Identification of novel myelodysplastic syndromes prognostic subgroups by integration of inflammation, cell-type composition, and immune signatures in the bone marrow

  1. Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, United Kingdom
  2. Department of Basic and Clinical Neuroscience, King’s College London, London, United Kingdom
  3. Department of Biostatistics and Health Informatics, King’s College London, London, United Kingdom
  4. NIHR BRC SLAM NHS Foundation Trust, London, United Kingdom
  5. Perron Institute for Neurological and Translational Science, University of Western Australia Medical School, Perth, WA 6009, Australia
  6. Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, and public reviews.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Satyajit Rath
    Indian Institute of Science Education and Research (IISER), Pune, India
  • Senior Editor
    Satyajit Rath
    Indian Institute of Science Education and Research (IISER), Pune, India

Reviewer #1 (Public Review):

In their manuscript, Gerlevik et al. performed an integrative analysis of clinical, genetic and transcriptomic data to identify MDS subgroups with distinct outcomes. The study was based on the building of an "immunoscore" and then combined with genotype and clinical data to analyze patient outcomes using multi-omics factor analysis.

Strengths: Integrative analysis of RNA-seq, genotyping and clinical data

Weaknesses: Validation of the bioinformatic pipeline is incomplete

Major comments:

(1) This study considered two RNA-seq data sets publicly available and generated in two distinct laboratories. Are they comparable in terms of RNA-seq technique: polyA versus rRNA depletion, paired-end sequencing, fragment length?

(2) Data quality control (figure 1): the authors must show in a graph whether the features (dimensions) of factor 1 were available for each BMMNC and CD34+ samples.

(3) How to validate the importance of "immunoscore"? If GSEA of RNA-seq data was performed in the entire cohort, in the SF3B1-mutated samples or SRSF2-mutated samples (instead of patients having a high versus low level of factor 1 shown in Sup Fig. 4), what would be the ranking of Hallmarks or Reactome inflammatory terms among the others?

(4) To decipher cell-type composition of BMMNC and CD34+ samples, the authors used van Galen's data (2019; supplementary table 3). Cell composition is expressed as the proportion of each cell population among the others. Surprisingly, the authors found that the promonocyte-like score was increased in SF3B1-mutated samples and not in SRSF2-mutated samples, which are frequently co-mutated with TET2 and associated with a CMML-like phenotype. Is there a risk of bias if bone marrow subpopulations such as megakaryocytic-erythroid progenitors or early erythroid precursors are not considered?

(5) Figures 2a and 2b indicated that the nature of retrotransposons identified in BMMNC and CD34+ was different. ERVs were not detected in CD34+ cells. Are ERVs not reactivated in CD34+ cells? Is there a bias in the sequencing or bioinformatic method?

(6) What is the impact of factor 1 on survival? Is it different between BMMNC and CD34+ cells considering the distinct composition of factor 1 in CD34+ and BMMNC?

(7) In Figure 1e, genotype contributed to the variance of in the CD34+ cell analyses more importantly than in the BMMNC. Because the patients are different in the two cohorts, differences in the variance could be explained either by a greater variability of the type of mutations in CD34 or an increased frequency of poor prognosis mutations in CD34+ compared to BMMNC. The genotyping data must be shown.

(8) Fig. 2a-b: Features with high weight are shown for each factor. For factor 9, features seemed to have a low weight (Fig. 1b and 1c). However, factor 9 was predictive of EFS and OS in the BMMNC cohort. What are the features driving the prognostic value of factor 9?

(9) The authors also provided microarray analyses of CD34+ cell. It could be interesting to test more broadly the correlation between features identified by RNA-seq or microarrays.

(10) The authors should discuss the relevance of immunosenescence features in the context of SRSF2 mutation and extend the discussion to the interest of their pipeline for patient diagnosis and follow up under treatments.

Reviewer #2 (Public Review):

The authors performed a Multi-Omics Factor Analysis (MOFA) on analysis of two published MDS patient cohorts-1 from bone marrow mononuclear cells (BMMNCs) and CD34 cells (ref 17) and another from CD34+ cells (ref 15) --with three data modalities (clinical, genotype, and transcriptomics). Seven different views, including immune profile, inflammation/aging, Retrotransposon (RTE) expression, and cell-type composition, were derived from these modalities to attempt to identify the latent factors with significant impact on MDS prognosis.

SF3B1 was found to be the only mutation among 13 mutations in the BMMNC cohort that indicated a significant association with high inflammation. This trend was also observed to a lesser extent in the CD34+ cohort. The MOFA factor representing inflammation showed a good prognosis for MDS patients with high inflammation. In contrast, SRSF2 mutant cases showed a granulocyte-monocyte progenitor (GMP) pattern and high levels of senescence, immunosenescence, and malignant myeloid cells, consistent with their poor prognosis. Also, MOFA identified RTE expression as a risk factor for MDS. They proposed that this work showed the efficacy of their integrative approach to assess MDS prognostic risk that 'goes beyond all the scoring systems described thus far for MDS'.

Several issues need clarification and response:

(1) The authors do not provide adequate known clinical and molecular information which demonstrates prognostic risk of their sample cohorts in order to determine whether their data and approach 'goes 'beyond all the scoring systems described thus far for MDS'. For example, what data have the authors that their features provide prognostic data independent of the prior known factors related to prognosis (eg, marrow blasts, mutational, cytogenetic features, ring sideroblasts, IPSS-R, IPSS-M, MDA-SS)?

(2) A major issue in analyzing this paper relates to the specific patient composition from whom the samples and data were obtained. The cells from the Shiozawa paper (ref 17) is comprised of a substantial number of CMML patients. Thus, what evidence have the authors that much of the data from the BMMNCs from these patients and mutant SRSF2 related predominantly to their monocytic differentiation state?

(3) In addition, as the majority of patients in the Shiozawa paper have ring sideroblasts (n=59), thus potentially skewing the data toward consideration mainly of these patients, for whom better outcomes are well known.

(4) Further, regarding this patient subset, what evidence have the authors that the importance of the SF3B1 mutation was merely related to the preponderance of sideroblastic patients from whom the samples were analyzed?

(5) An Erratum was reported for the Shiozawa paper (Shiozawa Y, Malcovati L, Gallì A, et al. Gene expression and risk of leukemic transformation in myelodysplasia. Blood. 2018 Aug 23;132(8):869-875. doi: 10.1182/blood-2018-07-863134) that resulted from a coding error in the construction of the logistic regression model for subgroup prediction based on the gene expression profiles of BMMNCs. This coding error was identified after the publication of the article. The authors should indicate the effect this error may have had on the data they now report.

(6) What information have the authors as to whether the differing RTE findings were not predominantly related to the differentiation state of the cell population analyzed (ie higher in BM MNCs vs CD34, Fig 1)? What control data have the authors regarding these values from normal (non-malignant) cell populations?

(7) The statement in the Discussion regarding the effects of SRSF2 mutation is speculative and should be avoided. Many other somatic gene mutations have known stronger effects on prognosis for MDS.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation