Searched for: in-biosketch:yes
person:morgag04
Genomic Profiling to Contextualize the Results of Intervention for Smoldering Multiple Myeloma
Kazandjian, Dickran; Diamond, Benjamin; Papadimitriou, Marios; Hill, Elizabeth; Sklavenitis-Pistofidis, Romanos; Ziccheddu, Bachisio; Blaney, Patrick; Chojnacka, Monika; Durante, Michael; Maclachlan, Kylee; Young, Ryan; Usmani, Saad; Davies, Faith; Getz, Gad; Ghobrial, Irene; Korde, Neha; Morgan, Gareth; Maura, Francesco; Landgren, Ola
PURPOSE/UNASSIGNED:Early intervention for high-risk smoldering multiple myeloma (HR-SMM) achieves deep and prolonged responses. It is unclear if beneficial outcomes are due to the treatment of less complex, susceptible disease or inaccuracy in clinical definition of cases entered. EXPERIMENTAL DESIGN/UNASSIGNED:In this study, we interrogated whole-genome and whole-exome sequencing for 54 patients across two HR-SMM interventional studies (NCT01572480 and NCT02279394). RESULTS/UNASSIGNED:We reveal that the genomic landscape of treated HR-SMM is generally simple as compared with newly diagnosed multiple myeloma counterparts with less inactivation of tumor suppressor genes, RAS pathway mutations, MYC disruption, and APOBEC contribution. The absence of these events parallels that of indolent precursor conditions, possibly explaining overall excellent outcomes. However, some patients harboring genomic complexity fail to sustain response and experience resistant, progressive disease. Overall, clinical risk scores do not effectively discriminate between genomically indolent and aggressive disease. CONCLUSIONS/UNASSIGNED:Genomic profiling can contextualize the advantage of early intervention in SMM and guide personalization of therapy. See related commentary by Weinhold and Rasche, p. 4263.
PMCID:11444893
PMID: 38652812
ISSN: 1557-3265
CID: 5713852
The biological and clinical impact of deletions before and after large chromosomal gains in multiple myeloma
Cirrincione, Anthony M; Poos, Alexandra M; Ziccheddu, Bachisio; Kaddoura, Marcella; Bärtsch, Marc-Andrea; Maclachlan, Kylee; Chojnacka, Monika; Diamond, Benjamin; John, Lukas; Reichert, Philipp; Huhn, Stefanie; Blaney, Patrick; Gagler, Dylan; Rippe, Karsten; Zhang, Yanming; Dogan, Ahmet; Lesokhin, Alexander M; Davies, Faith; Goldschmidt, Hartmut; Fenk, Roland; Weisel, Katja C; Mai, Elias K; Korde, Neha; Morgan, Gareth J; Usmani, Saad; Landgren, Ola; Raab, Marc S; Weinhold, Niels; Maura, Francesco
Acquisition of a hyperdiploid (HY) karyotype or immunoglobulin heavy chain (IgH) translocations are considered key initiating events in multiple myeloma (MM). To explore if other genomic events can precede these events, we analyzed whole-genome sequencing data from 1173 MM samples. By integrating molecular time and structural variants within early chromosomal duplications, we indeed identified pregain deletions in 9.4% of patients with an HY karyotype without IgH translocations, challenging acquisition of an HY karyotype as the earliest somatic event. Remarkably, these deletions affected tumor suppressor genes (TSGs) and/or oncogenes in 2.4% of patients with an HY karyotype without IgH translocations, supporting their role in MM pathogenesis. Furthermore, our study points to postgain deletions as novel driver mechanisms in MM. Using multiomics approaches to investigate their biologic impact, we found associations with poor clinical outcome in newly diagnosed patients and profound effects on both the oncogene and TSG activity despite the diploid gene status. Overall, this study provides novel insights into the temporal dynamics of genomic alterations in MM.
