1
|
Mina A, Komrokji R. How I treat higher-risk MDS. Blood 2025; 145:2002-2011. [PMID: 39808802 DOI: 10.1182/blood.2024025271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/05/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
ABSTRACT Myelodysplastic syndromes/neoplasms (MDS) are a widely heterogenous group of myeloid malignancies characterized by morphological dysplasia, a defective hematopoiesis, and recurrent genetic abnormalities. The original International Prognostic Scoring System (IPSS) and revised IPSS have been used to risk-stratify patients with MDS to guide treatment strategies. In higher-risk MDS, the therapeutic approach is geared toward delaying leukemic transformation and prolonging survival. For more than a decade, the hypomethylating agents azacitidine and decitabine have been the standard of care and, when feasible, an allogeneic hematopoietic stem cell transplantation should be considered. However, the IPSS scoring systems solely rely on clinical, morphological, and cytogenetic features and do not account for somatic mutations present in >80% of cases. These genetic abnormalities have been shown to play a crucial role in prognostication, prompting the development of molecular IPSS, and the integration of genomic features into MDS classification systems in recent years. In this review, we delineate our approach to higher-risk MDS in the context of updated classifications and the latest prognostication tools. We use illustrative clinical cases to support our discussion and share insights from recent clinical trials, highlighting lessons learned.
Collapse
Affiliation(s)
- Alain Mina
- Myeloid Malignancies Program, Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Rami Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
2
|
Hu J, Tian S, Liu Q, Hou J, wu J, Wang X, Shu Y. A prognostic signature of Glutathione metabolism-associated long non-coding RNAs for lung adenocarcinoma with immune microenvironment insights. Front Immunol 2025; 16:1477437. [PMID: 39995658 PMCID: PMC11847877 DOI: 10.3389/fimmu.2025.1477437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Background Glutathione (GSH) metabolism supports tumor redox balance and drug resistance, while long non-coding RNAs (lncRNAs) influence lung adenocarcinoma (LUAD) progression. This study developed a prognostic model using GSH-related lncRNAs to predict LUAD outcomes and assess tumor immunity. Methods This study analyzed survival data from The Cancer Genome Atlas (TCGA) and identified GSH metabolism-related lncRNAs using Pearson correlation. A prognostic model was built with Cox and Least Absolute Shrinkage and Selection Operator (LASSO) methods and validated by Kaplan-Meier analysis, Receiver Operating Characteristic (ROC) curves, and Principal Component Analysis (PCA). Functional analysis revealed immune infiltration and drug sensitivity differences. Quantitative PCR and experimental studies confirmed the role of lnc-AL162632.3 in LUAD. Results Our model included a total of nine lncRNAs, namely AL162632.3, AL360270.1, LINC00707, DEPDC1-AS1, GSEC, LINC01711, AL078590.2, AC026355.2, and AL096701.4. The model effectively forecasted patient survival, and the nomogram, incorporating additional clinical risk factors, satisfied clinical needs adequately. Patient stratification based on model scores revealed significant disparities in immune cell composition, functionality, and mutations between groups. Additionally, variations were noted in the IC50 values for key lung cancer medications such as Cisplatin, Docetaxel, and Paclitaxel. In vitro cell experiment results showed that AL162632.3 was markedly upregulated, while AC026355.2 tended to be downregulated across these cell lines. Ultimately, suppressing lnc-AL162632.3 markedly reduced the growth, mobility, and invasiveness of lung cancer cells. Conclusion This study identified GSH metabolism-related lncRNAs as key prognostic factors in LUAD and developed a model for risk stratification. High-risk patients showed increased tumor mutation burden (TMB) and stemness, emphasizing the potential of personalized immunotherapy to improve survival outcomes.
Collapse
Affiliation(s)
- Junxi Hu
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Shuyu Tian
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
- Dalian Medical University, Dalian, China
| | - Qingwen Liu
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
- Dalian Medical University, Dalian, China
| | - Jiaqi Hou
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
- Dalian Medical University, Dalian, China
| | - Jun wu
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Xiaolin Wang
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Yusheng Shu
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
| |
Collapse
|
3
|
Liu XZ, Zhou SJ, Huang J, Zhao CF, Jiang LX, Zhang YD, Mei C, Ma LY, Zhou XP, Shao YP, Wu GQ, Xiao XB, Yao RX, Du XH, Hu TL, Qian SX, Li Y, Yan XF, Huang L, Wang ML, Fu JP, Shou LH, Jiang WH, Jin WM, Li LJ, Le J, Luo WJ, Zhang Y, Zhou XJ, Zhang H, Lang XH, Zhou M, Jin J, Jiang HF, Zhang J, Ouyang GF, Tong HY. [Long-term hypomethylating agents in patients with myelodysplastic syndromes: a multi-center retrospective study]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:738-747. [PMID: 39307720 PMCID: PMC11535563 DOI: 10.3760/cma.j.cn121090-20240405-00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Indexed: 12/06/2024]
Abstract
Objective: To evaluate the efficacy and safety of hypomethylating agents (HMA) in patients with myelodysplastic syndromes (MDS) . Methods: A total of 409 MDS patients from 45 hospitals in Zhejiang province who received at least four consecutive cycles of HMA monotherapy as initial therapy were enrolled to evaluate the efficacy and safety of HMA. Mann-Whitney U or Chi-square tests were used to compare the differences in the clinical data. Logistic regression and Cox regression were used to analyze the factors affecting efficacy and survival. Kaplan-Meier was used for survival analysis. Results: Patients received HMA treatment for a median of 6 cycles (range, 4-25 cycles) . The complete remission (CR) rate was 33.98% and the overall response rate (ORR) was 77.02%. Multivariate analysis revealed that complex karyotype (P=0.02, OR=0.39, 95%CI 0.18-0.84) was an independent favorable factor for CR rate. TP53 mutation (P=0.02, OR=0.22, 95%CI 0.06-0.77) was a predictive factor for a higher ORR. The median OS for the HMA-treated patients was 25.67 (95%CI 21.14-30.19) months. HMA response (P=0.036, HR=0.47, 95%CI 0.23-0.95) was an independent favorable prognostic factor, whereas complex karyotype (P=0.024, HR=2.14, 95%CI 1.10-4.15) , leukemia transformation (P<0.001, HR=2.839, 95%CI 1.64-4.92) , and TP53 mutation (P=0.012, HR=2.19, 95%CI 1.19-4.07) were independent adverse prognostic factors. There was no significant difference in efficacy and survival between the reduced and standard doses of HMA. The CR rate and ORR of MDS patients treated with decitabine and azacitidine were not significantly different. The median OS of patients treated with decitabine was longer compared with that of patients treated with azacitidine (29.53 months vs 20.17 months, P=0.007) . The incidence of bone marrow suppression and pneumonia in the decitabine group was higher compared with that in the azacitidine group. Conclusion: Continuous and regular use of appropriate doses of hypomethylating agents may benefit MDS patients to the greatest extent if it is tolerated.
Collapse
Affiliation(s)
- X Z Liu
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - S J Zhou
- Department of Hematology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - J Huang
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China Department of Hematology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - C F Zhao
- Department of Hematology, Jinhua Municipal Central Hospital, Jinhua 321000, China
| | - L X Jiang
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Y D Zhang
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - C Mei
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - L Y Ma
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - X P Zhou
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Y P Shao
- Department of Hematology, Taizhou Hospital, Taizhou 317000, China
| | - G Q Wu
- Department of Hematology, Dongyang Hospital Affiliated to Wenzhou Medical University, Dongyang 322100, China
| | - X B Xiao
- Department of Hematology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China
| | - R X Yao
- Department of Hematology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - X H Du
- Department of Hematology, Ningbo Yinzhou People's Hospital, Ningbo 315040, China
| | - T L Hu
- Department of Hematology, Zhejiang Provincial Hospital of TCM, Hangzhou 310006, China
| | - S X Qian
- Department of Hematology, Hangzhou First People's Hospital, Hangzhou 310006, China
| | - Y Li
- Department of Hematology, First Hospital of Jiaxing, Jiaxing 314001, China
| | - X F Yan
- Department of Hematology, Quzhou People's Hospital, Quzhou 324000, China
| | - L Huang
- Department of Hematology, Jinhua People's Hospital, Jinhua 321000, China
| | - M L Wang
- Department of Hematology, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - J P Fu
- Department of Hematology, Shaoxing People's Hospital, Shaoxing 312000, China
| | - L H Shou
- Department of Hematology, Huzhou Central Hospital, Huzhou 313000, China
| | - W H Jiang
- Department of Hematology, Taizhou First People's Hospital, Taizhou 318020, China
| | - W M Jin
- Department of Hematology, Lishui Municipal People Hospital, Lishui 323000, China
| | - L J Li
- Department of Hematology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - J Le
- Department of Hematology, Li Huili Hospital, Ningbo 315040, China
| | - W J Luo
- Department of Hematology, The First People's Hospital of Xiaoshan District, Hangzhou 311200, China
| | - Y Zhang
- Department of Hematology, Yueqing People's Hospital, Wenzhou 325600, China
| | - X J Zhou
- Department of Hematology, Haining People's Hospital, Haining 314400, China
| | - H Zhang
- Department of Hematology, The Third Affiliated Hospital of Wenzhou Medical University, Rui'an 325200, China
| | - X H Lang
- Department of Hematology, Yongkang First People's Hospital, Yongkang 321300, China
| | - M Zhou
- Department of Hematology, Zhuji People's Hospital, Zhuji 311800, China
| | - J Jin
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - H F Jiang
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - J Zhang
- Department of Hematology, Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou 310016, China
| | - G F Ouyang
- Department of Hematology, The First Affiliated Hospital of Ningbo University School of Medicine, Ningbo 315010, China
| | - H Y Tong
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
4
|
Enjeti A, Ashraf A, Caillet V, Alam A, Silar J, Keer H, Castaldi F, Paine T. Real-world study of the use of azacitidine in myelodysplasia in Australia. EJHAEM 2024; 5:527-534. [PMID: 38895081 PMCID: PMC11182405 DOI: 10.1002/jha2.911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 06/21/2024]
Abstract
Hypomethylating agents are the most widely used upfront therapy for patients with myelodysplastic syndrome (MDS) who are not suitable for hematopoietic stem cell transplantation. In Australia, azacitidine was, until recently, the only approved and subsidized treatment for patients with intermediate-2 and high-risk MDS, chronic myelomonocytic leukemia, and low blast acute myeloid leukemia. We analyzed prescription data to evaluate the real-world persistence and overall survival (OS) of patients prescribed azacitidine for the first time in Australia. A retrospective cohort analysis of patients who had been prescribed Pharmaceutical Benefits Scheme (PBS)-listed azacitidine for the first time, between January 2016 and April 2021, was conducted using the PBS 10% dataset. Treatment persistence and OS were estimated using Kaplan-Meier methods. The impact of the number of treatment cycles and treatment adherence on OS was also estimated. There were 351 patients in the PBS 10% dataset who initiated treatment with azacitidine. The average age (standard deviation [SD]) at azacitidine initiation was 71.9 (11.1) years and the average number (SD) of azacitidine prescriptions was 5.6 (0.2). The median persistence on azacitidine was 15.6 months, and the OS was 13.4 months. The median OS for patients who had six or more cycles of azacitidine treatment was greater compared to patients who had five or less cycles of treatment. The data from this real-world study illustrate the unmet medical needs of patients with MDS treated with azacitidine in Australia. The majority of patients are not treated with the optimal number of cycles of azacitidine, which is negatively correlated with patient outcomes.
Collapse
Affiliation(s)
- Anoop Enjeti
- Faculty of Medicine and Public HealthUniversity of NewcastleNewcastleNew South WalesAustralia
- Department of HaematologyCalvary Mater HospitalNewcastleNew South WalesAustralia
| | - Asma Ashraf
- Department of HaematologyCalvary Mater HospitalNewcastleNew South WalesAustralia
| | | | - Arif Alam
- Prospection Pty Ltd.SydneyNew South WalesAustralia
| | - Jonathan Silar
- Faculty of Medicine and Public HealthUniversity of NewcastleNewcastleNew South WalesAustralia
- Department of HaematologyCalvary Mater HospitalNewcastleNew South WalesAustralia
| | - Harold Keer
- Astex Pharmaceuticals, Inc.PleasantonCaliforniaUSA
| | | | - Taleisha Paine
- Otsuka Australia PharmaceuticalChatswoodNew South WalesAustralia
| |
Collapse
|
5
|
Rodenbach RA, Thordardottir T, Brauer M, Hall AC, Ward E, Smith CB, Campbell TC. Communication Strategies of Transplant Hematologists in High-Risk Decision-Making Conversations. JCO Oncol Pract 2024; 20:538-548. [PMID: 38241601 PMCID: PMC11590740 DOI: 10.1200/op.23.00574] [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: 09/08/2023] [Revised: 11/06/2023] [Accepted: 12/08/2023] [Indexed: 01/21/2024] Open
Abstract
PURPOSE Shared decision making (SDM) is essential to empower patients with blood cancers to make goal-concordant decisions about allogeneic hematopoietic cell transplantation. This study characterizes communication strategies used by hematologists to discuss treatment options and facilitate SDM with patients in this high-risk, high-reward setting. METHODS AND MATERIALS We recruited US hematologists who routinely perform allogeneic hematopoietic cell transplant through email. Participants conducted up to an hour-long video-recorded encounter with an actor portraying a 67-year-old man with recently diagnosed high-risk myelodysplastic syndrome. We transcribed and qualitatively analyzed video-recorded data. RESULTS The mean age of participants (N = 37) was 44 years, 65% male, and 68% White. Many hematologists included similar key points in this initial consultation, although varied in how much detail they provided. Their discussion of treatment options included transplant and chemotherapy and less commonly supportive care or clinical trials. They often emphasized transplant's potential for cure, discussed transplant chronologically from pretransplant considerations through the post-transplant course, and outlined risks, complications, and major outcomes. Hematologists referred to several elements that formed the basis of treatment decision making. The strength of their treatment recommendations ranged from strong recommendations for transplant or chemotherapy to deferrals pending more information. Hematologists also varied in the extent to which they indicated the decision was physician-driven, patient-led, or shared. CONCLUSION The transplant decision-making discussion is complex. Identification of similar content areas used by hematologists can be used as the basis for a communication tool to help hematologists discuss allogeneic hematopoietic cell transplant with patients.
