159 results on '"Savona MR"'
Search Results
2. Special considerations in the management of adult patients with acute leukaemias and myeloid neoplasms in the COVID-19 era: recommendations from a panel of international experts
- Author
-
Zeidan, AM, Boddu, PC, Patnaik, MM, Bewersdorf, JP, Stahl, M, Rampal, RK, Shallis, RM, Steensma, DP, Savona, MR, Sekeres, MA, Roboz, GJ, DeAngelo, DJ, Schuh, AC, Padron, E, Zeidner, JF, Walter, RB, Onida, F, Fathi, AT, DeZern, A, Hobbs, G, Stein, EM, Vyas, P, Wei, AH, Bowen, DT, Montesinos, P, Griffiths, EA, Verma, AK, Keyzner, A, Bar-Natan, M, Navada, SC, Kremyanskaya, M, Goldberg, AD, Al-Kali, A, Heaney, ML, Nazha, A, Salman, H, Luger, S, Pratz, KW, Konig, H, Komrokji, R, Deininger, M, Cirici, BX, Bhatt, VR, Silverman, LR, Erba, HP, Fenaux, P, Platzbecker, U, Santini, V, Wang, ES, Tallman, MS, Stone, RM, and Mascarenhas, J
- Abstract
The ongoing COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 is a global public health crisis. Multiple observations indicate poorer post-infection outcomes for patients with cancer than for the general population. Herein, we highlight the challenges in caring for patients with acute leukaemias and myeloid neoplasms amid the COVID-19 pandemic. We summarise key changes related to service allocation, clinical and supportive care, clinical trial participation, and ethical considerations regarding the use of lifesaving measures for these patients. We recognise that these recommendations might be more applicable to high-income countries and might not be generalisable because of regional differences in health-care infrastructure, individual circumstances, and a complex and highly fluid health-care environment. Despite these limitations, we aim to provide a general framework for the care of patients with acute leukaemias and myeloid neoplasms during the COVID-19 pandemic on the basis of recommendations from international experts.
- Published
- 2020
3. Implications ofTP53allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes
- Author
-
Bernard, E, Nannya, Y, Hasserjian, RP, Devlin, SM, Tuechler, H, Medina-Martinez, JS, Yoshizato, T, Shiozawa, Y, Saiki, R, Malcovati, L, Levine, MF, Arango, JE, Zhou, YY, Sole, F, Cargo, CA, Haase, D, Creignou, M, Germing, U, Zhang, YM, Gundem, G, Sarian, A, van de Loosdrecht, AA, Jadersten, M, Tobiasson, M, Kosmider, O, Follo, MY, Thol, F, Pinheiro, RF, Santini, V, Kotsianidis, I, Boultwood, J, Santos, FPS, Schanz, J, Kasahara, S, Ishikawa, T, Tsurumi, H, Takaori-Kondo, A, Kiguchi, T, Polprasert, C, Bennett, JM, Klimek, VM, Savona, MR, Belickova, M, Ganster, C, Palomo, L, SANZ, G, Ades, L, Della Porta, MG, Smith, AG, Werner, Y, Patel, M, Viale, A, Vanness, K, Neuberg, DS, Stevenson, KE, Menghrajani, K, Bolton, KL, Fenaux, P, Pellagatti, A, Platzbecker, U, Heuser, M, Valent, P, Chiba, S, Miyazaki, Y, Finelli, C, Voso, MT, Shih, LY, Fontenay, M, Jansen, JH, Cervera, J, Atsuta, Y, Gattermann, N, Ebert, BL, Bejar, R, Greenberg, PL, Cazzola, M, Hellstrom-Lindberg, E, Ogawa, S, and Papaemmanuil, E
- Abstract
Clinical sequencing across a large prospective cohort of patients with myelodysplasic syndrome uncovers distinct associations between the mono- and biallelic states ofTP53and clinical presentation Tumor protein p53 (TP53) is the most frequently mutated gene in cancer(1,2). In patients with myelodysplastic syndromes (MDS),TP53mutations are associated with high-risk disease(3,4), rapid transformation to acute myeloid leukemia (AML)(5), resistance to conventional therapies(6-8)and dismal outcomes(9). Consistent with the tumor-suppressive role ofTP53, patients harbor both mono- and biallelic mutations(10). However, the biological and clinical implications ofTP53allelic state have not been fully investigated in MDS or any other cancer type. We analyzed 3,324 patients with MDS forTP53mutations and allelic imbalances and delineated two subsets of patients with distinct phenotypes and outcomes. One-third ofTP53-mutated patients had monoallelic mutations whereas two-thirds had multiple hits (multi-hit) consistent with biallelic targeting. Established associations with complex karyotype, few co-occurring mutations, high-risk presentation and poor outcomes were specific to multi-hit patients only.TP53multi-hit state predicted risk of death and leukemic transformation independently of the Revised International Prognostic Scoring System (IPSS-R)(11). Surprisingly, monoallelic patients did not differ fromTP53wild-type patients in outcomes and response to therapy. This study shows that consideration ofTP53allelic state is critical for diagnostic and prognostic precision in MDS as well as in future correlative studies of treatment response.
- Published
- 2020
4. TP53 mutation status divides myelodysplastic syndromes with complex karyotypes into distinct prognostic subgroups
- Author
-
Haase, D, Stevenson, KE, Neuberg, D, Maciejewski, JP, Nazha, A, Sekeres, MA, Ebert, BL, Garcia-Manero, G, Haferlach, C, Haferlach, T, Kern, W, Ogawa, S, Nagata, Y, Yoshida, K, Graubert, TA, Walter, MJ, List, AF, Komrokji, RS, Padron, E, Sallman, D, Papaemmanuil, E, Campbell, PJ, Savona, MR, Seegmiller, A, Adès, L, Fenaux, P, Shih, L-Y, Bowen, D, Groves, MJ, Tauro, S, Fontenay, M, Kosmider, O, Bar-Natan, M, Steensma, D, Stone, R, Heuser, M, Thol, F, Cazzola, M, Malcovati, L, Karsan, A, Ganster, C, Hellström-Lindberg, E, Boultwood, J, Pellagatti, A, Santini, V, Quek, L, Vyas, P, Tüchler, H, Greenberg, PL, Bejar, R, and Committee, International Working Group For Mds Molecular Prognostic
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Tp53 mutation ,medicine.disease_cause ,0302 clinical medicine ,hemic and lymphatic diseases ,80 and over ,Chromosome 7 (human) ,Aged, 80 and over ,Mutation ,Tumor ,Karyotype ,Hematology ,Middle Aged ,Prognosis ,Combined Modality Therapy ,myelodysplastic syndromes ,3. Good health ,Survival Rate ,030220 oncology & carcinogenesis ,Female ,medicine.medical_specialty ,Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,and over ,Article ,03 medical and health sciences ,Rare Diseases ,Internal medicine ,Complex Karyotype ,Genetics ,medicine ,Biomarkers, Tumor ,Humans ,Survival rate ,Gene ,Aged ,TP53 mutation , myelodysplastic syndromes ,Chromosome Aberrations ,International Working Group for MDS Molecular Prognostic Committee ,business.industry ,Myelodysplastic syndromes ,medicine.disease ,030104 developmental biology ,Karyotyping ,Myelodysplastic Syndromes ,Tumor Suppressor Protein p53 ,business ,Biomarkers ,Follow-Up Studies - Abstract
Risk stratification is critical in the care of patients with myelodysplastic syndromes (MDS). Approximately 10% have a complex karyotype (CK), defined as more than two cytogenetic abnormalities, which is a highly adverse prognostic marker. However, CK-MDS can carry a wide range of chromosomal abnormalities and somatic mutations. To refine risk stratification of CK-MDS patients, we examined data from 359 CK-MDS patients shared by the International Working Group for MDS. Mutations were underrepresented with the exception of TP53 mutations, identified in 55% of patients. TP53 mutated patients had even fewer co-mutated genes but were enriched for the del(5q) chromosomal abnormality (p 10%), abnormal 3q, abnormal 9, and monosomy 7 as having the greatest survival risk. The poor risk associated with CK-MDS is driven by its association with prognostically adverse TP53 mutations and can be refined by considering clinical and karyotype features.
- Published
- 2019
5. Therapeutic approaches in myelofibrosis and myelodysplastic/myeloproliferative overlap syndromes
- Author
-
Sochacki AL, Fischer MA, and Savona MR
- Subjects
MDS/MPN neoplasms ,hemic and lymphatic diseases ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Emerging therapy ,lcsh:RC254-282 - Abstract
Andrew L Sochacki,1 Melissa A Fischer,1 Michael R Savona1,2 1Department of Internal Medicine, Vanderbilt University Medical Center, 2Vanderbilt-Ingram Cancer Center, Nashville, TN, USA Abstract: The discovery of JAK2V617F a decade ago led to optimism for a rapidly developing treatment revolution in Ph- myeloproliferative neoplasms. Unlike BCR–ABL, however, JAK2 was found to have a more heterogeneous role in carcinogenesis. Therefore, for years, development of new therapies was slow, despite standard treatment options that did not address the overwhelming symptom burden in patients with primary myelofibrosis (MF), post-essential thrombocythemia MF, post-polycythemia vera MF, and myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN) syndromes. JAK–STAT inhibitors have changed this, drastically ameliorating symptoms and ultimately beginning to show evidence of impact on survival. Now, the genetic foundations of myelofibrosis and MDS/MPN are rapidly being elucidated and contributing to targeted therapy development. This has been empowered through updated response criteria for MDS/MPN and refined prognostic scoring systems in these diseases. The aim of this article is to summarize concisely the current and rationally designed investigational therapeutics directed at JAK–STAT, hedgehog, PI3K–Akt, bone marrow fibrosis, telomerase, and rogue epigenetic signaling. The revolution in immunotherapy and novel treatments aimed at previously untargeted signaling pathways provides hope for considerable advancement in therapy options for those with chronic myeloid disease. Keywords: MDS/MPN neoplasms, emerging therapy
- Published
- 2016
6. An international consortium proposal of uniform response criteria for myelodysplastic/myeloproliferative neoplasms (MDS/MPN) in adults
- Author
-
Savona MR, Malcovati L, Komrokji R, Tiu RV, Mughal TI, Orazi A, Kiladjian JJ, Padron E, Solary E, Tibes R, Itzykson R, Cazzola M, Mesa R, Maciejewski J, Fenaux P, Garcia-Manero G, Gerds A, Sanz G, Niemeyer CM, Cervantes F, Germing U, Cross NC, List AF, and MDS/MPN International Working Group
- Subjects
hemic and lymphatic diseases ,food and beverages - Abstract
Myelodysplastic syndromes (MDS) and myeloproliferative neoplasms (MPN) are hematologically diverse stem cell malignancies sharing phenotypic features of both myelodysplastic syndromes and myeloproliferative neoplasms. There are currently no standard treatment recommendations for most adult patients with MDS/MPN. To optimize efforts to improve the management and disease outcomes, it is essential to identify meaningful clinical and biologic end points and standardized response criteria for clinical trials. The dual dysplastic and proliferative features in these stem cell malignancies define their uniqueness and challenges. We propose response assessment guidelines to harmonize future clinical trials with the principal objective of establishing suitable treatment algorithms. An international panel comprising laboratory and clinical experts in MDS/MPN was established involving 3 independent academic MDS/MPN workshops (March 2013, December 2013, and June 2014). These recommendations are the result of this collaborative project sponsored by the MDS Foundation.
- Published
- 2015
7. Erythropoietin-stimulating agents in oncology.
- Author
-
Savona MR and Silver SM
- Abstract
Erythropoiesis stimulating agents (ESAs) are some of the most widely used agents in oncology. Yet, the use of ESAs to treat chemotherapy induced anemia in cancer patients has raised issues of concern since 2003, when the two initial studies that were powered to detect meaningful differences in survival with ESA therapy were either halted early, or concluded with deleterious effects on survival. Several trials since then have caused both guideline writers and the FDA to recommend that ESAs should not employed to drive the hemoglobin to greater than 12 g/dL. It is still unclear what role these agents will play in cancer patients with hemoglobin of less than 12 g/dL. In myelodysplastic syndrome, however, these agents are likely part of a core of supportive care measures for low-grade disease, and need to be readily available for these patients. In this review, we attempt to describe the relevant experimental data on ESAs, their current role in clinical oncology, and the research and discoveries that may widen the scope, and enhance the benefit of these agents. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
8. Methylation sequencing enhances interpretation of clonal hematopoiesis dynamics.
- Author
-
Parker AC, Van Amburg J, Heimlich JB, Pershad Y, Mickels NA, Mack T, Ferrell PB Jr, Savona MR, Jones A, and Bick AG
- Abstract
We have developed a cost-effective DNA methylation sequencing assay to improve monitoring of clonal hematopoiesis. By inferring cell-type proportions, this method enhances interpretation of clonal trajectories compared to interpretation based on variant allele fraction only., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
9. Germline genetics, disease, and exposure to medication influence longitudinal dynamics of clonal hematopoiesis.
- Author
-
Mack T, Pershad Y, Vlasschaert C, Bejan CA, Heimlich JB, Li Y, Mickels NA, Van Amburg JC, Ulloa J, Silver AJ, Luo LY, Jones A, Ferrell PB, Kishtagari A, Xu Y, Savona MR, and Bick AG
- Abstract
Not available.
- Published
- 2024
- Full Text
- View/download PDF
10. Sustained benefits of imetelstat on patient-reported fatigue in patients with lower-risk myelodysplastic syndromes ineligible for, or relapsed/refractory to, erythropoiesis-stimulating agents and high transfusion burden in the phase 3 IMerge study.
- Author
-
Sekeres MA, Santini V, Díez-Campelo M, Komrokji RS, Fenaux P, Savona MR, Madanat YF, Valcárcel-Ferreiras D, Oliva EN, Regnault A, Creel K, Sengupta N, Dougherty S, Shah S, Sun L, Wan Y, Navada S, Zeidan AM, and Platzbecker U
- Published
- 2024
- Full Text
- View/download PDF
11. Risk prediction for clonal cytopenia: multicenter real-world evidence.
- Author
-
Xie Z, Komrokji R, Al Ali N, Regelson A, Geyer S, Patel A, Saygin C, Zeidan AM, Bewersdorf JP, Mendez L, Kishtagari A, Zeidner JF, Coombs CC, Madanat YF, Chung S, Badar T, Foran J, Desai P, Tsai C, Griffiths EA, Al Malki MM, Amanam I, Lai C, Deeg HJ, Ades L, Arana Yi C, Osman AEG, Dinner S, Abaza Y, Taylor J, Chandhok N, Soong D, Brunner AM, Carraway HE, Singh A, Elena C, Ferrari J, Gallì A, Pozzi S, Padron E, Patnaik MM, Malcovati L, Savona MR, and Al-Kali A
- Subjects
- Humans, Female, Male, Aged, Middle Aged, Adult, Aged, 80 and over, Prognosis, Risk Factors, Risk Assessment methods, Clonal Hematopoiesis, Cytopenia, Mutation
- Abstract
Abstract: Clonal cytopenia of undetermined significance (CCUS) represents a distinct disease entity characterized by myeloid-related somatic mutations with a variant allele fraction of ≥2% in individuals with unexplained cytopenia(s) but without a myeloid neoplasm (MN). Notably, CCUS carries a risk of progressing to MN, particularly in cases featuring high-risk mutations. Understanding CCUS requires dedicated studies to elucidate its risk factors and natural history. Our analysis of 357 patients with CCUS investigated the interplay between clonality, cytopenia, and prognosis. Multivariate analysis identified 3 key adverse prognostic factors: the presence of splicing mutation(s) (score = 2 points), platelet count of <100 × 109/L (score = 2.5), and ≥2 mutations (score = 3). Variable scores were based on the coefficients from the Cox proportional hazards model. This led to the development of the clonal cytopenia risk score (CCRS), which stratified patients into low- (score of <2.5 points), intermediate- (score of 2.5 to <5), and high-risk (score of ≥5) groups. The CCRS effectively predicted 2-year cumulative incidence of MN for low- (6.4%), intermediate- (14.1%), and high-risk (37.2%) groups, respectively, by the Gray test (P < .0001). We further validated the CCRS by applying it to an independent CCUS cohort of 104 patients, demonstrating a c-index of 0.64 (P = .005) in stratifying the cumulative incidence of MN. Our study underscores the importance of integrating clinical and molecular data to assess the risk of CCUS progression, making the CCRS a valuable tool that is practical and easily calculable. These findings are clinically relevant, shaping the management strategies for CCUS and informing future clinical trial designs., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Data-driven, harmonised classification system for myelodysplastic syndromes: a consensus paper from the International Consortium for Myelodysplastic Syndromes.
