1
|
Adès L. Jumping barriers in clinical trials: release the brake. Blood 2025; 145:1335-1336. [PMID: 40146155 DOI: 10.1182/blood.2024027711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025] Open
Affiliation(s)
- Lionel Adès
- Hôpital Saint Louis, Université Paris Cité, and THEMA Institute
| |
Collapse
|
2
|
Jacoby MA, Duncavage ED, Khanna A, Chang GS, Nonavinkere Srivatsan S, Miller CA, Gao F, Robinson J, Shao J, Fulton RS, Fronick CC, O'Laughlin M, Heath SE, Brendel K, Chavez M, DiPersio JF, Abboud CN, Stockerl-Goldstein K, Westervelt P, Cashen A, Pusic I, Oh ST, Welch JS, Wells DA, Loken MR, Uy GL, Walter MJ. Monitoring clonal burden as an alternative to blast count for myelodysplastic neoplasm treatment response. Leukemia 2025; 39:166-177. [PMID: 39367170 DOI: 10.1038/s41375-024-02426-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024]
Abstract
Accurate assessment of therapy response in myelodysplastic neoplasm (MDS) has been challenging. Directly monitoring mutational disease burden may be useful, but is not currently included in MDS response criteria, and the correlation of mutational burden and traditional response endpoints is not completely understood. Here, we used genome-wide and targeted next-generation sequencing (NGS) to monitor clonal and subclonal molecular disease burden in 452 samples from 32 patients prospectively treated in a clinical trial. Molecular responses were compared with International Working Group (IWG) 2006-defined response assessments. We found that myeloblast percentage consistently underestimates MDS molecular disease burden and that mutational clearance patterns for marrow complete remission (mCR), which depends on myeloblast assessment, was not different than stable disease or bone marrow aplasia, underscoring a major limitation of using mCR. In contrast, achieving a complete remission (CR) was associated with the highest level of mutation clearance and lowest residual mutational burden in higher-risk MDS patients. A targeted gene panel approach was inferior to genome-wide sequencing in defining subclones and their molecular responses but may be adequate for monitoring molecular disease burden when a targeted gene is present in the founding clone. Our work supports incorporating serial NGS-based monitoring into prospective MDS clinical trials.
Collapse
Affiliation(s)
- Meagan A Jacoby
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Eric D Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Ajay Khanna
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Gue Su Chang
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | | | - Christopher A Miller
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Josh Robinson
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Jin Shao
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Robert S Fulton
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Catrina C Fronick
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Michelle O'Laughlin
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Sharon E Heath
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Kimberly Brendel
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Monique Chavez
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Camille N Abboud
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Keith Stockerl-Goldstein
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Peter Westervelt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- MaineHealth Cancer Center, Scarborough, ME, USA
| | - Amanda Cashen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Iskra Pusic
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Stephen T Oh
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - John S Welch
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- A2 Biotherapeutics Inc., Agoura Hills, CA, USA
| | | | | | - Geoffrey L Uy
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| | - Matthew J Walter
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
3
|
Armstrong EP, Franic DM, Malone DC, Mellors P, Pham SV, Masseria C, Mody L, Hogea C. Patient-centric care in myelodysplastic syndromes: A global systematic literature review and gap analysis. Leuk Res 2024; 147:107592. [PMID: 39341087 DOI: 10.1016/j.leukres.2024.107592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/19/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Disease progression and poor prognosis in higher-risk (HR) myelodysplastic syndrome (MDS) create an urgent need for interventions to improve the patient care experience in this vulnerable population. Patient-centric physician-supported strategies in conjunction with emerging therapies can help advance overall care and improve outcomes. The objective of this study was to evaluate patient-centric care (PCC) in the treatment of HR-MDS and identify opportunities to develop strategies to address care gaps for an optimal patient care experience. METHODS A global systematic literature review (SLR) was conducted by cross-referencing MDS/HR-MDS with PCC terms, using PubMed, Embase, and Cochrane Collaboration databases (2017-2022) in accordance with Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines. RESULTS In all, 59 MDS articles (45 empirical, 14 reviews) met the study inclusion criteria. Of these, 6 empirical articles focused on the HR-MDS population while none of the reviews did. Identified themes fell into 2 categories: health-related quality of life (HRQoL) and disparities. HRQoL was further categorized based on findings in the literature to include groupings of patient-reported outcomes (PROs), fatigue/frailty, and patient/preferences/treatment decisions/shared decision making (SDM). CONCLUSIONS With new treatments potentially on the horizon for HR-MDS, a call to action is timely to address the overall lack of empirical PCC data. The patient-centric approach presents critical opportunities for integration of physician-supported strategies with more effective first-line therapies to help optimize the journey of patients with HR-MDS and ensure meaningful outcomes by reducing patient/caregiver burden, aligning with and respecting patient preferences, and including patients as active participants in their treatment.
Collapse
Affiliation(s)
| | | | | | - Patrick Mellors
- Penn Medicine, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Sissi V Pham
- AESARA, Inc., PO Box 4266, Chapel Hill, NC 27517, USA.
| | | | - Lorie Mody
- AESARA, Inc., PO Box 4266, Chapel Hill, NC 27517, USA
| | | |
Collapse
|
4
|
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, Zeidan AM. 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). Hemasphere 2024; 8:e69. [PMID: 38774655 PMCID: PMC11106800 DOI: 10.1002/hem3.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/04/2024] [Accepted: 04/01/2024] [Indexed: 05/24/2024] Open
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.