Collapse
Affiliation(s)
- Rachel A. Rodenbach
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
- James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Markus Brauer
- University of Wisconsin Department of Psychology, Madison, Wisconsin, USA
| | - Aric C. Hall
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Earlise Ward
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
- University of Wisconsin School of Medicine and Public Health, Department of Family Medicine and Community Health, Madison, Wisconsin, USA
| | | | - Toby C. Campbell
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| |
Collapse
|
6
|
Saga T, Kanagawa M, Harada T, Lang L, Yamawaki F, Ishihara T. Prognostic Value of Pretreatment Fetal Hemoglobin Levels in Patients with Myelodysplastic Syndromes and Acute Myeloid Leukemia Treated with Azacitidine: A Single-center Retrospective Study. Intern Med 2024; 63:781-790. [PMID: 37495538 PMCID: PMC11008988 DOI: 10.2169/internalmedicine.1216-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 06/05/2023] [Indexed: 07/28/2023] Open
Abstract
Objective Azacitidine (AZA) has been the standard of care for elderly patients with high-risk myelodysplastic syndromes (MDS). However, reliable clinical predictors of outcome have yet to be identified. The prognostic value of fetal hemoglobin (HbF) levels has been reported for decitabine therapy. We evaluated pretreatment HbF levels in AZA monotherapy as a prognostic marker in MDS/acute myeloid leukemia (AML). Methods This study included chemotherapy-naïve patients who had received seven-day treatment schedules of AZA and whose HbF levels were measured at the onset of treatment between March 2011 and July 2020. Patients were grouped into HbF-normal (<1.0%) or HbF-elevated (≥1.0%) groups. Responses were classified according to the International Working Group 2006 criteria. Patients Twenty-nine patients were included and classified as having either MDS (n=21), chronic myelomonocytic leukemia (n=5), myelodysplastic/myeloproliferative neoplasm unclassifiable (n=1), or AML with <30% marrow blasts (n=2) based on the World Health Organization 2016 diagnostic criteria. According to the revised International Prognostic Scoring System classification, 20/29 patients were at intermediate, high, or very high risk. Pretreatment HbF levels were elevated in 13/29 patients. Results The median follow-up duration was 13.0 (range 1.5-93.5) months. The HbF-elevated group was associated with a significantly higher hematologic improvement rate (76.9% vs. 25%, p=0.009) and better overall survival (median, 21.0 vs. 13.0 months, p=0.048) than the HbF-normal group. Conclusion These results suggest that elevated pretreatment HbF levels can predict better outcomes in patients with MDS/AML treated with AZA.
Collapse
Affiliation(s)
- Tomoyuki Saga
- Department of Hematology, Kin-ikyo Chuo Hospital, Japan
| | | | - Tomoya Harada
- Department of Hematology, Kin-ikyo Chuo Hospital, Japan
| | - Lang Lang
- Department of Hematology, Kin-ikyo Chuo Hospital, Japan
| | | | | |
Collapse
|
7
|
Inoue Y, Okamoto H, Miyashita A, Kawaji-Kanayama Y, Chinen S, Fujino T, Tsukamoto T, Shimura Y, Mizutani S, Kaneko H, Kuwahara-Ota S, Fuchida SI, Nishiyama D, Hirakawa K, Uchiyama H, Uoshima N, Kawata E, Kuroda J. Clinical impacts of severe thrombocytopenia in the first cycle of azacitidine monotherapy and cytogenetics in patients with myelodysplastic syndrome: The Kyoto Conditional Survival Scoring System. Oncol Lett 2024; 27:62. [PMID: 38192677 PMCID: PMC10773215 DOI: 10.3892/ol.2023.14193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
Azacitidine (AZA) has been one of the standard treatments for transplantation-ineligible patients with myelodysplastic syndrome (MDS); however, hematological toxicities frequently cause treatment interruption in the early phase of the therapy. The present study conducted a multicenter retrospective study to investigate the prognostic impacts of various factors, including factors included in the Revised International Prognostic Scoring System (IPSS-R) and severe cytopenia in the early phase of AZA monotherapy in 212 patients with MDS. Severe cytopenia was evaluated after the initiation of therapy by absolute neutrophil counts on the 29th day after AZA (ANC29) initiation, and red cell concentrates (RCC) and platelet concentrate (PC) transfusion units required within 28 days from the start of AZA, designated in the present study as RCC28 and PC28, respectively. The survival period was determined from the 29th day of AZA treatment to death from any cause as the conditional survival period after the first cycle of AZA (CS-AZA1). Multivariate analysis demonstrated that severe thrombocytopenia defined by >30 units of PC28 and very poor risk cytogenetics according to IPSS-R were independent prognostic factors for CS-AZA1. The Kyoto Conditional Survival Scoring System was subsequently developed by incorporating severe thrombocytopenia defined by PC28 and very poor risk cytogenetics, which successfully stratified the risks of the patients in CS-AZA1. In conclusion, extreme PC transfusion dependency during the first cycle of AZA and very poor risk cytogenetics are important prognostic factors in AZA monotherapy for MDS.
Collapse
Affiliation(s)
- Yu Inoue
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Haruya Okamoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Akihiro Miyashita
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yuka Kawaji-Kanayama
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Shotaro Chinen
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takahiro Fujino
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Taku Tsukamoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yuji Shimura
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Shinsuke Mizutani
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hiroto Kaneko
- Department of Hematology, Aiseikai Yamashina Hospital, Kyoto 607-8086, Japan
| | - Saeko Kuwahara-Ota
- Department of Hematology, Japan Community Health Care Organization Kyoto Kuramaguchi Medical Center, Kyoto 603-8151, Japan
| | - Shin-Ichi Fuchida
- Department of Hematology, Japan Community Health Care Organization Kyoto Kuramaguchi Medical Center, Kyoto 603-8151, Japan
| | - Daichi Nishiyama
- Department of Hematology, Fukuchiyama City Hospital, Fukuchiyama, Kyoto 620-0056, Japan
| | - Koichi Hirakawa
- Department of Hematology, Fukuchiyama City Hospital, Fukuchiyama, Kyoto 620-0056, Japan
| | - Hitoji Uchiyama
- Department of Hematology, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto 605-0981, Japan
| | - Nobuhiko Uoshima
- Department of Hematology, Japanese Red Cross Kyoto Daini Hospital, Kyoto 602-8031, Japan
| | - Eri Kawata
- Department of Hematology, Matsushita Memorial Hospital, Moriguchi, Osaka 570-8540, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
8
|
WU J, HOU L, ZHANG X, Elizabeth G, GAO C, WANG J. Efficacy of Yisui granule on myelodysplastic syndromes in SKM-1 mouse xenograft model through suppressing Wnt/β-catenin signaling pathway. J TRADIT CHIN MED 2024; 44:78-87. [PMID: 38213242 PMCID: PMC10774724 DOI: 10.19852/j.cnki.jtcm.20231204.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/21/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVE To unmask the underlying mechanisms of Yisui granule (, YSG) for the treatment of Myelodysplastic syndromes (MDS). METHODS Our study used an SKM-1 mouse xenograft model of MDS to explore the anti-tumor potential of YSG and its safety, assess its effect on overall survival (OS), and evaluate whether its mechanism is associated with the demethylation of the secreted frizzled related protein 5 (sFRP5) gene and suppressing Wnt/β-catenin pathway. Bisulfite amplicon sequencing was applied to detect the level of methylation of the sFRP5 gene; western blotting, immunofluorescence staining, and real-time Polymerase Chain Reaction were performed to detect DNA methyltransferase 1 (DNMT1), sFRP5, and other Wnt/β-catenin pathway-related mRNA and protein expression. RESULTS The results showed that high-dosage YSG exerted an anti-tumor effect similar to that of decitabine, improved OS, and reduced long-term adverse effects in the long term. Mechanically, YSG reduced the expression of DNMT1 methyltransferase, decreased the methylation, and increased the expression of the Wnt/β-catenin pathway antagonist-sFRP5. Furthermore, components of the Wnt/β-catenin pathway, including Wnt3a, β-catenin, c-Myc, and cyclinD1, were down-regulated in response to YSG, suggesting that YSG could treat MDS by demethylating the sFRP5 gene and suppressing the Wnt/β-catenin pathway. CONCLUSIONS Our findings demonstrated that YSG could be used alone or in combination with decitabine to improve outcomes in the MDS animal model, providing an alternative solution for treating MDS.
Collapse
Affiliation(s)
- Jieya WU
- 1 Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Li HOU
- 1 Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiaoyuan ZHANG
- 1 Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Gullen Elizabeth
- 2 Department of Pharmacology, Yale Medical School, New Haven, CT 06510, USA
| | - Chong GAO
- 3 Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Jing WANG
- 1 Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| |
Collapse
|
9
|
Schmutz M, Zucknick M, Schlenk RF, Mertens D, Benner A, Weichenhan D, Mücke O, Döhner K, Plass C, Bullinger L, Claus R. Predictive value of DNA methylation patterns in AML patients treated with an azacytidine containing induction regimen. Clin Epigenetics 2023; 15:171. [PMID: 37885041 PMCID: PMC10601277 DOI: 10.1186/s13148-023-01580-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous disease with a poor prognosis. Dysregulation of the epigenetic machinery is a significant contributor to disease development. Some AML patients benefit from treatment with hypomethylating agents (HMAs), but no predictive biomarkers for therapy response exist. Here, we investigated whether unbiased genome-wide assessment of pre-treatment DNA-methylation profiles in AML bone marrow blasts can help to identify patients who will achieve a remission after an azacytidine-containing induction regimen. RESULTS A total of n = 155 patients with newly diagnosed AML treated in the AMLSG 12-09 trial were randomly assigned to a screening and a refinement and validation cohort. The cohorts were divided according to azacytidine-containing induction regimens and response status. Methylation status was assessed for 664,227 500-bp-regions using methyl-CpG immunoprecipitation-seq, resulting in 1755 differentially methylated regions (DMRs). Top regions were distilled and included genes such as WNT10A and GATA3. 80% of regions identified as a hit were represented on HumanMethlyation 450k Bead Chips. Quantitative methylation analysis confirmed 90% of these regions (36 of 40 DMRs). A classifier was trained using penalized logistic regression and fivefold cross validation containing 17 CpGs. Validation based on mass spectra generated by MALDI-TOF failed (AUC 0.59). However, discriminative ability was maintained by adding neighboring CpGs. A recomposed classifier with 12 CpGs resulted in an AUC of 0.77. When evaluated in the non-azacytidine containing group, the AUC was 0.76. CONCLUSIONS Our analysis evaluated the value of a whole genome methyl-CpG screening assay for the identification of informative methylation changes. We also compared the informative content and discriminatory power of regions and single CpGs for predicting response to therapy. The relevance of the identified DMRs is supported by their association with key regulatory processes of oncogenic transformation and support the idea of relevant DMRs being enriched at distinct loci rather than evenly distribution across the genome. Predictive response to therapy could be established but lacked specificity for treatment with azacytidine. Our results suggest that a predictive epigenotype carries its methylation information at a complex, genome-wide level, that is confined to regions, rather than to single CpGs. With increasing application of combinatorial regimens, response prediction may become even more complicated.
Collapse
Affiliation(s)
- Maximilian Schmutz
- Hematology and Oncology, Medical Faculty, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuela Zucknick
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway
| | - Richard F Schlenk
- NCT-Trial Center, National Center of Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel Mertens
- Cooperation Unit "Mechanisms of Leukemogenesis", German Cancer Research Center, Heidelberg, Germany
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, Ulm University Medical Center, Ulm, Germany
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dieter Weichenhan
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Mücke
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars Bullinger
- German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
- Department of Hematology, Oncology, and Cancer Immunology, Campus Virchow Klinikum, Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rainer Claus
- Hematology and Oncology, Medical Faculty, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany.