- Author
-
Komrokji RS, Lanino L, Ball S, Bewersdorf JP, Marchetti M, Maggioni G, Travaglino E, Al Ali NH, Fenaux P, Platzbecker U, Santini V, Diez-Campelo M, Singh A, Jain AG, Aguirre LE, Tinsley-Vance SM, Schwabkey ZI, Chan O, Xie Z, Brunner AM, Kuykendall AT, Bennett JM, Buckstein R, Bejar R, Carraway HE, DeZern AE, Griffiths EA, Halene S, Hasserjian RP, Lancet J, List AF, Loghavi S, Odenike O, Padron E, Patnaik MM, Roboz GJ, Stahl M, Sekeres MA, Steensma DP, Savona MR, Taylor J, Xu ML, Sweet K, Sallman DA, Nimer SD, Hourigan CS, Wei AH, Sauta E, D'Amico S, Asti G, Castellani G, Delleani M, Campagna A, Borate UM, Sanz G, Efficace F, Gore SD, Kim TK, Daver N, Garcia-Manero G, Rozman M, Orfao A, Wang SA, Foucar MK, Germing U, Haferlach T, Scheinberg P, Miyazaki Y, Iastrebner M, Kulasekararaj A, Cluzeau T, Kordasti S, van de Loosdrecht AA, Ades L, Zeidan AM, and Della Porta MG
- Subjects
- Humans, Consensus, Myelodysplastic Syndromes classification, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes genetics
- Abstract
The WHO and International Consensus Classification 2022 classifications of myelodysplastic syndromes enhance diagnostic precision and refine decision-making processes in these diseases. However, some discrepancies still exist and potentially cause inconsistency in their adoption in a clinical setting. We adopted a data-driven approach to provide a harmonisation between these two classification systems. We investigated the importance of genomic features and their effect on the cluster assignment process to define harmonised entity labels. A panel of expert haematologists, haematopathologists, and data scientists who are members of the International Consortium for Myelodysplastic Syndromes was formed and a modified Delphi consensus process was adopted to harmonise morphologically defined categories without a distinct genomic profile. The panel held regular online meetings and participated in a two-round survey using an online voting tool. We identified nine clusters with distinct genomic features. The cluster of highest hierarchical importance was characterised by biallelic TP53 inactivation. Cluster assignment was irrespective of blast count. Individuals with monoallelic TP53 inactivation were assigned to other clusters. Hierarchically, the second most important group included myelodysplastic syndromes with del(5q). Isolated del(5q) and less than 5% of blast cells in the bone marrow were the most relevant label-defining features. The third most important cluster included myelodysplastic syndromes with mutated SF3B1. The absence of isolated del(5q), del(7q)/-7, abn3q26.2, complex karyotype, RUNX1 mutations, or biallelic TP53 were the basis for a harmonised label of this category. Morphologically defined myelodysplastic syndrome entities showed large genomic heterogeneity that was not efficiently captured by single-lineage versus multilineage dysplasia, marrow blasts, hypocellularity, or fibrosis. We investigated the biological continuum between myelodysplastic syndromes with more than 10% bone marrow blasts and acute myeloid leukaemia, and found only a partial overlap in genetic features. After the survey, myelodysplastic syndromes with low blasts (ie, less than 5%) and myelodysplastic syndromes with increased blasts (ie, 5% or more) were recognised as disease entities. Our data-driven approach can efficiently harmonise current classifications of myelodysplastic syndromes and provide a reference for patient management in a real-world setting., Competing Interests: Declaration of interests UG reports speaker honoraria from Novartis, AbbVie, and BMS; and institutional research support from BMS, AbbVie, and Jazz Pharmaceuticals. FE reports consultancy or advisory roles for AbbVie, Incyte, Syros, Novartis, and Jazz Pharmaceuticals. SH reports research support from STORM Therapeutics and AstraZeneca. EAG reports honoraria from AAMDS, MedscapeLIVE!, MediCom Worldwide, MJH Life Sciences, ASH, MDS International Foundation, and Physicians’ Education Resource; consulting fees from AbbVie, Alexion, Apellis, Takeda Oncology, Astex/Taiho Oncology, Alexion/AstraZeneca Rare Disease, Celgene/BMS, CTI BioPharma, Novartis, Partner Therapeutics, Picnic Health, and Servier; and research funding from Alexion, Apellis, Astex /Otsuka/Taiho Oncology, Blueprint Medicines, Celldex Therapeutics, Genentech, and NextCure. MAS reports participation on advisory boards for BMS, Kurome, Schrödinger, and Karyopharm. MD-C reports participation on a data safety monitoring board or advisory board for BMS, Novartis, Blueprint Medicines, GSK, Agios, Hemavan, Syros, Keros, Curis, and Astex/Otsuka; and payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing, or educational events for BMS, Novartis, and Keros. UP reports research support and honoraria from BMS, Geron, Curis, AbbVie, and Janssen. RBe reports employment or equity from Aptose Biosciences; participation on advisory boards for BMS, Servier, NeoGenomics, and Geron; being Data Monitoring Committee Chair for Gilead, Ipsen, and Keros; and consultancy for TenSixteen. YM reports honoraria from Nippon Shinyaku, BMS, Novartis, Sumitomo Pharma, Kyowa Kirin, AbbVie, Daiichi Sankyo, Takeda, Janssen, Astellas, Pfizer, Eisai, and Otsuka; and research funding from Chugai. AED reports participation on advisory boards, consultancy, or honoraria from Celgene/BMS, Agios, Novartis, Astellas, and Gilead; and participation on clinical trial committees or data safety monitoring boards for Novartis, AbbVie, Kura, Geron, Servier, Keros, and Celgene/BMS. DPS reports employment by Ajax Therapeutics; former employment by Novartis; and minor equity in Arrowhead and Bluebird. TKK reports consultancy for Agenus and ImmunoBiome. AK reports research support from Novartis and BMS; consulting fees from Alexion, Novartis, Amgen, Agios, Pfizer, Samsung, Celgene, F Hoffmann-La Roche, and Sobi; honoraria from Alexion, Novartis, Pfizer, Amgen, Samsung, Celgene, F Hoffmann-La Roche, BMS, Sobi, and Silence Therapeutics; and speakers fees from Alexion, Novartis, Amgen, Pfizer, Celgene, F Hoffmann-La Roche, and Sobi. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
13. PheMIME: an interactive web app and knowledge base for phenome-wide, multi-institutional multimorbidity analysis.
- Author
-
Zhang S, Strayer N, Vessels T, Choi K, Wang GW, Li Y, Bejan CA, Hsi RS, Bick AG, Velez Edwards DR, Savona MR, Phillips EJ, Pulley JM, Self WH, Hopkins WC, Roden DM, Smoller JW, Ruderfer DM, and Xu Y
- Subjects
- Humans, Knowledge Bases, Software, Schizophrenia, Phenomics, User-Computer Interface, Internet, Phenotype, Multimorbidity, Electronic Health Records
- Abstract
Objectives: To address the need for interactive visualization tools and databases in characterizing multimorbidity patterns across different populations, we developed the Phenome-wide Multi-Institutional Multimorbidity Explorer (PheMIME). This tool leverages three large-scale EHR systems to facilitate efficient analysis and visualization of disease multimorbidity, aiming to reveal both robust and novel disease associations that are consistent across different systems and to provide insight for enhancing personalized healthcare strategies., Materials and Methods: PheMIME integrates summary statistics from phenome-wide analyses of disease multimorbidities, utilizing data from Vanderbilt University Medical Center, Mass General Brigham, and the UK Biobank. It offers interactive and multifaceted visualizations for exploring multimorbidity. Incorporating an enhanced version of associationSubgraphs, PheMIME also enables dynamic analysis and inference of disease clusters, promoting the discovery of complex multimorbidity patterns. A case study on schizophrenia demonstrates its capability for generating interactive visualizations of multimorbidity networks within and across multiple systems. Additionally, PheMIME supports diverse multimorbidity-based discoveries, detailed further in online case studies., Results: The PheMIME is accessible at https://prod.tbilab.org/PheMIME/. A comprehensive tutorial and multiple case studies for demonstration are available at https://prod.tbilab.org/PheMIME_supplementary_materials/. The source code can be downloaded from https://github.com/tbilab/PheMIME., Discussion: PheMIME represents a significant advancement in medical informatics, offering an efficient solution for accessing, analyzing, and interpreting the complex and noisy real-world patient data in electronic health records., Conclusion: PheMIME provides an extensive multimorbidity knowledge base that consolidates data from three EHR systems, and it is a novel interactive tool designed to analyze and visualize multimorbidities across multiple EHR datasets. It stands out as the first of its kind to offer extensive multimorbidity knowledge integration with substantial support for efficient online analysis and interactive visualization., (© The Author(s) 2024. Published by Oxford University Press on behalf of the American Medical Informatics Association.)
- Published
- 2024
- Full Text
- View/download PDF
14. Pathogenesis and inflammaging in myelodysplastic syndrome.
- Author
-
Villaume MT and Savona MR
- Abstract
Myelodysplastic syndromes (MDS) are a genetically complex and phenotypically diverse set of clonal hematologic neoplasms that occur with increasing frequency with age. MDS has long been associated with systemic inflammatory conditions and disordered inflammatory signaling is implicated in MDS pathogenesis. A rise in sterile inflammation occurs with ageing and the term "inflammaging" has been coined by to describe this phenomenon. This distinct form of sterile inflammation has an unknown role in in the pathogenesis of myeloid malignancies despite shared correlations with age and ageing-related diseases. More recent is a discovery that many cases of MDS arise from clonal hematopoiesis of indeterminate potential (CHIP), an age associated, asymptomatic pre-disease state. The interrelationship between ageing, inflammation and clonal CHIP is complex and likely bidirectional with causality between inflammaging and CHIP potentially instrumental to understanding MDS pathogenesis. Here we review the concept of inflammaging and MDS pathogenesis and explore their causal relationship by introducing a novel framing mechanism of "pre-clonal inflammaging" and "clonal inflammaging". We aim to harmonize research on ageing, inflammation and MDS pathogenesis by contextualizing the current understanding of inflammaging and the ageing hematopoietic system with what is known about the etiology of MDS via its progression from CHIP.
- Published
- 2024
- Full Text
- View/download PDF
15. Molecular taxonomy of myelodysplastic syndromes and its clinical implications.
- Author
-
Bernard E, Hasserjian RP, Greenberg PL, Arango Ossa JE, Creignou M, Tuechler H, Gutierrez-Abril J, Domenico D, Medina-Martinez JS, Levine M, Liosis K, Farnoud N, Sirenko M, Jädersten M, Germing U, Sanz G, van de Loosdrecht AA, Nannya Y, Kosmider O, Follo MY, Thol F, Zamora L, Pinheiro RF, Pellagatti A, Elias HK, Haase D, Ganster C, Ades L, Tobiasson M, Palomo L, Della Porta MG, Fenaux P, Belickova M, Savona MR, Klimek VM, Santos FPS, Boultwood J, Kotsianidis I, Santini V, Solé F, Platzbecker U, Heuser M, Valent P, Finelli C, Voso MT, Shih LY, Fontenay M, Jansen JH, Cervera J, Gattermann N, Ebert BL, Bejar R, Malcovati L, Ogawa S, Cazzola M, Hellström-Lindberg E, and Papaemmanuil E
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Aged, 80 and over, Mutation, Adult, Prognosis, Loss of Heterozygosity, DNA Copy Number Variations, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes classification, Myelodysplastic Syndromes pathology
- Abstract
Abstract: Myelodysplastic syndromes (MDS) are clonal hematologic disorders characterized by morphologic abnormalities of myeloid cells and peripheral cytopenias. Although genetic abnormalities underlie the pathogenesis of these disorders and their heterogeneity, current classifications of MDS rely predominantly on morphology. We performed genomic profiling of 3233 patients with MDS or related disorders to delineate molecular subtypes and define their clinical implications. Gene mutations, copy-number alterations, and copy-neutral loss of heterozygosity were derived from targeted sequencing of a 152-gene panel, with abnormalities identified in 91%, 43%, and 11% of patients, respectively. We characterized 16 molecular groups, encompassing 86% of patients, using information from 21 genes, 6 cytogenetic events, and loss of heterozygosity at the TP53 and TET2 loci. Two residual groups defined by negative findings (molecularly not otherwise specified, absence of recurrent drivers) comprised 14% of patients. The groups varied in size from 0.5% to 14% of patients and were associated with distinct clinical phenotypes and outcomes. The median bone marrow (BM) blast percentage across groups ranged from 1.5% to 10%, and the median overall survival ranged from 0.9 to 8.2 years. We validated 5 well-characterized entities, added further evidence to support 3 previously reported subsets, and described 8 novel groups. The prognostic influence of BM blasts depended on the genetic subtypes. Within genetic subgroups, therapy-related MDS and myelodysplastic/myeloproliferative neoplasms had comparable clinical and outcome profiles to primary MDS. In conclusion, genetically-derived subgroups of MDS are clinically relevant and might inform future classification schemas and translational therapeutic research., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Ziftomenib in relapsed or refractory acute myeloid leukaemia (KOMET-001): a multicentre, open-label, multi-cohort, phase 1 trial.
- Author
-
Wang ES, Issa GC, Erba HP, Altman JK, Montesinos P, DeBotton S, Walter RB, Pettit K, Savona MR, Shah MV, Kremyanskaya M, Baer MR, Foran JM, Schiller G, Adès L, Heiblig M, Berthon C, Peterlin P, Rodríguez-Arbolí E, Salamero O, Patnaik MM, Papayannidis C, Grembecka J, Cierpicki T, Clegg B, Ray J, Linhares BM, Nie K, Mitra A, Ahsan JM, Tabachri M, Soifer HS, Corum D, Leoni M, Dale S, and Fathi AT
- Subjects
- Humans, Middle Aged, Male, Female, Aged, Adult, Neoplasm Recurrence, Local drug therapy, Maximum Tolerated Dose, Drug Resistance, Neoplasm, Dose-Response Relationship, Drug, Aged, 80 and over, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Nucleophosmin
- Abstract
Background: Ziftomenib (KO-539) is an oral selective menin inhibitor with known preclinical activity in menin-dependent acute myeloid leukaemia models. The primary objective of this study was to determine the recommended phase 2 dose in patients with relapsed or refractory acute myeloid leukaemia based on safety, pharmacokinetics, pharmacodynamics, and preliminary activity., Methods: KOMET-001 is a multicentre, open-label, multi-cohort, phase 1/2 clinical trial of ziftomenib in adults with relapsed or refractory acute myeloid leukaemia. Results of the phase 1 study, conducted at 22 hospitals in France, Italy, Spain, and the USA, are presented here and comprise the dose-escalation (phase 1a) and dose-validation and expansion (phase 1b) phases. Eligible patients were aged 18 years or older, had relapsed or refractory acute myeloid leukaemia, and had an Eastern Cooperative Oncology Group performance status of 2 or less. For phase 1a, patients (all molecular subtypes) received ziftomenib (50-1000 mg) orally once daily in 28-day cycles. For phase 1b, patients with NPM1 mutations or with KMT2A rearrangements were randomly assigned (1:1) using third-party interactive response technology to two parallel dose cohorts (200 mg and 600 mg ziftomenib). Primary endpoints were maximum tolerated dose or recommended phase 2 dose in phase 1a, and safety, remission rates, and pharmacokinetics supporting recommended phase 2 dose determination in phase 1b. Analyses were performed in all patients who received at least one dose of ziftomenib (modified intention-to-treat population). Phase 1a/1b is complete. This trial is registered with ClinicalTrials.gov, NCT04067336, and the EU Clinical Trials register, EudraCT 2019-001545-41., Findings: From Sept 12, 2019, to Aug 19, 2022, 83 patients received 50-1000 mg ziftomenib (39 [47%] were male and 44 [53%] were female). Median follow-up was 22·3 months (IQR 15·4-30·2). Of 83 patients, the most common grade 3 or worse treatment-emergent adverse events were anaemia (20 [24%]), febrile neutropenia (18 [22%]), pneumonia (16 [19%]), differentiation syndrome (12 [15%]), thrombocytopenia (11 [13%]), and sepsis (ten [12%]). Overall, 68 of 83 patients had serious adverse events, with two reported treatment-related deaths (one differentiation syndrome and one cardiac arrest). Differentiation syndrome rate and severity influenced the decision to halt enrolment of patients with KMT2A rearrangements. In Phase 1b, no responses were reported in patients treated at the 200 mg dose level. At the recommended phase 2 dose of 600 mg, nine (25%) of 36 patients with KMT2A rearrangement or NPM1 mutation had complete remission or complete remission with partial haematologic recovery. Seven (35%) of 20 patients with NPM1 mutation treated at the recommended phase 2 dose had a complete remission., Interpretation: Ziftomenib showed promising clinical activity with manageable toxicity in heavily pretreated patients with relapsed or refractory acute myeloid leukaemia. Phase 2 assessment of ziftomenib combination therapy in the upfront and relapsed or refractory setting is ongoing., Funding: Kura Oncology., Competing Interests: Declaration of interests ESW declares honoraria for educational talks from Aptitude, Astellas, Bioascend, CEA, CCO, Curio Sciences, Dava Oncology, Medscape, MD Education, OncLive, PER, Peerview, RTP, and Pfizer; participation on Data Safety Monitoring Committees and research grant committees for AbbVie and Gilead; advisory board participation for Abbvie, Astellas, Blueprint, Bristol Myers Squibb, Daiichi Sankyo, Gilead, GlaxoSmithKline, Immunogen, Janssen, Jazz, Kite, Novartis, NuProbe, PharmaEssentia, Pfizer, Qiagen, Rigel, Schrodinger, Sumitomo, Syndax, and Takeda; other financial and non-financial interests with UptoDate; and medical writing support for this work from Kura Oncology. GCI declares research funding to their institution from Astex, Cullinan Oncology, Kura Oncology, Merck, Novartis, and Syndax Pharmaceuticals; consultancy fees from AbbVie, Kura Oncology, Novartis, and Syndax Pharmaceuticals; support for attending meetings and travel from Kura Oncology; steering committee role with Kura Oncology and Novartis; receipt of material for sample analysis from NuProbe; and medical writing support for this work from Kura Oncology. HPE declares a leadership role with AbbVie (Chair, Independent Review Committee for VIALE A and VIALE C), Bristol Myers Squibb (Chair, AML Registry Steering Committee), and Glycomimetics (Scientific Steering Committee); speakers bureau for AbbVie, Bristol Myers Squibb, Incyte, Jazz, Novartis, and Servier; contracted research from AbbVie, ALX Oncology, Amgen, Aptose, Ascentage, Daiichi Sankyo, Forma, Gilead, Glycomimetics, Immunogen, Jazz, Kura Oncology, MacroGenics, Novartis, PTC, and Sumitomo Pharma; consultancy fees from AbbVie, Astellas, Bristol Myers Squibb, Daiichi Sankyo, Glycomimetics, Incyte, Jazz, Kura Oncology, Novartis, Pfizer, Servier, Stemline, and Sumitomo Pharma; and medical writing support for this work from Kura Oncology. JKA declares a leadership role with the American Society of Hematology (guidelines panel), National Comprehensive Cancer Network (acute myeloid leukaemia panel vice-chair), and National Cancer Institute (co-chair on Leukemia Steering Committee); honoraria from Astellas, HMP Education, MD Education, and VJ HemOnc; advisory