Collapse
Affiliation(s)
- Fabio Efficace
- Italian Group for Adult Hematologic Diseases (GIMEMA), Health Outcomes Research UnitGIMEMA Data CenterRomeItaly
| | - Rena Buckstein
- Department of Medical Oncology/HematologySunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Gregory A. Abel
- Divisions of Population Sciences and Hematologic MalignanciesDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | | | - Pierre Fenaux
- Hôpital Saint LouisAssistance Publique Hôpitaux de Paris and Paris Cité UniversityParisFrance
| | - Jan Philipp Bewersdorf
- Leukemia Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Andrew M. Brunner
- Leukemia Program, Harvard Medical SchoolMassachusetts General Hospital Cancer CenterBostonMassachusettsUSA
| | - Rafael Bejar
- Division of Hematology and Oncology, Moores Cancer CenterUC San DiegoLa JollaCaliforniaUSA
| | - Uma Borate
- Ohio State University Comprehensive Cancer Center/James Cancer HospitalOhio State UniversityColumbusOhioUSA
| | - Amy E. DeZern
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins HospitalBaltimoreMarylandUSA
| | - Peter Greenberg
- Department of Medicine, Division of Hematology, Cancer InstituteStanford University School of MedicineStanfordCaliforniaUSA
| | - Gail J. Roboz
- Weill Cornell Medical College and New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Michael R. Savona
- Department of Medicine, Division of Hematology/OncologyVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Francesco Sparano
- Italian Group for Adult Hematologic Diseases (GIMEMA), Health Outcomes Research UnitGIMEMA Data CenterRomeItaly
| | - Jacqueline Boultwood
- Blood Cancer UK Molecular Haematology Unit, Radcliffe Department of MedicineNuffield Division of Clinical Laboratory SciencesUniversity of OxfordOxfordUK
| | - Rami Komrokji
- Department of Malignant HematologyH. Lee Moffitt Cancer CenterTampaFloridaUSA
| | - David A. Sallman
- Department of Malignant HematologyH. Lee Moffitt Cancer CenterTampaFloridaUSA
| | - Zhuoer Xie
- Department of Malignant HematologyH. Lee Moffitt Cancer CenterTampaFloridaUSA
| | - Guillermo Sanz
- Health Research Institute La Fe, Valencia, SpainHospital Universitario y Politécnico La FeValenciaSpain
| | - Hetty E. Carraway
- Leukemia Program, Hematology and Medical OncologyTaussig Cancer Institute, Cleveland ClinicClevelandOhioUSA
| | - Justin Taylor
- Sylvester Comprehensive Cancer CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Stephen D. Nimer
- Sylvester Comprehensive Cancer CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Matteo Giovanni Della Porta
- Department of Biomedical SciencesIRCCS Humanitas Clinical and Research Center & Humanitas UniversityMilanItaly
| | - Valeria Santini
- Myelodysplastic Syndromes Unit, Department of Experimental and Clinical Medicine, Hematology, Azienda Ospedaliero Universitaria CareggiUniversity of FlorenceFlorenceItaly
| | - Maximilian Stahl
- Department of Medical OncologyDana‐Farber Cancer Institute and Harvard Medical SchoolBostonMassachusettsUSA
| | - Uwe Platzbecker
- Department of Hematology and Cellular TherapyUniversity Hospital LeipzigLeipzigGermany
| | - Mikkael A. Sekeres
- Sylvester Comprehensive Cancer CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal MedicineYale University School of Medicine and Yale Cancer CenterNew HavenConnecticutUSA
| |
Collapse
|
5
|
Zeidan AM, Ando K, Rauzy O, Turgut M, Wang MC, Cairoli R, Hou HA, Kwong YL, Arnan M, Meers S, Pullarkat V, Santini V, Malek K, Kiertsman F, Niolat J, Ramos PM, Menssen HD, Fenaux P, Miyazaki Y, Platzbecker U. Sabatolimab plus hypomethylating agents in previously untreated patients with higher-risk myelodysplastic syndromes (STIMULUS-MDS1): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Haematol 2024; 11:e38-e50. [PMID: 38065203 DOI: 10.1016/s2352-3026(23)00333-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Sabatolimab is an immunotherapy targeting T-cell immunoglobulin domain and mucin domain-3 (TIM-3), an immuno-myeloid regulator expressed on immune cells and leukaemic stem cells. In this trial, we compared the efficacy and safety of sabatolimab plus hypomethylating agent with placebo plus hypomethylating agents in previously untreated patients with higher-risk myelodysplastic syndromes. METHODS STIMULUS-MDS1 was a multicentre, randomised, double-blind, placebo-controlled, phase 2 study done at 54 investigational sites in 17 countries. Adult patients (aged ≥18 years) with intermediate-risk, high-risk, and very high-risk myelodysplastic syndromes (according to Revised International Prognostic Scoring System criteria) who had not received previous treatment were included. Patients were randomly assigned (1:1) to intravenous sabatolimab (400 mg on day 8 and 22) or placebo plus a hypomethylating agent (intravenous decitabine 20 mg/m2 on day 1-5 or intravenous or subcutaneous azacitidine 75 mg/m2 on day 1-7 or day 1-5 and day 8 and 9) every 28 days until treatment discontinuation. The two primary endpoints were complete response rate and progression-free survival, assessed in the full analysis set, which included all randomly assigned patients. Complete response was analysed, as prespecified, 7 months after the last patient was randomly assigned. All other analyses presented, including progression-free survival, were done at the final data cutoff prespecified via a protocol amendment on Sept 2, 2021. Safety was assessed in in all patients who received at least one dose of study treatment. This study is registered with ClinicalTrials.gov, NCT03946670, and is ongoing. FINDINGS Between July 29, 2019, and Aug 10, 2020, 127 patients were randomly assigned to sabatolimab plus a hypomethylating agent group (sabatolimab group; n=65) or placebo plus a hypomethylating agent (placebo group; n=62). The median age of participants was 73 years (IQR 69-77), of whom 86 (68%) of 127 patients were male and 77 (61%) were White. The primary endpoints were not met. Complete response (cutoff date of March 10, 2021) was achieved in 14 (22%; 95% CI 12·3-33·5) of 65 patients in the sabatolimab group vs 11 (18%; 9·2-29·5) of 62 patients in the placebo group (p=0·77). At the cutoff date of the final analysis (March 1, 2022), median follow-up for progression-free survival was 17·8 months (IQR 16·6-19·4) in the sabatolimab group and 19·2 months (17·7-22·3) in the placebo group, and the median progression-free survival was 11·1 months (95% CI 7·6-17·6) in the sabatolimab group vs 8·5 months (6·9-11·3) in the placebo group (hazard ratio 0·75 [95% CI 0·48-1·17]; p=0·1022). The most common adverse events of any grade were neutropenia (35 [56%] of 62 patients in the sabatolimab group vs 43 [68%] of 63 patients in the placebo group), thrombocytopenia (30 [48%] vs 32 [51%]), constipation (29 [47%] vs 24 [38%]), diarrhoea (27 [44%] vs 14 [22%]), anaemia (22 [35%] vs 34 [54%]), febrile neutropenia (22 [35%] vs 15 [24%]), and leukopenia (15 [24%] vs 20 [32%]). One patient developed a serious potential treatment-related immune-mediated adverse event in the sabatolimab group. There was one treatment-related death in the sabatolimab group due to pneumonitis. INTERPRETATION The addition of sabatolimab to hypomethylating agents in this study did not result in a significant improvement in complete response rates or progression-free survival. Sabatolimab had a manageable safety in most patients with higher-risk myelodysplastic syndromes. A randomised phase 3 trial is ongoing to assess the potential benefit of sabatolimab plus azacitidine on overall survival in this setting. FUNDING Novartis Pharmaceuticals.