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany.
| |
Collapse
|
10
|
Sharplin K, Proudman W, Chhetri R, Tran ENH, Choong J, Kutyna M, Selby P, Sapio A, Friel O, Khanna S, Singhal D, Damin M, Ross D, Yeung D, Thomas D, Kok CH, Hiwase D. A Personalized Risk Model for Azacitidine Outcome in Myelodysplastic Syndrome and Other Myeloid Neoplasms Identified by Machine Learning Model Utilizing Real-World Data. Cancers (Basel) 2023; 15:4019. [PMID: 37627047 PMCID: PMC10452100 DOI: 10.3390/cancers15164019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Azacitidine is an approved therapy for higher-risk myelodysplastic syndrome (MDS). However, only 30-40% patients respond to azacitidine, and the responses may take up to six cycles to become evident. Delayed responses and the myelosuppressive effects of azacitidine make it challenging to predict which patients will benefit. This is further compounded by a lack of uniform prognostic tools to identify patients at risk of early treatment failure. Hence, we performed a retrospective analysis of 273 consecutive azacytidine-treated patients. The median overall survival was 16.25 months with only 9% alive at 5 years. By using pre-treatment variables incorporated into a random forest machine learning model, we successfully identified those patients unlikely to benefit from azacytidine upfront (7.99 vs. 22.8 months, p < 0.0001). This model also identified those who required significantly more hospitalizations and transfusion support. Notably, it accurately predicted survival outcomes, outperforming the existing prognostic scoring system. By integrating somatic mutations, we further refined the model and identified three distinct risk groups with significant differences in survival (5.6 vs. 10.5 vs. 43.5 months, p < 0.0001). These real-world findings emphasize the urgent need for personalized prediction tools tailored to hypomethylating agents, reducing unnecessary complications and resource utilization in MDS treatment.
Collapse
Affiliation(s)
- Kirsty Sharplin
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - William Proudman
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - Rakchha Chhetri
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Elizabeth Ngoc Hoa Tran
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Jamie Choong
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - Monika Kutyna
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Philip Selby
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Aidan Sapio
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - Oisin Friel
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Beaumont Hospital, D09 V2N0 Dublin, Ireland
| | - Shreyas Khanna
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Deepak Singhal
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Michelle Damin
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - David Ross
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
- Genetic and Molecular Pathology, SA Pathology, Adelaide, SA 5000, Australia
| | - David Yeung
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Daniel Thomas
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Chung H. Kok
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Devendra Hiwase
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| |
Collapse
|
11
|
Sauta E, Robin M, Bersanelli M, Travaglino E, Meggendorfer M, Zhao LP, Caballero Berrocal JC, Sala C, Maggioni G, Bernardi M, Di Grazia C, Vago L, Rivoli G, Borin L, D'Amico S, Tentori CA, Ubezio M, Campagna A, Russo A, Mannina D, Lanino L, Chiusolo P, Giaccone L, Voso MT, Riva M, Oliva EN, Zampini M, Riva E, Nibourel O, Bicchieri M, Bolli N, Rambaldi A, Passamonti F, Savevski V, Santoro A, Germing U, Kordasti S, Santini V, Diez-Campelo M, Sanz G, Sole F, Kern W, Platzbecker U, Ades L, Fenaux P, Haferlach T, Castellani G, Della Porta MG. Real-World Validation of Molecular International Prognostic Scoring System for Myelodysplastic Syndromes. J Clin Oncol 2023; 41:2827-2842. [PMID: 36930857 PMCID: PMC10414702 DOI: 10.1200/jco.22.01784] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/13/2023] [Indexed: 03/19/2023] Open
Abstract
PURPOSE Myelodysplastic syndromes (MDS) are heterogeneous myeloid neoplasms in which a risk-adapted treatment strategy is needed. Recently, a new clinical-molecular prognostic model, the Molecular International Prognostic Scoring System (IPSS-M) was proposed to improve the prediction of clinical outcome of the currently available tool (Revised International Prognostic Scoring System [IPSS-R]). We aimed to provide an extensive validation of IPSS-M. METHODS A total of 2,876 patients with primary MDS from the GenoMed4All consortium were retrospectively analyzed. RESULTS IPSS-M improved prognostic discrimination across all clinical end points with respect to IPSS-R (concordance was 0.81 v 0.74 for overall survival and 0.89 v 0.76 for leukemia-free survival, respectively). This was true even in those patients without detectable gene mutations. Compared with the IPSS-R based stratification, the IPSS-M risk group changed in 46% of patients (23.6% and 22.4% of subjects were upstaged and downstaged, respectively).In patients treated with hematopoietic stem cell transplantation (HSCT), IPSS-M significantly improved the prediction of the risk of disease relapse and the probability of post-transplantation survival versus IPSS-R (concordance was 0.76 v 0.60 for overall survival and 0.89 v 0.70 for probability of relapse, respectively). In high-risk patients treated with hypomethylating agents (HMA), IPSS-M failed to stratify individual probability of response; response duration and probability of survival were inversely related to IPSS-M risk.Finally, we tested the accuracy in predicting IPSS-M when molecular information was missed and we defined a minimum set of 15 relevant genes associated with high performance of the score. CONCLUSION IPSS-M improves MDS prognostication and might result in a more effective selection of candidates to HSCT. Additional factors other than gene mutations can be involved in determining HMA sensitivity. The definition of a minimum set of relevant genes may facilitate the clinical implementation of the score.
Collapse
Affiliation(s)
- Elisabetta Sauta
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Marie Robin
- Department of Hematology and Bone Marrow Transplantation, Hôpital Saint-Louis/Assistance Publique-Hôpitaux de Paris (AP-HP)/University Paris 7, Paris, France
| | - Matteo Bersanelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Erica Travaglino
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | | | - Lin-Pierre Zhao
- Department of Hematology and Bone Marrow Transplantation, Hôpital Saint-Louis/Assistance Publique-Hôpitaux de Paris (AP-HP)/University Paris 7, Paris, France
| | | | - Claudia Sala
- Experimental, Diagnostic and Specialty Medicine, DIMES, Bologna, Italy
| | - Giulia Maggioni
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Massimo Bernardi
- Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, University Vita-Salute San Raffaele, Milan, Italy
| | - Carmen Di Grazia
- Hematology and Transplant Center, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Luca Vago
- Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, University Vita-Salute San Raffaele, Milan, Italy
| | - Giulia Rivoli
- Hematology and Transplant Center, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Saverio D'Amico
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | | | - Marta Ubezio
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Alessia Campagna
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Antonio Russo
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Daniele Mannina
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Luca Lanino
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Patrizia Chiusolo
- Hematology, IRCCS Fondazione Policlinico Universitario Gemelli & Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luisa Giaccone
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Maria Teresa Voso
- Hematology, Policlinico Tor Vergata & Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Marta Riva
- Hematology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Esther Natalie Oliva
- Hematology, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Matteo Zampini
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Elena Riva
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | | | | | - Niccolo’ Bolli
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Alessandro Rambaldi
- Hematology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Francesco Passamonti
- Hematology, ASST Sette Laghi, Ospedale di Circolo of Varese, Varese, Italy
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Victor Savevski
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Armando Santoro
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Ulrich Germing
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich-Heine-University, University Clinic, Düsseldorf, Germany
| | - Shahram Kordasti
- Haematology, Guy's Hospital and Comprehensive Cancer Centre, King's College, London, United Kingdom
- Hematology Department and Stem Cell Transplant Unit, DISCLIMO-Università Politecnica delle Marche, Ancona, Italy
| | - Valeria Santini
- Hematology, Azienda Ospedaliero-Universitaria Careggi & University of Florence, Florence, Italy
| | - Maria Diez-Campelo
- Hematology Department, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Guillermo Sanz
- Hematology, Hospital Universitario La Fe, Valencia, Spain
| | - Francesc Sole
- Institut de Recerca Contra la Leucèmia Josep Carreras, Barcelona, Spain
| | | | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Lionel Ades
- Department of Hematology and Bone Marrow Transplantation, Hôpital Saint-Louis/Assistance Publique-Hôpitaux de Paris (AP-HP)/University Paris 7, Paris, France
| | - Pierre Fenaux
- Department of Hematology and Bone Marrow Transplantation, Hôpital Saint-Louis/Assistance Publique-Hôpitaux de Paris (AP-HP)/University Paris 7, Paris, France
| | | | | | - Matteo Giovanni Della Porta
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| |
Collapse
|
12
|
Abaza Y, Patel AA. Novel Therapies in Myelodysplastic Syndrome: Where Do Venetoclax and Isocitrate Dehydrogenase Inhibitors Fit in? Cancer J 2023; 29:188-194. [PMID: 37195775 DOI: 10.1097/ppo.0000000000000657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
ABSTRACT Myelodysplastic syndromes (MDSs) are a heterogeneous group of clonal hematopoietic stem cell disorders with treatment approaches tailored to the presence of cytopenias, disease risk, and molecular mutation profile. In higher-risk MDSs, the standard of care are DNA methyltransferase inhibitors, otherwise referred to as hypomethylating agents (HMAs), with consideration for allogeneic hematopoietic stem cell transplantation in appropriate candidates. Given modest complete remission rates (15%-20%) with HMA monotherapy and median overall survival of approximately 18 months, there is much interest in the investigation of combination and targeted treatment approaches. Furthermore, there is no standard treatment approach in patients with progression of disease after HMA therapy. In this review, we aim to summarize the current evidence for the B-cell lymphoma-2 inhibitor, venetoclax, and a variety of isocitrate dehydrogenase inhibitors in the treatment of MDSs along with discussing their potential role in the treatment paradigm of this disease.
Collapse
Affiliation(s)
- Yasmin Abaza
- From the Department of Hematology and Oncology, Northwestern University, Robert Lurie Cancer Center
| | - Anand Ashwin Patel
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, IL
| |
Collapse
|
13
|
Awada H, Gurnari C, Xie Z, Bewersdorf JP, Zeidan AM. What's Next after Hypomethylating Agents Failure in Myeloid Neoplasms? A Rational Approach. Cancers (Basel) 2023; 15:2248. [PMID: 37190176 PMCID: PMC10137017 DOI: 10.3390/cancers15082248] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/07/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Hypomethylating agents (HMA) such as azacitidine and decitabine are a mainstay in the current management of patients with myelodysplastic syndromes/neoplasms (MDS) and acute myeloid leukemia (AML) as either single agents or in multidrug combinations. Resistance to HMA is not uncommon, and it can result due to several tumor cellular adaptations. Several clinical and genomic factors have been identified as predictors of HMA resistance. However, the management of MDS/AML patients after the failure of HMA remains challenging in the absence of standardized guidelines. Indeed, this is an area of active research with several potential therapeutic agents currently under development, some of which have demonstrated therapeutic potential in early clinical trials, especially in cases with particular mutational characteristics. Here, we review the latest findings and give a rational approach for such a challenging scenario.
Collapse
Affiliation(s)
- Hussein Awada
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Zhuoer Xie
- Department of Hematology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Comprehensive Cancer Center, New York, NY 10065, USA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University and Yale Cancer Center, New Haven, CT 06511, USA
| |
Collapse
|
14
|
Wang C, Sallman DA. Current Therapeutic Landscape in Lower Risk Myelodysplastic Syndromes. Curr Treat Options Oncol 2023; 24:387-408. [PMID: 36966266 DOI: 10.1007/s11864-023-01062-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 03/27/2023]
Abstract
OPINION STATEMENT Lower risk myelodysplastic syndromes are typically characterized by an indolent disease course with a relatively low risk of transformation into acute myeloid leukemia. These patients are classically identified using the revised International Prognostic Scoring System and most likely its molecular version in the near future which may change the paradigm of treatment. The overall goals of care are symptomatic control to reduce transfusion requirements and improve quality of life. Symptomatic anemia is the most common indication to initiate disease-specific therapies after the optimization of supportive measures. Currently, erythropoiesis-stimulating agents remain the standard upfront therapy for anemia, and patients with del(5q) cytogenetic changes can benefit from lenalidomide monotherapy. Other therapeutic options after failure of upfront treatment include luspatercept, hypomethylating agents, and immunosuppressive therapies after taking into account of individualized disease features. Allogeneic hematopoietic stem cell transplant is the only potentially curative option and is usually reserved for medically fit patients with severe symptomatic cytopenias who failed all standard options and/or the disease is progressing toward higher risk categories. Fortunately, novel investigational therapies are rapidly emerging by targeting different biological processes contributing to MDS pathogenesis, and eligible patients should be managed in clinical trials if available.
