Skip to main content

EDITORIAL article

Front. Oncol., 07 August 2023
Sec. Cancer Metabolism
This article is part of the Research Topic Proteomic and Metabolic Reprogramming in Myeloma Cells within the Tumor Microenvironment View all 9 articles

Editorial: Proteomic and metabolic reprogramming in myeloma cells within the tumor microenvironment

  • 1Age Related Diseases Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
  • 2Faculty of Medicine and Surgery, University Vita-Salute San Raffaele, Milano, Italy
  • 3Experimental Hematology and Immunology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
  • 4Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, United States

Multiple myeloma (MM) is a hematological malignancy characterized by the dysregulated proliferation of transformed plasma cells (PCs) primarily in the bone marrow (BM) (1). For the highly specialized role as antibody secreting factories, PCs and their malignant counterpart must cope with different stressors linked to massive immunoglobulin synthesis (2). As such, they display an exquisite dependency on stress adaptive, protein degradative and metabolic pathways that ensure the sustainability of such a demanding process (35). While providing survival advantage, these “lineage” dependencies have similarly highlighted fundamental therapeutic vulnerabilities that have led to the introduction of the proteasome inhibitor (PI) bortezomib in the clinical setting (69). On the other hand, the strict interaction of MM cells with the immune and non-immune components of the BM milieu is a pathognomonic feature of the disease (10); and it is becoming clear how metabolic and proteomic reprogramming in MM cells can shape this mutual interaction, thus favoring a protective and MM-promoting environment (11, 12). The clinical success of immunomodulatory agents (IMiDs) and immunotherapy that alter the tumor-BM interface underscores the importance of its targeting (1317), prompting for a deeper understanding of the role of metabolic rewiring in this interaction to unveil novel therapeutic dependencies. In this scenario, the reviews and original articles of this Research Topic give insights on open questions on the tumor-intrinsic or tumor-host mechanisms driving disease progression, emphasizing the emerging role of metabolic dependencies as new potential therapeutic opportunities.

Specifically, two reviews discuss the role of the MM-milieu interplay in mediating resistance to PIs (Schwestermann et al.) and IMiDs (Chen and Gooding). While providing a detailed summary of the current knowledge on the interaction between MM cells and BM components, Schwestermann et al. have focused on how the metabolic rewire shapes MM cell ability to adapt, grow and resist to therapies, specifically PIs. Importantly, they highlight how metabolic switches occurring after treatment with PIs may be targeted to overcome PI-resistance and improve overall drug efficacy. On a similar line, Chen and Gooding summarize the current literature on the mechanisms underlying IMiDs resistance with a specific highlight at the gap existing in the characterization of Cereblon (CRBN)-independent mechanisms, including those related to immune microenvironment and metabolism. In a broader light, Solimando et al. provide an extensive overview on the mechanisms by which MM resist to different therapies, including epigenetic modification and cancer cell stemness; and discuss therapeutic strategies to improve the management of MM patients.

Of clinical relevance, Raimondi et al. discuss the importance of metabolic reprogramming of MM cells in the context of MM-related bone disease. Specifically, they focus on the role of glutamine addiction, in controlling osteoblast differentiation from mesenchymal stromal cells, and thus bone remodeling. While providing an exhaustive review of the implication of metabolic features in the pathophysiology of MM bone disease, they also emphasize the clinical implication of such findings that may allow for the identification of new tracers of active bone disease that are under investigation that may be more sensitive in patients with negativity at the PET/TC with 18F-fluorodeoxyglucose (FDG).

The local concentration of metabolites in the BM milieu may affect MM progression and drug sensitivity (12). The original article by Trudu et al. in this Research Topic, describes a novel intrinsic role of arginine shortage in directly promoting MM proliferation and resistance to therapies. The authors show that, beyond the established immunosuppressive role of arginine deprivation on T cells (18), the reduction in arginine availability may have a direct pro-survival effect, mediated by increased activation of AKT, that results in higher PI-resistance to in vitro and MM growth in vivo.

