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
  • My Cart

Search

  • Advanced search
Journal of Nuclear Medicine
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI
  • Subscribe
  • My alerts
  • Log in
  • 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
LetterLetters to the Editor

Potential Theranostic Role of Bone Marrow Glucose Metabolism on Baseline 18F-FDG PET/CT in Metastatic Melanoma

Romain-David Seban, Laurence Champion, Izza Muneer, Shwe Synn, Lawrence H. Schwartz and Laurent Dercle
Journal of Nuclear Medicine January 2022, 63 (1) 166; DOI: https://doi.org/10.2967/jnumed.121.262361
Romain-David Seban
*Columbia University Medical Center New York, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Laurence Champion
*Columbia University Medical Center New York, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Izza Muneer
*Columbia University Medical Center New York, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shwe Synn
*Columbia University Medical Center New York, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lawrence H. Schwartz
*Columbia University Medical Center New York, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Laurent Dercle
*Columbia University Medical Center New York, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Info & Metrics
  • PDF
Loading

TO THE EDITOR: We have read with great interest the article “Prognostic Value of Bone Marrow Metabolism on Pretreatment 18F-FDG PET/CT in Patients with Metastatic Melanoma Treated with anti-PD-1 Therapy” from Nakamoto et al. (1). Although The Journal of Nuclear Medicine has published several papers highlighting the clinical value of sequential 18F-FDG PET/CT for response assessment (2,3), this new article showed that baseline 18F-FDG PET/CT could also be used to guide treatment decisions.

To this end, the authors studied a population of 92 patients with a diagnosis of metastatic melanoma treated with immune checkpoint inhibitors (ICIs) (1). ICIs included anti-PD1 as single-agent (88%) or in combination (12%) with anticytotoxic T-lymphocyte antigen-4 or antilymphocyte activation gene-3. The authors have evaluated whether patients’ overall survival (OS) could be predicted by biomarkers such as demographic or clinical (n = 4), biologic (n = 1), pathologic (n = 1), and imaging variables extracted from baseline and on treatment 18F-FDG PET/CT (n = 6).

Confirming Prognostic Significance of BM Metabolism

In this population, the authors confirmed that noninvasive measurement of glucose metabolism on nontumoral bone marrow can be used to predict OS in patients with a diagnosis of advanced melanoma treated with ICIs (4). They demonstrated that mean bone marrow–to–liver uptake ratio (BLRmean = bone marrow SUVmean/liver SUVmean) was the most valuable prognostic imaging biomarker. High baseline BLRmean predicted a significantly poor progression-free survival and OS. The finding that bone marrow glucose metabolism is an imaging biomarker correlated with survival is of interest and aligns with findings in existing literature (4,5). Hence, this letter aims to share with the readership of The Journal of Nuclear Medicine (JNM) recent publications in the field to provide a deeper understanding on the relationship between bone marrow and clinical outcomes.

Association Between Bone Marrow (BM) Metabolism and Systemic Inflammation

In a recent review published in JNM, tumor and bone marrow glucose metabolism were analyzed in 12 studies including 2,588 cancer patients who underwent both 18F-FDG PET/CT scans and biochemical assessments with blood samples (6). Most studies showed that these imaging biomarkers were associated with clinical outcomes as well as systemic inflammatory responses (high C-reactive protein, low albumin, high neutrophils or leukocytes or platelets).

Association Between Bone Marrow (BM) Metabolism and Tumor Immune Environment

Because there is a cross talk between the tumor immune environment and BM, our team evaluated the association between bone marrow glucose metabolism and transcriptomics in patients with a diagnosis of metastatic cutaneous melanoma treated with ICIs (4). To this end, we assessed the tumor immune microenvironment using transcriptomics analysis on tumor tissues. Strikingly, high bone marrow metabolism was associated with an upregulation of genes related to dendritic cells, regulatory T cell activity, and memory T cells phenotypes (4).

Of note, this was a pilot study and the major molecular pathways determining the cross talk between the BM and tumor immune environment remain to be elucidated. For now, preclinical studies showed that tumor growth in melanoma seems to play a critical role in reprogramming the host immune system by regulating hematopoiesis, which might be associated with the expansion of immunosuppressive cells such as tumor-associated macrophages, regulatory T cells, and myeloid-derived suppressor cells (MDSCs) (7).

In patients with gynecologic cancer, high BM glucose metabolism was mainly due to the production of granulocyte colony-stimulating factor (G-CSF) by tumor cells (8). Patients with high BM glucose metabolism displayed an immunosuppressive phenotype with increased MDSCs and decreased CD8+ T cells (8).

Prospective studies and translational studies correlating BM glucose metabolism with antitumor immunity are warranted. Continued efforts need to be made and should focus on improving our understanding of physiopathologic concepts. We have to clarify the association between baseline bone marrow glucose metabolism and the presence of an immunosuppressive environment. This is necessary to unravel further cancer-related inflammation and immunosuppressive phenotypes associated with immunotherapy resistance through the use of quantitative transcriptome analyses of tumor, lymphoid tissue biopsies, and immuno-PET imaging.

Potential Theranostic Approaches

The demonstration of the prognostic value of BM glucose metabolism and of its association with tumor immune environment offers a springboard to exciting, new theranostic research. Novel therapies blocking immunosuppressive agents, such as MDSCs, are indeed under investigation (9) to potentiate ICIs. The underlying assumption is that glucose metabolism on tissues with medullary and extramedullary hematopoiesis could be associated with tumor-induced immune suppression. For instance, preimmunotherapy 18F-FDG PET/CT that explores bone marrow might be a relevant assay to predict response to MDSCs-blockade therapies, in combination with ICI (10).