Collapse
Affiliation(s)
- Chen Wang
- Department of Internal Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
15
|
Wang X, Jing H, Li H. A novel cuproptosis-related lncRNA signature to predict prognosis and immune landscape of lung adenocarcinoma. Transl Lung Cancer Res 2023; 12:230-246. [PMID: 36895935 PMCID: PMC9989802 DOI: 10.21037/tlcr-22-500] [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: 07/02/2022] [Accepted: 01/02/2023] [Indexed: 02/28/2023]
Abstract
Background Cuproptosis, a recently discovered type of programmed cell death (PCD), paves a new avenue for cancer treatment. It has been revealed that PCD-related lncRNAs play a critical role in various biological processes of lung adenocarcinoma (LUAD). However, the role of cuproptosis-related lncRNA (CuRLs) remains unclear. This study aimed to identify and validate a CuRLs-based signature for the prognostic prediction of patients with LUAD. Methods RNA sequencing data and clinical information of LUAD were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Pearson correlation analysis was used to identify CuRLs. Univariate Cox regression analysis, Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression, and stepwise multivariate Cox analysis were applied to construct a novel prognostic CuRLs signature. A nomogram was developed for the prediction of patient survival outcomes. Gene set variation analysis (GSVA), gene set enrichment analysis (GSEA), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were utilized to explore potential functions underlying the CuRLs signature. Patients were divided into low- and high-risk groups. Several algorithms, such as tumor immune estimation resource (TIMER), cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT), and QuanTIseq, were combined to comprehensively investigate the differences in immune landscape between different risk groups. Sensitivity to common anticancer drugs was analyzed using the pRRophetic algorithm. Results We constructed a novel prognostic signature based on 10 CuRLs, including CARD8-AS1, RUNDC3A-AS1, TMPO-AS1, MIR31HG, SEPSECS-AS1, DLGAP1-AS1, LINC01137, ZSCAN16-AS1, APTR, and ELOA-AS1. This 10-CuRLs risk signature showed great diagnostic accuracy combined with traditional clinical risk factors, and a nomogram was constructed for potential clinical translation. The tumor immune microenvironment was significantly different between different risk groups. Among drugs commonly used in the treatment of lung cancer, the sensitivity of cisplatin, docetaxel, gemcitabine, gefitinib, and paclitaxel was higher in low-risk patients, and patients in the low-risk group may benefit more from imatinib. Conclusions These results revealed the outstanding contribution of the CuRLs signature to the evaluation of prognosis and treatment modalities for patients with LUAD. The differences in characteristics between different risk groups provide an opportunity for better patient stratification and to explore novel drugs in different risk groups.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Jing
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Hasegawa K, Wei AH, Garcia-Manero G, Daver NG, Rajakumaraswamy N, Iqbal S, Chan RJ, Hu H, Tse P, Yan J, Zoratti MJ, Xie F, Sallman DA. Azacitidine Monotherapy in Patients With Treatment-Naïve Higher-risk Myelodysplastic Syndrome: A Systematic Literature Review and Meta-analysis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:127-137. [PMID: 36428152 DOI: 10.1016/j.clml.2022.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND The global incidence of myelodysplastic syndromes (MDS) has been estimated as 0.06 to 0.26/100,000. Since their introduction, hypomethylating agents have played a central role in the treatment of MDS, with heterogeneous real-world outcomes. MATERIALS AND METHODS We assessed and synthesized clinical outcomes of azacitidine (AZA) monotherapy in treatment-naïve patients with higher-risk MDS. A systematic literature review was conducted by searching MEDLINE, Embase, and CENTRAL to identify randomized clinical trials (RCTs) and observational studies, both prospective and retrospective, reporting complete remission (CR), partial remission (PR), overall survival (OS), duration of response (DOR), time-to-response (TTR), and myelosuppressive adverse events (AEs) for patients treated with AZA monotherapy. Noncomparative meta-analyses were used to summarize effects. RESULTS The search identified 3250 abstracts, of which 34 publications describing 16 studies (5 RCTs, 3 prospective, and 8 retrospective observational) were included. Across all studies, pooled CR was 16%; PR was 6%; Median OS was 16.4 months; median DOR was 10.1 months; median TTR was 4.6 months. Proportions of grade 3/4 anemia and thrombocytopenia AEs were 10% and 30%. CONCLUSIONS The effectiveness and efficacy of AZA monotherapy-as measured by CR and median OS-was limited. These findings highlight a significant unmet medical need for effective treatments for patients with higher-risk MDS.
Collapse
Affiliation(s)
| | - Andrew H Wei
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
| | | | - Naval G Daver
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | - Hao Hu
- Gilead Sciences, Inc., Foster City, CA
| | | | - Jiajun Yan
- McMaster University, Hamilton, ON, Canada
| | | | - Feng Xie
- McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
17
|
Koenig KL, Borate U. New investigational combinations for higher-risk MDS. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:368-374. [PMID: 36485141 PMCID: PMC9820246 DOI: 10.1182/hematology.2022000351] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myelodysplastic syndromes (MDS) are typically a hematologic malignancy of older adults characterized by dysplastic hematopoiesis, cytopenia(s), and risk of acute myeloid leukemia transformation. The treatment approach to MDS depends largely on risk stratification of an individual's disease, most commonly using the Revised International Prognostic Scoring System, which takes into account peripheral blood cytopenias and bone marrow blast percentage and cytogenetics. The current standard of care for patients with higher-risk MDS (HR-MDS) includes hypomethylating agents (HMAs), decitabine and azacitidine, and allogenic stem cell transplant for patients able to undergo this therapy. However, leukemic transformation remains a significant challenge, and outcomes with these current therapies are still dismal. There are several novel therapies in development aiming to improve upon the outcomes of single-agent HMA therapy using combination strategies with HMAs. Here we discuss the current standard of care for HR-MDS treatment and explore some of the most promising combination therapies coming out of the pipeline for HR-MDS.
Collapse
|
18
|
Komrokji RS, Singh AM, Ali NA, Chan O, Padron E, Sweet K, Kuykendall A, Lancet JE, Sallman DA. Assessing the role of venetoclax in combination with hypomethylating agents in higher risk myelodysplastic syndrome. Blood Cancer J 2022; 12:148. [PMID: 36329025 PMCID: PMC9633639 DOI: 10.1038/s41408-022-00744-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Rami S. Komrokji
- grid.468198.a0000 0000 9891 5233Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Avani M. Singh
- grid.170693.a0000 0001 2353 285XDepartment of Hematology/Oncology, University of South Florida & Moffitt Cancer Center, Tampa, FL USA
| | - Najla Al Ali
- grid.468198.a0000 0000 9891 5233Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Onyee Chan
- grid.468198.a0000 0000 9891 5233Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Eric Padron
- grid.468198.a0000 0000 9891 5233Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Kendra Sweet
- grid.468198.a0000 0000 9891 5233Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Andrew Kuykendall
- grid.468198.a0000 0000 9891 5233Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Jeffrey E. Lancet
- grid.468198.a0000 0000 9891 5233Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - David A. Sallman
- grid.468198.a0000 0000 9891 5233Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| |
Collapse
|
19
|
Mohty R, Al Hamed R, Bazarbachi A, Brissot E, Nagler A, Zeidan A, Mohty M. Treatment of myelodysplastic syndromes in the era of precision medicine and immunomodulatory drugs: a focus on higher-risk disease. J Hematol Oncol 2022; 15:124. [PMID: 36045390 PMCID: PMC9429775 DOI: 10.1186/s13045-022-01346-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous clonal disease of myeloid neoplasms characterized by ineffective hematopoiesis, variable degree of cytopenias, and an increased risk of progression to acute myeloid leukemia (AML). Molecular and genetic characterization of MDS has led to a better understanding of the disease pathophysiology and is leading to the development of novel therapies. Targeted and immune therapies have shown promising results in different hematologic malignancies. However, their potential use in MDS is yet to be fully defined. Here, we review the most recent advances in therapeutic approaches in MDS, focusing on higher-risk disease. Allogeneic hematopoietic cell transplantation is beyond the scope of this article.
Collapse
Affiliation(s)
- Razan Mohty
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Rama Al Hamed
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Eolia Brissot
- Department of Clinical Hematology and Cellular Therapy, Saint-Antoine Hospital, AP-HP, Sorbonne University, and INSERM, Saint-Antoine Research Centre, 75012, Paris, France
| | - Arnon Nagler
- Hematology and Bone Marrow Transplant Unit, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Amer Zeidan
- Division of Hematology/Oncology, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Mohamad Mohty
- Department of Clinical Hematology and Cellular Therapy, Saint-Antoine Hospital, AP-HP, Sorbonne University, and INSERM, Saint-Antoine Research Centre, 75012, Paris, France.
| |
Collapse
|
20
|
Prospective comparison of 5- and 7-day administration of azacitidine for myelodysplastic syndromes: a JALSG MDS212 trial. Int J Hematol 2022; 116:228-238. [PMID: 35508695 DOI: 10.1007/s12185-022-03347-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 10/18/2022]
Abstract
The hypomethylating agent azacitidine (AZA) significantly extends overall survival (OS) in patients with higher risk myelodysplastic syndromes (MDS), when compared with other conventional care regimens, including supportive care and low-dose and intensive chemotherapy. However, the effects of 5- and 7-day treatment schedules of AZA (AZA-5 and AZA-7, respectively) on the OS of MDS patients had not been compared prospectively. We started a phase 3 trial comparing the effects of AZA-7 and AZA-5 on MDS patients with refractory anemia with excess blasts (RAEB) and RAEB in transformation (RAEB-T). However, this trial was prematurely terminated because of poor recruitment. Using all data, there was no significant difference in the OS of patients between AZA-7 (92 patients) and AZA-5 (95 patients), with the 2-year OS rates of AZA-7 and AZA-5 at 36.4% and 25.8%, respectively (P = 0.293). Adverse event profiles were similar between the two groups. Interestingly, data of the centrally diagnosed RAEB and RAEB-T cases showed that AZA-7 significantly prolonged the time to leukemia transformation compared with AZA-5 (P = 0.022), confirmed by multivariate analysis. Although this trial could not provide definite evidence, the results support the use of AZA-7 for RAEB and RAEB-T. (UMIN Clinical Trials Registry UMIN000009633).
Collapse
|
21
|
Awada H, Gurnari C, Durmaz A, Awada H, Pagliuca S, Visconte V. Personalized Risk Schemes and Machine Learning to Empower Genomic Prognostication Models in Myelodysplastic Syndromes. Int J Mol Sci 2022; 23:2802. [PMID: 35269943 PMCID: PMC8911403 DOI: 10.3390/ijms23052802] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are characterized by variable clinical manifestations and outcomes. Several prognostic systems relying on clinical factors and cytogenetic abnormalities have been developed to help stratify MDS patients into different risk categories of distinct prognoses and therapeutic implications. The current abundance of molecular information poses the challenges of precisely defining patients' molecular profiles and their incorporation in clinically established diagnostic and prognostic schemes. Perhaps the prognostic power of the current systems can be boosted by incorporating molecular features. Machine learning (ML) algorithms can be helpful in developing more precise prognostication models that integrate complex genomic interactions at a higher dimensional level. These techniques can potentially generate automated diagnostic and prognostic models and assist in advancing personalized therapies. This review highlights the current prognostication models used in MDS while shedding light on the latest achievements in ML-based research.
Collapse
Affiliation(s)
- Hussein Awada
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (H.A.); (C.G.); (A.D.); (S.P.)
| | - Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (H.A.); (C.G.); (A.D.); (S.P.)
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Arda Durmaz
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (H.A.); (C.G.); (A.D.); (S.P.)
| | - Hassan Awada
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Simona Pagliuca
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (H.A.); (C.G.); (A.D.); (S.P.)
- Department of Clinical Hematology, CHRU Nancy, CEDEX, 54035 Nancy, France
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (H.A.); (C.G.); (A.D.); (S.P.)
| |
Collapse
|
22
|
A Randomized Phase 2 Trial of Azacitidine ± Durvalumab as First-line Therapy for Higher-Risk Myelodysplastic Syndromes. Blood Adv 2021; 6:2207-2218. [PMID: 34972214 PMCID: PMC9006291 DOI: 10.1182/bloodadvances.2021005487] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/29/2021] [Indexed: 11/20/2022] Open
Abstract
This is the first reported randomized trial of immune checkpoint inhibitor therapy in HR-MDS. Azacitidine combined with the PD-L1 inhibitor durvalumab was feasible but did not improve outcomes over azacitidine alone.
Azacitidine-mediated hypomethylation promotes tumor cell immune recognition but may increase the expression of inhibitory immune checkpoint molecules. We conducted the first randomized phase 2 study of azacitidine plus the immune checkpoint inhibitor durvalumab vs azacitidine monotherapy as first-line treatment for higher-risk myelodysplastic syndromes (HR-MDS). In all, 84 patients received 75 mg/m2 subcutaneous azacitidine (days 1-7 every 4 weeks) combined with 1500 mg intravenous durvalumab on day 1 every 4 weeks (Arm A) for at least 6 cycles or 75 mg/m² subcutaneous azacitidine alone (days 1-7 every 4 weeks) for at least 6 cycles (Arm B). After a median follow-up of 15.25 months, 8 patients in Arm A and 6 in Arm B remained on treatment. Patients in Arm A received a median of 7.9 treatment cycles and those in Arm B received a median of 7.0 treatment cycles with 73.7% and 65.9%, respectively, completing ≥4 cycles. The overall response rate (primary end point) was 61.9% in Arm A (26 of 42) and 47.6% in Arm B (20 of 42; P = .18), and median overall survival was 11.6 months (95% confidence interval, 9.5 months to not evaluable) vs 16.7 months (95% confidence interval, 9.8-23.5 months; P = .74). Durvalumab-related adverse events (AEs) were reported by 71.1% of patients; azacitidine-related AEs were reported by 82% (Arm A) and 81% (Arm B). Grade 3 or 4 hematologic AEs were reported in 89.5% (Arm A) vs 68.3% (Arm B) of patients. Patients with TP53 mutations tended to have a worse response than patients without these mutations. Azacitidine increased programmed cell death ligand 1 (PD-L1 [CD274]) surface expression on bone marrow granulocytes and monocytes, but not blasts, in both arms. In summary, combining azacitidine with durvalumab in patients with HR-MDS was feasible but with more toxicities and without significant improvement in clinical outcomes over azacitidine alone. This trial was registered at www.clinicaltrials.gov as #NCT02775903.