Growing evidence are showing that mitochondria are critical for MM development, progression, and sensitivity to therapies. Two reviews in this Research Topic analyzed the role of mitochondrial metabolism in general in MM pathobiology (Nair et al.), or in particular by focusing on the MM tumor suppressor TRAF3 (Jung et al.) a signal transducer that has been recently showed to be recruited to the mitochondria in B cells (19). Nair et al. extensively analyze how mitochondria metabolism influences MM development and drug resistance and vice versa how MM mutations affect metabolic pathways. The authors also discuss the role of mitochondria in regulating MM metabolism, redox homeostasis, iron and calcium fluxes, and the activation of the integrated stress response and intrinsic apoptosis. On this line, Jung et al., describe the role of TRAF3 in normal and malignant B cells. TRAF3 is a negative regulator of key B cell signaling pathways, whose loss induces enhanced antibody responses and increased B cell survival (20, 21). Moreover, recent findings have shown that it is recruited to the mitochondrial membrane through its interaction with the mitochondrial fission factor MFF (19). In their mini-review, the authors summarize the current knowledge on TRAF3 signaling activities and on these newly discovered roles in modulating mitochondrial morphology, cellular metabolism, and mitochondria-dependent apoptosis.

Finally, novel original culture methods for the analysis of the interplay between BM adipocyte-MM cells have been presented by Fairfield et al. The authors propose three in vitro culture systems allowing the study of MM-adipocyte crosstalk using primary BM adipocyte and not those obtained through the differentiation in vitro of BM-derived mesenchymal stromal cells. Of technological relevance, the authors describe in detail the protocols to perform 2D and 3D co-cultures and the following imaging and proteomic characterizations, providing novel tools to dissect how adipocytes and MM cells influence each other in the BM environment.

In summary, this Research Topic provides a critical overview and novel evidence on the role of proteomic and metabolic reprogramming of myeloma cells as key drivers of MM pathogenesis and drug resistance with important therapeutic implications. Open questions remain on how protein degradative and metabolic pathways affect immune activation and tumor sensitivity to immunotherapies. Further investigations will be instrumental in exploiting such metabolic and proteomic dependencies to improve the therapeutic efficacy of anti-MM agents, increase response durability and patient overall outcome.

Author contributions

EM: Writing – original draft, Writing – review & editing, AG: Writing – original draft, Writing – review & editing.

Acknowledgments

We thank Prof. Holger Auner (University of Lausanne, Switzerland, and Imperial College London, UK) for his role as guest editor. EM is supported by grants from Fondazione Cariplo (Giovani Ricercatori, 2018-0257), Fondazione Telethon and Fondazione Cariplo (Joint Grant on rare diseases, GJC21079/2022-0577) and the Italian Ministry of Health (GR-2018-12368387). AG is a Fellow of The Leukemia & Lymphoma Society and a Scholar of the American Society of Hematology; she received support from the International Myeloma Society (IMS); she is supported by an Individual Start-UP grant from the Italian Association for Cancer Research (AIRC) (project #27750); a FPRC “5xmille” 2019 Ministry of Health project (IDEE) and a FPRC “5xmille” 2021 Ministry of Health project (EMAGEN-FaBer).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Kumar SK, Rajkumar V, Kyle RA, van Duin M, Sonneveld P, Mateos M-V, et al. Multiple myeloma. Nat Rev Dis Primers (2017) 3:17046. doi: 10.1038/nrdp.2017.46

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Tellier J, Nutt SL. Plasma cells: The programming of an antibody-secreting machine. Eur J Immunol (2019) 49:30–7. doi: 10.1002/eji.201847517

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Bianchi G, Anderson KC. Contribution of inhibition of protein catabolism in myeloma. Cancer J (2019) 25:11–8. doi: 10.1097/PPO.0000000000000349

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Perini T, Materozzi M, Milan E. The Immunity-Malignancy equilibrium in multiple myeloma: lessons from oncogenic events in plasma cells. FEBS J (2022) 289:4383–97. doi: 10.1111/febs.16068

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Cenci S. The proteasome in terminal plasma cell differentiation. Semin Hematol (2012) 49:215–22. doi: 10.1053/j.seminhematol.2012.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Gulla A, Anderson KC. Multiple myeloma: the (r)evolution of current therapy and a glance into future. Haematologica (2020) 105:2358–67. doi: 10.3324/haematol.2020.247015