Collapse
Affiliation(s)
- Amer M Zeidan
- Yale Cancer Center, Yale University, New Haven, CT, USA.
| | - Kiyoshi Ando
- Tokai University School of Medicine, Isehara, Japan
| | - Odile Rauzy
- Institut Universitaire du Cancer Toulouse Oncopole, Toulouse University Hospital, Toulouse, France
| | | | - Ming-Chung Wang
- Kaohsiung-Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Roberto Cairoli
- ASST Niguarda Hospital, Milan, Italy; Università degli Studi di Milano Bicocca, Milan, Italy
| | - Hsin-An Hou
- National Taiwan University Hospital, Taipei, Taiwan
| | - Yok-Lam Kwong
- University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Montserrat Arnan
- Institut Català d'Oncologia-Hospital Duran i Reynals, IDIBELL, Hospitalet Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | - Pierre Fenaux
- Hôpital Saint-Louis, Université Paris Cité, Paris, France
| | | | | |
Collapse
|
6
|
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, Zeidan AM. 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). Blood Rev 2023; 62:101128. [PMID: 37704469 DOI: 10.1016/j.blre.2023.101128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/31/2023] [Accepted: 08/16/2023] [Indexed: 09/15/2023]
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.
Collapse
Affiliation(s)
- Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhuoer Xie
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Matteo Giovanni Della Porta
- IRCCS Humanitas Clinical and Research Center & Humanitas University, Department of Biomedical Sciences, via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Rami Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Mina L Xu
- Departments of Pathology & Laboratory Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - Omar Abdel-Wahab
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin Taylor
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mikkael A Sekeres
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Guillermo Sanz
- Health Research Institute La Fe, Valencia, Spain; Hospital Universitario y Politécnico La Fe, Valencia, Spain; CIBERONC, IS Carlos III, Madrid, Spain
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Gail J Roboz
- Weill Cornell Medical College and New York Presbyterian Hospital, New York, NY, USA
| | | | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Olatoyosi Odenike
- Leukemia Program, University of Chicago Medicine and University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | - Stephen D Nimer
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Aziz Nazha
- Department of Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ravi Majeti
- Division of Hematology, Department of Medicine, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard F Little
- National Cancer Institute, Cancer Therapy Evaluation Program, Rockville, MD, USA
| | - Alan F List
- Precision BioSciences, Inc., Durham, NC, USA
| | - Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, and Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | | | - Steven D Gore
- National Cancer Institute, Cancer Therapy Evaluation Program, Rockville, MD, USA
| | - Peter Greenberg
- Division of Hematology, Department of Medicine, Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria E Figueroa
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Pierre Fenaux
- Hôpital Saint Louis, Assistance Publique Hôpitaux de Paris and Paris Cité University, Paris, France
| | - Fabio Efficace
- Italian Group for Adult Hematologic Diseases (GIMEMA), Health Outcomes Research Unit, Rome, Italy
| | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jane E Churpek
- Department of Hematology, Oncology, and Palliative Care, Carbone Cancer Center, The University of Wisconsin-Madison, Madison, WI, USA
| | - Hetty E Carraway
- Leukemia Program, Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rena Buckstein
- Department of Medical Oncology/ Hematology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Andrew M Brunner
- Leukemia Program, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Jacqueline Boultwood
- Blood Cancer UK Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Uma Borate
- Ohio State University Comprehensive Cancer Center/ James Cancer Hospital, Ohio State University, Columbus, OH, USA
| | - Rafael Bejar
- Division of Hematology and Oncology, Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - John M Bennett
- University of Rochester Medical Center, Department of Pathology and Laboratory Medical Center, Rochester, NY, USA
| | - Andrew H Wei
- Department of Haematology, Peter MacCallum Cancer Centre, Royal Melbourne Hospital, Walter and Eliza Hall Institute of Medical Research and University of Melbourne, Victoria, Australia
| | | | - Michael R Savona
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
7
|
Pleyer L, Vaisband M, Drost M, Pfeilstöcker M, Stauder R, Heibl S, Sill H, Girschikofsky M, Stampfl-Mattersberger M, Pichler A, Hartmann B, Petzer A, Schreder M, Schmitt CA, Vallet S, Melchardt T, Zebisch A, Pichler P, Zaborsky N, Machherndl-Spandl S, Wolf D, Keil F, Hasenauer J, Larcher-Senn J, Greil R. Cox proportional hazards deep neural network identifies peripheral blood complete remission to be at least equivalent to morphologic complete remission in predicting outcomes of patients treated with azacitidine-A prospective cohort study by the AGMT. Am J Hematol 2023; 98:1685-1698. [PMID: 37548390 DOI: 10.1002/ajh.27046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/07/2023] [Accepted: 07/16/2023] [Indexed: 08/08/2023]
Abstract
The current gold standard of response assessment in patients with myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia (CMML), and acute myeloid leukemia (AML) is morphologic complete remission (CR) and CR with incomplete count recovery (CRi), both of which require an invasive BM evaluation. Outside of clinical trials, BM evaluations are only performed in ~50% of patients during follow-up, pinpointing a clinical need for response endpoints that do not necessitate BM assessments. We define and validate a new response type termed "peripheral blood complete remission" (PB-CR) that can be determined from the differential blood count and clinical parameters without necessitating a BM assessment. We compared the predictive value of PB-CR with morphologic CR/CRi in 1441 non-selected, consecutive patients diagnosed with MDS (n = 522; 36.2%), CMML (n = 132; 9.2%), or AML (n = 787; 54.6%), included within the Austrian Myeloid Registry (aMYELOIDr; NCT04438889). Time-to-event analyses were adjusted for 17 covariates remaining in the final Cox proportional hazards (CPH) model. DeepSurv, a CPH neural network model, and permutation-based feature importance were used to validate results. 1441 patients were included. Adjusted median overall survival for patients achieving PB-CR was 22.8 months (95%CI 18.9-26.2) versus 10.4 months (95%CI 9.7-11.2) for those who did not; HR = 0.366 (95%CI 0.303-0.441; p < .0001). Among patients achieving CR, those additionally achieving PB-CR had a median adjusted OS of 32.6 months (95%CI 26.2-49.2) versus 21.7 months (95%CI 16.9-27.7; HR = 0.400 [95%CI 0.190-0.844; p = .0161]) for those who did not. Our deep neural network analysis-based findings from a large, prospective cohort study indicate that BM evaluations solely for the purpose of identifying CR/CRi can be omitted.