Collapse
|
23
|
Ball S, Komrokji RS, Sallman DA. Prognostic scoring systems and risk stratification in myelodysplastic syndrome: focus on integration of molecular profile. Leuk Lymphoma 2021; 63:1281-1291. [PMID: 34933652 DOI: 10.1080/10428194.2021.2018579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Myelodysplastic syndromes (MDS) form a clinically and molecularly heterogeneous disease group. Precise risk stratification remains crucial for choosing optimal management strategies. Several conventional prognostic scoring systems have been developed and validated in the MDS population. These risk models divide patients into prognostic subgroups based on clinical and cytogenetic characteristics. Lack of dynamicity, variable risk estimate across models, and heterogeneity within intermediate-risk group are the limitations of traditional models like IPSS-R, with questionable relevance of these scoring systems in treated MDS patients. Recent progress in next-generation sequencing techniques has improved understanding of the distribution and prognostic importance of recurrent genetic mutations in MDS. Early studies have suggested that incorporating mutations in risk stratification could supplement IPSS-R in further refining the model's performance in predicting overall survival and risk of transformation to acute myeloid leukemia and should translate into a molecularly driven prognostication approach in the near future.
Collapse
Affiliation(s)
- Somedeb Ball
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Rami S Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
24
|
Volpe VO, Garcia-Manero G, Komrokji RS. Myelodysplastic Syndromes: A New Decade. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:1-16. [PMID: 34544674 DOI: 10.1016/j.clml.2021.07.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
Myelodysplastic syndromes (MDS) are a group of heterogeneous clonal hematopoietic stem cell disorders. The 2020 Surveillance, Epidemiology, and End Results data demonstrates the incidence rate of MDS increases with age especially in those greater than 70 years of age. Risk stratification that impact prognosis, survival, and rate of acute myeloid leukemia (AML) transformation in MDS is largely dependent on revised International Prognostic Scoring System along with molecular genetic testing as a supplement. Low risk MDS typically have a more indolent disease course in which treatment is only initiated to ameliorate symptoms of cytopenias. In many, anemia is the most common cytopenia requiring treatment and erythroid stimulating agents, are considered first line. In contrast, high risk MDS tend to behave more aggressively for which treatment should be initiated rapidly with Hypomethylating Agents (HMA) being in the frontline. In those with high risk MDS and eligible, evaluation for allogeneic stem cell transplant should be considered as this is the only potential curative option for MDS. With the use of molecular genetic testing, a personalized approach to therapy in MDS has ensued. As the treatment landscape in MDS continues to flourish with novel targeted agents, we ambitiously seek to improve survival rates especially among the relapsed/refractory and transplant ineligible.
Collapse
Affiliation(s)
- Virginia O Volpe
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | | | - Rami S Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL.
| |
Collapse
|
25
|
Komrokji R, Al Ali N, Padron E, Lancet J, Nazha A, Steensma D, DeZern A, Roboz G, Garcia-Manero G, Sekeres MA, Sallman D. What is the optimal time to initiate hypomethylating agents (HMAs) in higher risk myelodysplastic syndromes (MDSs)? Leuk Lymphoma 2021; 62:2762-2767. [PMID: 34114922 DOI: 10.1080/10428194.2021.1938028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Hypomethylating agents (HMAs) are the standard of care for higher risk MDS (HR-MDS) patients. The current dogma is to begin HMA therapy in all HR-MDS patients at the time of initial diagnosis. We investigated the impact of the timing of HMA initiation among HR-MDS patients presenting with adequate blood counts to discern the possible benefit of early treatment based solely on disease risk. We identified 320 HR-MDS patients with adequate hematopoiesis who were treated with HMA. The complete response rates were 21%, 26%, 23%, and 7% respectively for patients treated within 30, 31-60, 61-90, and more than 90 days from time of diagnosis (p=.046). The median OS from the date of diagnosis was 641, 550, 979, and 806 days, respectively (p=.2). A delay in initiating HMA therapy in HR-MDS patients with adequate blood counts is not associated with worsened outcomes.
Collapse
Affiliation(s)
- Rami Komrokji
- Malignant Hematology Department, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Najla Al Ali
- Malignant Hematology Department, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Eric Padron
- Malignant Hematology Department, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jeffrey Lancet
- Malignant Hematology Department, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Aziz Nazha
- Leukemia Program, Cleveland Clinic, Cleveland, OH, USA
| | | | - Amy DeZern
- Kimmel Cancer Center/Johns Hopkins University, Baltimore, MD, USA
| | - Gail Roboz
- Weill Cornell Medical College, New York, NY, USA
| | | | | | - David Sallman
- Malignant Hematology Department, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
26
|
Garcia JS, Swords RT, Roboz GJ, Jacoby MA, Garcia-Manero G, Hong WJ, Yang X, Zhou Y, Platzbecker U, Steensma DP, Wolff JE, Fenaux P. A systematic review of higher-risk myelodysplastic syndromes clinical trials to determine the benchmark of azacitidine and explore alternative endpoints for overall survival. Leuk Res 2021; 104:106555. [PMID: 33705966 DOI: 10.1016/j.leukres.2021.106555] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/29/2022]
Abstract
The hypomethylating agent azacitidine can prolong overall survival (OS) in patients with higher risk-myelodysplastic syndromes (HR-MDS) compared to conventional regimens. However, outcomes differ largely between studies, making it challenging to determine the contribution of novel therapies added to azacitidine. Further, a discrepancy is seen between complete (CR) or partial (PR) response rates and OS improvement with azacitidine, making it challenging to rely on earlier endpoints than OS. We conducted a systematic literature search and study-level systematic review of 237 clinical studies to better understand outcomes for HR-MDS patients treated with azacitidine. Pooled marrow CR was 9% (N = 2654; 95% CI: 6-13 %), CR rate was 17 % (N = 6943; 95% CI: 15-20 %), and median OS (mOS) was 18.6 months (N = 2820; 95% CI: 15.3-21.9). A weak correlation to mOS was detected with CR rate (207 patient cohorts, Pearson's r = 0.315; P < 0.0005), and a much stronger correlation with median progression-free survival (mPFS) (r=0.88, P = 3 × 10-14). Six-months progression-free survival rates correlated with 1-year OS rates but were only infrequently reported (N = 41 patient cohorts) therefore not allowing a robust recommendation for a surrogate to the established OS endpoint. Larger patient numbers and patient-level data appear necessary, especially for designing future clinical trials using azacitidine combinations.
Collapse
Affiliation(s)
| | | | - Gail J Roboz
- Weill Medical College of Cornell University and New York-Presbyterian Hospital, NY, NY, USA
| | - Meagan A Jacoby
- Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | | - Pierre Fenaux
- Hôpital Saint-Louis, Assistance Publique - Hôpitaux de Paris, Université de Paris, Paris, France
| |
Collapse
|
27
|
Hu N, Qin T, Du X, Wang B, Wang X, Xu Z, Pan L, Qu S, Xiao Z. Evaluation of Reduced-Dose Decitabine and Azacitidine for Treating Myelodysplastic Syndromes: A Retrospective Study. Med Sci Monit 2021; 27:e928454. [PMID: 33514682 PMCID: PMC7856837 DOI: 10.12659/msm.928454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Hypomethylating agents (HMA) are considered the first-line therapy for high-risk myelodysplastic syndromes (MDS). However, as the efficacy and safety of rational dosing regimens are lacking, we evaluated the effectiveness and safety of reduced-dose azacitidine (AZA) vs. decitabine (DAC) in adult MDS patients. Material/Methods This retrospective study was conducted at the Institute of Hematology & Blood Diseases Hospital, for hospitalized MDS patients diagnosed (WHO 2008 classification criteria) from May 2006 to February 2020. These AZA- and DCA-naive patients treated with AZA 100 mg/(m2·day) for 5 days to 7 days or DAC 20 mg/(m2·day) for 3 days to 4 days, or 20 mg/(m2·day) 1 day/week for 3 weeks/month were assessed for treatment responses and adverse events. Results Of the 158 enrolled MDS patients, 120 and 38 patients were administered reduced-dose DAC and AZA, respectively. All the patients received a median of 2 treatment cycles. The overall response rates (ORR) were 50.0% and 73.3% in the AZA and DAC groups, respectively (P=0.007). The percentage of platelet transfusion dependence in the AZA group was lower than the DAC group (P=0.026). The multivariate analysis demonstrated that the DAC treatment was a significant factor for improved responses (OR 2.928; 95% CI 1.267–6.896; P=0.012), and the absolute neutrophil count (ANC) was a predictor of the ORR (OR 0.725; 95% CI 0.558–0.898; P=0.008). Neutropenia (P=0.016) and infection (P=0.032) incidences were higher in the DAC group. Conclusions The reduced-dose DAC group demonstrated a better response than the AZA group in MDS patients with different prognostic risks. The patients’ pre-treatment ANC was a significant factor associated with the ORR.
Collapse
Affiliation(s)
- Naibo Hu
- Department of Myelodysplastic Syndromes, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (mainland).,Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Tiejun Qin
- Department of Myelodysplastic Syndromes, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (mainland)
| | - Xiaoyan Du
- Medical Department, Yidu Cloud (Beijing) Technology Co., Ltd., Beijing, China (mainland)
| | - Bingyi Wang
- Medical Department, Yidu Cloud (Beijing) Technology Co., Ltd., Beijing, China (mainland)
| | - Xiaoyun Wang
- Medical Department, Yidu Cloud (Beijing) Technology Co., Ltd., Beijing, China (mainland)
| | - Zefeng Xu
- Department of Myelodysplastic Syndromes, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (mainland)
| | - Lijuan Pan
- Department of Myelodysplastic Syndromes, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (mainland)
| | - Shiqiang Qu
- Department of Myelodysplastic Syndromes, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (mainland)
| | - Zhijian Xiao
- Department of Myelodysplastic Syndromes, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (mainland)
| |
Collapse
|
28
|
Lewis R, Bewersdorf JP, Zeidan AM. Clinical Management of Anemia in Patients with Myelodysplastic Syndromes: An Update on Emerging Therapeutic Options. Cancer Manag Res 2021; 13:645-657. [PMID: 33531837 PMCID: PMC7846829 DOI: 10.2147/cmar.s240600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 01/13/2021] [Indexed: 12/18/2022] Open
Abstract
For the majority of patients with lower-risk myelodysplastic syndrome (LR-MDS), one of the primary clinical goals is to alleviate the symptoms associated with the resultant cytopenias and to minimize the transfusion burden. While supportive red blood cell (RBC) transfusions and erythropoiesis-stimulating agents (ESAs) may lead to clinical improvement, frequent transfusions are often complicated by iron overload and decreased quality of life; furthermore, most patients either do not respond to ESAs or will eventually develop resistance. As such, there is a great need for further therapeutic options in the management of anemia related to MDS. Several additional therapeutics are now available in select patients with LR-MDS and symptomatic anemia including luspatercept, lenalidomide, and immunosuppressive therapy. Furthermore, several novel agents are currently in development to address this area of clinical need such as imetelstat and roxadustat. In this article, we review the currently available therapeutic options for symptomatic anemia in LR-MDS as well as review the therapeutic agents in development.
Collapse
Affiliation(s)
- Russell Lewis
- Department of Medicine, Section of Hematology, Yale University, New Haven, CT, USA
| | | | - Amer M Zeidan
- Department of Medicine, Section of Hematology, Yale University, New Haven, CT, USA
| |
Collapse
|
29
|
Stein EM, Bonifacio G, Latremouille-Viau D, Shi S, Guerin A, Wu EQ, Sadek I, Cao X. Treatment patterns and outcomes in patients with myelodysplastic syndromes treated with hypomethylating agents: a SEER-Medicare analysis. Leuk Lymphoma 2021; 62:1411-1421. [PMID: 33430673 DOI: 10.1080/10428194.2020.1869959] [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] [Indexed: 12/30/2022]
Abstract
To describe real-world treatment patterns and outcomes among adult patients with myelodysplastic syndromes (MDS) treated with hypomethylating agents (HMA), patients were identified in the SEER-Medicare database (01/2006-12/2016); 3,046 patients with MDS treated with HMA were included. An algorithm was developed to categorize patients into MDS risk groups: the majority of patients were classified as Higher-risk (70.9%), 8.0% as Intermediate-risk, and 21.1% as Unknown-risk. Overall, 77.4% of patients initiated azacitidine and 22.6% decitabine; they received an average of 5.1 index-HMA cycles, of which 90.9% were complete with a median cycle duration of 28 days. Median survival was 11.6, 18.4, and 19.1 months for the Higher-risk, Intermediate-risk, and Unknown-risk groups, respectively. Median time-to-AML transformation was 19.3 months for the Higher-risk group and 50.4 months for the Intermediate-risk group (not reached for Unknown-risk). Data highlight the unmet medical needs of patients with MDS treated with HMA, particularly for the Higher-risk MDS group.
