1
|
Javier López Rivera J, Gomez-Lopera N, Moreno-Garcia DJ, Orduz-Rodriguez R, Combariza-Vallejo JF, Isaza-Ruget M. Plasma Cell Enrichment and New Genomic Approaches in Multiple Myeloma: A Scoping Review. J Appl Lab Med 2025:jfaf044. [PMID: 40248905 DOI: 10.1093/jalm/jfaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/12/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Multiple myeloma (MM) is a genetically heterogeneous disease where specific genetic abnormalities have a significant impact on a patient's prognosis. Diagnostic and prognostic tools like fluorescence in situ hybridization (FISH), PCR, microarrays, and next-generation sequencing (NGS) have transformed MM management. However, the effectiveness of these techniques is often limited by the low concentration of plasma cells in bone marrow samples, which makes enrichment methods necessary. This review aims to clarify how these techniques enhance the detection of genetic abnormalities, reduce false-negative results, and facilitate more precise risk stratification for MM patients. CONTENT Following Preferred Reporting Items for Systematic reviews and Meta-Analyses Extension for Scoping Review (PRISMA-ScR) guidelines, the literature on plasma cell separation methods used in genetic studies of MM was systematically identified and mapped. Searches were conducted in the Medline and Embase databases using a structured strategy, supplemented by manual searches on Google Scholar. Of 399 publications evaluated, 69 met the inclusion criteria; 37% utilized FISH and 19% demonstrated an increasing use of NGS. Plasma cell enrichment significantly improved diagnostic accuracy, increasing the detection rates of genetic abnormalities from 61% in non-enriched samples to 95.5% in enriched samples. While FISH remains the gold standard, emerging technologies such as NGS offer superior sensitivity and the ability to identify critical genetic alterations to refine molecular subtypes. SUMMARY Clinically significant genetic alterations are detected more frequently with plasma cell enrichment techniques, contributing to improved prognosis and treatment strategies for MM patients.
Collapse
Affiliation(s)
- Juan Javier López Rivera
- Laboratorio Especializado en Biología Molecular, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
- Grupo de Genética Médica, Clínica Universitaria Colombia, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
| | - Natalia Gomez-Lopera
- Laboratorio Clínico y de Patología, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
| | | | - Rocío Orduz-Rodriguez
- Laboratorio Clínico y de Patología, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
| | - Juan F Combariza-Vallejo
- Servicio de Hematología, Clínica Universitaria Colombia, Clínica Colsanitas S.A., Grupo Keralty, Bogotá, Colombia
| | - Mario Isaza-Ruget
- Laboratorio Clínico y de Patología, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
- Unidad de Investigación, Fundación Universitaria Sanitas, Grupo de investigación INPAC, Grupo Keralty, Bogotá, Colombia
| |
Collapse
|
2
|
Hose D, Ray S, Rößler S, Thormann U, Schnettler R, de Veirman K, El Khassawna T, Heiss C, Hild A, Zahner D, Alagboso F, Henss A, Beck S, Emde-Rajaratnam M, Burhenne J, Bamberger J, Menu E, de Bruyne E, Gelinsky M, Kampschulte M, Rohnke M, Wenisch S, Vanderkerken K, Hanke T, Seckinger A, Alt V. Bortezomib-releasing silica-collagen xerogels for local treatment of osteolytic bone- and minimal residual disease in multiple myeloma. J Hematol Oncol 2024; 17:128. [PMID: 39695697 PMCID: PMC11657678 DOI: 10.1186/s13045-024-01636-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Accumulation of malignant plasma cells in the bone marrow causes lytic bone lesions in 80% of multiple myeloma patients. Frequently fracturing, they are challenging to treat surgically. Myeloma cells surviving treatment in the presumably protective environment of bone lesions impede their healing by continued impact on bone turnover and can explain regular progression of patients without detectable minimal residual disease (MRD). Locally applicable biomaterials could stabilize and foster healing of bone defects, simultaneously delivering anti-cancer compounds at systemically intolerable concentrations, overcoming drug resistance. METHODS We developed silica-collagen xerogels (sicXer) and bortezomib-releasing silica-collagen xerogels (boXer) for local treatment of osteolytic bone disease and MRD. In vitro and in vivo (tissue sections) release of bortezomib