board role with AbbVie, Aptitude Health, Astellas, BlueBird Bio, Curio, Daiichi Sankyo, Dark Blue Therapeutics, Gilead, Kura Oncology, Kymera, Rigel, Stemline Therapeutics, Syros, and Treadwell Therapeutics; meeting attendance and travel expenses from Astellas, Daiichi Sankyo, HMP Education, MD Education, and VJ HemOnc; medical writing support for this work from Kura Oncology; and participation on a Data Safety Committee for Glycomimetics. PM declares consultancy fees from Kura Oncology and Syndax Pharmaceuticals and medical writing support for this work from Kura Oncology. SDB declares honoraria from Astellas, Bristol Myers Squibb, Menarini, and Servier; consultancy fees from AbbVie, Bristol Myers Squibb, Forma, Remix, Rigel, and Servier; travel expenses from Janssen, Pfizer, Rigel, and Servier; and medical writing support for this work from Kura Oncology. RBW declares clinical trial support from Kura Oncology and medical writing support for this work from Kura Oncology. KP declares honoraria from Merck (investigator meeting lecture); advisory board role with AbbVie, Incyte, PharmaEssentia, Protagonist, and Sobi; and medical writing support for this work from Kura Oncology. MRS declares research funding to institution from ALX Oncology, Astex, Incyte, Takeda, and TG Therapeutics; consultancy fees from AbbVie, Bristol Myers Squibb, CTI BioPharma, Forma, Geron, GlaxoSmithKline, Karyopharm, Rigel, Ryvu, Taiho, and Treadwell; stock or stock options in Empath Bioscience, Karyopharm, and Ryvu; and medical writing support for this work from Kura Oncology. MVS declares research grant to their institution from AbbVie, Astellas, Celgene, Kura Oncology, and MRKR Therapeutics; travel expenses from Dava Oncology; and medical writing support for this work from Kura Oncology. MK declares consultancy fees from AbbVie, CTI Biopharma, Incyte, and Protagonist; advisory board role with CTI BioPharma, Incyte, Kura Oncology, and Morphosys; travel expenses from Protagonist; and medical writing support for this work from Kura Oncology. MRB declares research funding to their institution from AbbVie, Ascentage, Astellas, Gilead, Kura Oncology, and Takeda and medical writing support for this work from Kura Oncology. JMF declares leadership role with National Cancer Institute Leukemia Steering Committee and the National Heart, Lung, and Blood Institute national Myelodysplastic Syndrome Study Steering Committee; stock or stock options with Aurinia; honoraria from AmerisourceBergen/IntrinsiQ Specialty Solutions, Aptitude Health, and MJH LifeSciences; consultancy fees from Autolus, Bristol Myers Squibb, Remix, and Syndax; grants to institution for clinical trial support from Actinium, Astellas, Celgene, Chordia, Kura Oncology, Novartis, Pfizer, Roivant, Sellas, and Servier; and medical writing support for this work from Kura Oncology. GS declares contracts through their institution from AbbVie, Actinium, Actuate, Agios, Arog, Astellas, AlloVir, Amgen, Aptevo, AltruBio, AVM Bio, Bristol Myers Squibb/Celgene; BioMea, Biopath, Biosight, Cellularity, Celator, Constellation, Cogent, Cellectis, Cullinan, Daiichi Sankyo, Deciphera, Delta-Fly, Fate, Forma, FujiFilm, Gamida, Genentech-Roche, Rigel Glycomimetics, Geron, Gilead, Incyte, Janssen, Jazz, Karyopharm, Kite/Gilead, Kronos Bio, Kura Oncology, Immunogene, ImmuneOnc, Loxo, Marker, Mateon, Novartis, Onconova, Ono-UK, Orca, Pfizer, PrECOG, Regimmune, Samus, Sangamo, Sellas, Stemline, Syros, Takeda, Tolero, and Trovagene; consultancy fees from Bristol Myers Squibb, Curios, Daiichi, and Novartis; speakers bureau role for AbbVie, Agios, Amgen, Astellas, Blueprint Medicine, Bristol Myers Squibb, Celgene, Karyopharm, GlaxoSmithKline, Kite (Gilead), Jazz, Rigel, Seattle genetics, and Stemline; board or advisory committee membership for Agios, Autolus, AVM Biotech, Bristol Myers Squibb, Gamida, Gilead, GlaxoSmithKline, Incyte, Novartis, Orca, Rigel, and Stemline; board of trustees membership for Leukemia Lymphoma Society Los Angeles; secretary or treasurer membership for the American Society of Hematology Research Collaborative Board of Directors; and holds stock with Amgen, Bristol Myers Squibb, and Janssen/Johnson & Johnson. LA declares research funding to institution from AbbVie, Bristol Myers Squibb, Jazz Pharmaceuticals, and Novartis; consultancy fees from AbbVie, Jazz Pharmaceuticals, and Novartis; and medical writing support for this work from Kura Oncology. ER declares consulting fees from Astellas and Laboratories Delbert; honoraria from AbbVie, Astellas, Eurocept, and Jazz Pharmaceuticals; support for attending meetings and travel from AbbVie, Gilead, and Jazz Pharmaceuticals; and medical writing support for this work from Kura Oncology. MMP declares research funding to institution from Epigenetix, Kura Oncology, Polaris, Solutherapeutics, and Stem Line Pharma; medical writing support for this work from Kura Oncology; and Data Safety Monitoring Boards with CTI Biopharma. CP declares honoraria from AbbVie, Amgen, Astellas, Blueprint, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Incyte, Janssen, Jazz Pharmaceuticals, Laboratories Delbert, Menarini-Stemline, Novartis, Paladin Labs, Pfizer, and Servier; travel expenses from AbbVie, Amgen and Pfizer; medical writing support for this work from Kura Oncology; and Data Safety Monitoring Board or advisory board membership for AbbVie, Amgen, Astellas, Blueprint, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Incyte, Janssen, Jazz Pharmaceuticals, Laboratories Delbert, Paladin Labs, and Pfizer. JG declares research support to institution, consultancy fees, royalties, patents, and stock with Kura Oncology, and medical writing support for this work from Kura Oncology. TC declares research support to institution and royalties from, and patents and owns stock with Kura Oncology; and medical writing support for this work from Kura Oncology. BC, JR, BML, MH, PP, and OS declare medical writing support for this work from Kura Oncology. KN declares employment with Kura Oncology; received stock options from Kura Oncology; and medical writing support for this work from Kura Oncology. AM declares employment with Kura Oncology; owns stock with Kura Oncology; patents and patent applications with Kura Oncology; and medical writing support for this work from Kura Oncology. JMA declares employment and stock or stock options with and travel expenses from Kura Oncology, and medical writing support for this work from Kura Oncology. MT declares employment with Kura Oncology; stock and restricted stock units with Kura Oncology; reimbursement for conference fees, hotels and travel expenses from Kura Oncology; and medical writing support for this work from Kura Oncology. HSS declares employment, patents and patent applications, and owns stock with, Kura Oncology, and medical writing support for this work from Kura Oncology. DC declares employment with Kura Oncology; stock or stock options with Kura Oncology; and medical writing support for this work from Kura Oncology. ML declares employment, patents and patent applications, stock or stock options, support for attending meetings and travel, and other financial or non-financial interests with Kura Oncology; and medical writing support for this work from Kura Oncology. SD declares employment, patents and patent applications, stock or stock options, and other financial or non-financial interests with Kura Oncology; and medical writing support for this work from Kura Oncology. ATF declares consultancy fees from AbbVie, Amgen, Astellas, AstraZeneca, Autolus, Bristol Myers Squibb/Celgene, Daiichi Sankyo, EnClear, Forma, Genentech, Gilead, Immunogen, Ipsen, Kite, Mablytics, Menarini, Novartis, Orum, Pfizer, PureTech, Remix, Rigel, Servier, and Takeda; clinical trial support from AbbVie, Bristol Myers Squibb, and Servier; and medical writing support for this work from Kura Oncology. CB declares no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
17. Contemporary Approach to the Diagnosis and Classification of Myelodysplastic Neoplasms/Syndromes-Recommendations From the International Consortium for Myelodysplastic Neoplasms/Syndromes (MDS [icMDS]).
- Author
-
Aakash F, Gisriel SD, Zeidan AM, Bennett JM, Bejar R, Bewersdorf JP, Borate UM, Boultwood J, Brunner AM, Buckstein R, Carraway HE, Churpek JE, Daver NG, DeZern AE, Efficace F, Fenaux P, Figueroa ME, Garcia-Manero G, Gore SD, Greenberg PL, Griffiths EA, Halene S, Hourigan CS, Kim TK, Kim N, Komrokji RS, Kutchroo VK, List AF, Little RF, Majeti R, Nazha A, Nimer SD, Odenike O, Padron E, Patnaik MM, Platzbecker U, Della Porta MG, Roboz GJ, Sallman DA, Santini V, Sanz G, Savona MR, Sekeres MA, Stahl M, Starczynowski DT, Steensma DP, Taylor J, Abdel-Wahab O, Wei AH, Xie Z, Xu ML, Hasserjian RP, and Loghavi S
- Abstract
Myelodysplastic neoplasms/syndromes (MDS) are a heterogeneous group of biologically distinct entities characterized by variable degrees of ineffective hematopoiesis. Recently, 2 classification systems (the 5th edition of the World Health Organization Classification of Haematolymphoid tTumours and the International Consensus Classification) further subcharacterized MDS into morphologically and genetically defined groups. Accurate diagnosis and subclassification of MDS require a multistep systemic approach. The International Consortium for MDS (icMDS) summarizes a contemporary, practical, and multimodal approach to MDS diagnosis and classification., (Copyright © 2024 United States & Canadian Academy of Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Molecular and clinical presentation of UBA1-mutated myelodysplastic syndromes.
- Author
-
Sirenko M, Bernard E, Creignou M, Domenico D, Farina A, Arango Ossa JE, Kosmider O, Hasserjian R, Jädersten M, Germing U, Sanz G, van de Loosdrecht AA, Gurnari C, Follo MY, Thol F, Zamora L, Pinheiro RF, Pellagatti A, Elias HK, Haase D, Sander B, Orna E, Zoldan K, Eder LN, Sperr WR, Thalhammer R, Ganster C, Adès L, Tobiasson M, Palomo L, Della Porta MG, Huberman K, Fenaux P, Belickova M, Savona MR, Klimek VM, Santos FPS, Boultwood J, Kotsianidis I, Santini V, Solé F, Platzbecker U, Heuser M, Valent P, Finelli C, Voso MT, Shih LY, Ogawa S, Fontenay M, Jansen JH, Cervera J, Ebert BL, Bejar R, Greenberg PL, Gattermann N, Malcovati L, Cazzola M, Beck DB, Hellström-Lindberg E, and Papaemmanuil E
- Subjects
- Humans, Male, Middle Aged, Aged, Adult, Aged, 80 and over, Female, Young Adult, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes diagnosis, Ubiquitin-Activating Enzymes genetics, Mutation
- Abstract
Abstract: Mutations in UBA1, which are disease-defining for VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome, have been reported in patients diagnosed with myelodysplastic syndromes (MDS). Here, we define the prevalence and clinical associations of UBA1 mutations in a representative cohort of patients with MDS. Digital droplet polymerase chain reaction profiling of a selected cohort of 375 male patients lacking MDS disease-defining mutations or established World Health Organization (WHO) disease classification identified 28 patients (7%) with UBA1 p.M41T/V/L mutations. Using targeted sequencing of UBA1 in a representative MDS cohort (n = 2027), we identified an additional 27 variants in 26 patients (1%), which we classified as likely/pathogenic (n = 12) and of unknown significance (n = 15). Among the total 40 patients with likely/pathogenic variants (2%), all were male and 63% were classified by WHO 2016 criteria as MDS with multilineage dysplasia or MDS with single-lineage dysplasia. Patients had a median of 1 additional myeloid gene mutation, often in TET2 (n = 12), DNMT3A (n = 10), ASXL1 (n = 3), or SF3B1 (n = 3). Retrospective clinical review, where possible, showed that 82% (28/34) UBA1-mutant cases had VEXAS syndrome-associated diagnoses or inflammatory clinical presentation. The prevalence of UBA1 mutations in patients with MDS argues for systematic screening for UBA1 in the management of MDS., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
19. Expanded profiling of WD repeat domain 5 inhibitors reveals actionable strategies for the treatment of hematologic malignancies.
- Author
-
Meyer CT, Smith BN, Wang J, Teuscher KB, Grieb BC, Howard GC, Silver AJ, Lorey SL, Stott GM, Moore WJ, Lee T, Savona MR, Weissmiller AM, Liu Q, Quaranta V, Fesik SW, and Tansey WP
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Sulfonamides pharmacology, Sulfonamides therapeutic use, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Protein p53 genetics, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Hematologic Neoplasms drug therapy, Hematologic Neoplasms genetics, Hematologic Neoplasms metabolism, Xenograft Model Antitumor Assays
- Abstract
WD40 Repeat Domain 5 (WDR5) is a highly conserved nuclear protein that recruits MYC oncoprotein transcription factors to chromatin to stimulate ribosomal protein gene expression. WDR5 is tethered to chromatin via an arginine-binding cavity known as the "WIN" site. Multiple pharmacological inhibitors of the WDR5-interaction site of WDR5 (WINi) have been described, including those with picomolar affinity and oral bioavailability in mice. Thus far, however, WINi have only been shown to be effective against a number of rare cancer types retaining wild-type p53. To explore the full potential of WINi for cancer therapy, we systematically profiled WINi across a panel of cancer cells, alone and in combination with other agents. We report that WINi are unexpectedly active against cells derived from both solid and blood-borne cancers, including those with mutant p53. Among hematologic malignancies, we find that WINi are effective as a single agent against leukemia and diffuse large B cell lymphoma xenograft models, and can be combined with the approved drug venetoclax to suppress disseminated acute myeloid leukemia in vivo. These studies reveal actionable strategies for the application of WINi to treat blood-borne cancers and forecast expanded utility of WINi against other cancer types., Competing Interests: Competing interests statement:Fesik, S. W., Stauffer, S. R., Salovich, J. M., Tansey, W. P., Wang, F., Phan, J., Olejniczak, E. T., inventors. WDR5 inhibitors and modulators. United States Patent US 10,501,466. 10 December 2019. Fesik, S. W., Stauffer, S. R., Tansey, W. P., Olejniczak, E. T., Phan, J., Wang, F., Jeon, K., Gogliotti, R. D., inventors. WDR5 inhibitors and modulators. United States Patent US 10,160,763. 25 December 2018. Lee. T.; Alvarado, J.; Tian, J.; Meyers, K. M.; Han, C.; Mills, J. J.; Teuscher, K. B.; Fesik, S. W. WDR5 inhibitors and modulators. WO 2020086857. 30 April 2020. Lee. T.; Han, C.; Mills, J. J.; Teuscher, K. B.; Tian, J.; Meyers, K. M.; Chowdhury, S.; Fesik, S. W. WDR5 inhibitors and modulators. WO 2020247679. 10 December 2020. Lee. T.; Teuscher, K. B.; Tian, J.; Meyers, K. M.; Chowdhury, S.; Fesik, S. W. WDR5 Inhibitors and modulators. WO 2021092525. 14 May 2021. Lee. T.; Teuscher, K. B.; Chowdhury, S.; Tian, J.; Meyers, K. M.; Fesik, S. W. WDR5 Inhibitors and modulators. WO2022236101. 10 November 2022. C.T.M. and V.Q. are co-founders of Duet BioSystems. M.R.S. has stock in Karyopharm and Ryvu; serves on advisory boards or consults for BMS, CTI, Forma, Geron, GSK, Karyopharm, Rigel, Ryvu, Taiho and Treadwell. All other authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
20. Multiomic profiling of human clonal hematopoiesis reveals genotype and cell-specific inflammatory pathway activation.
- Author
-
Heimlich JB, Bhat P, Parker AC, Jenkins MT, Vlasschaert C, Ulloa J, Van Amburg JC, Potts CR, Olson S, Silver AJ, Ahmad A, Sharber B, Brown D, Hu N, van Galen P, Savona MR, Bick AG, and Ferrell PB
- Subjects
- Humans, Genotype, Mutation, Gene Expression Profiling, Dioxygenases, DNA Methyltransferase 3A metabolism, Male, Female, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Clonal Hematopoiesis, Inflammation genetics
- Abstract
Abstract: Clonal hematopoiesis (CH) is an age-associated phenomenon that increases the risk of hematologic malignancy and cardiovascular disease. CH is thought to enhance disease risk through inflammation in the peripheral blood.1 Here, we profile peripheral blood gene expression in 66 968 single cells from a cohort of 17 patients with CH and 7 controls. Using a novel mitochondrial DNA barcoding approach, we were able to identify and separately compare mutant Tet methylcytosine dioxygenase 2 (TET2) and DNA methyltransferase 3A (DNMT3A) cells with nonmutant counterparts. We discovered the vast majority of mutated cells were in the myeloid compartment. Additionally, patients harboring DNMT3A and TET2 CH mutations possessed a proinflammatory profile in CD14+ monocytes through previously unrecognized pathways such as galectin and macrophage inhibitory factor. We also found that T cells from patients with CH, although mostly unmutated, had decreased expression of GTPase of the immunity associated protein genes, which are critical to T-cell development, suggesting that CH impairs T-cell function., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. Rapid prototyping of perfusion cell culture devices for three-dimensional imaging of mesenchymal stem cell deposition and proliferation.
- Author
-
Scott TE, Boccarossa T, Florian D, Fischer MA, Peck SH, Savona MR, Pingen G, and Guelcher SA
- Abstract
Perfusion of porous scaffolds transports cells to the surface to yield cellular constructs for 3D models of disease and for tissue engineering applications. While ceramic scaffolds mimic the structure and composition of trabecular bone, their opacity and tortuous pores limit the penetration of light into the interior. Scaffolds that are both perfusable and amenable to fluorescence microscopy are therefore needed to visualize the spatiotemporal dynamics of cells in the bone microenvironment. In this study, a hybrid injection molding approach was designed to enable rapid prototyping of collector arrays with variable configurations that are amenable to longitudinal imaging of attached human mesenchymal stem cells (hMSCs) using fluorescence microscopy. Cylindrical collectors were arranged in an array that is permeable to perfusion in the xy -plane and to light in the z-direction for imaging from below. The effects of the collector radius, number, and spacing on the collection efficiency of perfused hMSCs was simulated using computational fluid dynamics (CFD) and measured experimentally using fluorescence microscopy. The effect of collector diameter on simulated and experimental cell collection efficiencies followed a trend similar to that predicted by interception theory corrected for intermolecular and hydrodynamic forces for the arrays with constant collector spacing. In contrast, arrays designed with constant collector number yielded collection efficiencies that poorly fit the trend with collector radius predicted by interception theory. CFD simulations of collection efficiency agreed with experimental measurements within a factor of two. These findings highlight the utility of CFD simulations and hybrid injection molding for rapid prototyping of collector arrays to optimize the longitudinal imaging of cells without the need for expensive and time-consuming tooling., Competing Interests: The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Scott Guelcher reports financial support was provided by Incyte Corporation. Michael Savona reports financial support was provided by Incyte Corporation. Sun Peck reports financial support was provided by 10.13039/100000738US Department of Veterans Affairs. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
22. Clonal hematopoiesis and inflammation in the vasculature: CHIVE, a prospective, longitudinal clonal hematopoiesis cohort and biorepository.