Collapse
Affiliation(s)
- Eytan M Stein
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gaetano Bonifacio
- Health Economics and Outcomes Research, US Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | - Sherry Shi
- Analysis Group, Inc., Montreal, QC, Canada
| | | | - Eric Q Wu
- Analysis Group, Inc., Boston, MA, USA
| | - Islam Sadek
- Health Economics and Outcomes Research, US Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Xiting Cao
- Health Economics and Outcomes Research, US Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| |
Collapse
|
30
|
Stein EM, Bonifacio G, Latrémouille-Viau D, Shi S, Guérin A, Wu EQ, Sadek I, Cao X. Healthcare resource utilization and costs in patients with myelodysplastic syndromes treated with hypomethylating agents: a SEER-Medicare analysis. J Med Econ 2021; 24:234-243. [PMID: 33472483 DOI: 10.1080/13696998.2021.1876714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS To describe healthcare resource utilization (HRU) and costs in patients with myelodysplastic syndromes (MDS) treated with hypomethylating agents (HMA) based on HMA-treatment response. MATERIALS AND METHODS SEER-Medicare data (January 2006-December 2016) were used to identify adults diagnosed with MDS (SEER: January 2009-December 2015) initiated on HMA (index date). HMA-treatment success (indicators: ≥7 HMA cycles, stem cell transplantation, and transfusion independence) or failure (indicators: acute myeloid leukemia [AML], AML-like treatment, and death) was determined using a claim-based algorithm. HRU and costs were assessed from the index date to 1-year post-index, overall and stratified by HMA-treatment success or failure. Among patients with HMA-treatment failure, HRU and costs were also assessed from failure to 1-year post-failure. RESULTS The study included 3,046 patients (mean age: 77.4 years; females: 36.8%). Rates of HMA-treatment success and failure were 44.4% and 76.2%, respectively (20.6% had HMA-treatment success then failure). Overall, patients had 15.2 inpatient admissions per-100-patients-per-month (median follow-up: 5.9 months). Patients with HMA-treatment success had 7.5 inpatient admissions per-100-patients-per-month (median follow-up: 12.0 months), while those with HMA-treatment failure had 20.4 and 35.3 admissions per-100-patients-per-month pre- and post-HMA-treatment failure, respectively (median follow-up: 4.3 and 1.8 months, pre- and post-HMA-treatment failure, respectively). Mean total healthcare costs were $12,494 per-patient-per-month overall, $8,069 per-patient-per-month among patients with HMA-treatment success, and $13,809 and $19,242 per-patient-per-month pre- and post-HMA-treatment failure, respectively. Outpatient costs (68.3%) were the main contributor of total healthcare costs overall, while inpatient costs (80.3%) were the main cost driver post-HMA-treatment failure. LIMITATIONS Without available laboratory test results, clinical indicators observed in claims were used to assess HMA-treatment response. CONCLUSIONS Over 75% of patients with MDS failed HMA-treatment within 6 months of initiation and were observed with more inpatient admissions than those with HMA-treatment success, translating into substantially higher healthcare costs. HMA-treatment failure results in an important economic burden in MDS patients.
Collapse
Affiliation(s)
- Eytan M Stein
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | - Eric Q Wu
- Analysis Group, Inc, Boston, MA, USA
| | - Islam Sadek
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Xiting Cao
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| |
Collapse
|
31
|
Papageorgiou SG, Kotsianidis I, Bouchla A, Symeonidis A, Galanopoulos A, Viniou NA, Hatzimichael E, Vassilakopoulos TP, Gogos D, Megalakaki A, Zikos P, Diamantopoulos P, Kourakli A, Giannoulia P, Papoutselis M, Poulakidas E, Arapaki M, Vardi A, Anagnostopoulos A, Mparmparousi D, Papaioannou M, Bouronikou E, Dimou M, Papadaki H, Panayiotidis P, Pappa V. Serum ferritin and ECOG performance status predict the response and improve the prognostic value of IPSS or IPSS-R in patients with high-risk myelodysplastic syndromes and oligoblastic acute myeloid leukemia treated with 5-azacytidine: a retrospective analysis of the Hellenic national registry of myelodysplastic and hypoplastic syndromes. Ther Adv Hematol 2020; 11:2040620720966121. [PMID: 33343854 PMCID: PMC7727043 DOI: 10.1177/2040620720966121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/23/2020] [Indexed: 11/16/2022] Open
Abstract
Background: 5-azacytidine (5-AZA) improves survival of patients with higher-risk myelodysplastic syndromes (MDSs) and oligoblastic acute myeloid leukemia (AML); however, predictive factors for response and outcome have not been consistently studied. Methods: This study of the Hellenic MDS Study Group included 687 consecutive patients with higher-risk MDS and oligoblastic AML treated with 5-AZA. Results: The International Prognostic Scoring System (IPSS) revised version (IPSS-R), Eastern Cooperative Oncology Group Performance Status (ECOG PS) (0 or 1 versus ⩾2) and baseline serum ferritin (SF) levels > 520 ng/ml were shown to independently predict response to 5-AZA. In the survival analysis, the IPSS and IPSS-R risk classification systems along with the ECOG PS and SF levels > 520 ng/ml proved to be independent prognosticators for overall survival (OS), as well as for leukemia-free survival (LFS). Next, we built new multivariate models for OS and LFS, incorporating only ECOG PS and SF levels besides IPSS or IPSS-R risk classification systems. Thereby, the new modified IPSS and IPSS-R risk classification systems (H-PSS, H-PSS-R) could each discriminate a low, an intermediate and a high-risk patient group regarding OS and LFS. The H-PSS and H-PSS-R proved to be better predictors of OS than their previous counterparts as well as the French prognostic score, while the most powerful OS predictor was the new, H-PSS-R system. Conclusions: ECOG PS and SF levels > 520 ng/ml independently predict response to 5-AZA, OS and LFS. Their incorporation in the IPSS and IPSS-R scores enhances these scores’ predictive power in 5-AZA-treated higher-risk MDS and oligoblastic AML patients.
Collapse
Affiliation(s)
- Sotirios G Papageorgiou
- Consultant of Hematology, Second Department of Internal Medicine and Research Unit, University General Hospital "Attikon", 1 Rimini St., Haidari, Athens, 12462, Greece
| | - Ioannis Kotsianidis
- Department of Hematology, Democritus University of Thrace Medical School, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Anthi Bouchla
- Second Department of Internal Medicine and Research Unit, University General Hospital "Attikon", Haidari, Athens, Greece
| | | | | | | | | | - Theodoros P Vassilakopoulos
- Department of Hematology, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | | | | | | | | | | | | | - Menelaos Papoutselis
- Department of Hematology, Democritus University of Thrace Medical School, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Elias Poulakidas
- "401" Army General Hospital of Athens, Mesogeion and Kanellopoulou 1, Athens, Greece
| | - Maria Arapaki
- Department of Hematology, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | - Anna Vardi
- Hematology Department, General Hospital of Thessaloniki "George Papanikolaou", Thessaloniki, Greece
| | - Achilles Anagnostopoulos
- Hematology Department, General Hospital of Thessaloniki "George Papanikolaou", Thessaloniki, Greece
| | - Despoina Mparmparousi
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria Papaioannou
- Hematology Department, University General Hospital of Thessaloniki AHEPA, Thessaloniki, Greece
| | - Eleni Bouronikou
- University General Hospital of Larissa, Mezourlo, Larissa, Greece
| | - Maria Dimou
- First Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Helen Papadaki
- University General Hospital of Heraklion, Voutes, Heraklion, Greece
| | - Panayiotis Panayiotidis
- First Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Pappa
- Second Department of Internal Medicine and Research Unit, University General Hospital "Attikon", Haidari, Athens, Greece
| |
Collapse
|
32
|
Kubasch AS, Platzbecker U. Patient stratification in myelodysplastic syndromes: how a puzzle may become a map. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:418-425. [PMID: 33275703 PMCID: PMC7727505 DOI: 10.1182/hematology.2020000126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Heterogeneity is the disease-defining epithet of myelodysplastic syndromes (MDS), a clonal disorder of hematopoietic stem and progenitor cells. During the last decade, significant progress has been made to better understand the diversity of clinical, molecular, cellular, and immunological factors that are bound to the prognosis and outcomes of patients with MDS. Despite the rapid generation of all of this biological information, how to implement it has fallen short. Redefining clinical tools to use this new information remains a challenge. The holistic integration of novel, high-impact individual risk parameters such as patient-reported outcomes or mutational and immunological data into conventional risk stratification systems may further refine patient subgroups, improve predictive power for survival, and provide a next-generation classification and prognosis system for patients with MDS. Dichotomic treatment strategies in patients with MDS according to their patient and disease profiles highlight the importance of precise risk stratification, which may be complemented by the definition of granular cohorts of patients with myeloid neoplasms and a druggable target (ie, IDH1/2 mutations) across conventional blast thresholds.
Collapse
Affiliation(s)
- Anne Sophie Kubasch
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany; German MDS Study Group (D-MDS), Leipzig, Germany; and European Myelodysplastic Syndromes Cooperative Group (EMSCO), Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany; German MDS Study Group (D-MDS), Leipzig, Germany; and European Myelodysplastic Syndromes Cooperative Group (EMSCO), Leipzig, Germany
| |
Collapse
|
33
|
Bewersdorf JP, Zeidan AM. Good but not good enough: Clinical trial participation of patients with myelodysplastic syndromes. Cancer 2020; 126:4664-4667. [PMID: 32767670 DOI: 10.1002/cncr.33106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 11/06/2022]
Affiliation(s)
- Jan Philipp Bewersdorf
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
34
|
Brierley CK, Zabor EC, Komrokji RS, DeZern AE, Roboz GJ, Brunner AM, Stone RM, Sekeres MA, Steensma DP. Low participation rates and disparities in participation in interventional clinical trials for myelodysplastic syndromes. Cancer 2020; 126:4735-4743. [PMID: 32767690 DOI: 10.1002/cncr.33105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 11/11/2022]
Abstract
BACKGROUND The development of novel therapies for the myelodysplastic syndromes (MDS) is hampered by inadequate trial recruitment. Factors contributing to low trial accrual are incompletely understood. METHODS This study analyzed a pooled patient database from institutions of the US MDS Clinical Research Consortium to compare the characteristics of participants in interventional trials with those of patients who did not enroll in a trial. RESULTS Data were identified for 1919 patients with MDS, and 449 of these patients (23%) participated in an interventional clinical trial. The median age of all patients was 68 years, and 64% were male. Patients who participated in trials were significantly younger than nonparticipants (P = .014), and men were more likely to participate in a trial (71% of trial participants were male, whereas 61% of nonparticipants were; P < .001). Race and ethnicity were not associated with trial enrollment. Patients in more affluent ZIP codes had a higher participation rate (P < .001). Patients with intermediate- and high-risk disease according to the revised International Prognostic Scoring System were overrepresented (P = .004), and trial participants less frequently had treatment-related disease (P < .001). In multivariable analyses, participation in a clinical trial was associated with a reduced hazard of death (P = .004). Even at large referral centers, only a minority of patients with MDS enrolled in interventional trials. CONCLUSIONS Restrictive trial eligibility criteria that exclude patients with MDS on account of age, comorbidities, or a history of another cancer are limit enrollment of MDS patients to clinical trials. Gaining insight into the barriers to trial accrual may help investigators and study sponsors to design trials that will accrue more rapidly and augment treatment options for patients with MDS.