was assessed by ultrahigh-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS) and time-of-flight secondary ion mass spectrometry (ToF-SIMS). Material impact on bone formation was assessed in vitro regarding osteoclast/osteoblast numbers and activity. In vivo, drilling defects in a rat- and the 5T33-myeloma mouse model were treated by both materials and assessed by immunohistochemistry, UPLC-MS/MS, µCT, and ToF-SIMS. The material's anti-myeloma activity was assessed using ten human myeloma cell lines (HMCLs) and eight primary myeloma cell samples including four patients refractory to systemic bortezomib treatment. RESULTS sicXer and boXer show primary stability comparable to trabecular bone. Granule size and preparation method tailor degradation as indicated by release of the xerogel components (silica and collagen) and bortezomib into culture medium. In vitro, both materials reduce osteoclast activity and do not negatively interfere with osteoblast differentiation and function. The presumed resulting net bone formation with maintained basic remodeling properties was validated in vivo in a rat bone defect model, showing significantly enhanced bone formation for boXer compared to non-treated defects. Both materials induce myeloma cell apoptosis in all HMCLs and primary myeloma cell samples. In the 5T33-myeloma mouse model, both materials stabilized drilling defects and locally controlled malignant plasma cell growth. CONCLUSIONS The combination of stabilization of fracture-prone lesions, stimulation of bone healing, and anti-tumor effect suggest clinical testing of sicXer and boXer as part of a combined systemic/local treatment strategy in multiple myeloma and non-malignant diseases.
Collapse
Affiliation(s)
- Dirk Hose
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium.
| | - Seemun Ray
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | - Sina Rößler
- Institut für Werkstoffwissenschaft, Max-Bergmann-Zentrum für Biomaterialien, Technische Universität Dresden, Budapester Straße 27, 01069, Dresden, Germany
| | - Ulrich Thormann
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | | | - Kim de Veirman
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Thaqif El Khassawna
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | - Christian Heiss
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | - Anne Hild
- Klinische Anatomie und Experimentelle Chirurgie C/O Institut für Veterinär-Anatomie, -Histologie und -Embryologie, Justus-Liebig-Universität Gießen, Frankfurter Straße 98, 35392, Gießen, Germany
| | - Daniel Zahner
- Justus-Liebig-Universität Gießen, Ludwigstraße 23, 35392, Gießen, Germany
| | - Francisca Alagboso
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | - Anja Henss
- I. Physikalisches Institut, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring 16, 35392, Gießen, Germany
| | - Susanne Beck
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Martina Emde-Rajaratnam
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Jürgen Burhenne
- Innere Medizin IX - Abteilung für Klinische Pharmakologie und Pharmakoepidemiologie, Medizinische Fakultät/Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Juliane Bamberger
- Labor Für Experimentelle Radiologie, Justus-Liebig-Universität Gießen, Carl-Maria-von-Weber-Straße 8, 35392, Gießen, Germany
| | - Eline Menu
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Elke de Bruyne
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Michael Gelinsky
- Zentrum für Translationale Knochen-, Gelenk- und Weichgewebeforschung, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Marian Kampschulte
- Labor Für Experimentelle Radiologie, Justus-Liebig-Universität Gießen, Carl-Maria-von-Weber-Straße 8, 35392, Gießen, Germany
| | - Marcus Rohnke
- Physikalisch-Chemisches Institut, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring 17, 35392, Gießen, Germany
| | - Sabine Wenisch
- Klinische Anatomie und Experimentelle Chirurgie C/O Institut für Veterinär-Anatomie, -Histologie und -Embryologie, Justus-Liebig-Universität Gießen, Frankfurter Straße 98, 35392, Gießen, Germany
| | - Karin Vanderkerken
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Thomas Hanke
- Institut für Werkstoffwissenschaft, Max-Bergmann-Zentrum für Biomaterialien, Technische Universität Dresden, Budapester Straße 27, 01069, Dresden, Germany
| | - Anja Seckinger
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Volker Alt
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany.
