All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit the International Myeloma Foundation or HealthTree for Multiple Myeloma.

The Multiple Myeloma Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your Multiple Myeloma Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The Multiple Myeloma Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the Multiple Myeloma Hub cannot guarantee the accuracy of translated content. The Multiple Myeloma Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2020-07-03T08:18:19.000Z

Defining the significance of MGUS: the bone marrow microenvironment is compromised from an early stage in MM

Bookmark this article

Multiple myeloma (MM) remains an incurable disease despite the ability to readily detect its precursor states: monoclonal gammopathy of unknown significance (MGUS), and smoldering MM (SMM). Although early intervention is an attractive approach to treatment, premalignancy does not always lead to progression. Therefore, comprehensive molecular characterization of the tumor microenvironment and the host immune response is required to understand disease progression further.1

In a recent study, Oksana Zavidij from the Dana-Farber Cancer Institute and colleagues aimed to elucidate the transcriptomic alterations within the immune microenvironment in disease progression, by performing single-cell RNA-seq (scRNA-seq) on patient samples across all stages of MM. This study was published in Nature Cancer.1

Methods

  • scRNA-seq was performed on bone marrow (BM) samples that were collected at the Dana-Farber Cancer Institute from patients with MGUS (n = 5 patients), low-risk SMM (n = 3), high-risk SMM (n = 8), or newly diagnosed MM (n = 7), as well as healthy donors (n = 9)
    • CD138 or CD45+ cell fractions were isolated using magnetic-activated cell sorting and in silico cell filtering based on gene expression
    • Approximately 19,000 CD45+/CD138 cells from the microenvironment were sequenced
    • 21 subpopulations of cells were isolated by clustering cells based on the gene expression profile
    • Using the expression of known marker genes and finding the top differentially expressed genes for each cluster, cells were classified into ten broad types, ranging from hematopoietic progenitor cells and pre-B cells to mature populations engaged in the immune response
  • Sequencing data was validated by performing mass cytometry by time-of-flight (CyTOF) on BM aspirates of patients with MGUS, SMM and MM (n = 13), and healthy BM controls (n = 4)

Results1

The tumor microenvironment changes considerably during myeloma progression, starting at the MGUS stage with increased populations of natural killer (NK) cells, T cells, and nonclassical monocytes (Figure 1). At the SMM stage, there is an early accumulation of regulatory and gamma-delta T cells, followed by loss of CD8+ memory populations, and an increase in interferon (IFN) signaling. At the MM stage, CD14+ monocytes show a loss of antigen presentation through dysregulation of major histocompatibility complex (MHC) type II genes, which leads to T cell suppression. Find a more detailed list of key findings in Table 1.


Figure 1. Adapted representation of immune cell composition changes between healthy and multiple myeloma bone marrow samples1

 

Table 1. Summary of key findings from scRNA-seq and CyTOF analysis in the microenvironment of bone marrow samples from patients with MGUS, SMM, and MM1

BM, bone marrow; CyTOF, cytometry by time-of-flight; HLA-DR, human leukocyte antigen DR isotype; MGUS, monoclonal gammopathy of unknown significance; MHC, major histocompatibility complex; MM, multiple myeloma; NK, natural killer; scRNA-seq, single-cell RNA-seq; SMM, smoldering MM

The composition of the BM microenvironment changes early in MM progression (Figure 1)

  • Significant enrichment in NK, T and CD16+ cells seen in the BM from patients, as well as a relative decrease in plasmacytoid dendritic cells, immature neutrophils, CD14+ monocytes, and other progenitor cells
    • BM samples from healthy donors universally had < 2% CD16+ macrophages and < 10% NK cells, while samples from patients showed a spectrum from unaltered compositions to approximately 12% CD16+ macrophages and approximately 40% NK cells (these changes were independent of tumor burden)
  • Increases in NK cells were accompanied by a shift in phenotype
    • three populations of NK cells identified: CXCR4+, CX3CR1+/CCL3+, and IL7R+CD62L+
    • positive association between NK cell frequency and enrichment for the CXCR4+ subset (Fisher exact test, p = 0.012; n = 23), while samples with lower NK-cell frequencies displayed a shift toward the CX3CR1+ subset
  • Except for neutrophil output, CyTOF data validated the results obtained by scRNA-seq

Increased Treg numbers

and patient-specific heterogeneity in noncytotoxic T cells

  • Significantly increased Treg numbers in the BM microenvironment from patients (q = 0.0005, degrees of freedom [d.f.] = 26.1, t-test of normal vs patient samples)
  • High expression of MHC type II proteins in Treg cells, which may inhibit T cell proliferation and cytokine production

Cytotoxic T cell populations shifted toward effector phenotype during disease progression

  • CD8+ cells with expression of granzyme K were more prevalent in healthy individuals and patients with MGUS (p = 0.0013) than in SMM and MM samples
  • CD8+ cells with expression of granzymes B and H were more prevalent in the SMM and MM stages (p = 3.1 × 10−4)
  • CyTOF data agreed with this, demonstrating a two-fold decrease in memory CD8+ cells in the BM aspirates of patients with SMM