Collapse
Affiliation(s)
| | - Emily C Zabor
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio.,Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Rami S Komrokji
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Gail J Roboz
- Weill Cornell Medical College, New York, New York
| | - Andrew M Brunner
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard M Stone
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mikkael A Sekeres
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio.,Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - David P Steensma
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Bewersdorf JP, Zeidan AM. Following in the footsteps of acute myeloid leukemia: are we witnessing the start of a therapeutic revolution for higher-risk myelodysplastic syndromes? Leuk Lymphoma 2020; 61:2295-2312. [PMID: 32421403 PMCID: PMC7670856 DOI: 10.1080/10428194.2020.1761968] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 12/21/2022]
Abstract
For most patients with higher-risk myelodysplastic syndromes (HR-MDS) the hypomethylating agents (HMA) azacitidine and decitabine remain the mainstay of therapy. However, the prognosis mostly remains poor and aside from allogeneic hematopoietic stem cell transplantation no curative treatment options exist. Unlike acute myeloid leukemia, which has seen a dramatic expansion of available therapies recently, no new agents have been approved for MDS in the United States since 2006. However, various novel HMAs, HMA in combination with venetoclax, immune checkpoint inhibitors, and targeted therapies for genetically defined patient subgroups such as APR-246 or IDH inhibitors, have shown promising results in early stages of clinical testing. Furthermore, the wider availability of genetic testing is going to allow for a more individualized treatment of MDS patients. Herein, we review the current treatment approach for HR-MDS and discuss recent therapeutic advances and the implications of genetic testing on management of HR-MDS.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT
| | - Amer M. Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
36
|
Papadopoulos V, Diamantopoulos PT, Papageorgiou SG, Papoutselis M, Vrachiolias G, Pappa V, Galanopoulos AG, Vassilakopoulos TP, Hatzimichael E, Zikos P, Papadaki HA, Bouchla A, Panayiotidis P, Megalakaki A, Papaioannou M, Liapis K, Dryllis G, Tsokanas D, Kourakli A, Symeonidis A, Viniou NA, Kotsianidis I. Estimated glomerular filtration rate independently predicts outcome of azacitidine therapy in higher-risk Myelodysplastic syndromes. Results from 536 patients of the Hellenic National Registry of Myelodysplastic and Hypoplastic syndromes. Hematol Oncol 2020; 38:541-553. [PMID: 32495951 DOI: 10.1002/hon.2756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/23/2022]
Abstract
Higher-risk Myelodysplastic syndromes (MDS) patients undergoing treatment with 5-azacytidine (AZA) are typically elderly with several comorbidities. However, the effect of comorbidities on the effectiveness and safety of AZA in real-world settings remains unclear. We analyzed data from 536 AZA-treated patients with higher-risk MDS, Myelodysplastic/Myeloproliferative neoplasms and low blast count Acute Myeloid Leukemia enrolled to the Hellenic National Registry of Myelodysplastic and Hypoplastic Syndromes. Multivariate analysis adjusted also for the International Prognostic Scoring System (IPSS), its revised version (IPSS-R) and the French Prognostic Scoring System (FPSS), demonstrated independent associations of overall and leukemia-free survival with estimated glomerular filtration rate (eGFR) <45 mL min-1 /1.73 m2 (P = .039, P = .023, respectively), ECOG performance status <2 (P = .015, P = .006), and presence of peripheral blood blasts (P = .008, P = .034), while secondary MDS also correlated with significantly shorter leukemia-free survival (P = .039). Addition of eGFR <45 mL min-1 /1.73 m2 , in IPSS-R and FPSS increased the predictive power of both models. Only FPSS ≤2 and eGFR <45 mL min-1 /1.73 m2 predicted worse response to AZA in multivariate analysis, whereas eGFR <45 mL min-1 /1.73 m2 correlated significantly with death from hemorrhage (P = .003) and cardiovascular complications (P = .006). In conclusion, in the second largest real-world series of AZA-treated MDS patients, we show that an eGFR <45 mL min-1 /1.73 m2 is an independent predictor of worse response and survival. This higher cut-off, instead of the commonly used serum creatinine >2 mg/dL, can be utilized as a more precise indicator of renal comorbidity during AZA therapy. Incorporation of eGFR in the prognostic assessment of AZA-treated MDS patients may prove useful not only in routine practice, but also for the appropriate patient stratification in clinical trials with AZA combinations.
Collapse
Affiliation(s)
- Vasileios Papadopoulos
- Department of Hematology, Democritus University of Thrace Medical School, Alexandroupolis, Greece
| | | | - Sotirios G Papageorgiou
- Second Department of Internal Medicine, Hematology Unit, Attikon University General Hospital, Athens, Greece
| | - Menelaos Papoutselis
- Department of Hematology, Democritus University of Thrace Medical School, Alexandroupolis, Greece
| | - George Vrachiolias
- Department of Hematology, Democritus University of Thrace Medical School, Alexandroupolis, Greece
| | - Vassiliki Pappa
- Second Department of Internal Medicine, Hematology Unit, Attikon University General Hospital, Athens, Greece
| | | | - Theodoros P Vassilakopoulos
- Department of Hematology, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Panagiotis Zikos
- Department of Hematology, General Hospital of Patras "Agios Andreas", Patras, Greece
| | - Helen A Papadaki
- Department of Hematology, University General Hospital of Heraklion, Heraklion, Greece
| | - Anthi Bouchla
- Second Department of Internal Medicine, Hematology Unit, Attikon University General Hospital, Athens, Greece
| | - Panayiotis Panayiotidis
- First Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Maria Papaioannou
- Department of Hematology, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Konstantinos Liapis
- Department of Hematology, Democritus University of Thrace Medical School, Alexandroupolis, Greece
| | - George Dryllis
- First Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Tsokanas
- Department of Internal Medicine, University Hospital of Patras, Rio, Greece
| | - Alexandra Kourakli
- Department of Internal Medicine, University Hospital of Patras, Rio, Greece
| | - Argiris Symeonidis
- Department of Internal Medicine, University Hospital of Patras, Rio, Greece
| | - Nora-Athina Viniou
- First Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Kotsianidis
- Department of Hematology, Democritus University of Thrace Medical School, Alexandroupolis, Greece
| |
Collapse
|
37
|
Davidoff AJ, Hu X, Bewersdorf JP, Wang R, Podoltsev NA, Huntington SF, Gore SD, Ma X, Zeidan AM. Hypomethylating agent (HMA) therapy use and survival in older adults with Refractory Anemia with Excess Blasts (RAEB) in the United States (USA): a large propensity score-matched population-based study †. Leuk Lymphoma 2020; 61:1178-1187. [PMID: 31878809 PMCID: PMC7735409 DOI: 10.1080/10428194.2019.1703970] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/16/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022]
Abstract
Hypomethylating agents (HMA) showed overall survival (OS) benefits in patients with higher-risk myelodysplastic syndromes (HR-MDS) in clinical trials. We conducted a retrospective cohort study of Surveillance, Epidemiology, and End Results (SEER)-Medicare data of patients ≥66 years diagnosed with refractory anemia with excess blasts (RAEB), a proxy for HR-MDS, in 01/2001-04/2004 (pre-period) or 01/2006-12/2011 (post-period). Association between post-period diagnosis and OS was examined using propensity scores (PS)-matched samples. Among 1876 RAEB patients, median OS was 9 months and 30.8% received HMAs (3.6% in pre-period; 43.0% in post-period) with no association between post-period diagnosis and OS. In the top PS quartile, post-period diagnosis was associated with a 74% lower risk of death (Hazard ratio [HR] = 0.26, 95%-CI: 0.10-0.69, p = 0.007), while outcomes were worse in the lowest PS quartile (HR = 2.80, 95%-CI: 1.06-7.36, p = 0.037). HMA lead to a 3-month OS benefit for patients most likely to receive HMA but not for unselected RAEB cohort.
Collapse
Affiliation(s)
- Amy J. Davidoff
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, CT
| | - Xin Hu
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| | | | - Rong Wang
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Health Policy and Management, School of Public Health, Yale University, New Haven, CT
| | - Nikolai A. Podoltsev
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT
| | - Scott F. Huntington
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT
| | - Steven D. Gore
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT
| | - Xiaomei Ma
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Health Policy and Management, School of Public Health, Yale University, New Haven, CT
| | - Amer M. Zeidan
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT
| |
Collapse
|
38
|
Evolving therapies for lower-risk myelodysplastic syndromes. Ann Hematol 2020; 99:677-692. [PMID: 32078008 DOI: 10.1007/s00277-020-03963-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 02/10/2020] [Indexed: 12/16/2022]
Abstract
The development in the therapeutic landscape of myelodysplastic syndromes (MDS) has substantially lagged behind other hematologic malignancies with no new drug approvals for MDS for 13 years since the approval of decitabine in the United States in 2006. While therapeutic concepts for MDS patients continue to be primarily defined by clinical-pathologic risk stratification tools such as the International Prognostic Scoring System (IPSS) and its revised version IPSS-R, our understanding of the genetic landscape and the molecular pathogenesis of MDS has greatly evolved over the last decade. It is expected that the therapeutic approach to MDS patients will become increasingly individualized based on prognostic and predictive genetic features and other biomarkers. Herein, we review the current treatment of lower-risk MDS patients and discuss promising agents in advanced clinical testing for the treatment of symptomatic anemia in lower-risk MDS patients such as luspatercept and imetelstat. Lastly, we review the clinical development of new agents and the implications of the wider availability of mutational analysis for the management of individual MDS patients.
Collapse
|
39
|
Lee IH, Kang BW, Kim JG, Bae WK, Ki MS, Park I, Jo JC, Kim JY, Koh SA, Lee KH, Cho YY, Ryoo HM, Kwak SG, Lee JL, Lee SA. Comparison of three risk stratification models for non-clear cell renal cell carcinoma patients treated with temsirolimus as first-line therapy. Korean J Intern Med 2020; 35:185-193. [PMID: 30301310 PMCID: PMC6960037 DOI: 10.3904/kjim.2018.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 06/01/2018] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS For metastatic renal cell carcinoma (RCC), various prognostic scoring systems have been developed. However, owing to the low prevalence of nonclear cell RCC, the three most commonly used tools were mainly developed based on patients with clear cell histology. Accordingly, this study applied three prognostic models to Korean non-clear cell RCC patients treated with first-line temsirolimus. METHODS This study analyzed data for 74 patients with non-clear cell RCC who were treated with temsirolimus as the first-line therapy at eight medical centers between 2011 and 2016. The receiver-operating characteristic (ROC) curves for the different prognostic models were analyzed. RESULTS Twenty-seven (36.5%), 24 (32.4%), and 44 patients (59.5%) were assigned to the poor prognosis groups of the Memorial Sloan-Kettering Cancer Center (MSKCC), International Metastatic RCC Database Consortium (IMDC), and Advanced Renal Cell Carcinoma (ARCC) risk stratification models, respectively. All three prognostic models reliably discriminated the risk groups to predict progression-free survival and overall survival (p < 0.001). The area under the ROC curve (AUC) for progression and survival was highest for the ARCC model (0.777; 0.734), followed by the IMDC (0.756; 0.724) and the MSKCC (0.742; 0.712) models. Furthermore, the sensitivity and specificity for predicting progression were highest with the ARCC model (sensitivity 63.6%, specificity 85.7%), followed by the MSKCC (sensitivity 58.2%, specificity 86.5%) and the IMDC models (sensitivity 56.4%, specificity 85.7%). CONCLUSION All three prognostic models accurately predicted the survival of the non-clear cell RCC patients treated with temsirolimus as the first-line therapy. Furthermore, the ARCC risk model performed better than the other risk models in predicting survival.
Collapse
Affiliation(s)
- In Hee Lee
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Byung Woog Kang
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
- Correspondence to Byung Woog Kang, M.D. Department of Hematology/Oncology, Kyungpook National University Chilgok Hospital, 807 Hoguk-ro, Buk-gu, Daegu 41404, Korea. Tel: +82-53-200-2622, Fax: +82-53-200-2029, E-mail:
| | - Jong Gwang Kim
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Woo Kyun Bae
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Myung Seo Ki
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Inkeun Park
- Division of Medical Oncology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Korea
- Inkeun Park, M.D. Division of Medical Oncology, Department of Internal Medicine, Gachon University Gil Medical Center, 21 Namdong-daero 774beon-gil, Namdong-gu, Incheon 21565, Korea. Tel: +82-32-460-3229, Fax: +82-32-460-2391, E-mail:
| | - Jae-Cheol Jo
- Department of Hematology and Oncology, Ulsan University Hospital, Ulsan, Korea
| | - Jin Young Kim
- Division of Hematology/Oncology, Department of Internal Medicine, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Sung Ae Koh
- Department of Hematology-Oncology, Yeungnam University College of Medicine, Daegu, Korea
| | - Kyung Hee Lee
- Department of Hematology-Oncology, Yeungnam University College of Medicine, Daegu, Korea
| | - Yoon Young Cho
- Department of HematologyOncology, Daegu Catholic University Medical Center, Daegu, Korea
| | - Hun Mo Ryoo
- Department of HematologyOncology, Daegu Catholic University Medical Center, Daegu, Korea
| | - Sang Gyu Kwak
- Department of Medical Statistics, Daegu Catholic University Medical Center, Daegu, Korea
| | - Jung Lim Lee
- Department of Oncology/Hematology, Daegu Fatima Hospital, Daegu, Korea
| | - Sun Ah Lee
- Department of Oncology/Hematology, Daegu Fatima Hospital, Daegu, Korea
| |
Collapse
|
40
|
Buckstein RJ. Integrating patient-centered factors in the risk assessment of MDS. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:373-380. [PMID: 31808887 PMCID: PMC6913474 DOI: 10.1182/hematology.2019000041] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Myelodysplastic syndromes are clonal myeloid neoplasms that primarily present in older adults. Although leukemia develops in approximately 25% to 30% of individuals, the significantly shortened survival in this population is attributed more commonly to nonleukemic causes. The current prognostic scoring systems for leukemia and overall survival based on disease characteristics are becoming increasingly sophisticated and accurate with the incorporation of molecular data. The addition of patient-related factors such as comorbidity, disability, frailty, and fatigue to these new models may improve their predictive power for overall survival, treatment toxicity, and health care costs. To improve the generalizability of clinical trial results to the real world, geriatric assessment testing should become a standard of care in MDS clinical trials.