- Klinik und Poliklinik für Unfallchirurgie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
3
|
Sima H, Shao W. Advancements in the design and function of bispecific CAR-T cells targeting B Cell-Associated tumor antigens. Int Immunopharmacol 2024; 142:113166. [PMID: 39298818 DOI: 10.1016/j.intimp.2024.113166] [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: 07/18/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Single-targeted CAR-T has exhibited notable success in treating B-cell tumors, effectively improving patient outcomes. However, the recurrence rate among patients remains above fifty percent, primarily attributed to antigen escape and the diminished immune persistence of CAR-T cells. Over recent years, there has been a surge of interest in bispecific CAR-T cell therapies, marked by an increasing number of research articles and clinical applications annually. This paper undertakes a comprehensive review of influential studies on the design of bispecific CAR-T in recent years, examining their impact on bispecific CAR-T efficacy concerning disease classification, targeted antigens, and CAR design. Notable distinctions in antigen targeting within B-ALL, NHL, and MM are explored, along with an analysis of how CAR scFv, transmembrane region, hinge region, and co-stimulatory region design influence Bi-CAR-T efficacy across different tumors. The summary provided aims to serve as a reference for designing novel and improved CAR-Ts, facilitating more efficient treatment for B-cell malignant tumors.
Collapse
Affiliation(s)
- Helin Sima
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, China.
| |
Collapse
|
4
|
Behsen AD, Holien T, Micci F, Rye M, Rasmussen JM, Andersen K, Hess ES, Børset M, Keats J, Våtsveen TK, Misund K. Cell surface marker heterogeneity in human myeloma cell lines for modeling of disease and therapy. Sci Rep 2024; 14:28805. [PMID: 39567630 PMCID: PMC11579509 DOI: 10.1038/s41598-024-80263-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 11/18/2024] [Indexed: 11/22/2024] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy originating from plasma cells. Genetically, MM is categorized into two subtypes: hyperdiploid and non-hyperdiploid tumors, with distinct chromosomal characteristics. Human myeloma cell lines (HMCLs) are instrumental in understanding MM and identifying therapeutic targets. However, their utility depends on their resemblance to patient-derived cells. We analyzed protein expression of cell surface markers, including myeloma drug targets and immunotherapy-relevant markers, in nine HMCLs using flow cytometry. RNAseq analysis was performed to correlate protein expression with transcriptomic data. While five cell surface markers (CD47, CD49d, CD138, CD269 (BCMA), and GPRC5D) were highly expressed and three (CD19, CD20, and CD117) were low or negative across cell lines, heterogenous expression was observed for the 16 remaining markers. A comparative study with patient-derived transcriptomic data indicated that four of our six in-house HMCLs closely resembled patient disease. Furthermore, our hyperdiploid HMCLs correlated better with hyperdiploid than non-hyperdiploid patient samples. Here, we also describe three previously uncharacterized HMCLs; IH-1, URVIN and FOLE. Our findings underscore the importance of evaluating surface protein expression in HMCLs when modeling MM. The observed variations in expression levels emphasize the need for a strategic selection of cell lines based on the study's objectives.
Collapse
Affiliation(s)
- Alenka Djarmila Behsen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Toril Holien
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Immunology and Transfusion Medicine, St. Olav's University Hospital, Trondheim, Norway
- Department of Hematology, St. Olav's University Hospital, Trondheim, Norway
| | - Francesca Micci
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Morten Rye
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Surgery, St. Olav's University Hospital, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Jenny Malm Rasmussen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kristin Andersen
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Eli Svorkdal Hess
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Magne Børset
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Immunology and Transfusion Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Jonathan Keats
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Thea Kristin Våtsveen
- K.G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Kristine Misund
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Medical Genetics, St. Olav's University Hospital, Trondheim, Norway.
| |
Collapse
|
5
|
Pallocca M, Betti M, Baldinelli S, Palombo R, Bucci G, Mazzarella L, Tonon G, Ciliberto G. Clinical bioinformatics desiderata for molecular tumor boards. Brief Bioinform 2024; 25:bbae447. [PMID: 39297878 PMCID: PMC11411775 DOI: 10.1093/bib/bbae447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/28/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Clinical Bioinformatics is a knowledge framework required to interpret data of medical interest via computational methods. This area became of dramatic importance in precision oncology, fueled by cancer genomic profiling: most definitions of Molecular Tumor Boards require the presence of bioinformaticians. However, all available literature remained rather vague on what are the specific needs in terms of digital tools and expertise to tackle and interpret genomics data to assign novel targeted or biomarker-driven targeted therapies to cancer patients. To fill this gap, in this article, we present a catalog of software families and human skills required for the tumor board bioinformatician, with specific examples of real-world applications associated with each element presented.