IFN response was seen across immune cell types in the MM samples

  • Including NK, nonclassical monocytes, and small numbers of B cells
  • Possible association between the signature of enriched IFN-responsive genes and myeloma progression

CD14+ monocytes in the SMM and MM microenvironment showed defective antigen presentation due to intracellular accumulation of HLA-DR

  • A population of CD14+ monocytes were significantly upregulated in all patient samples (t-test, n = 30, d.f. = 19.7, q = 1.3 × 10−6)
  • CyTOF analysis revealed that CD14+ cells from patients with SMM and MM exhibited significantly lower HLA-DR surface expression than healthy donors (p = 0.0087)
    • Phenotypic change promoted by the myeloma cells induced internalization of MHC class II in CD14+ cells, reducing their potential for antigen presentation
  • Posttranslational control of MHC class II is due to the upregulation of the E3 ubiquitin-protein ligase gene MARCHF1/MARCH1 in patients compared with healthy BM (p = 0.008)

CD14+ monocytes in the BM microenvironment can promote the proliferation of myeloma cells and suppress T cell activation

  • Coculture of myeloma cell lines with CD14+ HLA-DRlow cells, increases the fraction of cells in S-phase (p = 0.006) and G2M phase (p = 0.0012) of the cell cycle, compared with control cells, promoting MM cell proliferation
  • CD14+ HLA-DRlow cells can also alter T cell activity, suppressing its activation in SMM and MM

 

Conclusions and further discussion

During the 6th World Congress on Controversies in Multiple Myeloma (COMy) Meeting, Irene Ghobrial from the Dana-Farber Cancer Institute, and one of the corresponding authors of this study presented the results right before their publication.2 Figure 2 represents a visual summary of the alterations observed in the immune microenvironment during progression to MM. These results are heterogeneous across patients and, despite the small sample size, could help develop future strategies on immune-based biomarkers for patient stratification and therapeutic targets to prevent progression to active disease.1

In her talk, she highlighted the importance of developing a comprehensive analysis of MM evolution, combining the genetic and epigenetic events in the plasma cell with tumor microenvironment regulation of the disease activation through immune and non-immune cells. With that aim, her group is working on what they call “Precision Prevention”: identifying signatures of expression that include both, plasma cells and microenvironment, and could potentially lead to personalized early treatment decision.2,3

 Figure 2. Immune microenvironment dysregulation model from healthy BM to MM; adapted from Zavidij et al.1

It is essential to read these data in the context of an aging population with an increased cellular senescence burden. During the last COMy 2020, Gordon Cook, University of Leeds, gave us an overview of the immune status in MM and how the cellular senescence also affects the immune system.4

The so-called immunosenescence is a gradual deterioration of immune function brought on by the natural aging process. Changes in the number and function of hematopoietic stem cells have been described, entailing a decreased diversity of functional T cells, increasing pro-inflammatory cytokines, and leading to a chronic inflammatory state with advancing age. This state is one of the multiple reasons why in the elderly population there is an increased incidence of metabolic diseases, infections, frailty, and ultimately, cancer.4

Crucial advances on the evolution of MM will be achieved with the future results from the PROMISE study (NCT03689595): the first US cancer screening study for MM where investigators want to analyze samples from 50,000 high-risk individuals, defined as African American > 45 years, or people with a first-degree relative with MM. They estimate to be able to identify at least 3,000 positive cases to study thoroughly, and potentially prevent MM precursor progression to active disease.2   

In conclusion, it is possible now to confirm a multilayered immune modulation that can be present at the early stages of MM. The prognostic power of this signature needs to be further elucidated within the heterogeneity of patients with MGUS, SMM, and MM.

Expert Opinion

“This study showed that the immune microenvironment is compromised even at the MGUS stage. It helps us understand that MGUS is not as benign as we thought it was. It also helps us understand the steps of immune dysregulation that occur as the patient transitions from MGUS to smoldering MM to overt MM. The hope is that we use this knowledge to develop specific immunotherapy that can delay or prevent progression.”

  1. Zavidij O, Haradhvala NJ, Mouhieddine TH, et al. Single-cell RNA sequencing reveals compromised immune microenvironment in precursor stages of multiple myeloma. Nat Cancer. 2020;1:493-506. DOI: https://doi.org/10.1038/s43018-020-0053-3
  2. Gohbrial I. Role of the microenvironment. Session 2 (Immunotherapy: where do we stand?). 6thWorld Congresses on Controversies in Multiple Myeloma (COMy); May 14, 2020. Virtual.
  3. Bustoros M, Sklavenitis-Pistofidis R, Park J, et al. Genomic profiling of smoldering multiple myeloma identifies patients at a high risk of disease progression. J Clin Oncol. 2020. DOI: 1200/JCO.20.00437
  4. Cook G. Immune status in myeloma. Session 2 (Immunotherapy: where do we stand?). 6thWorld Congresses on Controversies in Multiple Myeloma (COMy); May 14, 2020. Virtual.

Your opinion matters

As a result of this content, I commit to reviewing the CARTITUDE clinical program to guide my understanding of cilta-cel in clinical practice.
27 votes - 6 days left ...

Newsletter

Subscribe to get the best content related to multiple myeloma delivered to your inbox