Skip to main content

Main menu

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Journal of Nuclear Medicine
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI
  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart
Journal of Nuclear Medicine

Advanced Search

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • View or Listen to JNM Podcast
  • Visit JNM on Facebook
  • Join JNM on LinkedIn
  • Follow JNM on Twitter
  • Subscribe to our RSS feeds
Research ArticleINVITED PERSPECTIVE

Synergy Between Radiopharmaceutical Therapy and Immune Response: Deciphering the Underpinning Mechanisms for Future Actions

Elif Hindié and Ravi B. Patel
Journal of Nuclear Medicine May 2025, 66 (5) 793-794; DOI: https://doi.org/10.2967/jnumed.125.269644
Elif Hindié
1Nuclear Medicine Department, Bordeaux University Hospital, Bordeaux, France;
2INCIA, University of Bordeaux, CNRS, UMR 5287, Bordeaux, France;
3Institut Universitaire de France, Paris, France; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ravi B. Patel
4Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Info & Metrics
  • PDF
Loading

Radiopharmaceutical therapy (RPT) has, in comparison to external-beam radiation therapy (EBRT), the unique advantage of addressing all tumor sites, including occult metastases. On the other hand, dose heterogeneity is the Achilles heel of RPT. Tumor-absorbed doses can vary widely between patients and in the same patient (1). Nonhomogeneous target expression also leads to intralesional dose heterogeneity, including in tiny metastases (2). To counter the impact of heterogeneity and maximize tumor cell killing, research in RPT has often investigated the maximum tolerated dose. However, this unavoidably leads to increased toxicity to healthy tissues. For example, lymphocyte decrease in blood was correlated with spleen and bone marrow–absorbed doses from [177Lu]Lu-DOTATATE (1). The concept of maximum tolerated dose also ignores the synergy that exists between radiation and the immune system, through radiation-induced immunostimulatory effects. In this regard, the use of higher activities might not be necessarily beneficial as it can hamper mounting an immune response, as shown in the article by Constanzo et al. reported in The Journal of Nuclear Medicine (3).

It has long been recognized that radiation stimulates phenotypic changes that modulate the immune susceptibility of tumor cells. This has also raised interest in using radiotherapy to promote greater response to immunotherapies. Preclinical and clinical data have shown that low-dose EBRT can reshape the tumor microenvironment of tumors with scarce immune infiltration and together with immunotherapy induce simultaneous mobilization of innate and adaptive immunity (4). Similar findings have also been reported with RPT (5,6). In one study, combined RPT and immune checkpoint inhibitors (ICI) activated production of proinflammatory cytokines, promoted tumor infiltration and clonal expansion of CD8+ T cells, and reduced metastases (5).

Antigen presentation by major histocompatibility complex (MHC) proteins is essential for adaptive immunity. The MHC-I system, also called human leukocyte antigen class I in humans, plays a pivotal role in immune surveillance by presenting processed antigens, including tumor neoantigens, to CD8+ T cells. Interaction between a T cell receptor and specific peptide–MHC complexes can trigger T cells to proliferate and mount a specific cellular immune response. One of the recognized effects of EBRT is the increases in cell surface expression of MHC-I molecules, intracellular peptide levels, and antigen presentation, with cytotoxic T lymphocyte recognition of irradiated cells (7). There have been some data showing that RPT could upregulate the expression of MHC-I (8), but these studies were performed in vitro or in immunocompromised animal models.

To investigate the role of MHC-I in RPT, Constanzo et al. used 2 different murine melanoma cell lines that express either low or high MHC-I levels (B16F10 MHC-I-low and B16K1 MHC-I-high) and engrafted them in C57BL/6J syngeneic mice as well as in athymic nude mice (3). The response to a single injection of [225Ac]Ac-DOTA-TA99 (monoclonal antibody targeting the melanosomal polypeptide TYRP-1/gp75), with 2 different levels of activities (9.25 or 18.5 kBq), was assessed and related to dosimetry (using the distribution of 111In-DTPA-TA99 as a surrogate).

Similar antitumor effects and delays in tumor growth were obtained with 4 times lower absorbed dose in melanoma grafts with high MHC-I expression than in melanoma grafts with low MHC-I expression (3). Another key finding is that tumor response to 9.25 kBq of [225Ac]Ac-DOTA-TA99 was better in C57BL/6J immunocompetent mice than in athymic nude (immunocompromised) mice, suggesting a crucial role of T cell–mediated immune response. Interestingly, increasing the administered activity of α-RPT (18.5 kBq instead of 9.25 kBq) resulted in white blood cell decreases from bone marrow toxicity but did not result in improved tumor control. The authors conclude that MHC-I expression may predict immunostimulatory effects, and that RPT regimens should take into account MHC-I expression level.