Collapse
Affiliation(s)
- Matteo Pallocca
- Institute of Experimental Endocrinology and Oncology, National Research Council, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Martina Betti
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| | - Sara Baldinelli
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| | - Ramona Palombo
- Institute of Experimental Endocrinology and Oncology, National Research Council, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Gabriele Bucci
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milan, Italy
| | - Luca Mazzarella
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IRCCS IEO - European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
- Department of Experimental Oncology, IRCCS IEO - European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Giovanni Tonon
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, and Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milan, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| |
Collapse
|
6
|
Hummel M, Hielscher T, Emde-Rajaratnam M, Salwender H, Beck S, Scheid C, Bertsch U, Goldschmidt H, Jauch A, Moreaux J, Seckinger A, Hose D. Quantitative Integrative Survival Prediction in Multiple Myeloma Patients Treated With Bortezomib-Based Induction, High-Dose Therapy and Autologous Stem Cell Transplantation. JCO Precis Oncol 2024; 8:e2300613. [PMID: 38986047 PMCID: PMC11371111 DOI: 10.1200/po.23.00613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/13/2024] [Accepted: 04/24/2024] [Indexed: 07/12/2024] Open
Abstract
PURPOSE Given the high heterogeneity in survival for patients with multiple myeloma, it would be clinically useful to quantitatively predict the individual survival instead of attributing patients to two to four risk groups as in current models, for example, revised International Staging System (R-ISS), R2-ISS, or Mayo-2022-score. PATIENTS AND METHODS Our aim was to develop a quantitative prediction tool for individual patient's 3-/5-year overall survival (OS) probability. We integrated established clinical and molecular risk factors into a comprehensive prognostic model and evaluated and validated its risk discrimination capabilities versus R-ISS, R2-ISS, and Mayo-2022-score. RESULTS A nomogram for estimating OS probabilities was built on the basis of a Cox regression model. It allows one to translate the individual risk profile of a patient into 3-/5-year OS probabilities by attributing points to each prognostic factor and summing up all points. The nomogram was externally validated regarding discrimination and calibration. There was no obvious bias or overfitting of the prognostic index on the validation cohort. Resampling-based and external evaluation showed good calibration. The c-index of the model was similar on the training (0.76) and validation cohort (0.75) and significantly higher than for the R-ISS (P < .001) or R2-ISS (P < .01). CONCLUSION In summary, we developed and validated individual quantitative nomogram-based OS prediction. Continuous risk assessment integrating molecular prognostic factors is superior to R-ISS, R2-ISS, or Mayo-2022-score alone.
Collapse
Affiliation(s)
- Manuela Hummel
- Deutsches Krebsforschungszentrum, Abteilung für Biostatistik, Heidelberg, Germany
| | - Thomas Hielscher
- Deutsches Krebsforschungszentrum, Abteilung für Biostatistik, Heidelberg, Germany
| | - Martina Emde-Rajaratnam
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), Jette, Belgium
| | - Hans Salwender
- Asklepios Tumorzentrum Hamburg, AK Altona and St Georg, Hamburg, Germany
| | - Susanne Beck
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), Jette, Belgium
- Universitätsklinikum Heidelberg, Molekularpathologisches Zentrum, Heidelberg, Germany
| | - Christof Scheid
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
| | - Uta Bertsch
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Heidelberg, Germany
| | - Hartmut Goldschmidt
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Heidelberg, Germany
- Nationales Centrum für Tumorerkrankungen, Heidelberg, Germany
| | - Anna Jauch
- Universität Heidelberg, Institut für Humangenetik, Heidelberg, Germany
| | - Jérôme Moreaux
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Anja Seckinger
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), Jette, Belgium
| | - Dirk Hose
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), Jette, Belgium
| |
Collapse
|
7
|