PMCID:11375460
PMID: 38728430
ISSN: 1528-0020
CID: 5697772
Progression-free survival as a surrogate endpoint in myeloma clinical trials: an evolving paradigm
Pawlyn, Charlotte; Schjesvold, Fredrik H; Cairns, David A; Wei, L J; Davies, Faith; Nadeem, Omar; Abdulhaq, Haifaa; Mateos, Maria-Victoria; Laubach, Jacob; Weisel, Katja; Ludwig, Heinz; Rajkumar, S Vincent; Sonneveld, Pieter; Jackson, Graham; Morgan, Gareth; Richardson, Paul G
Measurement of overall survival (OS) remains the gold standard for interpreting the impact of new therapies for multiple myeloma in phase 3 trials. However, as outcomes have improved, it is increasingly challenging to use OS as the primary endpoint if timely approval of novel agents is to be ensured to enable maximum benefit for patients. Surrogate endpoints of OS, such as progression-free survival (PFS) and response to treatment, have contributed to approval decisions by the Food and Drug Administration (FDA) and European Medicines Agency as endpoints demonstrating clinical benefit, and the FDA has recently supported the use of minimal residual disease (MRD) as an accelerated approval endpoint in multiple myeloma. This review aims to address situations in which the use of PFS as a surrogate endpoint warrants careful interpretation especially for specific subgroups of patients and considers ways to ensure that studies can be designed to account for possible discordance between PFS and OS. The utility of subgroup analyses, including the potential for those not pre-specified, to identify target populations for new agents is also discussed.
PMCID:11319634
PMID: 39134544
ISSN: 2044-5385
CID: 5697132
Deciphering the genetics and mechanisms of predisposition to multiple myeloma
Went, Molly; Duran-Lozano, Laura; Halldorsson, Gisli H; Gunnell, Andrea; Ugidos-Damboriena, Nerea; Law, Philip; Ekdahl, Ludvig; Sud, Amit; Thorleifsson, Gudmar; Thodberg, Malte; Olafsdottir, Thorunn; Lamarca-Arrizabalaga, Antton; Cafaro, Caterina; Niroula, Abhishek; Ajore, Ram; Lopez de Lapuente Portilla, Aitzkoa; Ali, Zain; Pertesi, Maroulio; Goldschmidt, Hartmut; Stefansdottir, Lilja; Kristinsson, Sigurdur Y; Stacey, Simon N; Love, Thorvardur J; Rognvaldsson, Saemundur; Hajek, Roman; Vodicka, Pavel; Pettersson-Kymmer, Ulrika; Späth, Florentin; Schinke, Carolina; Van Rhee, Frits; Sulem, Patrick; Ferkingstad, Egil; Hjorleifsson Eldjarn, Grimur; Mellqvist, Ulf-Henrik; Jonsdottir, Ingileif; Morgan, Gareth; Sonneveld, Pieter; Waage, Anders; Weinhold, Niels; Thomsen, Hauke; Försti, Asta; Hansson, Markus; Juul-Vangsted, Annette; Thorsteinsdottir, Unnur; Hemminki, Kari; Kaiser, Martin; Rafnar, Thorunn; Stefansson, Kari; Houlston, Richard; Nilsson, Björn
Multiple myeloma (MM) is an incurable malignancy of plasma cells. Epidemiological studies indicate a substantial heritable component, but the underlying mechanisms remain unclear. Here, in a genome-wide association study totaling 10,906 cases and 366,221 controls, we identify 35 MM risk loci, 12 of which are novel. Through functional fine-mapping and Mendelian randomization, we uncover two causal mechanisms for inherited MM risk: longer telomeres; and elevated levels of B-cell maturation antigen (BCMA) and interleukin-5 receptor alpha (IL5RA) in plasma. The largest increase in BCMA and IL5RA levels is mediated by the risk variant rs34562254-A at TNFRSF13B. While individuals with loss-of-function variants in TNFRSF13B develop B-cell immunodeficiency, rs34562254-A exerts a gain-of-function effect, increasing MM risk through amplified B-cell responses. Our results represent an analysis of genetic MM predisposition, highlighting causal mechanisms contributing to MM development.