For clinical relevance, they examined tissue samples from 12 patients with well-differentiated metastatic thyroid cancer or grade 2 midgut neuroendocrine tumors scheduled for RPT. MHC-I expression varied widely among patients. MHC-I expression correlated with the level of CD8+ infiltration (3). It will be interesting to see how these markers correlate with response to RPT and patient outcomes, as the authors are planning to investigate.

As the investigators note MHC-I expression is necessary for CD8+ T cell–mediated cytotoxicity. However, many tumors avoid immune surveillance by limiting MHC-I expression, which also leads to resistance to CD8+ T cell–based immunotherapies, such as checkpoint blockade (9). Immunogenic stimuli including EBRT or interferon-γ can induce MHC-I expression in many tumor models that have not undergone epigenetic silencing. RPT increased MHC-I in melanoma cells that already strongly expressed MHC-I but was ineffective in MHC-I-low tumors (3). Whether a combination with interferon-γ could be helpful in some instances, as suggested by the authors, warrants investigation.

An important aspect of this study is that relatively low absorbed doses of α-RPT induced surface MHC-I expression (3), whereas this was not seen in a study using low absorbed doses of β-RPT (5). Immune activation through the cyclic guanosine monophosphate–adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway has been shown to be an important pathway for β-RPT (5) as well as for α-RPT (6). Differences in immune activation between α- and β-RPT still need to be deciphered.

Due to prolonged blood circulation, radioimmunotherapy has less favorable tumor–to–bone marrow dose ratios than does RPT with small ligands, which might have contributed to the reduced immune response with increased administered activity (3). Limiting systemic lymphodepletion is important. In the context of combination therapy with ICI, lymphodepletion may blunt a systemic immune response (5). Agents that have off-target uptake in immune-rich organs such as the bone marrow, spleen, or lymph nodes may also cause systemic lymphodepletion. Therefore, as future clinical trials combining RPT with ICI are developed, systemic lymphopenia may be an important toxicity to consider during phase I development and dose-finding studies.

The reported findings are important as they suggest that immune response might compensate for a lower absorbed dose (3), thus reducing the impact of dose heterogeneity in RPT (1,2). The study opens new avenues and will stimulate further research. It will be important to understand the mechanisms that initiate and lead to enhanced MHC-I expression. Do these mechanisms depend on DNA damage, cGAS-STING pathways (10), or other radiation pathways (membrane, etc.)? Do they differ with the type of radiation (α, β−, Auger), cellular location of the radiopharmaceutical (e.g., intracytoplasmic or membrane), and half-life? Would these mechanisms involve only irradiated cells or also propagate to nonirradiated neighboring and distant cells? With EBRT, it has been shown that extracellular vesicles released from irradiated cells can upregulate MHC-I in nonirradiated tumor cells through activation of the ataxia telangiectasia mutated/ataxia telangiectasia mutated Rad-3 related/checkpoint kinase 1 axis and downstream Janus kinase–signal transducer and activator transcription protein signaling (11). Also, which types of peptides and neoantigens are presented with MHC-I enhancement? Are these peptides tumor-specific, radiation-specific, or both? Although there are some data with EBRT (12), investigations in specific settings of RPT are needed.

With its ability to deliver radiation to disseminated lesions, also stimulating immunogenicity at all disease sites and with various dose levels, RPT might lend itself, better than EBRT, to combination with ICIs. However, despite some encouraging results, many questions remain surrounding the optimal approach to clinical implementation. Understanding the underlying mechanisms will be key. Successful combinations will probably require more personalized RPT approaches with modifications of standard RPT regimens to optimally sustain immune fitness and enhance the antitumor immune response.

Early use of RPT to eradicate occult micrometastases might offer a cure. As absorbed doses in tiny lesions can be low and potentially heterogeneous (2), combination with ICIs could prove synergistic. Investigating this combination in the neoadjuvant setting, where ICIs are displaying high potential, should be encouraged.

We congratulate Constanzo et al. for having brought into light some aspects of the synergy between RPT and immune response, setting the basis for future studies.

DISCLOSURE

No potential conflict of interest relevant to this article was reported.

Footnotes

  • Published online Mar. 27, 2025.

  • © 2025 by the Society of Nuclear Medicine and Molecular Imaging.