Tagliari de Oliveira S, Binato R, Ellen Broto G, Tomie Takakura E, Navarro Gordan Ferreira Martins L, Abdelhay E, Panis C. Transcriptome of bone marrow-Derived stem cells reveals new inflammatory mediators related to increased survival in patients with multiple myeloma. Cytokine 2024; 179:156613. [PMID: 38643632 DOI: 10.1016/j.cyto.2024.156613] [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: 02/02/2024] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 04/23/2024]
Abstract
Although multiple myeloma (MM) is a neoplasm that leads affected individuals to death, little is known about why some patients survive much longer than others. In this context, we investigated the transcriptomic profile of bone marrow hematopoietic stem cells obtained from MM patients and compared the clinical outcomes of death and survival six months after bone marrow transplantation. The leukapheresis products of 39 patients with MM eligible for autologous transplantation were collected and analyzed. After extraction, the RNA was analyzed using the GeneChip Human Exon 1.0 Array method. The transcriptome profile was analyzed in silico, and the differentially expressed signaling pathways of interest were validated. The results showed a difference in the expression of inflammation-related genes, immune response processes, and the oxidative stress pathway. The in silico study also pointed out the involvement of the NFκB transcription factor in the possible modulation of these genes. We chose to validate molecules participating in these processes, including the cytokines TNF-α, IFN-γ, and TGF-β1; in addition, we measured the levels of oxidative stress mediators (pro-oxidant profile and the total antioxidant capacity). TNF-α levels were significantly reduced in patients who died and were over 50 years old at diagnosis, as well as in patients with plasmacytoma. Increased TNF-α was detected in patients with very high levels of β2-microglobulin. IFN-γ reduction was observed in patients with a complete response to treatment compared to those with a very good response. Patients with plasmacytoma who died also had an increased pro-oxidant profile. These data show the profile of inflammatory response markers that are altered in patients with MM who die quickly and serve as a basis for the development of future studies of markers to predict better survival in this disease.
Collapse
Affiliation(s)
- Stefania Tagliari de Oliveira
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Unioeste - Francisco Beltrão - Paraná, Brazil; Rede de Assistência a Saúde Metropolitana de Sarandi - Programa de Residência Médica em Clínica Médica - Sarandi - Paraná, Brazil
| | - Renata Binato
- Instituto Nacional de Câncer, Centro de Transplante de Medula Óssea, Rio de Janeiro, Brazil
| | - Geise Ellen Broto
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Unioeste - Francisco Beltrão - Paraná, Brazil
| | - Erika Tomie Takakura
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Unioeste - Francisco Beltrão - Paraná, Brazil
| | | | - Eliana Abdelhay
- Instituto Nacional de Câncer, Centro de Transplante de Medula Óssea, Rio de Janeiro, Brazil
| | - Carolina Panis
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Unioeste - Francisco Beltrão - Paraná, Brazil.
| |
Collapse
|
8
|
Seckinger A, Salwender H, Martin H, Scheid C, Hielscher T, Bertsch U, Hummel M, Jauch A, Knauf W, Emde-Rajaratnam M, Beck S, Neben K, Dührig J, Lindemann W, Schmidt-Wolf IGH, Hänel M, Blau IW, Weisel K, Weinhold N, Raab MS, Goldschmidt H, Choon-Quinones M, Hose D. Molecular Long-Term Analysis of the GMMG-HD4 Trial in Multiple Myeloma-Patterns of Association of Chromosomal Aberrations with Response and Proliferation Determining Survival in Selecting Treatments in View of Limited Resources in Low- and Middle-Income Countries. Int J Mol Sci 2024; 25:6431. [PMID: 38928138 PMCID: PMC11204152 DOI: 10.3390/ijms25126431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Based on the lack of differences in progression-free and overall survival after a median follow-up of 93 months in our HOVON-65/GMMG-HD4 trial (German part; n = 395) randomizing VAD induction (vincristin/adriamycin/dexamthasone)/tandem-transplantation/thalidomide-maintenance vs. PAD induction (bortezomib/adriamycin/dexamethasone)/tandem transplantation/bortezomib maintenance, we discern how chromosomal aberrations determine long-term prognosis by different patterns of association with proliferation and treatment-dependent response, whether responses achieved by different regimens are equal regarding