Collapse
Affiliation(s)
- Lisa Pleyer
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Marc Vaisband
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Manuel Drost
- Assign Data Management and Biostatistics GmbH, Innsbruck, Austria
| | - Michael Pfeilstöcker
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 3rd Medical Department for Hematology and Oncology, Hanusch Hospital, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University Vienna, Vienna, Austria
| | - Reinhard Stauder
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Sonja Heibl
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 4th Medical Department of Internal Medicine, Hematology, Internistic Oncology and Palliative Medicine, Klinikum Wels-Grieskirchen GmbH, Wels, Austria
| | - Heinz Sill
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Division of Hematology, Medical University of Graz, Graz, Austria
| | - Michael Girschikofsky
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 1st Medical Department, Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz GmbH Elisabethinen, Linz, Austria
| | - Margarete Stampfl-Mattersberger
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Internal Medicine 2, Wiener Gesundheitsverbund, Klinik Donaustadt, Vienna, Austria
| | - Angelika Pichler
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Internal Medicine, Hematology and Internal Oncology, LKH Hochsteiermark, Leoben, Austria
| | - Bernd Hartmann
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Internal Medicine 2, Landeskrankenhaus Feldkirch, Feldkirch, Austria
| | - Andreas Petzer
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Internal Medicine I: Medical Oncology and Hematology, Ordensklinikum Linz GmbH, Barmherzige Schwestern, Linz, Austria
| | - Martin Schreder
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 1st Department of Internal Medicine, Center for Oncology and Hematology, Wiener Gesundheitsverbund, Klinik Ottakring, Vienna, Austria
| | - Clemens A Schmitt
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Hematology and Internal Oncology, Kepler University Hospital, Johannes Kepler University, Linz, Austria
- Charité-University Medical Center, Molecular Cancer Research Center, Berlin, Germany
| | - Sonia Vallet
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- University Hospital Krems, Department of Internal Medicine 2, Karl Landsteiner Private University of Health Sciences, Krems, Austria
| | - Thomas Melchardt
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Armin Zebisch
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Division of Hematology, Medical University of Graz, Graz, Austria
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Petra Pichler
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Clinical Department for Internal Medicine, University Hospital St Poelten, Karl Landsteiner University of Health Sciences, St Poelten, Austria
| | - Nadja Zaborsky
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Laboratory of Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria
| | - Sigrid Machherndl-Spandl
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 1st Medical Department, Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz GmbH Elisabethinen, Linz, Austria
| | - Dominik Wolf
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Felix Keil
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 3rd Medical Department for Hematology and Oncology, Hanusch Hospital, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University Vienna, Vienna, Austria
| | - Jan Hasenauer
- Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | | | - Richard Greil
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
8
|
Zeidan AM, Giagounidis A, Sekeres MA, Xiao Z, Sanz GF, Hoef MV, Ma F, Hertle S, Santini V. STIMULUS-MDS2 design and rationale: a phase III trial with the anti-TIM-3 sabatolimab (MBG453) + azacitidine in higher risk MDS and CMML-2. Future Oncol 2023; 19:631-642. [PMID: 37083373 DOI: 10.2217/fon-2022-1237] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
Patients with higher-risk myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML) unfit for hematopoietic stem cell transplantation have poor outcomes. Novel therapies that provide durable benefit with favorable tolerability and clinically meaningful improvement in survival are needed. T-cell immunoglobulin domain and mucin domain-3 (TIM-3) is an immuno-myeloid regulator expressed on immune and leukemic stem cells in myeloid malignancies. Sabatolimab is a novel immunotherapy targeting TIM-3 with a potential dual mechanism of reactivating the immune system and directly targeting TIM-3+ leukemic blasts suppressing the growth of cancer cells. Here, we describe the aims and design of the phase III STIMULUS-MDS2 trial, which aims to demonstrate the potential for sabatolimab plus azacitidine to improve survival for patients with higher-risk MDS and CMML-2 (NCT04266301). Clinical Trial Registration: NCT04266301 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Amer M Zeidan
- Yale University & Yale Cancer Center, New Haven, CT 06510, USA
| | | | - Mikkael A Sekeres
- Division of Hematology, Sylvester Cancer Center, University of Miami, Miami, FL 33065, USA
| | - Zhijian Xiao
- Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin, 300020, China
| | - Guillermo F Sanz
- Hospital Universitario y Politécnico La Fe, Valencia, 46026, Spain
- Health Research Institute La Fe (IIS La Fe), Valencia, 46026, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, 28029, Spain
| | | | - Fei Ma
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| | | | - Valeria Santini
- MDS Unit, Hematology, University of Florence, Florence, 50121, Italy
| |
Collapse
|
9
|
Efficace F, Koinig K, Cottone F, Bowen D, Mittelman M, Sommer K, Langemeijer S, Culligan D, Filanovsky K, Storck M, Smith A, van Marrewijk C, Dugas M, Stojkov I, Siebert U, de Witte T, Stauder R. Raising the standards of patient‐centered outcomes research in myelodysplastic syndromes: Clinical utility and validation of the subscales of the QUALMS from the MDS‐RIGHT project. Cancer Med 2022; 12:7529-7539. [PMID: 36533415 PMCID: PMC10067097 DOI: 10.1002/cam4.5487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 09/30/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Clinical decision-making for patients with myelodysplastic syndromes (MDS) is challenging, and both disease and treatment effects heavily impact health-related quality of life (HRQoL) of these patients. Therefore, disease-specific HRQoL measures can be critical to harness the patient voice in MDS research. METHODS We report a prospective international validation study of the Quality of Life in Myelodysplasia Scale (QUALMS) with a main focus on providing information on the psychometric characteristics of its three subscales: physical burden (QUALMS-P), emotional burden (QUALMS-E), and benefit finding (QUALMS-BF). The analysis is based on patients enrolled from three European countries and Israel, participating to the MDS-RIGHT Project. The scale structure and psychometric properties of the QUALMS were assessed. RESULTS Overall, 270 patients with a median age of 74 years were analyzed and the majority of them (60.3%) had a low MDS-Comorbidity Index score. Results of the confirmatory factor analysis supported the underlying scale structure of the QUALMS, which, in addition to a total score, includes three subscales: QUALMS-P, QUALMS-E, and the QUALMS-BF. The QUALMS-P exhibited the highest Cronbach's alpha coefficients. Discriminant validity analysis indicated good results with the QUALMS-P and QUALMS-E distinguishing between patients with different performance status, comorbidity, anemia, and transfusion dependency status. No floor and ceiling effects were observed. Responsiveness to change analysis supported the validity of the measure. Patients with a hemoglobin (Hb) level of <11 g/dL at study entry, who subsequently showed an improvement in their Hb levels, also reported a mean score change of 9 and 8 points (scales ranging between 0 and 100) in the expected direction of the QUALMS-E and QUALMS-P, respectively. CONCLUSIONS Our study provides additional validation data on the QUALMS from the international MDS-RIGHT Project. The use of this disease-specific HRQoL measure may contribute to raise quality standards of patient-centered outcomes research in MDS.
Collapse
Affiliation(s)
- Fabio Efficace
- Italian Group for Adult Hematologic Diseases (GIMEMA)Data Center and Health Outcomes Research UnitRomeItaly
| | - Karin Koinig
- Department of Internal Medicine V (Hematology and Oncology)Medical University InnsbruckInnsbruckAustria
| | - Francesco Cottone
- Italian Group for Adult Hematologic Diseases (GIMEMA)Data Center and Health Outcomes Research UnitRomeItaly
| | - David Bowen
- St. James's Institute of Oncology, Leeds Teaching HospitalsLeedsUK
| | - Moshe Mittelman
- Department of HematologyTel Aviv Sourasky (Ichilov) Medical Center, Sackler Faculty of Medicine, Tel Aviv UniversityTel Aviv‐YafoIsrael
| | - Kathrin Sommer
- Italian Group for Adult Hematologic Diseases (GIMEMA)Data Center and Health Outcomes Research UnitRomeItaly
| | - Saskia Langemeijer
- Department of HematologyRadboud University Medical CenterNijmegenThe Netherlands
| | | | | | - Michael Storck
- Institute of Medical Informatics, University of MünsterMünsterGermany
| | - Alexandra Smith
- Epidemiology and Cancer Statistics Group, Department of Health SciencesUniversity of YorkYorkUK
| | - Corine van Marrewijk
- Department of HematologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Martin Dugas
- Institute of Medical Informatics, Heidelberg University HospitalHeidelbergGermany
| | - Igor Stojkov
- Department of Public Health, Health Services Research and Health Technology AssessmentInstitute of Public Health, Medical Decision Making and Health Technology Assessment, UMIT ‐ University for Health Sciences, Medical Informatics and TechnologyHall in TirolAustria
| | - Uwe Siebert
- Department of Public Health, Health Services Research and Health Technology AssessmentInstitute of Public Health, Medical Decision Making and Health Technology Assessment, UMIT ‐ University for Health Sciences, Medical Informatics and TechnologyHall in TirolAustria
- Departments of Epidemiology and Health Policy & ManagementCenter for Health Decision Science, Harvard Chan School of Public HealthBostonMassachusettsUSA
- Institute for Technology Assessment and Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Theo de Witte
- Department of Tumor ImmunologyNijmegen Center for Molecular Life Sciences, Radboud University Medical CenterNijmegenThe Netherlands
| | - Reinhard Stauder
- Department of Internal Medicine V (Hematology and Oncology)Medical University InnsbruckInnsbruckAustria
| |
Collapse
|
10
|
Brunner AM, Gavralidis A, Ali NA, Hunter A, Komrokji R, Zeidan A, Sallman DA. Evaluating complete remission with partial hematologic recovery (CRh) as a response criterion in myelodysplastic syndromes (MDS). Blood Cancer J 2022; 12:153. [PMID: 36379923 PMCID: PMC9666661 DOI: 10.1038/s41408-022-00748-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
Myelodysplastic syndromes (MDS) treated with DNMTI therapy have responses according to the 2006 IWG response criteria. CR responses have had the strongest association with OS. Recently, CR with partial hematologic recovery (CRh; i.e. blasts <5%, ANC > 500, platelets > 50) has been evaluated in AML, but its relevance is unknown in MDS. We identified adult patients with MDS treated with DNMTIs. We assessed best overall response to therapy according to IWG 2006 criteria, and subsequently identified patients meeting CRh criteria from the subgroup with SD or mCR. We evaluated duration of therapy and overall survival according to response. We identified 311 patients with MDS who received treatment between 2007 and 2018. The median age at the time of therapy was 69 years (range 23-91). Median follow up was 60 months. According to IWG 2006, responses included CR (n = 43, 14%), PR (n = 2, 1%), mCR (n = 57, 18%), SD (n = 149, 48%) and PD (n = 60, 19%). 79 patients (25%) achieved HI. A total of 62 patients (20%) met CRh criteria leading to reclassification of mCR (now n = 26, 8%) or SD (now n = 118, 38%). Patients achieving CR had similar time on therapy (median 8.1mo) compared to CRh (median 6mo, HR 1.4, 95% CI 0.9-2.0), and longer than other responses (p < 0.001). OS varied according to response; median OS was similar between CR (23.3mo) and CRh (25mo, HR 1.28 [0.79-2.08]), which was longer than those with mCR (17.2mo, HR 1.71 [0.96-3.05]), SD (16.3mo, HR 1.61 [1.04-2.48]), and PD (8.7mo, HR 3.04 [1.91-4.83]) (p < 0.001). OS associations with CR/CRh were confirmed in multivariable analysis accounting for allogeneic transplant. MDS patients who achieve a CRh response had similar survival and duration on therapy as patients who achieve CR response and superior to other IWG responses. These data support further evaluation of CRh into future response criteria and clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Amer Zeidan
- Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
11
|
Pleyer L, Sekeres MA. An early glimpse at azacitidine plus venetoclax for myelodysplastic syndromes. Lancet Haematol 2022; 9:e714-e716. [PMID: 36063831 DOI: 10.1016/s2352-3026(22)00252-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Lisa Pleyer
- Austrian Group for Medical Tumor Therapy Study Group, Vienna, Austria; Salzburg Cancer Research Institute, Center for Clinical Cancer and Immunology Trials, Salzburg, Austria; Cancer Cluster Salzburg, Salzburg, Austria; 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria.