Collapse
Affiliation(s)
- Rena J Buckstein
- Odette Cancer Center, Sunnybrook Health Sciences Center, Toronto, ON, Canada
| |
Collapse
|
41
|
Zeidan AM, Hu X, Zhu W, Stahl M, Wang R, Huntington SF, Giri S, Bewersdorf JP, Podoltsev NA, Gore SD, Ma X, Davidoff AJ. Association of provider experience and clinical outcomes in patients with myelodysplastic syndromes receiving hypomethylating agents. Leuk Lymphoma 2019; 61:397-408. [PMID: 31570040 DOI: 10.1080/10428194.2019.1663423] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Population level survival of patients with myelodysplastic syndromes (MDS) treated with hypomethylating agents (HMA) is inferior to clinical trials. Using SEER-Medicare data, we identified 2086 MDS patients diagnosed in 2004-13, aged ≥66 years at diagnosis, and receiving ≥1 HMA cycle after 2005. We used multivariate logistic regression and Cox proportional hazards models to assess the impact of provider experience on persistent HMA therapy and overall survival (OS), respectively. Median number of HMA cycles was 4 and median OS was 10 months. Thirty-two percent of patients were treated by providers with ≥1 prior HMA initiation in the last 2 years and were more likely to receive ≥4 cycles of HMA therapy (OR = 1.29, 95% CI = 1.06-1.57; p = .01). No significant association was found between MDS or HMA initiation volume and survival. In conclusion, while HMA initiation volume was associated with persistent HMA treatment, neither MDS nor HMA initiation volumes were associated with OS in older MDS patients.
Collapse
Affiliation(s)
- Amer M Zeidan
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| | - Xin Hu
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| | - Weiwei Zhu
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| | - Maximilian Stahl
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Rong Wang
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA.,Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, CT, USA
| | - Scott F Huntington
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| | - Smith Giri
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| | - Jan Philipp Bewersdorf
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Nikolai A Podoltsev
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| | - Steven D Gore
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| | - Xiaomei Ma
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA.,Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, CT, USA
| | - Amy J Davidoff
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA.,Department of Health Policy and Management, School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
42
|
Nazha A, Sekeres MA, Bejar R, Rauh MJ, Othus M, Komrokji RS, Barnard J, Hilton CB, Kerr CM, Steensma DP, DeZern A, Roboz G, Garcia-Manero G, Erba H, Ebert BL, Maciejewski JP. Genomic Biomarkers to Predict Resistance to Hypomethylating Agents in Patients With Myelodysplastic Syndromes Using Artificial Intelligence. JCO Precis Oncol 2019; 3. [PMID: 31663066 PMCID: PMC6818517 DOI: 10.1200/po.19.00119] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
PURPOSE We developed an unbiased framework to study the association of several mutations in predicting resistance to hypomethylating agents (HMAs) in patients with myelodysplastic syndromes (MDS), analogous to consumer and commercial recommender systems in which customers who bought products A and B are likely to buy C: patients who have a mutation in gene A and gene B are likely to respond or not respond to HMAs. METHODS We screened a cohort of 433 patients with MDS who received HMAs for the presence of common myeloid mutations in 29 genes that were obtained before the patients started therapy. The association between mutations and response was evaluated by the Apriori market basket analysis algorithm. Rules with the highest confidence (confidence that the association exists) and the highest lift (strength of the association) were chosen. We validated our biomarkers in samples from patients enrolled in the S1117 trial. RESULTS Among 433 patients, 193 (45%) received azacitidine, 176 (40%) received decitabine, and 64 (15%) received HMA alone or in combination. The median age was 70 years (range, 31 to 100 years), and 28% were female. The median number of mutations per sample was three (range, zero to nine), and 176 patients (41%) had three or more mutations per sample. Association rules identified several genomic combinations as being highly associated with no response. These molecular signatures were present in 30% of patients with three or more mutations/sample with an accuracy rate of 87% in the training cohort and 93% in the validation cohort. CONCLUSION Genomic biomarkers can identify, with high accuracy, approximately one third of patients with MDS who will not respond to HMAs. This study highlights the importance of machine learning technologies such as the recommender system algorithm in translating genomic data into useful clinical tools.
Collapse
Affiliation(s)
| | | | - Rafael Bejar
- University of California San Diego, San Diego, CA
| | | | - Megan Othus
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | | | | | - David P Steensma
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Amy DeZern
- The Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | | | - Benjamin L Ebert
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
43
|
Chokr N, Pine AB, Bewersdorf JP, Shallis RM, Stahl M, Zeidan AM. Getting personal with myelodysplastic syndromes: is now the right time? Expert Rev Hematol 2019; 12:215-224. [PMID: 30977414 PMCID: PMC6540985 DOI: 10.1080/17474086.2019.1592673] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/06/2019] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Commonly used scoring systems rely on blood counts, histological and cytological examination of bone marrow and peripheral blood as well as cytogenetic assessments to estimate prognosis of patients with myelodysplastic syndromes (MDS) and guide therapy decisions. Next-generation sequencing (NGS) has identified recurrent genetic abnormalities in up to 90% of patients with MDS and may provide important information regarding the pathogenesis of the disease, diagnostic and prognostic evaluation, and therapy selection. Areas covered: Herein, the authors review the role of NGS in diagnosis, treatment, and prognosis of MDS at various disease stages, and discuss advantages and caveats of incorporating molecular genetics in routine management of MDS. While a vast majority of patients harbor recurrent mutations implicated in MDS pathogenesis, similar mutations can be detected in otherwise healthy individuals with other hematologic malignancies. Besides establishing a diagnosis, NGS may be used to monitor minimal residual disease following treatment. Expert opinion: As more targeted therapies become available, assessment of genetic mutations will become central to individualized therapy selection and may improve diagnostic accuracy and further guide management for each patient. However, multiple challenges remain before NGS can be incorporated into routine clinical practice.
Collapse
Affiliation(s)
- Nora Chokr
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA
| | - Alexander B. Pine
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA
| | - Jan Philipp Bewersdorf
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA
| | - Rory M. Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA
| | - Maximilian Stahl
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, USA
| |
Collapse
|
44
|
Zeidan AM, Shallis RM, Wang R, Davidoff A, Ma X. Epidemiology of myelodysplastic syndromes: Why characterizing the beast is a prerequisite to taming it. Blood Rev 2019; 34:1-15. [DOI: 10.1016/j.blre.2018.09.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/01/2018] [Accepted: 09/17/2018] [Indexed: 02/08/2023]
|
45
|
Rethinking clinical trial endpoints in myelodysplastic syndromes. Leukemia 2019; 33:570-575. [DOI: 10.1038/s41375-018-0367-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 11/08/2022]
|
46
|
Counseling patients with higher-risk MDS regarding survival with azacitidine therapy: are we using realistic estimates? Blood Cancer J 2018; 8:55. [PMID: 29891916 PMCID: PMC5995881 DOI: 10.1038/s41408-018-0081-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 03/21/2018] [Accepted: 04/03/2018] [Indexed: 11/30/2022] Open
|
47
|
The impact of anaemia, transfusion dependency, comorbidities and polypharmacy in elderly patients with low-risk myelodysplastic syndromes. Med Oncol 2018; 35:33. [PMID: 29417235 DOI: 10.1007/s12032-018-1094-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 01/31/2018] [Indexed: 10/18/2022]
Abstract
Myelodysplastic syndromes (MDS) are heterogeneous clonal disorders ranging from indolent conditions with a near-normal life expectancy to forms approaching acute myeloid leukaemia. Comorbid conditions have rarely been systematically studied among patients with MDS. Older age per se has a negative impact on survival of MDS patients, in particular of those with lower risk. However, age indirectly affects also the survival of higher-risk patients by limiting their eligibility to intensive treatments. In addition, ageing is associated with an increasingly high risk of developing comorbidity, and a high prevalence of comorbid diseases has indeed been reported in MDS patients. The impact of multi-morbidities/comorbidities and polypharmacy in patients with low-risk MDS patients is a poorly explored topic. We focused on medications, multi-morbidities and comorbidities of 155 low-risk MDS patients followed in the haematological outpatients clinics or in medical/oncology wards of our University Hospital. One or more comorbidities were present at diagnosis in 24 younger patients with MDS syndromes (31%), whereas 56 older patients with MDS (75%) presented 1 or more comorbidities (P < 0.001).The most frequent comorbidity was cardiac comorbidity 18% in younger patients and 25% in older patients. With no statistical significance between older and younger patients, congestive heart failure was the most frequent observed disease. Our study has shown a statistical correlation between transfusion dependency and polypathology (P = 0.0014). These data were also confirmed in a subanalysis of the younger group of patients. Our study has shown that comorbidity is very common among patients with MDS, potentially affecting the clinical course and outcome of MDS patients.
Collapse
|
48
|
Shallis RM, Zeidan AM. More is less, less is more, or does it really matter? The curious case of impact of azacitidine administration schedules on outcomes in patients with myelodysplastic syndromes. BMC HEMATOLOGY 2018; 18:4. [PMID: 29435332 PMCID: PMC5796398 DOI: 10.1186/s12878-018-0095-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/04/2018] [Indexed: 12/16/2022]
Abstract
Myelodysplastic syndromes (MDS) encompass a diverse group of hematologic disorders characterized by ineffective and malignant hematopoiesis, peripheral cytopenias and significantly increased risk of progression to acute myeloid leukemia (AML). The hypomethylating agents (HMA) azacitidine and decitabine induce meaningful clinical responses in a significant subset of patients with MDS. Though never compared directly with decitabine, only azacitidine has improved overall survival (OS) compared to conventional care in a randomized trial in patients with higher-risk MDS. The azacitidine regimen used in this pivotal trial AZA-001 included administration at 75 mg/m2/day for 7 consecutive days in 28-day cycles (7-0 regimen). Given the logistical difficulties of weekend administration in the 7-0 regimen, as well as in efforts to improve response rates, alternative dosing schedules have been used. In a typical 28-day cycle, administration schedules of 3, 5, 10, and (with the oral version of azacitidine) 14 and 21 days have been used in clinical trials. Most trials that evaluated alternative administration schedules of azacitidine did so in lower-risk MDS and did not directly compare to the 7-0 schedule. Given the lack of randomized prospective studies comparing the 7-0 schedule to the other regimens of azacitidine in MDS, Shapiro et al. conducted a systematic review in an attempt to answer this question. Here we place the findings of this important work in clinical context and review the current knowledge and unresolved issues regarding the impact of administration schedules of azacitidine on outcomes of patients with both lower-risk and higher-risk MDS.
Collapse
Affiliation(s)
- Rory M. Shallis
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT USA
- Section of Hematology, Department of Internal Medicine, Yale University, 333 Cedar Street, PO Box 208028, New Haven, CT 06520-8028 USA
| | - Amer M. Zeidan
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT USA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT USA
- Section of Hematology, Department of Internal Medicine, Yale University, 333 Cedar Street, PO Box 208028, New Haven, CT 06520-8028 USA
| |
Collapse
|
49
|
Long-term survival of older patients with MDS treated with HMA therapy without subsequent stem cell transplantation. Blood 2017; 131:818-821. [PMID: 29259002 DOI: 10.1182/blood-2017-10-811729] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/15/2017] [Indexed: 12/23/2022] Open
|
50
|
Haider M, Duncavage EJ, Afaneh KF, Bejar R, List AF. New Insight Into the Biology, Risk Stratification, and Targeted Treatment of Myelodysplastic Syndromes. Am Soc Clin Oncol Educ Book 2017; 37:480-494. [PMID: 28561687 DOI: 10.1200/edbk_175397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In myelodysplastic syndromes (MDS), somatic mutations occur in five major categories: RNA splicing, DNA methylation, activated cell signaling, myeloid transcription factors, and chromatin modifiers. Although many MDS cases harbor more than one somatic mutation, in general, there is mutual exclusivity of mutated genes within a class. In addition to the prognostic significance of individual somatic mutations, more somatic mutations in MDS have been associated with poor prognosis. Prognostic assessment remains a critical component of the personalization of care for patient with MDS because treatment is highly risk adapted. Multiple methods for risk stratification are available with the revised International Prognostic Scoring System (IPSS-R), currently considered the gold standard. Increasing access to myeloid gene panels and greater evidence for the diagnostic and predictive value of somatic mutations will soon make sequencing part of the standard evaluation of patients with MDS. In the absence of formal guidelines for their prognostic use, well-validated mutations can still refine estimates of risk made with the IPSS-R. Not only are somatic gene mutations advantageous in understanding the biology of MDS and prognosis, they also offer potential as biomarkers and targets for the treatment of patients with MDS. Examples include deletion 5q, spliceosome complex gene mutations, and TP53 mutations.
Collapse
Affiliation(s)
- Mintallah Haider
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Eric J Duncavage
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Khalid F Afaneh
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Rafael Bejar
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Alan F List
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|