PMCID:11300596
PMID: 39103364
ISSN: 2041-1723
CID: 5696742
Anthropometric traits and risk of multiple myeloma: differences by race, sex and diagnostic clinical features
Arnold, Kevin D; Ong, Krystle L; Ravi, Gayathri; Cutshall, Hannah; Purnell, Kalyn; Wessel, Meredith C; Godby, Kelly N; Bal, Susan; Giri, Smith; Rogers, Laura Q; Demark-Wahnefried, Wendy; Davies, Faith E; Costa, Luciano J; Morgan, Gareth J; Birmann, Brenda M; Brown, Elizabeth E
BACKGROUND:Obesity is an established modifiable risk factor for multiple myeloma (MM). However, associations of obesity and MM risk in Black populations, for whom obesity and MM are more common, is less clear. METHODS:Using participants enrolled in the Integrative Molecular And Genetic Epidemiology study, we evaluated the association of anthropometric traits with MM risk overall, stratified by race and sex. Among cases, we assessed the association of BMI with the presence of myeloma-defining events. RESULTS:) were more likely to present at diagnosis with low renal function (OR = 1.62, 95% CI 1.09-2.40), deletion 13q (OR = 1.73, 95% CI 1.08-2.76) and lytic lesions or compression fractures (OR = 2.39, 95% CI 0.82-7.01) and less likely to present with severe diffuse osteopenia (OR = 0.51, 95% CI 0.31-0.81). CONCLUSIONS:Findings underscore the importance of obesity as a modifiable risk factor for MM, particularly in high-risk populations, and for the clinical presentation of disease.
PMID: 38849476
ISSN: 1532-1827
CID: 5665902
1q amplification and PHF19 expressing high-risk cells are associated with relapsed/refractory multiple myeloma
Johnson, Travis S; Sudha, Parvathi; Liu, Enze; Becker, Nathan; Robertson, Sylvia; Blaney, Patrick; Morgan, Gareth; Chopra, Vivek S; Dos Santos, Cedric; Nixon, Michael; Huang, Kun; Suvannasankha, Attaya; Zaid, Mohammad Abu; Abonour, Rafat; Walker, Brian A
Multiple Myeloma is an incurable plasma cell malignancy with a poor survival rate that is usually treated with immunomodulatory drugs (iMiDs) and proteosome inhibitors (PIs). The malignant plasma cells quickly become resistant to these agents causing relapse and uncontrolled growth of resistant clones. From whole genome sequencing (WGS) and RNA sequencing (RNA-seq) studies, different high-risk translocation, copy number, mutational, and transcriptional markers can be identified. One of these markers, PHF19, epigenetically regulates cell cycle and other processes and is already studied using RNA-seq. In this study, we generate a large (325,025 cells and 49 patients) single cell multi-omic dataset and jointly quantify ATAC- and RNA-seq for each cell and matched genomic profiles for each patient. We identify an association between one plasma cell subtype with myeloma progression that we call relapsed/refractory plasma cells (RRPCs). These cells are associated with chromosome 1q alterations, TP53 mutations, and higher expression of PHF19. We also identify downstream regulation of cell cycle inhibitors in these cells, possible regulation by the transcription factor (TF) PBX1 on chromosome 1q, and determine that PHF19 may be acting primarily through this subset of cells.
PMID: 38755140
ISSN: 2041-1723
CID: 5656262
The genomic landscape of Vk*MYC myeloma highlights shared pathways of transformation between mice and humans
Maura, Francesco; Coffey, David G; Stein, Caleb K; Braggio, Esteban; Ziccheddu, Bachisio; Sharik, Meaghen E; Du, Megan T; Tafoya Alvarado, Yuliza; Shi, Chang-Xin; Zhu, Yuan Xiao; Meermeier, Erin W; Morgan, Gareth J; Landgren, Ola; Bergsagel, P Leif; Chesi, Marta
Multiple myeloma (MM) is a heterogeneous disease characterized by frequent MYC translocations. Sporadic MYC activation in the germinal center of genetically engineered Vk*MYC mice is sufficient to induce plasma cell tumors in which a variety of secondary mutations are spontaneously acquired and selected over time. Analysis of 119 Vk*MYC myeloma reveals recurrent copy number alterations, structural variations, chromothripsis, driver mutations, apolipoprotein B mRNA-editing enzyme, catalytic polypeptide (APOBEC) mutational activity, and a progressive decrease in immunoglobulin transcription that inversely correlates with proliferation. Moreover, we identify frequent insertional mutagenesis by endogenous retro-elements as a murine specific mechanism to activate NF-kB and IL6 signaling pathways shared with human MM. Despite the increased genomic complexity associated with progression, advanced tumors remain dependent on MYC. In summary, here we credential the Vk*MYC mouse as a unique resource to explore MM genomic evolution and describe a fully annotated collection of diverse and immortalized murine MM tumors.
