1
|
Park S, Park YH, Huh J, Baik SM, Park DJ. Deep learning model for differentiating acute myeloid and lymphoblastic leukemia in peripheral blood cell images via myeloblast and lymphoblast classification. Digit Health 2024; 10:20552076241258079. [PMID: 38812848 PMCID: PMC11135107 DOI: 10.1177/20552076241258079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
Objective Acute leukemia (AL) is a life-threatening malignant disease that occurs in the bone marrow and blood, and is classified as either acute myeloid leukemia (AML) or acute lymphoblastic leukemia (ALL). Diagnosing AL warrants testing methods, such as flow cytometry, which require trained professionals, time, and money. We aimed to develop a model that can classify peripheral blood images of 12 cell types, including pathological cells associated with AL, using artificial intelligence. Methods We acquired 42,386 single-cell images of peripheral blood slides from 282 patients (82 with AML, 40 with ALL, and 160 with immature granulocytes). Results The performance of EfficientNet-V2 (B2) using the original image size exhibited the greatest accuracy (accuracy, 0.8779; precision, 0.7221; recall, 0.7225; and F1 score, 0.7210). The next-best accuracy was achieved by EfficientNet-V1 (B1), with a 256 × 256 pixels image. F1 score was the greatest for EfficientNet-V1 (B1) with the original image size. EfficientNet-V1 (B1) and EfficientNet-V2 (B2) were used to develop an ensemble model, and the accuracy (0.8858) and F1 score (0.7361) were improved. The classification performance of the developed ensemble model for the 12 cell types was good, with an area under the receiver operating characteristic curve above 0.9, and F1 scores for myeloblasts and lymphoblasts of 0.8873 and 0.8006, respectively. Conclusions The performance of the developed ensemble model for the 12 cell classifications was satisfactory, particularly for myeloblasts and lymphoblasts. We believe that the application of our model will benefit healthcare settings where the rapid and accurate diagnosis of AL is difficult.
Collapse
Affiliation(s)
- Sholhui Park
- Department of Laboratory Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Young Hoon Park
- Division of Hematology-Oncology, Department of Internal Medicine, Ewha Womans University Mokdong Hospital, Seoul, Korea
| | - Jungwon Huh
- Department of Laboratory Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Seung Min Baik
- Division of Critical Care Medicine, Department of Surgery, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Dong Jin Park
- Department of Laboratory Medicine, Eunpyeong St Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
2
|
Dozzo A, Galvin A, Shin JW, Scalia S, O'Driscoll CM, Ryan KB. Modelling acute myeloid leukemia (AML): What's new? A transition from the classical to the modern. Drug Deliv Transl Res 2022:10.1007/s13346-022-01189-4. [PMID: 35930221 DOI: 10.1007/s13346-022-01189-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/24/2022]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous malignancy affecting myeloid cells in the bone marrow (BM) but can spread giving rise to impaired hematopoiesis. AML incidence increases with age and is associated with poor prognostic outcomes. There has been a disconnect between the success of novel drug compounds observed in preclinical studies of hematological malignancy and less than exceptional therapeutic responses in clinical trials. This review aims to provide a state-of-the-art overview on the different preclinical models of AML available to expand insights into disease pathology and as preclinical screening tools. Deciphering the complex physiological and pathological processes and developing predictive preclinical models are key to understanding disease progression and fundamental in the development and testing of new effective drug treatments. Standard scaffold-free suspension models fail to recapitulate the complex environment where AML occurs. To this end, we review advances in scaffold/matrix-based 3D models and outline the most recent advances in on-chip technology. We also provide an overview of clinically relevant animal models and review the expanding use of patient-derived samples, which offer the prospect to create more "patient specific" screening tools either in the guise of 3D matrix models, microphysiological "organ-on-chip" tools or xenograft models and discuss representative examples.
Collapse
Affiliation(s)
| | - Aoife Galvin
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, 909 S. Wolcott Ave, Chicago, IL, 5091 COMRB, USA
| | - Santo Scalia
- Università degli Studi di Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Caitriona M O'Driscoll
- School of Pharmacy, University College Cork, Cork, Ireland.,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland
| | - Katie B Ryan
- School of Pharmacy, University College Cork, Cork, Ireland. .,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
3
|
Chen F, Licarete E, Wu X, Petrusca D, Maguire C, Jacobsen M, Colter A, Sandusky GE, Czader M, Capitano ML, Ropa JP, Boswell HS, Carta F, Supuran CT, Parkin B, Fishel ML, Konig H. Pharmacological inhibition of Carbonic Anhydrase IX and XII to enhance targeting of acute myeloid leukaemia cells under hypoxic conditions. J Cell Mol Med 2021; 25:11039-11052. [PMID: 34791807 PMCID: PMC8650039 DOI: 10.1111/jcmm.17027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 09/19/2021] [Indexed: 01/02/2023] Open
Abstract
Acute myeloid leukaemia (AML) is an aggressive form of blood cancer that carries a dismal prognosis. Several studies suggest that the poor outcome is due to a small fraction of leukaemic cells that elude treatment and survive in specialised, oxygen (O2)‐deprived niches of the bone marrow. Although several AML drug targets such as FLT3, IDH1/2 and CD33 have been established in recent years, survival rates remain unsatisfactory, which indicates that other, yet unrecognized, mechanisms influence the ability of AML cells to escape cell death and to proliferate in hypoxic environments. Our data illustrates that Carbonic Anhydrases IX and XII (CA IX/XII) are critical for leukaemic cell survival in the O2‐deprived milieu. CA IX and XII function as transmembrane proteins that mediate intracellular pH under low O2 conditions. Because maintaining a neutral pH represents a key survival mechanism for tumour cells in O2‐deprived settings, we sought to elucidate the role of dual CA IX/XII inhibition as a novel strategy to eliminate AML cells under hypoxic conditions. Our findings demonstrate that the dual CA IX/XII inhibitor FC531 may prove to be of value as an adjunct to chemotherapy for the treatment of AML.