- Author
-
Shannon ML, Heimlich JB, Olson S, Debevec A, Copeland Z, Kishtagari A, Vlasschaert C, Snider C, Silver AJ, Brown D, Spaulding T, Bhatta M, Pugh K, Stockton SS, Ulloa J, Xu Y, Baljevic M, Moslehi J, Jahangir E, Ferrell PB, Slosky D, Bick AG, and Savona MR
- Subjects
- Humans, Prospective Studies, Female, Male, Middle Aged, Aged, Registries, Hematologic Neoplasms genetics, Mutation, Adult, Clonal Hematopoiesis, Inflammation
- Abstract
Abstract: Clonal hematopoiesis (CH) is an age-associated phenomenon leading to an increased risk of both hematologic malignancy and nonmalignant organ dysfunction. Increasingly available genetic testing has made the incidental discovery of CH clinically common yet evidence-based guidelines and effective management strategies to prevent adverse CH health outcomes are lacking. To address this gap, the prospective CHIVE (clonal hematopoiesis and inflammation in the vasculature) registry and biorepository was created to identify and monitor individuals at risk, support multidisciplinary CH clinics, and refine taxonomy and standards of practice for CH risk mitigation. Data from the first 181 patients enrolled in this prospective registry recapitulate the molecular epidemiology of CH from biobank-scale retrospective studies, with DNMT3A, TET2, ASXL1, and TP53 as the most commonly mutated genes. Blood counts across all hematopoietic lineages trended lower in patients with CH. In addition, patients with CH had higher rates of end organ dysfunction, in particular chronic kidney disease. Among patients with CH, variant allele frequency was independently associated with the presence of cytopenias and progression to hematologic malignancy, whereas other common high-risk CH clone features were not clear. Notably, accumulation of multiple distinct high-risk clone features was also associated with cytopenias and hematologic malignancy progression, supporting a recently published CH risk score. Surprisingly, ∼30% of patients enrolled in CHIVE from CH clinics were adjudicated as not having clonal hematopoiesis of indeterminate potential, highlighting the need for molecular standards and purpose-built assays in this field. Maintenance of this well-annotated cohort and continued expansion of CHIVE to multiple institutions are underway and will be critical to understanding how to thoughtfully care for this patient population., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
23. Cost-Effective and Scalable Clonal Hematopoiesis Assay Provides Insight into Clonal Dynamics.
- Author
-
Mack T, Vlasschaert C, von Beck K, Silver AJ, Heimlich JB, Poisner H, Condon HR, Ulloa J, Sochacki AL, Spaulding TP, Kishtagari A, Bejan CA, Xu Y, Savona MR, Jones A, and Bick AG
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Adult, High-Throughput Nucleotide Sequencing methods, High-Throughput Nucleotide Sequencing economics, Cost-Benefit Analysis, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells cytology, Clonal Evolution genetics, Aged, 80 and over, Hematopoiesis genetics, Clonal Hematopoiesis genetics, Mutation
- Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is a common age-related phenomenon in which hematopoietic stem cells acquire mutations in a select set of genes commonly mutated in myeloid neoplasia which then expand clonally. Current sequencing assays to detect CHIP mutations are not optimized for the detection of these variants and can be cost-prohibitive when applied to large cohorts or to serial sequencing. In this study, an affordable (approximately US $8 per sample), accurate, and scalable sequencing assay for CHIP is introduced and validated. The efficacy of the assay was demonstrated by identifying CHIP mutations in a cohort of 456 individuals with DNA collected at multiple time points in Vanderbilt University's biobank and quantifying clonal expansion rates over time. A total of 101 individuals with CHIP/clonal cytopenia of undetermined significance were identified, and individual-level clonal expansion rate was calculated using the variant allele fraction at both time points. Differences in clonal expansion rate by driver gene were observed, but there was also significant individual-level heterogeneity, emphasizing the multifactorial nature of clonal expansion. Additionally, mutation co-occurrence and clonal competition between multiple driver mutations were explored., Competing Interests: Disclosure Statement M.R.S. has received honoraria for advisory board membership or consultancy from Bristol Myers Squibb, CTI, Forma, Geron, GlaxoSmithKline/Sierra Oncology, Karyopharm, Ryvu Therapeutics, and Taiho Pharmaceutical; has received research funding from ALX Oncology, Astex Pharmaceuticals, Incyte Corporation, Takeda, and TG Therapeutics; holds equity in Empath Biosciences, Karyopharm, and Ryvu Therapeutics; and has been reimbursed for travel expenses by Astex. A.G.B. has received honoraria for advisory board membership from, and holds equity in, TenSixteen Bio., (Copyright © 2024 Association for Molecular Pathology and American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. TLR3 agonism augments CD47 inhibition in acute myeloid leukemia.
- Author
-
Ramsey HE, Gorska AE, Smith BN, Monteith AJ, Fuller L, Arrate MP, and Savona MR
- Subjects
- Humans, Animals, Mice, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Xenograft Model Antitumor Assays, Cell Line, Tumor, Macrophages metabolism, Macrophages drug effects, Sulfonamides pharmacology, Receptors, Immunologic metabolism, Receptors, Immunologic antagonists & inhibitors, Antigens, Differentiation metabolism, Phagocytosis drug effects, Poly I-C pharmacology, CD47 Antigen metabolism, CD47 Antigen antagonists & inhibitors, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology
- Abstract
CD47-SIRPa is a myeloid check point pathway that promotes phagocytosis of cells lacking markers for self-recognition. Tumor cells can overexpress CD47 and bind to SIRPa on macrophages, preventing phagocytosis. CD47 expression is enhanced and correlated with a negative prognosis in acute myeloid leukemia (AML), with its blockade leading to cell clearance. ALX90 is an engineered fusion protein with high affinity for CD47. Composed of the N-terminal D1 domain of SIRPα genetically linked to an inactive Fc domain from human immunoglobulin (Ig) G, ALX90 is designed to avoid potential toxicity of CD47-expressing red blood cells. Venetoclax (VEN) is a specific B-cell lymphoma-2 (BCL-2) inhibitor that can restore apoptosis in malignant cells. In AML, VEN is combined with azanucleosides to induce superior remission rates, however treatment for refractory/relapse is an unmet need. We questioned whether the anti-tumor activity of a VENbased regimen can be augmented through CD47 inhibition (CD47i) in AML and how this triplet may be enhanced. Human AML cell lines were sensitive to ALX90 and its addition increased efficacy of a VEN plus azacitidin (VEN+AZA) regimen in vivo. However, CD47i failed to clear bone marrow tumor burden in PDX models. We hypothesized that the loss of resident macrophages in the bone marrow in AML reduced efficiency of CD47i. Therefore, we attempted to enhance this medullary macrophage population with agonism of TLR3 via polyinosinic:polycytidylic acid (poly(I:C)), which led to expansion and activation of medullary macrophages in in vivo AML PDX models and potentiated CD47i. In summary, the addition of poly(I:C) can enhance medullary macrophage populations to potentiate the phagocytosis merited by therapeutic inhibition of CD47.
- Published
- 2024
- Full Text
- View/download PDF
25. Solid Organ Transplant Recipients Exhibit More TET2-Mutant Clonal Hematopoiesis of Indeterminate Potential Not Driven by Increased Transplantation Risk.
- Author
-
Silver AJ, Vlasschaert C, Mack T, Sharber B, Xu Y, Bick AG, Pinson CW, and Savona MR
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Hematologic Neoplasms genetics, Hematologic Neoplasms epidemiology, Hematologic Neoplasms etiology, Hematologic Neoplasms pathology, Proto-Oncogene Proteins genetics, Risk Factors, Transplant Recipients, Clonal Hematopoiesis genetics, Dioxygenases, DNA-Binding Proteins genetics, Mutation, Organ Transplantation adverse effects
- Abstract
Purpose: Solid organ transplant recipients comprise a unique population of immunosuppressed patients with increased risk of malignancy, including hematologic neoplasms. Clonal hematopoiesis of indeterminate potential (CHIP) represents a known risk factor for hematologic malignancy and this study describes the prevalence and patterns of CHIP mutations across several types of solid organ transplants., Experimental Design: We use two national biobank cohorts comprised of >650,000 participants with linked genomic and longitudinal phenotypic data to describe the features of CHIP across 2,610 individuals who received kidney, liver, heart, or lung allografts., Results: We find individuals with an allograft before their biobank enrollment had an increased prevalence of TET2 mutations (OR, 1.90; P = 4.0e-4), but individuals who received transplants post-enrollment had a CHIP mutation spectrum similar to that of the general population, without enrichment of TET2. In addition, we do not observe an association between CHIP and risk of incident transplantation among the overall population (HR, 1.02; P = 0.91). And in an exploratory analysis, we do not find evidence for a strong association between CHIP and rates of transplant complications such as rejection or graft failure., Conclusions: These results demonstrate that recipients of solid organ transplants display a unique pattern of clonal hematopoiesis with enrichment of TET2 driver mutations, the causes of which remain unclear and are deserving of further study., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
26. A randomized, double-blind study of zinpentraxin alfa in patients with myelofibrosis who were previously treated with or ineligible for ruxolitinib: stage 2 of a phase II trial.
- Author
-
Verstovsek S, Talpaz M, Wadleigh M, Isidori A, Te Boekhorst P, Savona MR, Bose P, Pozdnyakova O, Mesa R, El-Galaly TC, O'Sullivan J, Gamel K, Higgins B, Katakam S, Todorov B, Trunzer K, and Harrison CN
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Double-Blind Method, Treatment Outcome, Nitriles therapeutic use, Primary Myelofibrosis drug therapy, Pyrazoles therapeutic use, Pyrimidines therapeutic use
- Published
- 2024
- Full Text
- View/download PDF
27. Interoperability of phenome-wide multimorbidity patterns: a comparative study of two large-scale EHR systems.
- Author
-
Strayer N, Vessels T, Choi K, Zhang S, Li Y, Han L, Sharber B, Hsi RS, Bejan CA, Bick AG, Balko JM, Johnson DB, Wheless LE, Wells QS, Philips EJ, Pulley JM, Self WH, Chen Q, Hartert T, Wilkins CH, Savona MR, Shyr Y, Roden DM, Smoller JW, Ruderfer DM, and Xu Y
- Abstract
Background: Electronic health records (EHR) are increasingly used for studying multimorbidities. However, concerns about accuracy, completeness, and EHRs being primarily designed for billing and administrative purposes raise questions about the consistency and reproducibility of EHR-based multimorbidity research., Methods: Utilizing phecodes to represent the disease phenome, we analyzed pairwise comorbidity strengths using a dual logistic regression approach and constructed multimorbidity as an undirected weighted graph. We assessed the consistency of the multimorbidity networks within and between two major EHR systems at local (nodes and edges), meso (neighboring patterns), and global (network statistics) scales. We present case studies to identify disease clusters and uncover clinically interpretable disease relationships. We provide an interactive web tool and a knowledge base combining data from multiple sources for online multimorbidity analysis., Findings: Analyzing data from 500,000 patients across Vanderbilt University Medical Center and Mass General Brigham health systems, we observed a strong correlation in disease frequencies (Kendall's τ = 0.643) and comorbidity strengths (Pearson ρ = 0.79). Consistent network statistics across EHRs suggest similar structures of multimorbidity networks at various scales. Comorbidity strengths and similarities of multimorbidity connection patterns align with the disease genetic correlations. Graph-theoretic analyses revealed a consistent core-periphery structure, implying efficient network clustering through threshold graph construction. Using hydronephrosis as a case study, we demonstrated the network's ability to uncover clinically relevant disease relationships and provide novel insights., Interpretation: Our findings demonstrate the robustness of large-scale EHR data for studying phenome-wide multimorbidities. The alignment of multimorbidity patterns with genetic data suggests the potential utility for uncovering shared biology of diseases. The consistent core-periphery structure offers analytical insights to discover complex disease interactions. This work also sets the stage for advanced disease modeling, with implications for precision medicine., Funding: VUMC Biostatistics Development Award, the National Institutes of Health, and the VA CSRD., Competing Interests: JWS is a member of the Scientific Advisory Board of Sensorium Therapeutics (with equity) and has received grant support from Biogen, Inc. He is the principal investigator of a collaborative study of the genetics of depression and bipolar disorder sponsored by 23andMe, for which 23andMe provides analysis time as in-kind support but no payments. DMR has served on advisory boards for Illumina and Alkermes and has received research funds unrelated to this work from PTC Therapeutics. All other authors declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
28. Toward a more patient-centered drug development process in clinical trials for patients with myelodysplastic syndromes/neoplasms (MDS): Practical considerations from the International Consortium for MDS (icMDS).
- Author
-
Efficace F, Buckstein R, Abel GA, Giesinger JM, Fenaux P, Bewersdorf JP, Brunner AM, Bejar R, Borate U, DeZern AE, Greenberg P, Roboz GJ, Savona MR, Sparano F, Boultwood J, Komrokji R, Sallman DA, Xie Z, Sanz G, Carraway HE, Taylor J, Nimer SD, Della Porta MG, Santini V, Stahl M, Platzbecker U, Sekeres MA, and Zeidan AM
- Abstract
Notable treatment advances have been made in recent years for patients with myelodysplastic syndromes/neoplasms (MDS), and several new drugs are under development. For example, the emerging availability of oral MDS therapies holds the promise of improving patients' health-related quality of life (HRQoL). Within this rapidly evolving landscape, the inclusion of HRQoL and other patient-reported outcomes (PROs) is critical to inform the benefit/risk assessment of new therapies or to assess whether patients live longer and better, for what will likely remain a largely incurable disease. We provide practical considerations to support investigators in generating high-quality PRO data in future MDS trials. We first describe several challenges that are to be thoughtfully considered when designing an MDS-focused clinical trial with a PRO endpoint. We then discuss aspects related to the design of the study, including PRO assessment strategies. We also discuss statistical approaches illustrating the potential value of time-to-event analyses and their implications within the estimand framework. Finally, based on a literature review of MDS randomized controlled trials with a PRO endpoint, we note the PRO items that deserve special attention when reporting future MDS trial results. We hope these practical considerations will facilitate the generation of rigorous PRO data that can robustly inform MDS patient care and support treatment decision-making for this patient population., Competing Interests: Fabio Efficace had consultancy or advisory role for AbbVie, Incyte, Syros, Novartis, and JAZZ Pharmaceuticals outside the submitted work. Rena Buckstein: Research funding and honoraria for advisory boards and speaking engagements from BMS, TAIHO, and Abbvie. Gregory A. Abel has consulted for Novartis and Geron outside the submitted work. Pierre Fenaux received research funding from BMS, Abbvie, Jazz Pharmaceuticals, Novartis, and Janssen; and had a consultancy with and received honoraria from BMS, Abbvie, Jazz Pharmaceuticals, and Novartis. Andrew M. Brunner received consulting or advisory board honoraria from Novartis, Acceleron, Agios, Abbvie, Takeda, Celgene/BMS, Keros Therapeutics, Taiho, Gilead; and has research support from the NIH SPORE in Myeloid Malignancies, and from the Edward P. Evans Foundation. Rafael Bejar owns equity in and is employed by Aptose Biosciences; he has served as an advisor to BMS, Servier, Gilead, and Ipsen; he is on the SAB for NeoGenomics. Amy E DeZern participated in advisory boards, and/or had a consultancy with and received honoraria from Celgene/BMS, Agios, Regenergon, Sobi, Novartis, Astellas, and Gilead. Amy E. DeZern served on clinical trial committees for Novartis, Abbvie, Kura, Geron, and Celgene/BMS. Gail J. Roboz: Consultancy: Abbvie, Amgen, Astra Zeneca, Bristol‐Myers Squibb, Caribou Biosciences, Celgene, Daiichi Sankyo, Ellipses Pharma, Genoptix, Geron, GlaxoSmithKline, Janssen, Jasper Pharmaceuticals, Jazz Pharmaceuticals, Molecular Partners, Novartis, Pfizer, Oncoverity, OncoPrecision, Rigel, Roche, Syndax, Takeda (IRC Chair), Telix Pharma; and research support: Janssen. Michael R. Savona: Membership on a board or advisory committee: Bristol Myers Squibb, CTI, Forma, Geron, GSK, Karyopharm, Rigel Ryvu, Taiho, Takeda, Treadwell; patents and royalties: Boehringer Ingelheim, Empath Biosciences; research funding: ALX Oncology, Astex, Incyte, Takeda, TG Therapeutics; equity ownership: Empath Biosciences, Karyopharm, Ryvu; consultancy: Forma, Geron, Karyopharm, Ryvu. Rami Komrokji: Abbvie: Speaker Bureau, Advisory board; BMS: Research grant, Advisory board; DSI: Advisory board; Geron: Consultancy; Janssen: Consultancy; Jazz: Speaker Bureau, Advisory board; Pharma Essentia: Speaker Bureau, Advisory board; Rigel: Speaker Bureau, Advisory board; Servio: Speaker Bureau, Advisory board; Sobi: Speaker Bureau, Advisory board; Sumitomo Pharma: consultancy, Advisory board. David A. Sallman served on the advisory board or panel for Agios, Avencell, BlueBird Bio, BMS, Dark Blue, Jasper Therapeutics, Kite, Magenta Therapeutics, NKARTA, Novartis, Rigel Shattuck Labs, Servier, Syndax, Syros; and had a consultancy with AbbBie, Gilead, Molecular Partners AG, Takeda. Guillermo Sanz received honoraria, advisory board membership, or consultation fees from AbbVie, BMS, ExCellThera, Novartis, Roche, and Takeda and participated in sponsored speaker's bureau for BMS, Novartis, and Takeda. Hetty E. Carraway: Consultancy: Abbvie, Amgen, Bristol‐Myers Squibb, Celgene, Daiichi Sankyo, Jazz Pharmaceuticals, Novartis, Rigel, Syndax, Servier; and research support: Celgene. Maximilian Stahl consulted for Curis Oncology and Boston Consulting; served on the advisory board for Novartis and Kymera, GSK, Rigel, and Sierra Oncology; and participated in GME activity for Novartis, Curis Oncology, Haymarket Media, and Clinical care options (CCO). Mikkael A. Sekeres has served on advisory boards for BMS, Novartis, Kurome, and Gilead. Amer M. Zeidan received research funding (institutional) from Celgene/BMS, Abbvie, Astex, Pfizer, Medimmune/Astra Zeneca, Boehringer‐Ingelheim, Cardiff oncology, Incyte, Takeda, Novartis, Aprea, and ADC Therapeutics. He participated in advisory boards, and/or had a consultancy with and received honoraria from AbbVie, Otsuka, Pfizer, Celgene/BMS, Jazz, Incyte, Agios, Boehringer‐Ingelheim, Novartis, Acceleron, Astellas, Daiichi Sankyo, Cardinal Health, Taiho, Seattle Genetics, BeyondSpring, Cardiff Oncology, Takeda, Ionis, Amgen, Janssen, Epizyme, Syndax, Gilead, Kura, Chiesi, ALX Oncology, BioCryst, Notable, Orum, and Tyme. He served on clinical trial committees for Novartis, Abbvie, Gilead, BioCryst, Abbvie, ALX Oncology, Geron, and Celgene/BMS. Fabio Efficace, Rena Buckstein, and Gregory A. Abel report being involved in the development and validation of the QUALMS, one of the PROs discussed in this paper. The QUALMS is copyrighted by Dana‐Farber Cancer Institute (DFCI) and the Children's Hospital of Eastern Ontario (CHEO). The remaining authors declare no conflict of interest., (© 2024 The Authors. HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)
- Published
- 2024
- Full Text
- View/download PDF
29. Ribosome subunit attrition and activation of the p53-MDM4 axis dominate the response of MLL-rearranged cancer cells to WDR5 WIN site inhibition.