prognosis, and whether subpopulations of patients could be defined as treatable without upfront "novel agents" in cases of limited resources, e.g., in low- or middle-income countries. Serum parameters and risk factors were assessed in 395 patients. CD138-purified plasma cells were subjected to fluorescence in situ hybridization (n = 354) and gene expression profiling (n = 204). We found chromosomal aberrations to be associated in four patterns with survival, proliferation, and response: deletion (del) del17p13, del8p21, del13q14, (gain) 1q21+, and translocation t(4;14) (all adverse) associate with higher proliferation. Of these, del17p is associated with an adverse response (pattern 1), and 1q21+, t(4;14), and del13q14 with a treatment-dependent better response (pattern 2). Hyperdiploidy associates with lower proliferation without impacting response or survival (pattern 3). Translocation t(11;14) has no association with survival but a treatment-dependent adverse response (pattern 4). Significantly fewer patients reach a near-complete response or better with "conventional" (VAD) vs. bortezomib-based treatment after induction or high-dose melphalan. These patients, however, show significantly better median progression-free and overall survival. Molecularly, patients responding to the two regimens differ in gene expression, indicating distinct biological properties of the responding myeloma cells. Patients with normal renal function (89.4%), low cytogenetic risk (72.5%), or low proliferation rate (37.9%) neither benefit in progression-free nor overall survival from bortezomib-based upfront treatment. We conclude that response level, the treatment by which it is achieved, and molecular background determine long-term prognosis. Chromosomal aberrations are associated in four patterns with proliferation and treatment-dependent responses. Associations with faster and deeper responses can be deceptive in the case of prognostically adverse aberrations 1q21+ and t(4;14). Far from advocating a return to "outdated" treatments, if resources do not permit state-of-the-art-treatment, normal renal function and/or molecular profiling identifies patient subpopulations doing well without upfront "novel agents".
Collapse
Affiliation(s)
- Anja Seckinger
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), 1090 Jette, Belgium
- Independent Myeloma Alliance, 8808 Pfäffikon, SZ, Switzerland
| | - Hans Salwender
- Department of Internal Medicine II, Asklepios Klinik Altona, 22763 Hamburg, Germany
| | - Hans Martin
- Department of Medicine, Hematology/Oncology, Goethe-University of Frankfurt, 60590 Frankfurt, Germany
| | - Christof Scheid
- Department I of Internal Medicine, University of Cologne, 50923 Köln, Germany
| | - Thomas Hielscher
- Abteilung für Biostatistik, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| | - Uta Bertsch
- Medizinische Klinik V, Universitätsklinikum Heidelberg, 69120 Heidelberg, Germany
| | - Manuela Hummel
- Abteilung für Biostatistik, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| | - Anna Jauch
- Institut für Humangenetik, Universität Heidelberg, 69120 Heidelberg, Germany
| | - Wolfgang Knauf
- Onkologische Gemeinschaftspraxis, Agaplesion Bethanien Krankenhaus, 60389 Frankfurt, Germany
| | - Martina Emde-Rajaratnam
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), 1090 Jette, Belgium
| | - Susanne Beck
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), 1090 Jette, Belgium
| | - Kai Neben
- Klinikum Mittelbaden, Medizinische Klinik 2, 76530 Baden-Baden, Germany
| | - Jan Dührig
- Katholisches Krankenhaus Hagen, 58099 Hagen, Germany
| | - Walter Lindemann
- Department of Hematology, University Hospital Essen, 45147 Essen, Germany
| | | | - Mathias Hänel
- Department of Internal Medicine III, Klinikum Chemnitz GmbH, 09113 Chemnitz, Germany
| | - Igor W. Blau
- Medical Clinic III Hematology and Oncology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Niels Weinhold
- Medizinische Klinik V, Universitätsklinikum Heidelberg, 69120 Heidelberg, Germany
| | - Marc S. Raab
- Medizinische Klinik V, Universitätsklinikum Heidelberg, 69120 Heidelberg, Germany
| | - Hartmut Goldschmidt
- Medizinische Klinik V, Universitätsklinikum Heidelberg, 69120 Heidelberg, Germany
- Nationales Centrum für Tumorerkrankungen, 69120 Heidelberg, Germany
| | | | - Dirk Hose
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), 1090 Jette, Belgium
- Independent Myeloma Alliance, 8808 Pfäffikon, SZ, Switzerland
| |
Collapse
|