Collapse
Affiliation(s)
- Fangli Chen
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana, USA
| | - Emilia Licarete
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana, USA.,Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Xue Wu
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana, USA
| | - Daniela Petrusca
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana, USA
| | - Callista Maguire
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Max Jacobsen
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Austyn Colter
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana, USA
| | - George E Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Magdalena Czader
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Maegan L Capitano
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana, USA
| | - James P Ropa
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana, USA
| | - H Scott Boswell
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana, USA
| | - Fabrizio Carta
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Firenze, Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Firenze, Italy
| | - Brian Parkin
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Melissa L Fishel
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana, USA.,Department of Pediatrics, Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA.,Department of Pharmacology & Toxicology, Indiana University, Indianapolis, Indiana, USA
| | - Heiko Konig
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
4
|
Horibata S, Gui G, Lack J, DeStefano CB, Gottesman MM, Hourigan CS. Heterogeneity in refractory acute myeloid leukemia. Proc Natl Acad Sci U S A 2019; 116:10494-10503. [PMID: 31064876 PMCID: PMC6535032 DOI: 10.1073/pnas.1902375116] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Successful clinical remission to therapy for acute myeloid leukemia (AML) is required for long-term survival to be achieved. Despite trends in improved survival due to better supportive care, up to 40% of patients will have refractory disease, which has a poorly understood biology and carries a dismal prognosis. The development of effective treatment strategies has been hindered by a general lack of knowledge about mechanisms of chemotherapy resistance. Here, through transcriptomic analysis of 154 cases of treatment-naive AML, three chemorefractory patient groups with distinct expression profiles are identified. A classifier, four key refractory gene signatures (RG4), trained based on the expression profile of the highest risk refractory patients, validated in an independent cohort (n = 131), was prognostic for overall survival (OS) and refined an established 17-gene stemness score. Refractory subpopulations have differential expression in pathways involved in cell cycle, transcription, translation, metabolism, and/or stem cell properties. Ex vivo drug sensitivity to 122 small-molecule inhibitors revealed effective group-specific targeting of pathways among these three refractory groups. Gene expression profiling by RNA sequencing had a suboptimal ability to correctly predict those individuals resistant to conventional cytotoxic induction therapy, but could risk-stratify for OS and identify subjects most likely to have superior responses to a specific alternative therapy. Such personalized therapy may be studied prospectively in clinical trials.
Collapse
Affiliation(s)
- Sachi Horibata
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892;
| | - Gege Gui
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20814
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702
| | - Christin B DeStefano
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20814
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Michael M Gottesman
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892;
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20814;
| |
Collapse
|
5
|
Candoni A, Papayannidis C, Martinelli G, Simeone E, Gottardi M, Iacobucci I, Gherlinzoni F, Visani G, Baccarani M, Fanin R. Flai (fludarabine, cytarabine, idarubicin) plus low-dose Gemtuzumab Ozogamicin as induction therapy in CD33-positive AML: Final results and long term outcome of a phase II multicenter clinical trial. Am J Hematol 2018; 93:655-663. [PMID: 29396857 DOI: 10.1002/ajh.25057] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 11/12/2022]
Abstract
The aim of this prospective clinical trial was to evaluate the efficacy and safety of a combination of Gemtuzumab-Ozogamicin (GO) and FLAI scheme (fludarabine, cytarabine, idarubicin) as a first-line therapy in CD33 positive AML. We treated 130 patients, aged <65, with a median age of 52 years. FLAI-GO induction regimen included fludarabine (30 mg/sqm) and cytarabine (2 g/sqm) on days 1-5; idarubicin (10 mg/sqm) on days 1, 3, and 5; and GO (3 mg/sqm) on day 6. SCT was planned for all high-risk AML patients, after consolidation with intermediate doses of cytarabine and idarubicin and a high dose of cytarabine. CD33 expression exceeded 20% in all cases. Primary endpoints of the study included feasibility, overall response rate (ORR) and toxicity. Secondary endpoints included the evaluation of MRD by WT1 expression, feasibility and outcome of consolidation with SCT, overall survival (OS) and disease-free survival (DFS). After induction with FLAI-GO, complete remission (CR) rate was 82%. Four patients achieved partial remission (PR) and 12% were resistant (ORR 85%); death during induction (DDI) was 3%. The hematological and extra hematological toxicity of FLAI-GO was manageable; 45% of patients experienced transient and reversible GO infusion related adverse events. In the setting of patients who achieved a cytological CR after FLAI-GO, the mean of WT1 copies dropped from 8337±9936 copies/104 ABL (diagnosis) to 182 ± 436 copies after induction therapy (p = 0.0001) showing a very good disease debulking. After a median follow-up of 54 months, 67/130 (52%) patients were alive. The probability of 1, 2, and 5-year OS was 80%, 63%, and 52%, respectively. The probability of 1, 2, and 5-year DFS was 77%, 58%, and 52%, respectively. Allogeneic and autologous SCT was performed in 60 (46%) and 23 (18%) patients, respectively. In summary, the final results of this trial confirm that FLAI-GO is an active and safe treatment strategy for CD33-positive AML patients aged ≤ 65 years, allowing a high ORR, a good disease debulking, favorable safety profile, low DDI, and subsequent high SCT rate. The encouraging results of this trial, consolidated by a long follow-up, support the reintroduction of GO in clinical practice.