- Author
-
Howard GC, Wang J, Rose KL, Jones C, Patel P, Tsui T, Florian AC, Vlach L, Lorey SL, Grieb BC, Smith BN, Slota MJ, Reynolds EM, Goswami S, Savona MR, Mason FM, Lee T, Fesik S, Liu Q, and Tansey WP
- Subjects
- Humans, Antineoplastic Agents pharmacology, Cell Cycle Proteins metabolism, Cell Cycle Proteins genetics, Cell Line, Tumor, Histone-Lysine N-Methyltransferase metabolism, Histone-Lysine N-Methyltransferase genetics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Peptidomimetics pharmacology, Intracellular Signaling Peptides and Proteins antagonists & inhibitors, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Myeloid-Lymphoid Leukemia Protein metabolism, Myeloid-Lymphoid Leukemia Protein genetics, Nuclear Proteins metabolism, Nuclear Proteins genetics, Ribosomes drug effects, Ribosomes metabolism, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Protein p53 genetics
- Abstract
The chromatin-associated protein WD Repeat Domain 5 (WDR5) is a promising target for cancer drug discovery, with most efforts blocking an arginine-binding cavity on the protein called the 'WIN' site that tethers WDR5 to chromatin. WIN site inhibitors (WINi) are active against multiple cancer cell types in vitro, the most notable of which are those derived from MLL-rearranged (MLLr) leukemias. Peptidomimetic WINi were originally proposed to inhibit MLLr cells via dysregulation of genes connected to hematopoietic stem cell expansion. Our discovery and interrogation of small-molecule WINi, however, revealed that they act in MLLr cell lines to suppress ribosome protein gene (RPG) transcription, induce nucleolar stress, and activate p53. Because there is no precedent for an anticancer strategy that specifically targets RPG expression, we took an integrated multi-omics approach to further interrogate the mechanism of action of WINi in human MLLr cancer cells. We show that WINi induce depletion of the stock of ribosomes, accompanied by a broad yet modest translational choke and changes in alternative mRNA splicing that inactivate the p53 antagonist MDM4. We also show that WINi are synergistic with agents including venetoclax and BET-bromodomain inhibitors. Together, these studies reinforce the concept that WINi are a novel type of ribosome-directed anticancer therapy and provide a resource to support their clinical implementation in MLLr leukemias and other malignancies., Competing Interests: GH, JW, KR, CJ, PP, TT, AF, LV, SL, BG, BS, MS, ER, SG, FM, QL No competing interests declared, MS Receives research funding from ALX Oncology, Astex, Incyte, Takeda and TG Therapeutics; has stock in Karyopharm and Ryvu; serves on advisory boards or consults for BMS, CTI, Forma, Geron, GSK, Karyopharm, Rigel, Ryvu, Taiho and Treadwell, TL, SF Patents: Lee T, Alvarado J, Tian J, Meyers KM, Han C, Mills JJ, Teuscher KB, Stauffer SR, Fesik SW. WDR5 inhibitors and modulators. WO 2020086857. 30 April 2020; Lee T, Han C, Mills JJ, Teuscher KB, Tian J, Meyers KM, Chowdhury S, Fesik SW. WDR5 inhibitors and modulators. WO 2020247679. 10 December 2020; Lee T, Teuscher KB, Tian J, Meyers KM, Chowdhury S, Fesik SW. WDR5 Inhibitors and modulators. WO 2021092525. 14 May 2021; Lee T, Teuscher KB, Chowdhury S, Tian J, Meyers KM, Fesik SW. WDR5 Inhibitors and modulators. WO2022236101. 10 November 2022, WT Patents: Fesik SW, Stauffer SR, Salovich JM, Tansey WP, Wang F, Phan J, Olejniczak ET, inventors. WDR5 inhibitors and modulators. United States Patent US 10,501,466. 10 December 2019; Fesik SW, Stauffer SR, Tansey WP, Olejniczak ET, Phan J, Wang F, Jeon K, Gogliotti RD, inventors. WDR5 inhibitors and modulators. United States Patent US 10,160,763. 25 December 2018, (© 2023, Howard et al.)
- Published
- 2024
- Full Text
- View/download PDF
30. Repair of leukemia-associated single nucleotide variants via interallelic gene conversion.
- Author
-
Silver AJ, Brown DJ, Olmstead SD, Watke JM, Gorska AE, Tanner L, Ramsey HE, and Savona MR
- Abstract
CRISPR-Cas9 is a useful tool for inserting precise genetic alterations through homology-directed repair (HDR), although current methods rely on provision of an exogenous repair template. Here, we tested the possibility of repairing heterozygous single nucleotide variants (SNVs) using the cell's own wild-type allele rather than an exogenous template. Using high-fidelity Cas9 to perform allele-specific CRISPR across multiple human leukemia cell lines as well as in primary hematopoietic cells from patients with leukemia, we find high levels of reversion to wild-type in the absence of exogenous template. Moreover, we demonstrate that bulk treatment to revert a truncating mutation in ASXL1 using CRISPR-mediated interallelic gene conversion (IGC) is sufficient to prolong survival in a human cell line-derived xenograft model (median survival 33 days vs 27.5 days; p = 0.0040). These results indicate that IGC can be applied to numerous types of leukemia and can meaningfully alter cellular phenotypes at scale. Because our method targets single-base mutations, rather than larger variants targeted by IGC in prior studies, it greatly expands the pool of risk-increasing genetic lesions which could potentially be targeted by IGC. This technique may reduce cost and complexity for experiments modeling phenotypic consequences of SNVs. The principles of SNV-specific IGC demonstrated in this proof-of-concept study could be applied to investigate the phenotypic effects of targeted clonal reduction of leukemogenic SNV driver mutations.
- Published
- 2024
- Full Text
- View/download PDF
31. Lactate Utilization Enables Metabolic Escape to Confer Resistance to BET Inhibition in Acute Myeloid Leukemia.
- Author
-
Monteith AJ, Ramsey HE, Silver AJ, Brown D, Greenwood D, Smith BN, Wise AD, Liu J, Olmstead SD, Watke J, Arrate MP, Gorska AE, Fuller L, Locasale JW, Stubbs MC, Rathmell JC, and Savona MR
- Subjects
- Humans, Animals, Mice, Transcription Factors metabolism, Lactic Acid, Cell Line, Tumor, Lactate Dehydrogenases, Bromodomain Containing Proteins, Cell Cycle Proteins, Nuclear Proteins metabolism, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute metabolism
- Abstract
Impairing the BET family coactivator BRD4 with small-molecule inhibitors (BETi) showed encouraging preclinical activity in treating acute myeloid leukemia (AML). However, dose-limiting toxicities and limited clinical activity dampened the enthusiasm for BETi as a single agent. BETi resistance in AML myeloblasts was found to correlate with maintaining mitochondrial respiration, suggesting that identifying the metabolic pathway sustaining mitochondrial integrity could help develop approaches to improve BETi efficacy. Herein, we demonstrated that mitochondria-associated lactate dehydrogenase allows AML myeloblasts to utilize lactate as a metabolic bypass to fuel mitochondrial respiration and maintain cellular viability. Pharmacologically and genetically impairing lactate utilization rendered resistant myeloblasts susceptible to BET inhibition. Low-dose combinations of BETi and oxamate, a lactate dehydrogenase inhibitor, reduced in vivo expansion of BETi-resistant AML in cell line and patient-derived murine models. These results elucidate how AML myeloblasts metabolically adapt to BETi by consuming lactate and demonstrate that combining BETi with inhibitors of lactate utilization may be useful in AML treatment., Significance: Lactate utilization allows AML myeloblasts to maintain metabolic integrity and circumvent antileukemic therapy, which supports testing of lactate utilization inhibitors in clinical settings to overcome BET inhibitor resistance in AML. See related commentary by Boët and Sarry, p. 950., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
32. Imetelstat in patients with lower-risk myelodysplastic syndromes who have relapsed or are refractory to erythropoiesis-stimulating agents (IMerge): a multinational, randomised, double-blind, placebo-controlled, phase 3 trial.
- Author
-
Platzbecker U, Santini V, Fenaux P, Sekeres MA, Savona MR, Madanat YF, Díez-Campelo M, Valcárcel D, Illmer T, Jonášová A, Bělohlávková P, Sherman LJ, Berry T, Dougherty S, Shah S, Xia Q, Sun L, Wan Y, Huang F, Ikin A, Navada S, Feller F, Komrokji RS, and Zeidan AM
- Subjects
- Humans, Male, Female, Adolescent, Adult, Treatment Outcome, Erythropoiesis, Double-Blind Method, Antineoplastic Combined Chemotherapy Protocols, Myelodysplastic Syndromes drug therapy, Thrombocytopenia drug therapy, Oligonucleotides
- Abstract
Background: Unmet medical needs remain in patients with red blood cell transfusion-dependent (RBC-TD) lower-risk myelodysplastic syndromes (LR-MDS) who are not responding to or are ineligible for erythropoiesis-stimulating agents (ESAs). Imetelstat, a competitive telomerase inhibitor, showed promising results in a phase 2 trial. We aimed to compare the RBC transfusion independence (RBC-TI) rate with imetelstat versus placebo in patients with RBC-TD LR-MDS., Methods: In phase 3 of IMerge, a double-blind, placebo-controlled trial conducted in 118 sites including university hospitals, cancer centres, and outpatient clinics in 17 countries, patients (aged ≥18 years) with ESA-relapsed, ESA-refractory, or ESA-ineligible LR-MDS (low or intermediate-1 risk disease as per International Prognostic Scoring System [IPSS] criteria) were randomly assigned via a computer-generated schedule (2:1) to receive imetelstat 7·5 mg/kg or placebo, administered as a 2-h intravenous infusion, every 4 weeks until disease progression, unacceptable toxic effects, or withdrawal of consent. Randomisation was stratified by previous RBC transfusion burden and IPSS risk group. Patients, investigators, and those analysing the data were masked to group assignment. The primary endpoint was 8-week RBC-TI, defined as the proportion of patients without RBC transfusions for at least 8 consecutive weeks starting on the day of randomisation until subsequent anti-cancer therapy, if any. Primary efficacy analyses were performed in the intention-to-treat population, and safety analyses were conducted in patients who received at least one dose of trial medication or placebo. This trial is registered with ClinicalTrials.gov (NCT02598661; substudy active and recruiting)., Findings: Between Sept 11, 2019, and Oct 13, 2021, 178 patients were enrolled and randomly assigned (118 to imetelstat and 60 to placebo). 111 (62%) were male and 67 (38%) were female. 91 (77%) of 118 patients had discontinued treatment by data cutoff in the imetelstat group versus 45 (75%) in the placebo group; a further one patient in the placebo group did not receive treatment. Median follow-up was 19·5 months (IQR 12·0-23·4) in the imetelstat group and 17·5 months (12·1-22·7) in the placebo group. In the imetelstat group, 47 (40% [95% CI 30·9-49·3]) patients had an RBC-TI of at least 8 weeks versus nine (15% [7·1-26·6]) in the placebo group (rate difference 25% [9·9 to 36·9]; p=0·0008). Overall, 107 (91%) of 118 patients receiving imetelstat and 28 (47%) of 59 patients receiving placebo had grade 3-4 treatment-emergent adverse events. The most common treatment-emergent grade 3-4 adverse events in patients taking imetelstat were neutropenia (80 [68%] patients who received imetelstat vs two [3%] who received placebo) and thrombocytopenia (73 [62%] vs five [8%]). No treatment-related deaths were reported., Interpretation: Imetelstat offers a novel mechanism of action with durable transfusion independence (approximately 1 year) and disease-modifying activity for heavily transfused patients with LR-MDS who are not responding to or are ineligible for ESAs., Funding: Janssen Research & Development before April 18, 2019, and Geron Corporation thereafter., Competing Interests: Declaration of interests UP received honoraria consultancy fees and research funding from Geron Corporation, BMS/Celgene, AbbVie, Jazz, Janssen, Syros, Servier, Silence Therapeutics, and Takeda. VS served on advisory boards with AbbVie, CTI, Geron Corporation, Gilead, BMS/Celgene, Novartis, Otsuka, Servier, and Syros, and received a travel grant from Janssen. PF received consultancy fees, research funding, and honoraria from BMS/Celgene, and honoraria and research funding from Celgene. MAS received advisory board fees from Geron Corporation, BMS/Celgene, Novartis, and Kurome. MRS received research funding from ALX Oncology, Astex, Incyte, Takeda, and TG Therapeutics; consults or serves on advisory or data safety monitoring boards for AbbVie, BMS/Celgene, Forma, Geron Corporation, Karyopharm, Novartis, Ryvu, Sierra Oncology, Taiho, Takeda, and TG Therapeutics; and has equity in Karyopharm and Ryvu. YFM received honoraria or consulting fees from Blueprint Medicines, Geron Corporation, and OncLive; participated in advisory boards and received honoraria from Sierra Oncology, Stemline Therapeutics, Blueprint Medicines, Morphosys, Taiho Oncology, and Novartis; and received travel reimbursement from Blueprint Medicines and Morphosys. MD-C consulted or participated in an advisory role with BMS/Celgene, Novartis, GlaxoSmithKline, and Blueprint Medicines; received travel and accommodation expenses from Gilead Sciences; and received honoraria from BMS/Celgene and Novartis. DV consulted or participated in an advisory role with BMS/Celgene, Amgen, GlaxoSmithKline, Novartis, Takeda, Pfizer, BMS/Celgene, Sanofi, Jazz Pharmaceuticals, and SOBI; participated in speakers' bureau at Agios, Novartis, Amgen, GlaxoSmithKline, Astellas Pharma, Pfizer, Jazz Pharmaceuticals, Sanofi/Aventis, BMS/Celgene, Astellas Pharma, Kyte, and Gebro Pharma; and received travel, accommodations, and expenses from BMS/Celgene, Amgen, Pfizer, GlaxoSmithKline, and Jazz Pharmaceuticals. TI consulted or participated in an advisory role at Novartis, AstraZeneca, and AbbVie. AJ consulted or participated in an advisory role at AbbVie, BMS/Celgene, and Novartis; and received travel, accommodations, and expenses from AbbVie and BMS/Celgene. RSK participated on a speaker bureau with Jazz, Servier, AbbVie, CTI, and PharmaEssentia; received advisory board fees or honoraria from BMS/Celgene, Novartis, AbbVie, Jazz, Servier, PharmaEssentia, Taiho, Takeda, Geron Corporation, Gilead/Forty Seven, and CTI; received travel, accommodations, expenses from Jazz, BMS/Celgene, and PharmaEssentia; has stock and other ownership interests in AbbVie; and received research funding from BMS/Celgene. AMZ received research funding (institutional) from BMS/Celgene, AbbVie, Astex, Pfizer, Kura, Medimmune/AstraZeneca, Boehringer Ingelheim, Incyte, Takeda, Novartis, Shattuck Labs, Geron Corporation, Foran, and Aprea; participated in advisory boards, had a consultancy with, or received honoraria from AbbVie, Pfizer, BMS/Celgene, Jazz, Incyte, Agios, Servier, Boehringer Ingelheim, Novartis, Astellas, Daiichi Sankyo, Geron Corporation, Taiho, Seattle Genetics, BeyondSpring, Takeda, Ionis, Amgen, Janssen, Genentech, Epizyme, Syndax, Gilead, Kura, Chiesi, ALX Oncology, BioCryst, Notable, Orum, Mendus, Zentalis, Schrodinger, Regeneron, Syros, Schrodinger, and Tyme; and served on clinical trial committees for Novartis, AbbVie, Gilead, Syros, BioCryst, ALX Oncology, Kura, Geron Corporation, and BMS/Celgene. LS, TB, SD, SS, QX, LJS, YW, FH, AI, SN, and FF are employees of Geron Corporation. PB declares no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
33. Driver mutation zygosity is a critical factor in predicting clonal hematopoiesis transformation risk.
- Author
-
Kishtagari A, Khan MAW, Li Y, Vlasschaert C, Marneni N, Silver AJ, von Beck K, Spaulding T, Stockton S, Snider C, Sochacki A, Dorand D, Mack TM, Ferrell PB Jr, Xu Y, Bejan CA, Savona MR, and Bick AG
- Subjects
- Humans, Mutation, Point Mutation, Chromosome Aberrations, Clonal Hematopoiesis, Hematologic Neoplasms genetics
- Abstract
Clonal hematopoiesis (CH) can be caused by either single gene mutations (eg point mutations in JAK2 causing CHIP) or mosaic chromosomal alterations (e.g., loss of heterozygosity at chromosome 9p). CH is associated with a significantly increased risk of hematologic malignancies. However, the absolute rate of transformation on an annualized basis is low. Improved prognostication of transformation risk is urgently needed for routine clinical practice. We hypothesized that the co-occurrence of CHIP and mCAs at the same locus (e.g., transforming a heterozygous JAK2 CHIP mutation into a homozygous mutation through concomitant loss of heterozygosity at chromosome 9) might have important prognostic implications for malignancy transformation risk. We tested this hypothesis using our discovery cohort, the UK Biobank (n = 451,180), and subsequently validated it in the BioVU cohort (n = 91,335). We find that individuals with a concurrent somatic mutation and mCA were at significantly increased risk of hematologic malignancy (for example, In BioVU cohort incidence of hematologic malignancies is higher in individuals with co-occurring JAK2 V617F and 9p CN-LOH; HR = 54.76, 95% CI = 33.92-88.41, P < 0.001 vs. JAK2 V617F alone; HR = 44.05, 95% CI = 35.06-55.35, P < 0.001). Currently, the 'zygosity' of the CHIP mutation is not routinely reported in clinical assays or considered in prognosticating CHIP transformation risk. Based on these observations, we propose that clinical reports should include 'zygosity' status of CHIP mutations and that future prognostication systems should take mutation 'zygosity' into account., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
34. Oral decitabine-cedazuridine versus intravenous decitabine for myelodysplastic syndromes and chronic myelomonocytic leukaemia (ASCERTAIN): a registrational, randomised, crossover, pharmacokinetics, phase 3 study.