PMCID:11076575
PMID: 38714690
ISSN: 2041-1723
CID: 5658362
Round Table Discussion on Optimal Clinical Trial Design in Precursor Multiple Myeloma
Ghobrial, Irene M; Gormley, Nicole; Kumar, Shaji K; Mateos, Maria-Victoria; Bergsagel, P Leif; Chesi, Marta; Dhodapkar, Madhav V; Dispenzieri, Angela; Fonseca, Rafael; Getz, Gad; Kastritis, Efstathios; Kristinsson, Sigurdur Y; Martinez-Climent, Jose Angel; Manier, Salomon; Marinac, Catherine R; Maura, Francesco; Morgan, Gareth J; Davies, Faith E; Nadeem, Omar; Nuvolone, Mario; Paiva, Bruno; O'Donnell, Elizabeth; Prosper, Felipe; Shah, Urvi A; Sklavenitis-Pistofidis, Romanos; Sperling, Adam S; Vassiliou, George S; Munshi, Nikhil C; Castle, Philip E; Anderson, Kenneth C; San Miguel, Jesus F
While the current approach to precursor hematologic conditions is to "watch and wait," this may change with the development of therapies that are safe and extend survival or delay the onset of symptomatic disease. The goal of future therapies in precursor hematologic conditions is to improve survival and prevent or delay the development of symptomatic disease while maximizing safety. Clinical trial considerations in this field include identifying an appropriate at-risk population, safety assessments, dose selection, primary and secondary trial endpoints including surrogate endpoints, control arms, and quality-of-life metrics, all of which may enable more precise benefit-risk assessment.
PMCID:11061588
PMID: 38441243
ISSN: 2643-3249
CID: 5657122
Genomic Classification and Individualized Prognosis in Multiple Myeloma
Maura, Francesco; Rajanna, Arjun Raj; Ziccheddu, Bachisio; Poos, Alexandra M; Derkach, Andriy; Maclachlan, Kylee; Durante, Michael; Diamond, Benjamin; Papadimitriou, Marios; Davies, Faith; Boyle, Eileen M; Walker, Brian; Hultcrantz, Malin; Silva, Ariosto; Hampton, Oliver; Teer, Jamie K; Siegel, Erin M; Bolli, Niccolò; Jackson, Graham H; Kaiser, Martin; Pawlyn, Charlotte; Cook, Gordon; Kazandjian, Dickran; Stein, Caleb; Chesi, Marta; Bergsagel, Leif; Mai, Elias K; Goldschmidt, Hartmut; Weisel, Katja C; Fenk, Roland; Raab, Marc S; Van Rhee, Fritz; Usmani, Saad; Shain, Kenneth H; Weinhold, Niels; Morgan, Gareth; Landgren, Ola
PURPOSE/OBJECTIVE:Outcomes for patients with newly diagnosed multiple myeloma (NDMM) are heterogenous, with overall survival (OS) ranging from months to over 10 years. METHODS:To decipher and predict the molecular and clinical heterogeneity of NDMM, we assembled a series of 1,933 patients with available clinical, genomic, and therapeutic data. RESULTS:translocations, APOBEC mutational signatures, and copy-number signatures (reflecting the complex structural variant chromothripsis). IRMMa accuracy and superiority compared with other prognostic models were validated on 256 patients enrolled in the GMMG-HD6 (ClinicalTrials.gov identifier: NCT02495922) clinical trial. Individualized patient risks were significantly affected across the 12 genomic groups by different treatment strategies (ie, treatment variance), which was used to identify patients for whom HDM-ASCT is particularly effective versus patients for whom the impact is limited. CONCLUSION/CONCLUSIONS:Integrating clinical, demographic, genomic, and therapeutic data, to our knowledge, we have developed the first individualized risk-prediction model enabling personally tailored therapeutic decisions for patients with NDMM.
PMID: 38194610
ISSN: 1527-7755
CID: 5655472
Approach to High-Risk Multiple Myeloma
Chen, Xiaoyi; Varma, Gaurav; Davies, Faith; Morgan, Gareth
Improving the outcome of high-risk myeloma (HRMM) is a key therapeutic aim for the next decade. To achieve this aim, it is necessary to understand in detail the genetic drivers underlying this clinical behavior and to target its biology therapeutically. Advances have already been made, with a focus on consensus guidance and the application of novel immunotherapeutic approaches. Cases of HRMM are likely to have impaired prognosis even with novel strategies. However, if disease eradication and minimal disease states are achieved, then cure may be possible.
PMID: 38195306
ISSN: 1558-1977
CID: 5628622