Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Improved 223Ra Therapy with Combination Epithelial Sodium Channel Blockade

Abou, Diane S. ; Fears, Amanda ; Summer, Lucy ; Longtine, Mark ; Benabdallah, Nadia ; Riddle, Ryan C. ; Ulmert, David LU ; Michalski, Jeff M. ; Wahl, Richard L. and Chesner, Denise , et al. (2021) In Journal of Nuclear Medicine 62(12). p.1751-1758
Abstract

[223Ra]RaCl2 is the first approved a-particle-emitting therapy and is indicated for treatment of bonemetastatic castration-resistant prostate cancer. Approximately half the dose is absorbed into the gastrointestinal tract within minutes of administration, limiting disease-site uptake and contributing to toxicity. Here,we investigated the role of enteric ion channels and their modulation for improved therapeutic efficacy and reduced side effects. Methods: Using primary human duodenal organoids (enteroids) asin vitromodelsof the functionalgastrointestinal epithelium, we found that amiloride (epithelial sodium ion channel blocker) and NS-1619 (K+ channel activator) presented significant effects in 223Ramembranal... (More)

[223Ra]RaCl2 is the first approved a-particle-emitting therapy and is indicated for treatment of bonemetastatic castration-resistant prostate cancer. Approximately half the dose is absorbed into the gastrointestinal tract within minutes of administration, limiting disease-site uptake and contributing to toxicity. Here,we investigated the role of enteric ion channels and their modulation for improved therapeutic efficacy and reduced side effects. Methods: Using primary human duodenal organoids (enteroids) asin vitromodelsof the functionalgastrointestinal epithelium, we found that amiloride (epithelial sodium ion channel blocker) and NS-1619 (K+ channel activator) presented significant effects in 223Ramembranal transport.Radioactivedrugdistributionwas evaluated for lead combinations in vivo and in osteosarcoma and prostate cancermodels.Results:Amiloride shifted 223Ra uptake in vivo fromthe gut and nearly doubled the uptake at sites of bone remodeling. Bone tumor growth inhibition with the combination as measured by bioluminescent imaging and radiographywas significantly greater than that with single agents alone, and the combination resulted in noweight loss.Conclusion: This combination of approved agentsmay readily be implemented as a clinical approach to improve the outcomes of bonemetastatic cancer patients with the benefit of ameliorated tolerability. COPYRIGHT

(Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; ; ; ; and , et al. (More)
; ; ; ; ; ; ; ; ; ; ; ; and (Less)
organization
publishing date
type
Contribution to journal
publication status
published
subject
keywords
Ra, amiloride, bone, gastrointestinal, ion channel
in
Journal of Nuclear Medicine
volume
62
issue
12
pages
8 pages
publisher
Society of Nuclear Medicine
external identifiers
  • pmid:33837069
  • scopus:85127983419
ISSN
0161-5505
DOI
10.2967/jnumed.121.261977
language
English
LU publication?
yes
id
7052b1f3-523c-4668-8f59-c3ce8ff21e36
date added to LUP
2022-06-14 14:05:57
date last changed
2024-04-18 02:01:14
@article{7052b1f3-523c-4668-8f59-c3ce8ff21e36,
  abstract     = {{<p>[223Ra]RaCl2 is the first approved a-particle-emitting therapy and is indicated for treatment of bonemetastatic castration-resistant prostate cancer. Approximately half the dose is absorbed into the gastrointestinal tract within minutes of administration, limiting disease-site uptake and contributing to toxicity. Here,we investigated the role of enteric ion channels and their modulation for improved therapeutic efficacy and reduced side effects. Methods: Using primary human duodenal organoids (enteroids) asin vitromodelsof the functionalgastrointestinal epithelium, we found that amiloride (epithelial sodium ion channel blocker) and NS-1619 (K+ channel activator) presented significant effects in 223Ramembranal transport.Radioactivedrugdistributionwas evaluated for lead combinations in vivo and in osteosarcoma and prostate cancermodels.Results:Amiloride shifted 223Ra uptake in vivo fromthe gut and nearly doubled the uptake at sites of bone remodeling. Bone tumor growth inhibition with the combination as measured by bioluminescent imaging and radiographywas significantly greater than that with single agents alone, and the combination resulted in noweight loss.Conclusion: This combination of approved agentsmay readily be implemented as a clinical approach to improve the outcomes of bonemetastatic cancer patients with the benefit of ameliorated tolerability. COPYRIGHT </p>}},
  author       = {{Abou, Diane S. and Fears, Amanda and Summer, Lucy and Longtine, Mark and Benabdallah, Nadia and Riddle, Ryan C. and Ulmert, David and Michalski, Jeff M. and Wahl, Richard L. and Chesner, Denise and Doucet, Michele and Zachos, Nicholas C. and Simons, Brian W. and Thorek, Daniel L.J.}},
  issn         = {{0161-5505}},
  keywords     = {{Ra; amiloride; bone; gastrointestinal; ion channel}},
  language     = {{eng}},
  number       = {{12}},
  pages        = {{1751--1758}},
  publisher    = {{Society of Nuclear Medicine}},
  series       = {{Journal of Nuclear Medicine}},
  title        = {{Improved <sup>223</sup>Ra Therapy with Combination Epithelial Sodium Channel Blockade}},
  url          = {{http://dx.doi.org/10.2967/jnumed.121.261977}},
  doi          = {{10.2967/jnumed.121.261977}},
  volume       = {{62}},
  year         = {{2021}},
}