1
|
Jakobsen MZ, Brøndum RF, Gregersen H, Due H, Dybkær K. A systematic literature review on clonal evolution events preceding relapse in multiple myeloma. Crit Rev Oncol Hematol 2025; 205:104560. [PMID: 39549892 DOI: 10.1016/j.critrevonc.2024.104560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
Despite considerable treatment advances, multiple myeloma (MM) remains an incurable hematological cancer due to treatment resistance. A systematic literature search was conducted to identify determinants for clonal evolution driving relapse and drug resistance in MM. A total of 631 non-duplicate publications were screened of which 28 articles were included for data extraction. Genetic alterations, mutational signatures, evolutionary trajectories, and non-genetic determinants were identified as key topics to characterize clonal evolution in relapsed MM. A variety of factors led to clonal diversification and increased tumor mutation burden, such as MAPK-Ras mutations and incremental changes related to chromosomal bands 1 and 17, while mutational signature analyses revealed that APOBEC activity and melphalan treatment leave a distinct impact on the clonal composition in MM genomes. To capture and dissect tumor heterogeneity, our review suggests combining methods or using technical approaches with high resolution to assess the impact of clonal evolution.
Collapse
Affiliation(s)
- Maja Zimmer Jakobsen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Rasmus Froberg Brøndum
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Center for Clinical Data Science, Aalborg University, and Aalborg University Hospital, Aalborg, Denmark
| | - Henrik Gregersen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Hanne Due
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Karen Dybkær
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark.
| |
Collapse
|
2
|
Gutiérrez-González A, Del Hierro I, Cariaga-Martínez AE. Advancements in Multiple Myeloma Research: High-Throughput Sequencing Technologies, Omics, and the Role of Artificial Intelligence. BIOLOGY 2024; 13:923. [PMID: 39596878 PMCID: PMC11592186 DOI: 10.3390/biology13110923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/01/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024]
Abstract
Multiple myeloma is a complex and challenging type of blood cancer that affects plasma cells in the bone marrow. In recent years, the development of advanced research techniques, such as omics approaches-which involve studying large sets of biological data like genes and proteins-and high-throughput sequencing technologies, has allowed researchers to analyze vast amounts of genetic information rapidly and gain new insights into the disease. Additionally, the advent of artificial intelligence tools has accelerated data analysis, enabling more accurate predictions and improved treatment strategies. This review aims to highlight recent research advances in multiple myeloma made possible by these novel techniques and to provide guidance for researchers seeking effective approaches in this field.
Collapse
Affiliation(s)
| | | | - Ariel Ernesto Cariaga-Martínez
- DS-OMICS—Data Science and Omics, AI-Driven Biomedicine Group, Universidad Alfonso X el Sabio, 28619 Villanueva de la Cañada, Spain; (A.G.-G.); (I.D.H.)
| |
Collapse
|
3
|
Morawska M, Kiełbus M, Paziewska M, Szelest M, Karczmarczyk A, Zaleska J, Własiuk P, Giannopoulos K, Grząśko N. Mutations of ARID1B, PIK3C2B, KMT2B, and FAT1 genes influence clinical outcome in newly diagnosed myeloma. Cancer Genet 2024; 288-289:118-125. [PMID: 39536425 DOI: 10.1016/j.cancergen.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/24/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
The study aimed to elucidate the mutational profile of patients with newly diagnosed multiple myeloma to understand correlations of alterations with clinical outcomes. A cohort of 20 patients was enrolled, and mutational analysis was conducted using the TruSight Oncology 500 DNA Kit. Identified genetic alterations were related to clinicopathologic features and treatment outcomes. A total of 724 high-quality variants were validated. All patients harbored mutations associated with the RTK-RAS pathway, with over half having alterations in PI3 K, NOTCH, and WNT pathways. Several gene mutations were associated with specific clinical characteristics and prognostic indicators, revealing a complex interplay between genetic alterations and myeloma type, standard prognostic indicators, biochemical parameters, and renal function. Genetic alterations significantly influencing progression-free survival concerned PIK3C2B, ARID1B genes, and concomitant mutations in KMT2B, FAT1, and ARID1B. The findings underscore the potential of gene mutation-based prognostic tools in enhancing clinical decision-making and suggest that further exploration of identified genetic markers could pave the way for improved prognostic stratification and targeted therapeutic interventions in multiple myeloma.