| | - Mikkael A Sekeres
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, USA
| |
Collapse
|
12
|
Effect of thrombopoietin receptor agonist on health-related quality of life and platelet transfusion burden for patients with myelodysplastic syndromes: a systematic review and meta-analysis. Ann Hematol 2022; 101:2219-2229. [PMID: 35976414 DOI: 10.1007/s00277-022-04950-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 07/27/2022] [Indexed: 11/01/2022]
Abstract
Thrombocytopenia is a common and unsolved problem in myelodysplastic syndrome (MDS) patients; we aimed to summarize the evidence of TPO-RA treatment for heath-related quality of life (HRQoL) and platelet transfusion burden of MDS patients. We searched Pubmed, Web of Science, EMBASE, and CENTRAL for randomized clinical trials (RCTs) comparing TPO-RA to placebo in MDS published until July 31, 2021. A random-effect model was used. Eight RCTs with 908 patients were identified. Only three RCTs involving eltrombopag reported HRQoL, and all three studies treated HRQoL as a secondary outcome. In these three RCTs, the HRQoL instruments used in each study were different. However, this outcome cannot be meta-analyzed because some studies did not provide complete data. Subsequent clinical trials should pay more attention to this. Compared to placebo, TPO-RA did not affect platelet transfusion incidence 0.83 (95% CI 0.60-1.15). There was no evidence for subgroup differences in the analyses of different types of TPO-RA, different additional agent, and different types of MDS risk groups. However, platelet transfusion units (RR = 0.68, 95% CI 0.53 to 0.84) were significantly decreased. The RR of patients who did not require platelet transfusion for 56 or more consecutive days was not different between groups (RR = 0.98, 95% CI 0.41 to 2.34). TPO-RA may decrease platelet transfusion units in MDS patients with thrombocytopenia. But the significance of this finding should be interpreted with caution, because too few studies were meta-analyzed.
Collapse
|
13
|
Kim N, Pavletic S, Norsworthy KJ. Meaningful response criteria for myelodysplastic syndromes. Br J Haematol 2021; 196:1137-1148. [PMID: 34628648 DOI: 10.1111/bjh.17838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/13/2021] [Accepted: 09/02/2021] [Indexed: 01/11/2023]
Abstract
Standardizing response criteria for myelodysplastic syndromes (MDS), a clinically and biologically heterogeneous group of disorders, has been historically challenging. The International Working Group (IWG) response criteria, first proposed in 2000 and modified in 2006 and 2018, represent the best effort by a group of international experts to define a set of clinically meaningful end-points in MDS. These criteria have been adopted in many MDS clinical trials, allowing for comparisons of response across trials. However, clinical experience has also revealed some limitations of these criteria, and most of the end-points proposed by the IWG require further validation. In this review, we present a critical analysis of the current MDS response criteria from both a practical standpoint and based on currently available clinical trial data. Potential areas for improvement in the criteria are highlighted, which may be considered in future iterations of the response criteria.
Collapse
Affiliation(s)
- Nina Kim
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Steven Pavletic
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kelly J Norsworthy
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
14
|
Duetz C, Cucchi DGJ, Polak TB, Janssen JJWM, Ossenkoppele GJ, Estey EH, van de Loosdrecht AA. The wider perspective: twenty years of clinical trials in myelodysplastic syndromes. Br J Haematol 2021; 196:329-335. [PMID: 34632583 DOI: 10.1111/bjh.17892] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/23/2021] [Accepted: 09/29/2021] [Indexed: 12/21/2022]
Abstract
Most patients with myelodysplastic syndromes (MDS) require therapeutic intervention. However, there are few approved treatments for MDS. To explore reasons, we searched clinicaltrials.gov and clinicaltrialsregister.eu for MDS trials from 2000 to 2020. We assessed which agents were under investigation and analysed clinical trial characteristics and continuation rates from phase I to II to III to approval. As such, we identified 384 unique agents in 426 phase I, 430 phase II and 48 phase III trials. Success rates for phase III trials and agents were low, and MDS trials took markedly longer to complete than the average clinical trial. Although success rates were higher when MDS-specific phase I trials were conducted, 52% of the agents had not been evaluated in a phase I trial for MDS. MDS trials often failed to include quality of life, an especially important outcome for older MDS patients. Our work identifies factors potentially contributing to the paucity of available agents for MDS. We suggest a framework to improve clinical research in MDS that might ultimately augment the number of available agents.