- Author
-
Garcia-Manero G, McCloskey J, Griffiths EA, Yee KWL, Zeidan AM, Al-Kali A, Deeg HJ, Patel PA, Sabloff M, Keating MM, Zhu N, Gabrail NY, Fazal S, Maly J, Odenike O, Kantarjian H, DeZern AE, O'Connell CL, Roboz GJ, Busque L, Buckstein R, Amin H, Randhawa J, Leber B, Shastri A, Dao KH, Oganesian A, Hao Y, Keer HN, Azab M, and Savona MR
- Subjects
- Male, Humans, Female, Decitabine adverse effects, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Myelomonocytic, Chronic drug therapy, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes etiology, Pneumonia etiology
- Abstract
Background: The DNA methyltransferase inhibitors azacitidine and decitabine for individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia are available in parenteral form. Oral therapy with similar exposure for these diseases would offer potential treatment benefits. We aimed to compare the safety and pharmacokinetics of oral decitabine plus the cytidine deaminase inhibitor cedazuridine versus intravenous decitabine., Methods: We did a registrational, multicentre, open-label, crossover, phase 3 trial of individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia and individuals with acute myeloid leukaemia, enrolled as separate cohorts; results for only participants with myelodysplastic syndromes or chronic myelomonocytic leukaemia are reported here. In 37 academic and community-based clinics in Canada and the USA, we enrolled individuals aged 18 years or older who were candidates to receive intravenous decitabine, with Eastern Cooperative Oncology Group performance status 0 or 1 and a life expectancy of at least 3 months. Participants were randomly assigned (1:1) to receive 5 days of oral decitabine-cedazuridine (one tablet once daily containing 35 mg decitabine and 100 mg cedazuridine as a fixed-dose combination) or intravenous decitabine (20 mg/m
2 per day by continuous 1-h intravenous infusion) in a 28-day treatment cycle, followed by 5 days of the other formulation in the next treatment cycle. Thereafter, all participants received oral decitabine-cedazuridine from the third cycle on until treatment discontinuation. The primary endpoint was total decitabine exposure over 5 days with oral decitabine-cedazuridine versus intravenous decitabine for cycles 1 and 2, measured as area under the curve in participants who received the full treatment dose in cycles 1 and 2 and had decitabine daily AUC0-24 for both oral decitabine-cedazuridine and intravenous decitabine (ie, paired cycles). On completion of the study, all patients were rolled over to a maintenance study. This study is registered with ClinicalTrials.gov, NCT03306264., Findings: Between Feb 8, 2018, and June 7, 2021, 173 individuals were screened, 138 (80%) participants were randomly assigned to a treatment sequence, and 133 (96%) participants (87 [65%] men and 46 [35%] women; 121 [91%] White, four [3%] Black or African-American, three [2%] Asian, and five [4%] not reported) received treatment. Median follow-up was 966 days (IQR 917-1050). Primary endpoint of total exposure of oral decitabine-cedazuridine versus intravenous decitabine was 98·93% (90% CI 92·66-105·60), indicating equivalent pharmacokinetic exposure on the basis of area under the curve. The safety profiles of oral decitabine-cedazuridine and intravenous decitabine were similar. The most frequent adverse events of grade 3 or worse were thrombocytopenia (81 [61%] of 133 participants), neutropenia (76 [57%] participants), and anaemia (67 [50%] participants). The incidence of serious adverse events in cycles 1-2 was 31% (40 of 130 participants) with oral decitabine-cedazuridine and 18% (24 of 132 participants) with intravenous decitabine. There were five treatment-related deaths; two deemed related to oral therapy (sepsis and pneumonia) and three to intravenous treatment (septic shock [n=2] and pneumonia [n=1])., Interpretation: Oral decitabine-cedazuridine was pharmacologically and pharmacodynamically equivalent to intravenous decitabine. The results support use of oral decitabine-cedazuridine as a safe and effective alternative to intravenous decitabine for treatment of individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia., Funding: Astex Pharmaceuticals., Competing Interests: Declaration of interests GG-M has consulted for Acceleron; received honoraria and research funding from AbbVie, Astex, Bristol Myers Squibb, Curis, Genentech, and Novartis; received honoraria from Aprea; and received research funding from Gilead. JMc has received research funding from Astex; consulted for AbbVie, Apellis, BMS, CTI BioPharma, Novartis, Pfizer, and Takeda; received honoraria from BMS, Blueprint, Incyte, Jazz, Novartis, Stemline, and Takeda; and is an equity holder in COTA Healthcare. EAG has received research funding from Astex, Blueprint, and Celldex; received research funding from and consulted and served on advisory committees for Alexion and BMS/Celgene; consulted for and served on advisory committees for AbbVie, Apellis, AstraZeneca, CTI BioPharma, Genentech, Novartis, Taiho, and Takeda; and received honoraria from Aplastic Anemia and MDS International Foundation, Medicom, Physician Educational Resource, and Picnic Health. KWLY has consulted for BMS/Celgene, GSK, Jazz, Novartis, Pfizer, Shattuck, Taiho, and Takeda; received research funding from Astex, Forma, Genentech, Geron, Gilead, Janssen, Jazz, Novartis, Roche, Pfizer, and Treadwell; and received honoraria from AbbVie and Novartis. AMZ has received research funding from AbbVie, ADC Therapeutics, Amgen, Aprea, Astex, AstraZeneca, BMS, Boehringer-Ingelheim, Cardiff Oncology, Celgene, Incyte, Medimmune, Novartis, Otsuka, Pfizer, Takeda, and Trovagene; consulted for AbbVie, Acceleron, Agios, Amgen, Aprea, Astellas, BeyondSpring, BMS, Boehringer-Ingelheim, Cardiff, Cardinal Health, Celgene, Daiichi Sankyo, Epizyme, Geron, Gilead, Incyte, Ionis, Janssen, Jazz, Kura, Novartis, Pfizer, Seattle Genetics, Syndax, Taiho, Takeda, Trovagene, and Tyme; received honoraria from AbbVie, Acceleron, Agios, Amgen, Aprea, Astellas, BeyondSpring, BMS, Boehringer-Ingelheim, Cardiff, Cardinal, Celgene, Daiichi Sankyo, Epizyme, Geron, Gilead, Incyte, Ionis, Janssen, Jazz, Kura, Novartis, Otsuka, Pfizer, Seattle Genetics, Syndax, Taiho, Takeda, Trovagene, and Tyme; received travel expenses from Cardiff, Novartis, and Pfizer; and had leadership roles in boards or committees for AbbVie, BMS, Celgene, Geron, Gilead, Kura, and Novartis. AA-K's institution has received research support from Astex. HJD, NZ, NYG, JMa, HA, and JR declare no competing interests. PAP has served on an advisory committee for Agios. MS has served on advisory committees for AbbVie, BMS, Celgene, Jazz, Novartis, Pfizer, Roche, and Taiho and served on advisory committees for and received research funding from Actinium and Astellas. M-MK has served on advisory boards for AstraZeneca, BeiGene, BMS, Seattle Genetics, and Taiho. SF has consulted for and received honoraria from Blueprint, CTI, Gilead, GSK, Incyte, Janssen, Jazz, Karyopharm, Novartis, PharmaEssentia, Sanofi, Servier, Stemline, Taiho, and Takeda. OO has consulted for AbbVie, BMS/Celgene, CTI, Impact, Novartis, and Taiho and his institution has received research funding from AbbVie, Agios, Aprea, Astex, AstraZeneca, BMS/Celgene, CTI, Incyte, Janssen, Kartos, NS Pharma, and OncoTherapy. HK has received grants from AbbVie, Amgen, Ascentage, BMS, Daiichi-Sankyo, Immunogen, Jazz, and Novartis and received honoraria from AbbVie, Amgen, Amphista, Ascentage, Astellas, Biologix, Curis, Ipsen, KAHR, Labcorp, Novartis, Pfizer, Shenzhen Target Rx, Stemline, and Takeda. AED has consulted for Taiho; received honoraria from BMS, Novartis, and Gilead; served as chair of a data safety monitoring board for Geron; and is the 2023 American Society of Hematology Education Chair. CLO has received research funding from Astex and Genentech. GJR has consulted or served on advisory boards or data and safety monitoring committees for AbbVie, Actiunuim, Agios, Amgen, Astellas, AstraZeneca, Bluebird, Blueprint, Bluebird Bio, BMS, Catamaran, Celgene, Daiichi, GSK, Helsinn, Janssen, Jasper, Jazz, Mesoblast, Novartis, Pfizer, Roche, Syndax, Takeda, and Trovagene and received research support from Janssen. LB has consulted for Novartis. RB has received honoraria and research funding from BMS and Taiho; received research funding from Takeda; and received honoraria from AbbVie. BL has received honoraria and served on advisory committees and speakers bureaus for AbbVie, Amgen, and BMS and consulted for, received honoraria, and served on advisory committees and speakers bureaus for Novartis and Pfizer. AS has consulted for and received research funding from Kymera, consulted for Janssen and Rigel, and received honoraria from National Association for Continuing Education. K-HD, AO, and HNK are employed by Astex. YH and MA were employed in the past 24 months by and consult for Astex. MRS has consulted and served on advisory boards for and received travel expenses from BMS, CTI, Forma, Geron, GSK, and Taiho; received travel expenses from Astex; is an equity holder in and served on advisory committees for Karyopharm and Ryvu; and received research funding from ALX Oncology, Astex, Incyte, and Takeda., (Copyright © 2024 Elsevier Ltd. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
35. LAIR-1 agonism as a therapy for acute myeloid leukemia.
- Author
-
Lovewell RR, Hong J, Kundu S, Fielder CM, Hu Q, Kim KW, Ramsey HE, Gorska AE, Fuller LS, Tian L, Kothari P, Paucarmayta A, Mason EF, Meza I, Manzanarez Y, Bosiacki J, Maloveste K, Mitchell N, Barbu EA, Morawski A, Maloveste S, Cusumano Z, Patel SJ, Savona MR, Langermann S, Myint H, Flies DB, and Kim TK
- Subjects
- Animals, Humans, Hematopoietic Stem Cells metabolism, Signal Transduction, Disease Models, Animal, Neoplastic Stem Cells metabolism, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute metabolism
- Abstract
Effective eradication of leukemic stem cells (LSCs) remains the greatest challenge in treating acute myeloid leukemia (AML). The immune receptor LAIR-1 has been shown to regulate LSC survival; however, the therapeutic potential of this pathway remains unexplored. We developed a therapeutic LAIR-1 agonist antibody, NC525, that induced cell death of LSCs, but not healthy hematopoietic stem cells in vitro, and killed LSCs and AML blasts in both cell- and patient-derived xenograft models. We showed that LAIR-1 agonism drives a unique apoptotic signaling program in leukemic cells that was enhanced in the presence of collagen. NC525 also significantly improved the activity of azacitidine and venetoclax to establish LAIR-1 targeting as a therapeutic strategy for AML that may synergize with standard-of-care therapies.
- Published
- 2023
- Full Text
- View/download PDF
36. Cost-effective and scalable clonal hematopoiesis assay provides insight into clonal dynamics.
- Author
-
Mack T, Vlasschaert C, von Beck K, Silver AJ, Heimlich JB, Poisner H, Condon HR, Ulloa J, Sochacki AL, Spaulding TP, Kishtagari A, Bejan CA, Xu Y, Savona MR, Jones A, and Bick A
- Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is a common age-related phenomenon that occurs when hematopoietic stem cells acquire mutations in a select set of genes commonly mutated in myeloid neoplasia which then expand clonally. Current sequencing assays to detect CHIP are not optimized for the detection of these variants and can be cost-prohibitive when applied to large cohorts or serial sequencing. Here, we present and validate a CHIP targeted sequencing assay that is affordable (∼$8/sample), accurate and highly scalable. To demonstrate the utility of this assay, we detected CHIP in a cohort of 456 individuals with DNA collected at multiple timepoints in the Vanderbilt BioVU biobank and quantified clonal expansion rates over time. A total of 101 individuals with CHIP were identified, and individual-level clonal expansion rate was calculated using the variant allele fraction (VAF) at both timepoints. Differences in clonal expansion rate by driver gene were observed, but there was also significant individual-level heterogeneity, emphasizing the multifactorial nature of clonal expansion. We further describe the mutation co-occurrence and clonal competition between multiple driver mutations.
- Published
- 2023
- Full Text
- View/download PDF
37. Classification, risk stratification and response assessment in myelodysplastic syndromes/neoplasms (MDS): A state-of-the-art report on behalf of the International Consortium for MDS (icMDS).
- Author
-
Stahl M, Bewersdorf JP, Xie Z, Porta MGD, Komrokji R, Xu ML, Abdel-Wahab O, Taylor J, Steensma DP, Starczynowski DT, Sekeres MA, Sanz G, Sallman DA, Roboz GJ, Platzbecker U, Patnaik MM, Padron E, Odenike O, Nimer SD, Nazha A, Majeti R, Loghavi S, Little RF, List AF, Kim TK, Hourigan CS, Hasserjian RP, Halene S, Griffiths EA, Gore SD, Greenberg P, Figueroa ME, Fenaux P, Efficace F, DeZern AE, Daver NG, Churpek JE, Carraway HE, Buckstein R, Brunner AM, Boultwood J, Borate U, Bejar R, Bennett JM, Wei AH, Santini V, Savona MR, and Zeidan AM
- Subjects
- Humans, Risk Assessment, Quality of Life, Prognosis, Neoplasms, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes therapy
- Abstract
The guidelines for classification, prognostication, and response assessment of myelodysplastic syndromes/neoplasms (MDS) have all recently been updated. In this report on behalf of the International Consortium for MDS (icMDS) we summarize these developments. We first critically examine the updated World Health Organization (WHO) classification and the International Consensus Classification (ICC) of MDS. We then compare traditional and molecularly based risk MDS risk assessment tools. Lastly, we discuss limitations of criteria in measuring therapeutic benefit and highlight how the International Working Group (IWG) 2018 and 2023 response criteria addressed these deficiencies and are endorsed by the icMDS. We also address the importance of patient centered care by discussing the value of quality-of-life assessment. We hope that the reader of this review will have a better understanding of how to classify MDS, predict clinical outcomes and evaluate therapeutic outcomes., Competing Interests: Declaration of Competing Interest Maximilian Stahl consulted for Curis Oncology and Boston Consulting; served on the advisory board for Novartis and Kymera, GSK, Rigel, Sierra Oncology; and participated in GME activity for Novartis, Curis Oncology, Haymarket Media and Clinical care options (CCO). Elizabeth A. Griffiths has received honoraria for advisory board membership from AbbVie, Alexion Pharmaceuticals, Apellis, Celgene/BMS, CTI Biopharma, Genentech, Novartis, Picnic Health, Takeda Oncology, Taiho Oncology. EAG has received research funding from Astex Pharmaceuticals, AstraZeneca Rare Disease, Alexion Pharmaceuticals, Apellis Pharmaceuticals, Blueprint Medicines, Genentech Inc., and honoraria for CME activities from Physicians Educational Resource, MediComWorldwide, American Society of Hematology, AAMDS International Foundation. Ravindra Majeti is on the Advisory Boards of Kodikaz Therapeutic Solutions, Syros Pharmaceuticals, TenSixteen Bio, Roche, and Cullgen Inc. and is an inventor on several patents related to CD47 cancer immunotherapy licensed to Gilead Sciences. R.M. receives research support from Gilead Sciences. Ravindra Majeti. is a co-founder and equity holder of Pheast Therapeutics, MyeloGene, and Orbital Therapeutics. Stephanie Halene consulted for Forma Therapeutics. Daniel T. Starczynowski is a consultant and received research funding from Kymera Therapeutics, Kurome Therapeutics, Captor Therapeutics, and Tolero Therapeutics. Daniel T. Starczynowski has equity in Kurome Therapeutics. Gail Roboz: Consultancy: Abbvie, Amgen, Argenx, Astra Zeneca, Bluebird Bio, Blueprint Medicines, Bristol-Myers Squibb, Caribou Biosciences, Celgene, Daiichi Sankyo, Ellipses Pharma, GlaxoSmithKline, Janssen, Jasper Pharmaceuticals, Jazz Pharmaceuticals, Molecular Partners, Novartis, Pfizer, Rigel, Roche, Syndax, Takeda (IRC Chair), Telix Pharma Research support: Janssen. David A. Sallman served on the advisory board of Aprea, AvenCell, BlueBird Bio, BMS, Intellia, Kite, Novartis, Shattuck Labs, Servier, Syndax. David A. Sallman served as a consultant for AbbVie, Magenta, Molecular Partners AG, Takeda and on the speakers' bureau for BMS, Incyte, Servier; David A. Sallman received research funding from Aprea, Jazz. Mrinal Patnaik received research funding from Kura Oncology and StemLine Pharmaceuticals. Andrew Brunner received consulting or advisory board honoraria from Novartis, Acceleron, Agios, Abbvie, Takeda, Celgene/BMS, Keros Therapeutics, Taiho, Gilead. Andrew Brunner has research support from the NIH SPORE in Myeloid Malignancies, and from the Edward P. Evans Foundation. Tae Kon Kim received research funding from Nextcure and is a consultant for Agenus. Alan List is employed by and has equity in Precision BioSciences, and has served as a consultant for Halia Therapeutics, CTI Biopharma, Aileron. Naval Daver has received research funding from Daiichi-Sankyo, Bristol-Myers Squibb, Pfizer, Gilead, Sevier, Genentech, Astellas, Daiichi-Sankyo, Abbvie, Hanmi, Trovagene, FATE therapeutics, Amgen, Novimmune, Glycomimetics, Trillium, and ImmunoGen and has served in a consulting or advisory role for Daiichi-Sankyo, Bristol-Myers Squibb, Arog, Pfizer, Novartis, Jazz, Celgene, AbbVie, Astellas, Genentech, Immunogen, Servier, Syndax, Trillium, Gilead, Amgen, Shattuck labs, and Agios. Guillermo Sanz received honoraria, advisory board membership or consultation fees from AbbVie, BMS, ExCellThera, Novartis, Roche, and Takeda and participated in sponsored speaker's bureau for BMS, Novartis, and Takeda. Mikkael A. Sekeres has served on advisory boards for BMS, Novartis, Kurome, and Gilead. Pierre Fenaux received research funding from BMS, Abbvie, Jazz Pharmaceuticals, Novartis, and Janssen. Pierre Fenaux had a consultancy with and received honoraria from BMS, Abbvie, Jazz Pharmaceuticals, and Novartis. Fabio Efficace had a consultancy or advisory role for AbbVie, Incyte, Janssen, and Syros, outside the submitted work. Omar Abdel-Wahab has served as a consultant for H3B Biomedicine, Foundation Medicine Inc., Merck, Prelude Therapeutics, and Janssen, and is on the Scientific Advisory Board of Envisagenics Inc., AIChemy, Harmonic Discovery Inc., and Pfizer Boulder; Omar Abdel-Wahab has received prior research funding from H3B Biomedicine and LOXO Oncology unrelated to the current manuscript. Andrew H.Wei has served on advisory boards for Novartis, Astra Zeneca, Astellas, Janssen, Jazz, Amgen, Roche, Pfizer, Abbvie, Servier, Gilead, BMS, Shoreline, Macrogenics, Novartis and Agios; receives research funding to the Institution from Novartis, Abbvie, Servier, Janssen, BMS, Syndax, Astex, Astra Zeneca, Amgen; serves on speaker's bureaus for Abbvie, Novartis, BMS, Servier, Astellas; Andrew H.Wei is an employee of the Walter and Eliza Hall Institute (WEHI). WEHI receives milestone and royalty payments related to the development of Venetoclax. Current and past employees of Walter and Eliza Hall Institute may be eligible for financial benefits related to these payments. Andrew H. Wei receives such a financial benefit. Amer M. Zeidan received research funding (institutional) from Celgene/BMS, Abbvie, Astex, Pfizer, Medimmune/AstraZeneca, Boehringer-Ingelheim, Cardiff oncology, Incyte, Takeda, Novartis, Aprea, and ADC Therapeutics. AMZ participated in advisory boards, and/or had a consultancy with and received honoraria from AbbVie, Otsuka, Pfizer, Celgene/BMS, Jazz, Incyte, Agios, Boehringer-Ingelheim, Novartis, Acceleron, Astellas, Daiichi Sankyo, Cardinal Health, Taiho, Seattle Genetics, BeyondSpring, Cardiff Oncology, Takeda, Ionis, Amgen, Janssen, Epizyme, Syndax, Gilead, Kura, Chiesi, ALX Oncology, BioCryst, Notable, Orum, and Tyme. AMZ served on clinical trial committees for Novartis, Abbvie, Gilead, BioCryst, Abbvie, ALX Oncology, Geron and Celgene/BMS. The National Heart, Lung, and Blood Institute receives research funding for the laboratory of Dr. Hourigan from Sellas and from the Foundation of the NIH AML MRD Biomarkers Consortium. All other authors have no conflicts of interest to disclose., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
38. Phase 1/2 Study of the Pan-PIM Kinase Inhibitor INCB053914 Alone or in Combination With Standard-of-Care Agents in Patients With Advanced Hematologic Malignancies.
