44 research outputs found

    2021 European Myeloma Network review and consensus statement on smoldering multiple myeloma: how to distinguish (and manage) Dr. Jekyll and Mr. Hyde

    Full text link
    According to the updated International Myeloma Working Group criteria, smoldering multiple myeloma (SMM) is an asymptomatic plasma cell disorder characterized by an M-component >3 g/dL, bone marrow plasma cell infiltration >10% and <60%, and absence of any myeloma-defining event. Active multiple myeloma is preceded by SMM, with a median time to progression of approximately 5 years. Cases of SMM range from the extremes of monoclonal gammopathy of undetermined significance-like, in which patients never progress during their lifetimes, to early multiple myeloma, in which transformation into symptomatic disease, based on genomic evolution, may be rapid and devastating. Such a split personality makes the prognosis and management of individual patients challenging, particularly with regard to the identification and possible early treatment of high-risk SMM. Outside of clinical trials, the conventional approach to SMM generally remains close observation until progression to active multiple myeloma. However, two prospective, randomized trials have recently demonstrated a significant clinical benefit in terms of time to progression, and of overall survival in one of the two studies, for some patients with higher-risk SMM treated with lenalidomide +/- dexamethasone, raising the question of whether such an approach should be considered a new standard of care. In this paper, experts from the European Myeloma Network describe current biological and clinical knowledge on SMM, focusing on novel insights into its molecular pathogenesis, new prognostic scoring systems proposed to identify SMM patients at higher risk of early transformation, and updated results of completed or ongoing clinical trials. Finally, some practical recommendations for the real-life management of these patients, based on Delphi consensus methodology, are provided

    Signatures of mutational processes in human cancer.

    Get PDF
    All cancers are caused by somatic mutations; however, understanding of the biological processes generating these mutations is limited. The catalogue of somatic mutations from a cancer genome bears the signatures of the mutational processes that have been operative. Here we analysed 4,938,362 mutations from 7,042 cancers and extracted more than 20 distinct mutational signatures. Some are present in many cancer types, notably a signature attributed to the APOBEC family of cytidine deaminases, whereas others are confined to a single cancer class. Certain signatures are associated with age of the patient at cancer diagnosis, known mutagenic exposures or defects in DNA maintenance, but many are of cryptic origin. In addition to these genome-wide mutational signatures, hypermutation localized to small genomic regions, 'kataegis', is found in many cancer types. The results reveal the diversity of mutational processes underlying the development of cancer, with potential implications for understanding of cancer aetiology, prevention and therapy

    Biology of peripheral T cell lymphomas – Not otherwise specified: Is something finally happening?

    Get PDF
    Introduction: Peripheral T-cell lymphomas represent a rare, heterogeneous group of nodal and extra-nodal mature T-cell lymphomas. Among those, the subtype of PTCL not otherwise specified (PTCL-NOS) account for about 25% of all PTCL. While other PTCL subtypes are increasingly recognized as discrete entities based on specific genotypic and phenotypic alterations, the diagnosis of PTCL-NOS is currently performed on an “exclusion criteria” model, since PTCL-NOS lack pathognomonic features. Methods: In this review, we describe the classical pathological features of PTCL-NOS and integrate them with the most recent molecular findings. Results: Thanks to gene expression profiling and next generation sequencing approaches, we have recently improved our knowledge of PTCL in general and PTCL-NOS in particular. Indeed, specific patterns of gene expression were reported to segregate PTCL into more homogeneous subtypes associated with distinct clinical outcome. Furthermore, we describe how immunophenotypic, expression and mutational data helped to better define a new subgroup of PTCL-NOS displaying a global profile close to T Follicular Helper cell elements. Finally, we review how these new acquisitions are changing the current diagnostic approach to PTCL-NOS, and how phenotypic features and oncogenic pathways operative in these lymphomas are becoming targets of novel treatments. Conclusion: Although no recurrent and specific biological aberrations have been discovered yet, novel integrated genomic and transcriptomics approaches are significantly improving our knowledge of PTCL biology and support the development of new powerful diagnostic and prognostic markers, as well as targets of future therapies

    The Route of the Malignant Plasma Cell in Its Survival Niche: Exploring &ldquo;Multiple Myelomas&rdquo;

    No full text
    Growing evidence points to multiple myeloma (MM) and its stromal microenvironment using several mechanisms to subvert effective immune and anti-tumor responses. Recent advances have uncovered the tumor-stromal cell influence in regulating the immune-microenvironment and have envisioned targeting these suppressive pathways to improve therapeutic outcomes. Nevertheless, some subgroups of patients include those with particularly unfavorable prognoses. Biological stratification can be used to categorize patient-, disease- or therapy-related factors, or alternatively, these biological determinants can be included in a dynamic model that customizes a given treatment to a specific patient. Genetic heterogeneity and current knowledge enforce a systematic and comprehensive bench-to-bedside approach. Given the increasing role of cancer stem cells (CSCs) in better characterizing the pathogenesis of solid and hematological malignancies, disease relapse, and drug resistance, identifying and describing CSCs is of paramount importance in the management of MM. Even though the function of CSCs is well-known in other cancer types, their role in MM remains elusive. With this review, we aim to provide an update on MM homing and resilience in the bone marrow micro milieu. These data are particularly interesting for clinicians facing unmet medical needs while designing novel treatment approaches for MM

    Genomics of Smoldering Multiple Myeloma: Time for Clinical Translation of Findings?

    No full text
    Smoldering multiple myeloma (SMM) is an asymptomatic disorder of clonal bone marrow (BM) plasma cells (PCs) in between the premalignant condition known as monoclonal gammopathy of undetermined significance and overt multiple myeloma (MM). It is characterized by a deep biological heterogeneity that is reflected in a markedly variable progression risk among patients. Recently proposed risk stratification models mainly rely on indirect markers of disease burden and are unable to identify cases in whom clonal PCs have already undergone the “malignant switch” but major clonal expansion has not occurred yet. In the last years, the application of next-generation sequencing (NGS) techniques has led to profound advances in the understanding of the molecular bases of SMM progression, and in all likelihood, it will contribute to the needed improvement of SMM prognostication. In this Review, we describe the recent advances in characterizing the genomic landscape of SMM and intrinsic determinants of its progression, highlighting their implications in terms of understanding of tumor evolution and prognostication. We also review the main studies investigating the role of the microenvironment in this early disease stage. Finally, we mention the results of the first randomized clinical trials and discuss the potential clinical translability of the genomic insights

    A Journey Through Myeloma Evolution: From the Normal Plasma Cell to Disease Complexity

    No full text
    Abstract. The knowledge of cancer origin and the subsequent tracking of disease evolution represent unmet needs that will soon be within clinical reach. This will provide the opportunity to improve patient's stratification and to personalize treatments based on cancer biology along its life history. In this review, we focus on the molecular pathogenesis of multiple myeloma (MM), a hematologic malignancy with a well-known multi-stage disease course, where such approach can sooner translate into a clinical benefit. We describe novel insights into modes and timing of disease initiation. We dissect the biology of the preclinical and pre-malignant phases, elucidating how knowledge of the genomics of the disease and the composition of the microenvironment allow stratification of patients based on risk of disease progression. Then, we explore cell-intrinsic and cell-extrinsic drivers of MM evolution to symptomatic disease. Finally, we discuss how this may relate to the development of refractory disease after treatment. By integrating an evolutionary view of myeloma biology with the recent acquisitions on its clonal heterogeneity, we envision a way to drive the clinical management of the disease based on its detailed biological features more than surrogates of disease burden
    corecore