Collapse
Affiliation(s)
- Carolien Duetz
- Department of Hematology, Amsterdam UMC, location VUmc, Vrije Universiteit, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - David G J Cucchi
- Department of Hematology, Amsterdam UMC, location VUmc, Vrije Universiteit, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Tobias B Polak
- Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands
| | - Jeroen J W M Janssen
- Department of Hematology, Amsterdam UMC, location VUmc, Vrije Universiteit, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gert J Ossenkoppele
- Department of Hematology, Amsterdam UMC, location VUmc, Vrije Universiteit, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Elihu H Estey
- Department of Hematology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Arjan A van de Loosdrecht
- Department of Hematology, Amsterdam UMC, location VUmc, Vrije Universiteit, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Oliva EN, Platzbecker U, Fenaux P, Garcia-Manero G, LeBlanc TW, Patel BJ, Kubasch AS, Sekeres MA. Targeting health-related quality of life in patients with myelodysplastic syndromes - Current knowledge and lessons to be learned. Blood Rev 2021; 50:100851. [PMID: 34088518 DOI: 10.1016/j.blre.2021.100851] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022]
Abstract
Using a range of health-related quality of life (HRQoL) instruments, most - but not all - studies of myelodysplastic syndromes (MDS) have reported that lower hemoglobin levels and red blood cell transfusion dependency are associated with worse HRQoL. In addition, some MDS treatments may significantly improve HRQoL, particularly among those patients who respond to therapy; however, the majority of these studies were underpowered for this secondary endpoint. Furthermore, decreased HRQoL has been associated with worse survival outcomes, and HRQoL scores can be used to refine classical prognostic systems. Despite the subjective nature of HRQoL, the importance and validity of measuring it in trials and clinical practice are increasingly being recognized, but properly validated MDS-specific instruments are required. We describe what is currently known about HRQoL in patients with MDS, and the limitations of measuring HRQoL, and we provide some recommendations to improve the measurement of this outcome in future trials.
Collapse
Affiliation(s)
- Esther N Oliva
- Grande Ospedale Metropolitano Bianchi Melacrino Morelli, Reggio Calabria, Italy.
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany.
| | - Pierre Fenaux
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Université Paris 7, Paris, France.
| | | | | | | | - Anne Sophie Kubasch
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany.
| | - Mikkael A Sekeres
- Sylvester Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
16
|
Nachtkamp K, Stark J, Kündgen A, Schroeder T, Strupp C, Strapatsas J, Schuler E, Kaivers J, Giagounidis A, Rautenberg C, Aul C, Runde V, Haas R, Kobbe G, Gattermann N, Germing U. Eligibility for clinical trials is unsatisfactory for patients with myelodysplastic syndromes, even at a tertiary referral center. Leuk Res 2021; 108:106611. [PMID: 33990002 DOI: 10.1016/j.leukres.2021.106611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 12/01/2022]
Abstract
Participation in clinical trials may allow patients with MDS to gain access to therapies not otherwise available. However, access is limited by strict inclusion and exclusion criteria, reflecting academic or regulatory questions addressed by the respective studies. We performed a simulation in order to estimate the average proportion of MDS patients eligible for participation in a clinical trial. The simulation drew upon 1809 patients in the Düsseldorf MDS Registry whose clinical data allowed eligibility screening for a wide range of clinical trials. This cohort was assumed to be alive and available for study participation. The simulation also posited that all MDS trials (n = 47) conducted in our center between 1987 and 2016 were open for recruitment. In addition, study activities in the year 2016 were analyzed to determine the proportion of patients eligible for at least one of the 9 MDS trials open at that time. On average, each clinical trial was suitable for about 18 % of patients in the simulation cohort. Conversely, 34 % of the patients were eligible for at least one of the 9 clinical studies in 2016. Inclusion/exclusion criteria of studies initiated by the pharmaceutical industry excluded more than twice the fraction of patients compared with investigator initiated trials (potential inclusion of 10 % vs. 21 %, respectively). Karyotype (average exclusion rate 58 %), comorbidities (40 %), and prior therapies (55 %) were the main reasons for exclusion. We suggest that in- and exclusion criteria should be less restrictive, in order to meet the needs of the real-life population of elderly MDS patients.
Collapse
Affiliation(s)
- Kathrin Nachtkamp
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany.
| | - Josefine Stark
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Andrea Kündgen
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Corinna Strupp
- Department of Oncology, Rheinland Klinikum Dormagen, Dr.-Geldmacher-Straße 20, 41540 Dormagen, Germany
| | - Judith Strapatsas
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Esther Schuler
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Jennifer Kaivers
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Aristoteles Giagounidis
- Department of Oncology and Hematology, VKKD Marienhospital Duesseldorf, Rochusstr. 2, 40479 Düsseldorf, Germany
| | - Christina Rautenberg
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Carlo Aul
- Department of Oncology and Hematology, VKKD Marienhospital Duesseldorf, Rochusstr. 2, 40479 Düsseldorf, Germany
| | - Volker Runde
- Department of Hematology and Oncology, Katholisches Karl-Leisner-Klinikum, Voßheider Str. 214, 47574 Goch, Germany
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Norbert Gattermann
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany
| |
Collapse
|
17
|
Trudeau JJ, He J, Rose E, Panter C, Randhawa S, Gater A. Content validity of patient-reported outcomes for use in lower-risk myelodysplastic syndromes. J Patient Rep Outcomes 2020; 4:69. [PMID: 32851569 PMCID: PMC7450032 DOI: 10.1186/s41687-020-00235-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The lower-risk (low and intermediate-1 risk based on IPSS) myelodysplastic syndrome (MDS) has a negative impact on patients' health-related quality of life (HRQoL). Patient Reported Outcomes (PROs) instruments, which are used to collect patients' HRQoL data, should have established content validity in the target population to ensure that the instrument is comprehensive and comprehensible. The present study was conducted to evaluate the content validity of the Quality of Life in Myelodysplasia Scale (QUALMS) and the Functional Assessment of Cancer Therapy-Anemia (FACT-An) PRO instruments in patients with lower-risk MDS. METHODS In this cross-sectional, qualitative study, 16 patients aged ≥18 years with lower-risk MDS, who were RBC transfusion dependent, literate and fluent in US-English were interviewed. Interviews were semi-structured comprising of two parts: concept elicitation (CE) explored symptoms and impacts important to patients, and cognitive debriefing (CD) assessed understanding and relevance of the QUALMS and FACT-An. A conceptual model was developed, which was used to map the concepts that emerged during CE onto the QUALMS and FACT-An to assess concept coverage and suitability of the instruments. RESULTS The median age of participants was 67.5 years (range: 51-91), with half being female (n = 8). Nine (56.2%) participants had intermediate-1-risk MDS and 10 (62.5%) were relapsed or refractory to erythropoiesis-stimulating agent treatment. Fatigue/tiredness (100.0%), shortness of breath (87.5%), weakness (81.2%), and low energy (75.0%) were reported most commonly and were the most bothersome symptoms as well. Of seven high-level HRQoL domains identified, activities of daily living (n = 16, 100.0%), physical functioning (n = 15, 93.8%), emotional wellbeing (n = 13, 81.3%), social functioning (n = 12, 75.0%), sleep disturbance (n = 9, 56.3%), and impact on work (n = 9, 56.3%) were the most commonly reported. For CD, the QUALMS and FACT-An were found to be mostly relevant and very well understood; response options were easy to use, and recall period was appropriate. CONCLUSION Both QUALMS and FACT-An demonstrated a strong face and content validity in patients with lower-risk MDS, suggesting that these instruments are appropriate for assessing HRQoL in this population.