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Richardson PG, Sonneveld P, Schuster M, Irwin D, Stadtmauer E, Facon T, et al. Extended follow-up of a phase 3 trial in relapsed multiple myeloma: final time-to-event results of the APEX trial. Blood (2007) 110:3557–60. doi: 10.1182/blood-2006-08-036947

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T, et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. New Engl J Med (2005) 352:2487–98. doi: 10.1056/NEJMoa043445

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D, et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. New Engl J Med (2003) 348:2609–17. doi: 10.1056/NEJMoa030288

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Bianchi G, Munshi NC. Pathogenesis beyond the cancer clone(s) in multiple myeloma. Blood (2015) 125:3049–58. doi: 10.1182/blood-2014-11-568881

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Wolska-Washer A, Smolewski P. Targeting protein degradation pathways in tumors: focusing on their role in hematological Malignancies. Cancers (Basel) (2022) 14(15):3778. doi: 10.3390/cancers14153778

PubMed Abstract | CrossRef Full Text | Google Scholar

12. ROman-Trufero M, Auner HW, Edwards CM. Multiple myeloma metabolism - a treasure trove of therapeutic targets? Front Immunol (2022) 13:897862. doi: 10.3389/fimmu.2022.897862

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Dimopoulos M, Spencer A, Attal M, Prince HM, Harousseau J-L, Dmoszynska A, et al. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. New Engl J Med (2007) 357:2123–32. doi: 10.1056/NEJMoa070594

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Benboubker L, Dimopoulos MA, Dispenzieri A, Catalano J, Belch AR, Cavo M, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. New Engl J Med (2014) 371:906–17. doi: 10.1056/NEJMoa1402551

PubMed Abstract | CrossRef Full Text | Google Scholar

15. McCarthy PL, Owzar K, Hofmeister CC, Hurd DD, Hassoun H, Richardson PG, et al. Lenalidomide after stem-cell transplantation for multiple myeloma. New Engl J Med (2012) 366:1770–81. doi: 10.1056/NEJMoa1114083

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Munshi NC, Anderson LD, Shah N, Madduri D, Berdeja J, Lonial S, et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med (2021) 384:705–16. doi: 10.1056/NEJMoa2024850

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Moreau P, Garfall AL, van de Donk NWCJ, Nahi H, San-Miguel JF, Oriol A, et al. Teclistamab in relapsed or refractory multiple myeloma. N Engl J Med (2022) 387:495–505. doi: 10.1056/NEJMoa2203478

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Fletcher M, Ramirez ME, Sierra RA, Raber P, Thevenot P, Al-Khami AA, et al. l-arginine depletion blunts antitumor T-cell responses by inducing myeloid-derived suppressor cells. Cancer Res (2015) 75:275–83. doi: 10.1158/0008-5472.CAN-14-1491

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Liu Y, Gokhale S, Jung J, Zhu S, Luo C, Saha D, et al. Mitochondrial fission factor is a novel interacting protein of the critical B cell survival regulator TRAF3 in B lymphocytes. Front Immunol (2021) 12:670338. doi: 10.3389/fimmu.2021.670338

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Bishop GA, Stunz LL, Hostager BS. TRAF3 as a multifaceted regulator of B lymphocyte survival and activation. Front Immunol (2018) 9:2161. doi: 10.3389/fimmu.2018.02161

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Xie P, Stunz LL, Larison KD, Yang B, Bishop GA. Tumor necrosis factor receptor-associated factor 3 is a critical regulator of B cell homeostasis in secondary lymphoid organs. Immunity (2007) 27:253–67. doi: 10.1016/j.immuni.2007.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: multiple myeloma, bone marrow, metabolism, mitochondria, proteasome inhibitors (PI), IMiDs, microenvironment

Citation: Milan E and Gullà A (2023) Editorial: Proteomic and metabolic reprogramming in myeloma cells within the tumor microenvironment. Front. Oncol. 13:1264740. doi: 10.3389/fonc.2023.1264740

Received: 21 July 2023; Accepted: 28 July 2023;
Published: 07 August 2023.

Edited and Reviewed by:

Ubaldo Emilio Martinez-Outschoorn, Thomas Jefferson University, United States

Copyright © 2023 Milan and Gullà. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Enrico Milan, milan.enrico@hsr.it; Annamaria Gullà, annamaria.gulla@ircc.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.