- Author
-
Patel MR, Donnellan W, Byrne M, Asch AS, Zeidan AM, Baer MR, Fathi AT, Kuykendall AT, Zheng F, Walker C, Cheng L, Marando C, and Savona MR
- Subjects
- Animals, Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Agents adverse effects, Antineoplastic Agents therapeutic use, Hematologic Neoplasms drug therapy, Hematologic Neoplasms etiology, Leukemia, Myeloid, Acute drug therapy, Primary Myelofibrosis drug therapy
- Abstract
Background: The Proviral Integration site of Moloney murine leukemia virus (PIM) kinases are implicated in tumorigenesis; the pan-PIM kinase inhibitor, INCB053914, demonstrated antitumor activity in hematologic malignancy preclinical models., Patients and Methods: This phase 1/2 study evaluated oral INCB053914 alone or combined with standard-of-care agents for advanced hematologic malignancies (NCT02587598). In Parts 1/2 (monotherapy), patients (≥18 years) had acute leukemia, high-risk myelodysplastic syndrome (MDS), MDS/myeloproliferative neoplasm, myelofibrosis (MF), multiple myeloma, or lymphoproliferative neoplasms. In Parts 3/4 (combination therapy), patients had relapsed/refractory or newly diagnosed (≥65 years, unfit for intensive chemotherapy) acute myeloid leukemia (AML) or MF with suboptimal ruxolitinib response., Results: Parts 1/2 (n = 58): 6 patients experienced dose-limiting toxicities (DLTs), most commonly aspartate aminotransferase/alanine aminotransferase-elevated (AST/ALT; each n = 4). Fifty-seven patients (98.3%) had treatment-emergent adverse events (TEAEs), most commonly ALT-elevated and fatigue (36.2% each); 48 (82.8%) had grade ≥3 TEAEs, most commonly anemia (31.0%); 8 (13.8%) had grade ≥3 ALT/AST-elevated TEAEs. Parts 3/4 (n = 39): for INCB053914 + cytarabine (AML; n = 6), 2 patients experienced DLTs (grade 3 maculopapular rash, n = 1; grade 3 ALT-elevated and grade 4 hypophosphatemia, n = 1); for INCB053914 + azacitidine (AML; n = 16), 1 patient experienced a DLT (grade 3 maculopapular rash). Two complete responses were observed (1 with incomplete count recovery). For INCB053914 + ruxolitinib (MF; n = 17), no DLTs occurred; 3 patients achieved best reduction of >25% spleen volume at week 12 or 24., Conclusion: INCB053914 was generally well tolerated as monotherapy and in combinations; TEAEs were most commonly ALT/AST-elevated. Limited responses were observed with combinations. Future studies are needed to identify rational, effective combination strategies., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
39. Genomic Data Heterogeneity across Molecular Diagnostic Laboratories: A Real-World Connect Myeloid Disease Registry Perspective on Variabilities in Genomic Assay Methodology and Reporting.
- Author
-
Patel JL, Erba HP, Savona MR, Grinblatt DL, Clark M, Clive TC, Smart TB, Makinde AY, DeGutis IS, Yu E, Eggington JM, and George TI
- Subjects
- Humans, Prospective Studies, Pilot Projects, Genomics, Registries, Laboratories, Pathology, Molecular
- Abstract
Genomic data variability from laboratory reports can impact clinical decisions and population-level analyses; however, the extent of this variability and the impact on the data's value are not well characterized. This pilot study used anonymized genetic and genomic test reports from the Connect Myeloid Disease Registry (NCT01688011), a multicenter, prospective, observational cohort study of patients with newly diagnosed myelodysplastic syndromes, acute myeloid leukemia, or idiopathic cytopenia of undetermined significance, to analyze laboratory test variabilities and limitations. Results for 56 randomly selected patients enrolled in the Registry were independently extracted and evaluated (data cutoff, January 2020). Ninety-five reports describing 113 assay results from these 56 patients were analyzed for discrepancies. Almost all assay results [101 (89%)] identified the sequencing technology applied, and 94 (83%) described the test limitations; 95 (84%) described the limits of detection, but none described the limit of blank for detecting false positives. RNA transcript identifiers were not provided for 20 (43%) variants analyzed by next-generation sequencing and reported by the same laboratory. Of 42 variants with variant allele frequencies ≥30%, 16 (38%) of the variants did not have report text indicating that the variants might be germline. Variabilities and lack of standardization present challenges for incorporating this information into clinical care and render data collation ineffective and unreliable for large-scale use in centralized databases for therapeutic discovery., (Copyright © 2023 Association for Molecular Pathology and American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
40. PheMIME: An Interactive Web App and Knowledge Base for Phenome-Wide, Multi-Institutional Multimorbidity Analysis.
- Author
-
Zhang S, Strayer N, Vessels T, Choi K, Wang GW, Li Y, Bejan CA, Hsi RS, Bick AG, Velez Edwards DR, Savona MR, Philips EJ, Pulley J, Self WH, Hopkins WC, Roden DM, Smoller JW, Ruderfer DM, and Xu Y
- Abstract
Motivation: Multimorbidity, characterized by the simultaneous occurrence of multiple diseases in an individual, is an increasing global health concern, posing substantial challenges to healthcare systems. Comprehensive understanding of disease-disease interactions and intrinsic mechanisms behind multimorbidity can offer opportunities for innovative prevention strategies, targeted interventions, and personalized treatments. Yet, there exist limited tools and datasets that characterize multimorbidity patterns across different populations. To bridge this gap, we used large-scale electronic health record (EHR) systems to develop the Phenome-wide Multi-Institutional Multimorbidity Explorer (PheMIME), which facilitates research in exploring and comparing multimorbidity patterns among multiple institutions, potentially leading to the discovery of novel and robust disease associations and patterns that are interoperable across different systems and organizations., Results: PheMIME integrates summary statistics from phenome-wide analyses of disease multimorbidities. These are currently derived from three major institutions: Vanderbilt University Medical Center, Mass General Brigham, and the UK Biobank. PheMIME offers interactive exploration of multimorbidity through multi-faceted visualization. Incorporating an enhanced version of associationSubgraphs, PheMIME enables dynamic analysis and inference of disease clusters, promoting the discovery of multimorbidity patterns. Once a disease of interest is selected, the tool generates interactive visualizations and tables that users can delve into multimorbidities or multimorbidity networks within a single system or compare across multiple systems. The utility of PheMIME is demonstrated through a case study on schizophrenia., Availability and Implementation: The PheMIME knowledge base and web application are accessible at https://prod.tbilab.org/PheMIME/. A comprehensive tutorial, including a use-case example, is available at https://prod.tbilab.org/PheMIME_supplementary_materials/. Furthermore, the source code for PheMIME can be freely downloaded from https://github.com/tbilab/PheMIME., Data Availability Statement: The data underlying this article are available in the article and in its online web application or supplementary material., Competing Interests: Competing Interest Statement The authors have declared no competing interest.
- Published
- 2023
- Full Text
- View/download PDF
41. PI3K Inhibition Restores and Amplifies Response to Ruxolitinib in Patients with Myelofibrosis.
- Author
-
Moyo TK, Kishtagari A, Villaume MT, McMahon B, Mohan SR, Stopczynski T, Chen SC, Fan R, Huo Y, Moon H, Tang Y, Bejan CA, Childress M, Anderson I, Rawling K, Simons RM, Moncrief A, Caza R, Dugger L, Collins A, Dudley CV, Ferrell PB, Byrne M, Strickland SA, Ayers GD, Landman BA, Mason EF, Mesa RA, Palmer JM, Michaelis LC, and Savona MR
- Subjects
- Humans, Aged, Phosphatidylinositol 3-Kinases, Pyrimidines therapeutic use, Nitriles therapeutic use, Primary Myelofibrosis drug therapy, Primary Myelofibrosis metabolism, Janus Kinase Inhibitors therapeutic use
- Abstract
Purpose: Treatment options are limited beyond JAK inhibitors for patients with primary myelofibrosis (MF) or secondary MF. Preclinical studies have revealed that PI3Kδ inhibition cooperates with ruxolitinib, a JAK1/2 inhibitor, to reduce proliferation and induce apoptosis of JAK2V617F-mutant cell lines., Patients and Methods: In a phase I dose-escalation and -expansion study, we evaluated the safety and efficacy of a selective PI3Kδ inhibitor, umbralisib, in combination with ruxolitinib in patients with MF who had a suboptimal response or lost response to ruxolitinib. Enrolled subjects were required to be on a stable dose of ruxolitinib for ≥8 weeks and continue that MTD at study enrollment. The recommended dose of umbralisib in combination with ruxolitinib was determined using a modified 3+3 dose-escalation design. Safety, pharmacokinetics, and efficacy outcomes were evaluated, and spleen size was measured with a novel automated digital atlas., Results: Thirty-seven patients with MF (median age, 67 years) with prior exposure to ruxolitinib were enrolled. A total of 2 patients treated with 800 mg umbralisib experienced reversible grade 3 asymptomatic pancreatic enzyme elevation, but no dose-limiting toxicities were seen at lower umbralisib doses. Two patients (5%) achieved a durable complete response, and 12 patients (32%) met the International Working Group-Myeloproliferative Neoplasms Research and Treatment response criteria of clinical improvement. With a median follow-up of 50.3 months for censored patients, overall survival was greater than 70% after 3 years of follow-up., Conclusions: Adding umbralisib to ruxolitinib in patients was well tolerated and may resensitize patients with MF to ruxolitinib without unacceptable rates of adverse events seen with earlier generation PI3Kδ inhibitors. Randomized trials testing umbralisib in the treatment of MF should be pursued., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
42. Current landscape of translational and clinical research in myelodysplastic syndromes/neoplasms (MDS): Proceedings from the 1 st International Workshop on MDS (iwMDS) Of the International Consortium for MDS (icMDS).
- Author
-
Bewersdorf JP, Xie Z, Bejar R, Borate U, Boultwood J, Brunner AM, Buckstein R, Carraway HE, Churpek JE, Daver NG, Porta MGD, DeZern AE, Fenaux P, Figueroa ME, Gore SD, Griffiths EA, Halene S, Hasserjian RP, Hourigan CS, Kim TK, Komrokji R, Kuchroo VK, List AF, Loghavi S, Majeti R, Odenike O, Patnaik MM, Platzbecker U, Roboz GJ, Sallman DA, Santini V, Sanz G, Sekeres MA, Stahl M, Starczynowski DT, Steensma DP, Taylor J, Abdel-Wahab O, Xu ML, Savona MR, Wei AH, and Zeidan AM
- Subjects
- Animals, Humans, Epigenomics, Cell- and Tissue-Based Therapy, Protein Processing, Post-Translational, Neoplasms, Myelodysplastic Syndromes etiology, Myelodysplastic Syndromes therapy
- Abstract
Biological events that contribute to the pathogenesis of myelodysplastic syndromes/neoplasms (MDS) are becoming increasingly characterized and are being translated into rationally designed therapeutic strategies. Herein, we provide updates from the first International Workshop on MDS (iwMDS) of the International Consortium for MDS (icMDS) detailing recent advances in understanding the genetic landscape of MDS, including germline predisposition, epigenetic and immune dysregulation, the complexities of clonal hematopoiesis progression to MDS, as well as novel animal models of the disease. Connected to this progress is the development of novel therapies targeting specific molecular alterations, the innate immune system, and immune checkpoint inhibitors. While some of these agents have entered clinical trials (e.g., splicing modulators, IRAK1/4 inhibitors, anti-CD47 and anti-TIM3 antibodies, and cellular therapies), none have been approved for MDS. Additional preclinical and clinical work is needed to develop a truly individualized approach to the care of MDS patients., Competing Interests: Declaration of Competing Interest Maximilian Stahl consulted for Curis Oncology and Boston Consulting; served on the advisory board for Novartis and Kymera; and participated in GME activity for Novartis, Curis Oncology, Haymarket Media and Clinical care options (CCO). Elizabeth A. Griffiths has received honoraria for advisory board membership from AbbVie, Alexion Pharmaceuticals, Apellis, Celgene/BMS, CTI Biopharma, Genentech, Novartis, Picnic Health, Takeda Oncology, Taiho Oncology. EAG has received research funding from Astex Pharmaceuticals, AstraZeneca Rare Disease, Alexion Pharmaceuticals, Apellis Pharmaceuticals, Blueprint Medicines, Genentech Inc., and honoraria for CME activities from Physicians Educational Resource, MediComWorldwide, American Society of Hematology, AAMDS International Foundation. Ravindra Majeti is on the Advisory Boards of Kodikaz Therapeutic Solutions, Syros Pharmaceuticals, TenSixteen Bio, Roche, and Cullgen Inc. and is an inventor on a number of patents related to CD47 cancer immunotherapy licensed to Gilead Sciences. R.M. receives research support from Gilead Sciences. Ravindra Majeti. is a co-founder and equity holder of Pheast Therapeutics, MyeloGene, and Orbital Therapeutics. Stephanie Halene consulted for Forma Therapeutics. Daniel T. Starczynowski is a consultant and received research funding from Kymera Therapeutics, Kurome Therapeutics, Captor Therapeutics, and Tolero Therapeutics. Daniel T. Starczynowski has equity in Kurome Therapeutics. David A. Sallman served on the advisory board of Aprea, AvenCell, BlueBird Bio, BMS, Intellia, Kite, Novartis, Shattuck Labs, Servier, Syndax. David A. Sallman served as a consultant for AbbVie, Magenta, Molecular Partners AG, Takeda and on the speakers' bureau for BMS, Incyte, Servier; David A. Sallman received research funding from Aprea, Jazz. Mrinal Patnaik received research funding from Kura Oncology and StemLine Pharmaceuticals. Andrew Brunner received consulting or advisory board honoraria from Novartis, Acceleron, Agios, Abbvie, Takeda, Celgene/BMS, Keros Therapeutics, Taiho, Gilead. Andrew Brunner has research support from the NIH SPORE in Myeloid Malignancies, and from the Edward P. Evans Foundation. Tae Kon Kim received research funding from Nextcure and is a consultant for Agenus. Alan List is employed by and has equity in Precision BioSciences, and has served as a consultant for Halia Therapeutics, CTI Biopharma, Aileron. Naval Daver has received research funding from Daiichi-Sankyo, Bristol-Myers Squibb, Pfizer, Gilead, Sevier, Genentech, Astellas, Daiichi-Sankyo, Abbvie, Hanmi, Trovagene, FATE therapeutics, Amgen, Novimmune, Glycomimetics, Trillium, and ImmunoGen and has served in a consulting or advisory role for Daiichi-Sankyo, Bristol-Myers Squibb, Arog, Pfizer, Novartis, Jazz, Celgene, AbbVie, Astellas, Genentech, Immunogen, Servier, Syndax, Trillium, Gilead, Amgen, Shattuck labs, and Agios. Guillermo Sanz received honoraria, advisory board membership or consultation fees from AbbVie, BMS, ExCellThera, Novartis, Roche, and Takeda and participated in sponsored speaker's bureau for BMS, Novartis, and Takeda. Mikkael A. Sekeres has served on advisory boards for BMS, Novartis, Kurome, and Gilead. Pierre Fenaux received research funding from BMS, Abbvie, Jazz Pharmaceuticals, Novartis, and Janssen. Pierre Fenaux had a consultancy with and received honoraria from BMS, Abbvie, Jazz Pharmaceuticals, and Novartis. Omar Abdel-Wahab has served as a consultant for H3B Biomedicine, Foundation Medicine Inc., Merck, Prelude Therapeutics, and Janssen, and is on the Scientific Advisory Board of Envisagenics Inc., AIChemy, Harmonic Discovery Inc., and Pfizer Boulder; Omar Abdel-Wahab has received prior research funding from H3B Biomedicine and LOXO Oncology unrelated to the current manuscript. Andrew H.Wei has served on advisory boards for Novartis, Astra Zeneca, Astellas, Janssen, Jazz, Amgen, Roche, Pfizer, Abbvie, Servier, Gilead, BMS, Shoreline, Macrogenics, Novartis and Agios; receives research funding to the Institution from Novartis, Abbvie, Servier, Janssen, BMS, Syndax, Astex, Astra Zeneca, Amgen; serves on speaker's bureaus for Abbvie, Novartis, BMS, Servier, Astellas; Andrew H.Wei is an employee of the Walter and Eliza Hall Institute (WEHI). WEHI receives milestone and royalty payments related to the development of Venetoclax. Current and past employees of Walter and Eliza Hall Institute may be eligible for financial benefits related to these payments. Andrew H.Wei receives such a financial benefit. Valeria Santini served in advisory boards from Abbvie, BMS, Geron, Gilead,Menarini, Novartis, Servier, Syros, ad received research support from BMS. Amer M. Zeidan received research funding (institutional) from Celgene/BMS, Abbvie, Astex, Pfizer, Medimmune/AstraZeneca, Boehringer-Ingelheim, Cardiff oncology, Incyte, Takeda, Novartis, Aprea, and ADC Therapeutics. AMZ participated in advisory boards, and/or had a consultancy with and received honoraria from AbbVie, Otsuka, Pfizer, Celgene/BMS, Jazz, Incyte, Agios, Boehringer-Ingelheim, Novartis, Acceleron, Astellas, Daiichi Sankyo, Cardinal Health, Taiho, Seattle Genetics, BeyondSpring, Cardiff Oncology, Takeda, Ionis, Amgen, Janssen, Epizyme, Syndax, Gilead, Kura, Chiesi, ALX Oncology, BioCryst, Notable, Orum, and Tyme. AMZ served on clinical trial committees for Novartis, Abbvie, Gilead, BioCryst, Abbvie, ALX Oncology, Geron and Celgene/BMS. The National Heart, Lung, and Blood Institute receives research funding for the laboratory of Dr. Hourigan from Sellas and from the Foundation of the NIH AML MRD Biomarkers Consortium. All other authors have no conflicts of interest to disclose., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
43. Transplantation Referral Patterns for Patients with Newly Diagnosed Higher-Risk Myelodysplastic Syndromes and Acute Myeloid Leukemia at Academic and Community Sites in the Connect® Myeloid Disease Registry: Potential Barriers to Care.