Collapse
Affiliation(s)
| | - Jianming He
- Janssen Global Services LLC, 700 US 202 South, Raritan, NJ, 08869, USA
| | - Esther Rose
- Janssen Pharmaceuticals LLC, Raritan, NJ, 08869, USA
| | | | | | - Adam Gater
- Adelphi Values, Bollington, Cheshire, UK
| |
Collapse
|
18
|
Menssen AJ, Walter MJ. Genetics of progression from MDS to secondary leukemia. Blood 2020; 136:50-60. [PMID: 32430504 PMCID: PMC7332895 DOI: 10.1182/blood.2019000942] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022] Open
Abstract
Our understanding of the genetics of acute myeloid leukemia (AML) development from myelodysplastic syndrome (MDS) has advanced significantly as a result of next-generation sequencing technology. Although differences in cell biology and maturation exist between MDS and AML secondary to MDS, these 2 diseases are genetically related. MDS and secondary AML cells harbor mutations in many of the same genes and functional categories, including chromatin modification, DNA methylation, RNA splicing, cohesin complex, transcription factors, cell signaling, and DNA damage, confirming that they are a disease continuum. Differences in the frequency of mutated genes in MDS and secondary AML indicate that the order of mutation acquisition is not random during progression. In almost every case, disease progression is associated with clonal evolution, typically defined by the expansion or emergence of a subclone with a unique set of mutations. Monitoring tumor burden and clonal evolution using sequencing provides advantages over using the blast count, which underestimates tumor burden, and could allow for early detection of disease progression prior to clinical deterioration. In this review, we outline advances in the study of MDS to secondary AML progression, with a focus on the genetics of progression, and discuss the advantages of incorporating molecular genetic data in the diagnosis, classification, and monitoring of MDS to secondary AML progression. Because sequencing is becoming routine in the clinic, ongoing research is needed to define the optimal assay to use in different clinical situations and how the data can be used to improve outcomes for patients with MDS and secondary AML.
Collapse
Affiliation(s)
- Andrew J Menssen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO; and
| | - Matthew J Walter
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO; and
- Siteman Cancer Center, Washington University, St. Louis, MO
| |
Collapse
|
19
|
Canaani J. Emerging Therapies for the Myelodysplastic Syndromes. Clin Hematol Int 2020; 2:13-17. [PMID: 34595438 PMCID: PMC8432342 DOI: 10.2991/chi.d.191202.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/27/2019] [Indexed: 01/11/2023] Open
Abstract
Despite considerable advances in our understanding of the molecular and epigenetic underpinnings of the myelodysplastic syndromes (MDS), this diverse group of myeloid neoplasms remains a significant clinical challenge. Considerable barriers to timely development of effective therapy include the diverse molecular landscape encountered in MDS patients, the difficulty in translating specific molecular aberration into a clinically meaningful animal model, as well as challenges in patient recruitment into clinical trials. These speak to the need to discover efficacious novel therapeutic targets which would in turn translate into improved patient outcomes in terms of both survival and quality of life. In this review, we outline recently published data pertaining to therapeutic advances in TGF-β pathway inhibition, STAT3, Hedgehog signaling, and additional therapeutic venues being actively explored in MDS.
Collapse
Affiliation(s)
- Jonathan Canaani
- Hematology Division, Chaim Sheba Medical Center, Tel Aviv University, Tel Hashomer, Israel
| |
Collapse
|
20
|
Patient-Reported Outcomes in Myelodysplastic Syndromes: the Move from Life Span to Health Span. Curr Hematol Malig Rep 2020; 15:149-154. [DOI: 10.1007/s11899-020-00562-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
21
|
Sekeres MA, Patel BJ. Lowering the boom on lower-risk myelodysplastic syndromes. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:367-372. [PMID: 31808873 PMCID: PMC6913460 DOI: 10.1182/hematology.2019000040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Lower-risk myelodysplastic syndromes are defined using prognostic scoring systems that incorporate data on bone marrow blast percentage, degree and numbers of cytopenias, and cytogenetic abnormalities. Increasingly, these are incorporating molecular abnormalities to further refine risk. Therapy is geared toward predominating cytopenias, with erythropoiesis-stimulating agents luspatercept and lenalidomide used to ameliorate anemia, romiplostim and eltrombopag tackling thrombocytopenia, and hypomethylating agents and antithymocyte globulin palliating pancytopenia. Newer agents on the horizon are abrogating the downstream sequelae of specific molecular mutations. One challenge for the future is in further modifying response criteria to align with improvements that are clinically meaningful to patients.
Collapse
|
22
|
Lin Y, Buckstein R. Outpatient transfusions: time to study what matters to patients. Transfusion 2019; 59:1887-1890. [DOI: 10.1111/trf.15382] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 05/13/2019] [Indexed: 01/28/2023]
Affiliation(s)
- Yulia Lin
- Department of Laboratory Medicine and Molecular Diagnostics; Sunnybrook Health Sciences Centre; Toronto Canada
- Department of Laboratory Medicine and Pathobiology; University of Toronto; Toronto Canada
- University of Toronto Quality in Utilization, Efficacy, & Safety in Transfusion (QUEST) Research Program; Toronto Canada
| | - Rena Buckstein
- Division of Hematology; Odette Cancer Centre, Sunnybrook Health Sciences Centre; Toronto Canada
- Department of Medicine; University of Toronto; Toronto Canada
| |
Collapse
|