regardless of location.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available