REFERENCES

  1. 1.↵
    1. Hebert K,
    2. Santoro L,
    3. Monnier M,
    4. et al
    . Absorbed dose-response relationship in patients with gastroenteropancreatic neuroendocrine tumors treated with [177Lu]Lu-DOTATATE: one step closer to personalized medicine. J Nucl Med. 2024;65:923–930.
    OpenUrlAbstract/FREE Full Text
  2. 2.↵
    1. Larouze A,
    2. Alcocer-Ávila M,
    3. Morgat C,
    4. Champion C,
    5. Hindié E
    . Membrane and nuclear absorbed doses from 177Lu and 161Tb in tumor clusters: effect of cellular heterogeneity and potential benefit of dual targeting—a Monte Carlo study. J Nucl Med. 2023;64:1619–1624.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    1. Constanzo J,
    2. Parach A,
    3. David T,
    4. et al
    . MHC-I-driven anti-tumor immunity counterbalances low absorbed doses of radiopharmaceutical therapy. J Nucl Med. 2025;66:785–792.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Herrera FG,
    2. Ronet C,
    3. Ochoa de Olza M,
    4. et al
    . Low-dose radiotherapy reverses tumor immune desertification and resistance to immunotherapy. Cancer Discov. 2022;12:108–133.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Patel RB,
    2. Hernandez R,
    3. Carlson P,
    4. et al
    . Low-dose targeted radionuclide therapy renders immunologically cold tumors responsive to immune checkpoint blockade. Sci Transl Med. 2021;13:eabb3631.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    1. Lejeune P,
    2. Cruciani V,
    3. Berg-Larsen A,
    4. et al
    . Immunostimulatory effects of targeted thorium-227 conjugates as single agent and in combination with anti-PD-L1 therapy. J Immunother Cancer. 2021;9:e002387.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    1. Reits EA,
    2. Hodge JW,
    3. Herberts CA,
    4. et al
    . Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203:1259–1271.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    1. Chakraborty M,
    2. Wansley EK,
    3. Carrasquillo JA,
    4. et al
    . The use of chelated radionuclide (samarium-153-ethylenediaminetetramethylenephosphonate) to modulate phenotype of tumor cells and enhance T cell-mediated killing. Clin Cancer Res. 2008;14:4241–4249.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    1. Sari G,
    2. Rock KL
    . Tumor immune evasion through loss of MHC class-I antigen presentation. Curr Opin Immunol. 2023;83:102329.
    OpenUrlPubMed
  10. 10.↵
    1. Deng L,
    2. Liang H,
    3. Xu M,
    4. et al
    . STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41:843–852.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Deng S,
    2. Wang J,
    3. Hu Y,
    4. et al
    . Irradiated tumour cell-derived microparticles upregulate MHC-I expression in cancer cells via DNA double-strand break repair pathway. Cancer Lett. 2024;592:216898.
    OpenUrlPubMed
  12. 12.↵
    1. Tailor A,
    2. Estephan H,
    3. Parker R,
    4. et al
    . Ionizing radiation drives key regulators of antigen presentation and a global expansion of the immunopeptidome. Mol Cell Proteomics. 2022;21:100410.
    OpenUrlCrossRefPubMed
  • Received for publication March 4, 2025.
  • Accepted for publication March 11, 2025.
PreviousNext
Back to top

In this issue

Journal of Nuclear Medicine: 66 (5)
Journal of Nuclear Medicine
Vol. 66, Issue 5
May 1, 2025
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Complete Issue (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Journal of Nuclear Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Synergy Between Radiopharmaceutical Therapy and Immune Response: Deciphering the Underpinning Mechanisms for Future Actions
(Your Name) has sent you a message from Journal of Nuclear Medicine
(Your Name) thought you would like to see the Journal of Nuclear Medicine web site.
Citation Tools
Synergy Between Radiopharmaceutical Therapy and Immune Response: Deciphering the Underpinning Mechanisms for Future Actions
Elif Hindié, Ravi B. Patel
Journal of Nuclear Medicine May 2025, 66 (5) 793-794; DOI: 10.2967/jnumed.125.269644

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Synergy Between Radiopharmaceutical Therapy and Immune Response: Deciphering the Underpinning Mechanisms for Future Actions
Elif Hindié, Ravi B. Patel
Journal of Nuclear Medicine May 2025, 66 (5) 793-794; DOI: 10.2967/jnumed.125.269644
Twitter logo Facebook logo LinkedIn logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One
Bookmark this article

Jump to section

  • Article
    • DISCLOSURE
    • Footnotes
    • REFERENCES
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Gastrin-Releasing Peptide Receptor Imaging and Therapy in the Era of Personalized Medicine
  • Perspective on Pattern of Failure in Patients with Biochemical Recurrence After PSMA Radioguided Surgery
Show more INVITED PERSPECTIVE

Similar Articles

SNMMI

© 2025 SNMMI

Powered by HighWire