Collapse
Affiliation(s)
- Marta Morawska
- Department of Experimental Haematooncology, Medical University of Lublin, Lublin, Poland.
| | - Michał Kiełbus
- Department of Experimental Haematooncology, Medical University of Lublin, Lublin, Poland
| | - Magdalena Paziewska
- Department of Experimental Haematooncology, Medical University of Lublin, Lublin, Poland
| | - Monika Szelest
- Department of Experimental Haematooncology, Medical University of Lublin, Lublin, Poland
| | - Agnieszka Karczmarczyk
- Department of Experimental Haematooncology, Medical University of Lublin, Lublin, Poland
| | - Joanna Zaleska
- Department of Experimental Haematooncology, Medical University of Lublin, Lublin, Poland
| | - Paulina Własiuk
- Department of Experimental Haematooncology, Medical University of Lublin, Lublin, Poland
| | - Krzysztof Giannopoulos
- Department of Experimental Haematooncology, Medical University of Lublin, Lublin, Poland
| | - Norbert Grząśko
- Department of Experimental Haematooncology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
4
|
Forster S, Radpour R, Ochsenbein AF. Molecular and immunological mechanisms of clonal evolution in multiple myeloma. Front Immunol 2023; 14:1243997. [PMID: 37744361 PMCID: PMC10516567 DOI: 10.3389/fimmu.2023.1243997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by the proliferation of clonal plasma cells in the bone marrow (BM). It is known that early genetic mutations in post-germinal center B/plasma cells are the cause of myelomagenesis. The acquisition of additional chromosomal abnormalities and distinct mutations further promote the outgrowth of malignant plasma cell populations that are resistant to conventional treatments, finally resulting in relapsed and therapy-refractory terminal stages of MM. In addition, myeloma cells are supported by autocrine signaling pathways and the tumor microenvironment (TME), which consists of diverse cell types such as stromal cells, immune cells, and components of the extracellular matrix. The TME provides essential signals and stimuli that induce proliferation and/or prevent apoptosis. In particular, the molecular pathways by which MM cells interact with the TME are crucial for the development of MM. To generate successful therapies and prevent MM recurrence, a thorough understanding of the molecular mechanisms that drive MM progression and therapy resistance is essential. In this review, we summarize key mechanisms that promote myelomagenesis and drive the clonal expansion in the course of MM progression such as autocrine signaling cascades, as well as direct and indirect interactions between the TME and malignant plasma cells. In addition, we highlight drug-resistance mechanisms and emerging therapies that are currently tested in clinical trials to overcome therapy-refractory MM stages.