In conclusion, the scientific community has demonstrated that BM glucose metabolism measured on 18F-FDG PET is associated with immunotherapy outcomes in patients with metastatic melanoma. The next step is to pursue efforts with prospective and large multicenter studies that would ensure a deeper understanding on how this specific biomarker could be used as a clinical decision support tool in patients with metastatic melanoma treated with ICIs.

DISCLOSURE

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

  • © 2022 by the Society of Nuclear Medicine and Molecular Imaging.

REFERENCES

  1. 1.↵
    1. Nakamoto R,
    2. Zaba LC,
    3. Liang T,
    4. et al
    . Prognostic value of bone marrow metabolism on pretreatment 18 F-FDG PET/CT in patients with metastatic melanoma treated with anti-PD-1 therapy. J Nucl Med. February 5, 2021 [Epub ahead of print].
  2. 2.↵
    1. Seban R-D,
    2. Schwartz LH,
    3. Bonardel G,
    4. Dercle L
    . Diagnosis of hyperprogressive disease in patients treated with checkpoint inhibitors using 18F-FDG PET/CT. J Nucl Med. 2020;61:1404–1405.
    OpenUrl
  3. 3.↵
    1. Dercle L,
    2. Seban R-D,
    3. Lazarovici J,
    4. et al
    . 18F-FDG PET and CT scans detect new imaging patterns of response and progression in patients with Hodgkin lymphoma treated by anti-programmed death 1 immune checkpoint inhibitor. J Nucl Med. 2018;59:15–24.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Seban R-D,
    2. Nemer JS,
    3. Marabelle A,
    4. et al
    . Prognostic and theranostic 18F-FDG PET biomarkers for anti-PD1 immunotherapy in metastatic melanoma: association with outcome and transcriptomics. Eur J Nucl Med Mol Imaging. 2019;46:2298–2310.
    OpenUrl
  5. 5.↵
    1. Prévost S,
    2. Boucher L,
    3. Larivée P,
    4. Boileau R,
    5. Bénard F
    . Bone marrow hypermetabolism on 18F-FDG PET as a survival prognostic factor in non-small cell lung cancer. J Nucl Med. 2006;47:559–565.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    1. Dolan RD,
    2. McLees NG,
    3. Irfan A,
    4. et al
    . The relationship between tumor glucose metabolism and host systemic inflammatory responses in patients with cancer: a systematic review. J Nucl Med. 2019;60:467–471.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    1. Kamran N,
    2. Li Y,
    3. Sierra M,
    4. et al
    . Melanoma induced immunosuppression is mediated by hematopoietic dysregulation. OncoImmunology. 2017;7:e1408750.
    OpenUrl
  8. 8.↵
    1. Shimura K,
    2. Mabuchi S,
    3. Komura N,
    4. et al
    . Prognostic significance of bone marrow FDG uptake in patients with gynecological cancer. Sci Rep. 2021;11:2257.
    OpenUrl
  9. 9.↵
    1. Holtzhausen A,
    2. Harris W,
    3. Ubil E,
    4. et al
    . TAM family receptor kinase inhibition reverses MDSC-mediated suppression and augments anti-PD-1 therapy in melanoma. Cancer Immunol Res. 2019;7:1672–1686.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    1. Liu Y,
    2. Wei G,
    3. Cheng WA,
    4. et al
    . Targeting myeloid-derived suppressor cells for cancer immunotherapy. Cancer Immunol Immunother. 2018;67:1181–1195.
    OpenUrl
PreviousNext
Back to top

In this issue

Journal of Nuclear Medicine: 63 (1)
Journal of Nuclear Medicine
Vol. 63, Issue 1
January 1, 2022
  • 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.
Potential Theranostic Role of Bone Marrow Glucose Metabolism on Baseline 18F-FDG PET/CT in Metastatic Melanoma
(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
Potential Theranostic Role of Bone Marrow Glucose Metabolism on Baseline 18F-FDG PET/CT in Metastatic Melanoma
Romain-David Seban, Laurence Champion, Izza Muneer, Shwe Synn, Lawrence H. Schwartz, Laurent Dercle
Journal of Nuclear Medicine Jan 2022, 63 (1) 166; DOI: 10.2967/jnumed.121.262361

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Potential Theranostic Role of Bone Marrow Glucose Metabolism on Baseline 18F-FDG PET/CT in Metastatic Melanoma
Romain-David Seban, Laurence Champion, Izza Muneer, Shwe Synn, Lawrence H. Schwartz, Laurent Dercle
Journal of Nuclear Medicine Jan 2022, 63 (1) 166; DOI: 10.2967/jnumed.121.262361
Twitter logo Facebook logo LinkedIn logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One
Bookmark this article

Jump to section

  • Article
    • DISCLOSURE
    • 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

  • Business Model Beats Science and Logic: Dosimetry and Paucity of Its Use
  • Determining PSMA-617 Mass and Molar Activity in Pluvicto Doses
  • The Value of Functional PET in Quantifying Neurotransmitter Dynamics
Show more Letters to the Editor

Similar Articles

SNMMI

© 2025 SNMMI

Powered by HighWire