- Author
-
Tomlinson B, de Lima M, Cogle CR, Thompson MA, Grinblatt DL, Pollyea DA, Komrokji RS, Roboz GJ, Savona MR, Sekeres MA, Abedi M, Garcia-Manero G, Kurtin SE, Maciejewski JP, Patel JL, Revicki DA, George TI, Flick ED, Kiselev P, Louis CU, DeGutis IS, Nifenecker M, Erba HP, Steensma DP, and Scott BL
- Subjects
- Humans, Aged, Registries, Health Services Accessibility, Myelodysplastic Syndromes therapy, Leukemia, Myeloid, Acute therapy, Hematopoietic Stem Cell Transplantation
- Abstract
Hematopoietic stem cell transplantation (HCT) is indicated for patients with higher-risk (HR) myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Age, performance status, patient frailty, comorbidities, and nonclinical factors (eg, cost, distance to site) are all recognized as important clinical factors that can influence HCT referral patterns and patient outcomes; however, the proportion of eligible patients referred for HCT in routine clinical practice is largely unknown. This study aimed to assess patterns of consideration for HCT among patients with HR-MDS and AML enrolled in the Connect® Myeloid Disease Registry at community/government (CO/GOV)- or academic (AC)-based sites, as well as to identify factors associated with rates of transplantation referral. We assessed patterns of consideration for and completion of HCT in patients with HR-MDS and AML enrolled between December 12, 2013, and March 6, 2020, in the Connect Myeloid Disease Registry at 164 CO/GOV and AC sites. Registry sites recorded whether patients were considered for transplantation at baseline and at each follow-up visit. The following answers were possible: "considered potentially eligible," "not considered potentially eligible," or "not assessed." Sites also recorded whether patients subsequently underwent HCT at each follow-up visit. Rates of consideration for HCT between CO/GOV and AC sites were compared using multivariable logistic regression analysis with covariates for age and comorbidity. Among the 778 patients with HR-MDS or AML enrolled in the Connect Myeloid Disease Registry, patients at CO/GOV sites were less likely to be considered potentially eligible for HCT than patients at AC sites (27.9% versus 43.9%; P < .0001). Multivariable logistic regression analysis with factors for age (<65 versus ≥65 years) and ACE-27 comorbidity grade (<2 versus ≥2) showed that patients at CO/GOV sites were significantly less likely than those at AC sites to be considered potentially eligible for HCT (odds ratio, 1.6, 95% confidence interval, 1.1 to 2.4; P = .0155). Among patients considered eligible for HCT, 45.1% (65 of 144) of those at CO/GOV sites and 35.7% (41 of 115) of those at AC sites underwent transplantation (P = .12). Approximately one-half of all patients at CO/GOV (50.1%) and AC (45.4%) sites were not considered potentially eligible for HCT; the most common reasons were age at CO/GOV sites (71.5%) and comorbidities at AC sites (52.1%). Across all sites, 17.4% of patients were reported as not assessed (and thus not considered) for HCT by their treating physician (20.7% at CO/GOV sites and 10.7% at AC sites; P = .0005). These findings suggest that many patients with HR-MDS and AML who may be candidates for HCT are not receiving assessment or consideration for transplantation in clinical practice. In addition, treatment at CO/GOV sites and age remain significant barriers to ensuring that all potentially eligible patients are assessed for HCT., (Copyright © 2023 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
44. An agenda to advance research in myelodysplastic syndromes: a TOP 10 priority list from the first international workshop in MDS.
- Author
-
Stahl M, Abdel-Wahab O, Wei AH, Savona MR, Xu ML, Xie Z, Taylor J, Starczynowski D, Sanz GF, Sallman DA, Santini V, Roboz GJ, Patnaik MM, Padron E, Odenike O, Nazha A, Nimer SD, Majeti R, Little RF, Gore S, List AF, Kutchroo V, Komrokji RS, Kim TK, Kim N, Hourigan CS, Hasserjian RP, Halene S, Griffiths EA, Greenberg PL, Figueroa M, Fenaux P, Efficace F, DeZern AE, Della Porta MG, Daver NG, Churpek JE, Carraway HE, Brunner AM, Borate U, Bennett JM, Bejar R, Boultwood J, Loghavi S, Bewersdorf JP, Platzbecker U, Steensma DP, Sekeres MA, Buckstein RJ, and Zeidan AM
- Subjects
- Humans, Chromosome Aberrations, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes therapy
- Published
- 2023
- Full Text
- View/download PDF
45. Targeting RARA overexpression with tamibarotene, a potent and selective RARα agonist, is a novel approach in AML.
- Author
-
de Botton S, Cluzeau T, Vigil C, Cook RJ, Rousselot P, Rizzieri DA, Liesveld JL, Fenaux P, Braun T, Banos A, Jurcic JG, Sekeres MA, Savona MR, Roboz GJ, Bixby D, Madigan K, Volkert A, Stephens K, Kang-Fortner Q, Baker K, Paul S, McKeown M, Carulli J, Eaton M, Hodgson G, Fiore C, Kelly MJ, Roth DA, and Stein EM
- Subjects
- Humans, Retinoic Acid Receptor alpha, Azacitidine adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute chemically induced, Myelodysplastic Syndromes drug therapy, Leukemia, Promyelocytic, Acute drug therapy
- Abstract
A superenhancer at the retinoic acid receptor alpha (RARA) gene is associated with RARA mRNA overexpression in ∼30% of non-acute promyelocytic leukemia acute myeloid leukemia (AML) and in ∼50% of myelodysplastic syndromes (MDS). RARA overexpression is an actionable target for treatment with tamibarotene, an oral potent and selective RARα agonist. Sensitivity to the RARα agonist tamibarotene was demonstrated in RARA-high but not RARA-low preclinical AML models. The combination of oral tamibarotene plus azacitidine was evaluated in a phase 2 clinical study in 51 newly diagnosed unfit patients with AML identified as RARA-positive (n = 22) or RARA-negative (n = 29) for RARA mRNA overexpression in peripheral blasts using a blood-based biomarker test. In 18 response-evaluable RARA-positive patients, complete remission (CR)/CR with incomplete hematologic recovery rate was 61%, CR rate was 50%, and time to initial composite CR was rapid at 1.2 months. Transfusion independence was attained by 72% of RARA-positive patients. In contrast, 28 response-evaluable RARA-negative patients had response rates that were consistent with azacitidine monotherapy. Tamibarotene in combination with azacitidine was well tolerated. The majority of nonhematologic adverse events were low grade and hematologic adverse events were comparable to single-agent azacitidine, demonstrating that there was no additional myelosuppression when tamibarotene was combined with azacitidine. These results support further evaluation of tamibarotene-based treatment strategies in patients with AML or MDS with RARA overexpression to provide a targeted approach with the goal of improving patient outcomes. This trial was registered at www.clinicaltrials.gov as #NCT02807558., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
46. Patient-specific comorbidities as prognostic variables for survival in myelofibrosis.
- Author
-
Sochacki AL, Bejan CA, Zhao S, Patel A, Kishtagari A, Spaulding TP, Silver AJ, Stockton SS, Pugh K, Dorand RD, Bhatta M, Strayer N, Zhang S, Snider CA, Stricker T, Nazha A, Bick AG, Xu Y, and Savona MR
- Subjects
- Humans, Prognosis, Proportional Hazards Models, Risk Factors, DNA, Primary Myelofibrosis diagnosis, Primary Myelofibrosis epidemiology, Primary Myelofibrosis genetics
- Abstract
Treatment decisions in primary myelofibrosis (PMF) are guided by numerous prognostic systems. Patient-specific comorbidities have influence on treatment-related survival and are considered in clinical contexts but have not been routinely incorporated into current prognostic models. We hypothesized that patient-specific comorbidities would inform prognosis and could be incorporated into a quantitative score. All patients with PMF or secondary myelofibrosis with available DNA and comprehensive electronic health record (EHR) data treated at Vanderbilt University Medical Center between 1995 and 2016 were identified within Vanderbilt's Synthetic Derivative and BioVU Biobank. We recapitulated established PMF risk scores (eg, Dynamic International Prognostic Scoring System [DIPSS], DIPSS plus, Genetics-Based Prognostic Scoring System, Mutation-Enhanced International Prognostic Scoring System 70+) and comorbidities through EHR chart extraction and next-generation sequencing on biobanked peripheral blood DNA. The impact of comorbidities was assessed via DIPSS-adjusted overall survival using Bonferroni correction. Comorbidities associated with inferior survival include renal failure/dysfunction (hazard ratio [HR], 4.3; 95% confidence interval [95% CI], 2.1-8.9; P = .0001), intracranial hemorrhage (HR, 28.7; 95% CI, 7.0-116.8; P = 2.83e-06), invasive fungal infection (HR, 41.2; 95% CI, 7.2-235.2; P = 2.90e-05), and chronic encephalopathy (HR, 15.1; 95% CI, 3.8-59.4; P = .0001). The extended DIPSS model including all 4 significant comorbidities showed a significantly higher discriminating power (C-index 0.81; 95% CI, 0.78-0.84) than the original DIPSS model (C-index 0.73; 95% CI, 0.70-0.77). In summary, we repurposed an institutional biobank to identify and risk-classify an uncommon hematologic malignancy by established (eg, DIPSS) and other clinical and pathologic factors (eg, comorbidities) in an unbiased fashion. The inclusion of comorbidities into risk evaluation may augment prognostic capability of future genetics-based scoring systems., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
47. Nanoparticle STING Agonist Reprograms the Bone Marrow to an Antitumor Phenotype and Protects Against Bone Destruction.
- Author
-
Florian DC, Bennett NE, Odziomek M, Baljon JJ, Wehbe M, Merkel AR, Fischer MA, Savona MR, Rhoades JA, Guelcher SA, and Wilson JT
- Subjects
- Mice, Animals, Bone Marrow, Cytokines, Phenotype, Tumor Microenvironment, Neoplasms, Nanoparticles
- Abstract
When breast cancer metastasizes to bone, treatment options are limited. Failure to treat bone metastases is thought to be due to therapy-resistant features of the bone marrow microenvironment. Using a murine model of bone metastatic mammary carcinoma, we demonstrate that systemic delivery of polymer nanoparticles loaded with cyclic dinucleotide (CDN) agonists of stimulator of interferon genes (STING) inhibited tumor growth and bone destruction after 7 days of treatment. Each dose of STING-activating nanoparticles trafficked to the bone marrow compartment and was retained within the tumor microenvironment for over 24 hours, enhancing antitumor immunity through proinflammatory cytokine production and early T-cell activation. While acquired resistance mechanisms, including increased levels of immunosuppressive cytokines and the infiltration of regulatory T cells, ultimately limited antitumor efficacy after 2 weeks of treatment, bone protective effects remained. Overall, these studies demonstrate that STING pathway activation, here enabled using a nanomedicine approach to enhance CDN delivery to bone metastatic sites, can reprogram the immune contexture of the bone marrow to an antitumor phenotype that inhibits bone colonization of metastatic breast cancer cells and protects from tumor-mediated bone destruction., Significance: Bone metastases are difficult to treat due to the inaccessibility of the bone marrow compartment and the immunosuppressive microenvironment that protects resident stem cells. Packaging a STING agonist into a nanoparticle that enables systemic administration and drug accumulation at tumor sites overcomes both barriers to stymie metastatic breast cancer growth., Competing Interests: N.E. Bennett reports grants from NIH during the conduct of the study. A.R. Merkel reports grants from Verteran's Administration and non-financial support from Vanderbilt University Medical Center during the conduct of the study; grants from NIH Clinical and Translational Science Award and National Center for Advancing Translational Sciences outside the submitted work. M.A. Fischer reports grants from Incyte Corporation during the conduct of the study. M.R. Savona reports grants from ALX Oncology, Astex, Incyte, Takeda, TG Therapeutics; personal fees from BMS, CTI, Forma, Geron, Novartis, Sierra Oncology, Taiho; personal fees and other from Ryvu and Karyopharm outside the submitted work. J.A. Rhoades reports grants from Veterans Administration during the conduct of the study. S.A. Guelcher reports grants from Incyte Corporation during the conduct of the study. J.T. Wilson reports a patent to 10,696,985 issued and a patent to PCT/US2019/058945 pending. No disclosures were reported by the other authors., (© 2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
48. Selective inhibition of MCL1 overcomes venetoclax resistance in a murine model of myelodysplastic syndromes.
- Author
-
Fischer MA, Song Y, Arrate MP, Gbyli R, Villaume MT, Smith BN, Childress MA, Stricker TP, Halene S, and Savona MR
- Subjects
- Animals, Mice, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Proto-Oncogene Proteins c-bcl-2, Disease Models, Animal, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Apoptosis, Cell Line, Tumor, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes metabolism
- Abstract
Treatment for myelodysplastic syndromes (MDS) remains insufficient due to clonal heterogeneity and lack of effective clinical therapies. Dysregulation of apoptosis is observed across MDS subtypes regardless of mutations and represents an attractive therapeutic opportunity. Venetoclax (VEN), a selective inhibitor of anti-apoptotic protein B-cell lymphoma- 2 (BCL2), has yielded impressive responses in older patients with acute myeloid leukemia (AML) and high risk MDS. BCL2 family anti-apoptotic proteins BCL-XL and induced myeloid cell leukemia 1 (MCL1) are implicated in leukemia survival, and upregulation of MCL1 is seen in VEN-resistant AML and MDS. We determined in vitro sensitivity of MDS patient samples to selective inhibitors of BCL2, BCL-XL and MCL1. While VEN response positively correlated with MDS with excess blasts, all MDS subtypes responded to MCL1 inhibition. Treatment with combined VEN + MCL1 inhibtion was synergistic in all MDS subtypes without significant injury to normal hematopoiesis and reduced MDS engraftment in MISTRG6 mice, supporting the pursuit of clinical trials with combined BCL2 + MCL1 inhibition in MDS.
- Published
- 2023
- Full Text
- View/download PDF
49. Interactive network-based clustering and investigation of multimorbidity association matrices with associationSubgraphs.
- Author
-
Strayer N, Zhang S, Yao L, Vessels T, Bejan CA, Hsi RS, Shirey-Rice JK, Balko JM, Johnson DB, Phillips EJ, Bick A, Edwards TL, Velez Edwards DR, Pulley JM, Wells QS, Savona MR, Cox NJ, Roden DM, Ruderfer DM, and Xu Y
- Subjects
- Algorithms, Cluster Analysis, Phenomics, Software, Multimorbidity
- Abstract
Motivation: Making sense of networked multivariate association patterns is vitally important to many areas of high-dimensional analysis. Unfortunately, as the data-space dimensions grow, the number of association pairs increases in O(n2); this means that traditional visualizations such as heatmaps quickly become too complicated to parse effectively., Results: Here, we present associationSubgraphs: a new interactive visualization method to quickly and intuitively explore high-dimensional association datasets using network percolation and clustering. The goal is to provide an efficient investigation of association subgraphs, each containing a subset of variables with stronger and more frequent associations among themselves than the remaining variables outside the subset, by showing the entire clustering dynamics and providing subgraphs under all possible cutoff values at once. Particularly, we apply associationSubgraphs to a phenome-wide multimorbidity association matrix generated from an electronic health record and provide an online, interactive demonstration for exploring multimorbidity subgraphs., Availability and Implementation: An R package implementing both the algorithm and visualization components of associationSubgraphs is available at https://github.com/tbilab/associationsubgraphs. Online documentation is available at https://prod.tbilab.org/associationsubgraphs_info/. A demo using a multimorbidity association matrix is available at https://prod.tbilab.org/associationsubgraphs-example/., (© The Author(s) 2022. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
50. Acly Deficiency Enhances Myelopoiesis through Acetyl Coenzyme A and Metabolic-Epigenetic Cross-Talk.
- Author
-
Greenwood DL, Ramsey HE, Nguyen PTT, Patterson AR, Voss K, Bader JE, Sugiura A, Bacigalupa ZA, Schaefer S, Ye X, Dahunsi DO, Madden MZ, Wellen KE, Savona MR, Ferrell PB, and Rathmell JC
- Subjects
- Animals, Mice, Acetyl Coenzyme A genetics, Acetyl Coenzyme A metabolism, Chromatin metabolism, ATP Citrate (pro-S)-Lyase deficiency, ATP Citrate (pro-S)-Lyase genetics, Epigenesis, Genetic, Myelopoiesis genetics, Chromatin Assembly and Disassembly
- Abstract
Hematopoiesis integrates cytokine signaling, metabolism, and epigenetic modifications to regulate blood cell generation. These processes are linked, as metabolites provide essential substrates for epigenetic marks. In this study, we demonstrate that ATP citrate lyase (Acly), which metabolizes citrate to generate cytosolic acetyl-CoA and is of clinical interest, can regulate chromatin accessibility to limit myeloid differentiation. Acly was tested for a role in murine hematopoiesis by small-molecule inhibition or genetic deletion in lineage-depleted, c-Kit-enriched hematopoietic stem and progenitor cells from Mus musculus. Treatments increased the abundance of cell populations that expressed the myeloid integrin CD11b and other markers of myeloid differentiation. When single-cell RNA sequencing was performed, we found that Acly inhibitor-treated hematopoietic stem and progenitor cells exhibited greater gene expression signatures for macrophages and enrichment of these populations. Similarly, the single-cell assay for transposase-accessible chromatin sequencing showed increased chromatin accessibility at genes associated with myeloid differentiation, including CD11b, CD11c, and IRF8. Mechanistically, Acly deficiency altered chromatin accessibility and expression of multiple C/EBP family transcription factors known to regulate myeloid differentiation and cell metabolism, with increased Cebpe and decreased Cebpa and Cebpb. This effect of Acly deficiency was accompanied by altered mitochondrial metabolism with decreased mitochondrial polarization but increased mitochondrial content and production of reactive oxygen species. The bias to myeloid differentiation appeared due to insufficient generation of acetyl-CoA, as exogenous acetate to support alternate compensatory pathways to produce acetyl-CoA reversed this phenotype. Acly inhibition thus can promote myelopoiesis through deprivation of acetyl-CoA and altered histone acetylome to regulate C/EBP transcription factor family activity for myeloid differentiation., (Copyright © 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.