Collapse
Affiliation(s)
- Stefan Forster
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Adrian F. Ochsenbein
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Marx A, Osváth M, Szikora B, Pipek O, Csabai I, Nagy Á, Bödör C, Matula Z, Nagy G, Bors A, Uher F, Mikala G, Vályi-Nagy I, Kacskovics I. Liquid biopsy-based monitoring of residual disease in multiple myeloma by analysis of the rearranged immunoglobulin genes-A feasibility study. PLoS One 2023; 18:e0285696. [PMID: 37235573 DOI: 10.1371/journal.pone.0285696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The need for sensitive monitoring of minimal/measurable residual disease (MRD) in multiple myeloma emerged as novel therapies led to deeper responses. Moreover, the potential benefits of blood-based analyses, the so-called liquid biopsy is prompting more and more studies to assess its feasibility. Considering these recent demands, we aimed to optimize a highly sensitive molecular system based on the rearranged immunoglobulin (Ig) genes to monitor MRD from peripheral blood. We analyzed a small group of myeloma patients with the high-risk t(4;14) translocation, using next-generation sequencing of Ig genes and droplet digital PCR of patient-specific Ig heavy chain (IgH) sequences. Moreover, well established monitoring methods such as multiparametric flow cytometry and RT-qPCR of the fusion transcript IgH::MMSET (IgH and multiple myeloma SET domain-containing protein) were utilized to evaluate the feasibility of these novel molecular tools. Serum measurements of M-protein and free light chains together with the clinical assessment by the treating physician served as routine clinical data. We found significant correlation between our molecular data and clinical parameters, using Spearman correlations. While the comparisons of the Ig-based methods and the other monitoring methods (flow cytometry, qPCR) were not statistically evaluable, we found common trends in their target detection. Regarding longitudinal disease monitoring, the applied methods yielded complementary information thus increasing the reliability of MRD evaluation. We also detected indications of early relapse before clinical signs, although this implication needs further verification in a larger patient cohort.
Collapse
Affiliation(s)
- Anita Marx
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Doctoral School of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Magdolna Osváth
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Doctoral School of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Bence Szikora
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Orsolya Pipek
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - István Csabai
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Ákos Nagy
- Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research Group, Semmelweis University, Budapest, Hungary
| | - Csaba Bödör
- Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research Group, Semmelweis University, Budapest, Hungary
| | - Zsolt Matula
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Ginette Nagy
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - András Bors
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Ferenc Uher
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Gábor Mikala
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - István Vályi-Nagy
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Imre Kacskovics
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
6
|
CRL4 CRBN E3 Ligase Complex as a Therapeutic Target in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14184492. [PMID: 36139651 PMCID: PMC9496858 DOI: 10.3390/cancers14184492] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Immunomodulatory drugs (IMiDs) are effective in the treatment of multiple myeloma (MM) and other hematological malignancies. Cereblon (CRBN), a target of IMiDs, forms the CRL4 E3 ubiquitin ligase complex (CRL4CRBN) with DDB1, CUL4A and RBX1. The insight into the molecular mechanism of IMiDs action has advanced dramatically since the identification of cereblon (CRBN) as their direct target. Targeting CRBN by IMiDs modifies CRL4CRBN substrate specificity towards non-physiological protein targets which are subsequently ubiquitinated and degraded by the proteasome. To date, IMiDs are the only known group of protein degraders used in clinical practice. This review provides the current state of knowledge about thalidomide and its derivatives’ mechanisms of action, and highlights the future perspectives for targeted protein degraders. Abstract Multiple myeloma (MM) is the second most common hematological malignancy with a recurrent clinical course. The introduction of immunomodulatory drugs (IMiDs) was one of the milestones in MM therapy leading to a significant improvement in patients’ prognosis. Currently, IMiDs are the backbone of MM therapy in newly diagnosed and relapsed/refractory settings. It is now known that IMiDs exert their anti-myeloma activity mainly by binding cereblon (CRBN), the substrate receptor protein of the CRL4 E3 ubiquitin ligase (CRL4CRBN) complex. By binding CRBN, IMiDs alter its substrate specificity, leading to ubiquitination and proteasomal degradation of proteins essential for MM cell survival. Following the success of IMiDs, it is not surprising that the possibility of using the CRL4CRBN complex’s activity to treat MM is being further explored. In this review, we summarize the current state of knowledge about novel players in the MM therapeutic landscape, namely the CRBN E3 ligase modulators (CELMoDs), the next generation of IMiDs with broader biological activity. In addition, we discuss a new strategy of tailored proteolysis called proteolysis targeting chimeras (PROTACs) using the CRL4CRBN to degrade typically undruggable proteins, which may have relevance for the treatment of MM and other malignancies in the future.
Collapse
|