Collapse
Affiliation(s)
- Anna Candoni
- Division of Hematology and SCT; University of Udine; Udine Italy
| | - Cristina Papayannidis
- Institute of Hematology and Oncology L. and A. Seràgnoli, University of Bologna; Bologna Italy
| | - Giovanni Martinelli
- Institute of Hematology and Oncology L. and A. Seràgnoli, University of Bologna; Bologna Italy
| | - Erica Simeone
- Division of Hematology and SCT; University of Udine; Udine Italy
| | | | - Ilaria Iacobucci
- Institute of Hematology and Oncology L. and A. Seràgnoli, University of Bologna; Bologna Italy
| | | | - Giuseppe Visani
- Hematology and SCT Center; San Salvatore Hospital; Pesaro Italy
| | - Michele Baccarani
- Institute of Hematology and Oncology L. and A. Seràgnoli, University of Bologna; Bologna Italy
| | - Renato Fanin
- Division of Hematology and SCT; University of Udine; Udine Italy
| |
Collapse
|
6
|
Immune therapies in acute myeloid leukemia: a focus on monoclonal antibodies and immune checkpoint inhibitors. Curr Opin Hematol 2018; 25:136-145. [PMID: 29206680 DOI: 10.1097/moh.0000000000000401] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE OF REVIEW This review discusses the rationale, efficacy, and toxicity of a variety of immune approaches being evaluated in the therapy of acute myeloid leukemia (AML) including naked and conjugated monoclonal antibodies, bispecific T-cell engager antibodies, and immune checkpoint blockade via antibodies targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed-death 1 (PD-1). RECENT FINDINGS The stellar success of immune therapies that harness the power of T cells in solid tumors and an improved understanding of the immune system in patients with hematologic malignancies have resulted in major efforts to develop immune therapies for the treatment of patients with AML. Monoclonal antibodies in AML therapy include naked antibodies against AML surface antigens such as CD33 (e.g. lintuzumab) or CD38 (e.g. daratumumab), antibodies conjugated to toxins in various anti-CD33 (gemtuzumab ozogamicin, SGN33A, IMGN779) and anti-CD123 (SL-401, SGN-CD123A) formulations, and antibodies conjugated to radioactive particles such as I or Ac-labeled anti-CD33 or anti-CD45 antibodies. Additional antigenic targets of interest in AML include CLL1, CD38, CD25, TIM3, FLT3, and others. Approaches to harness the body's own T cells against AML include antibodies that recruit and induce cytotoxicity of tumor cells by T cells (bispecific T-cell engager [BiTE] such as CD33 x CD3 (e.g. AMG 330) or CD123 x CD3 (e.g. flotetuzumab, JNJ-63709178) or antibodies that block immune checkpoint receptors CTLA4 (e.g. ipilimumab) or PD1/PD-L1 (e.g. nivolumab, pembrolizumab, avelumab) on T cells, unleashing the patients' T cells against leukemic cells. SUMMARY The ongoing trials and well designed correlative interrogation of the immune system in patients treated on such trials will further enhance our understanding and clinical application of immune therapies as single-agent and combination approaches for the treatment of AML.
Collapse
|
7
|
Brinda B, Khan I, Parkin B, Konig H. The rocky road to personalized medicine in acute myeloid leukaemia. J Cell Mol Med 2018; 22:1411-1427. [PMID: 29327808 PMCID: PMC5824388 DOI: 10.1111/jcmm.13478] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/11/2017] [Indexed: 12/26/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a malignant disorder of the myeloid blood lineage characterized by impaired differentiation and increased proliferation of hematopoietic precursor cells. Recent technological advances have led to an improved understanding of AML biology but also uncovered the enormous cytogenetic and molecular heterogeneity of the disease. Despite this heterogeneity, AML is mostly managed by a 'one-size-fits-all' approach consisting of intensive, highly toxic induction and consolidation chemotherapy. These treatment protocols have remained largely unchanged for the past several decades and only lead to a cure in approximately 30-35% of cases. The advent of targeted therapies in chronic myeloid leukaemia and other malignancies has sparked hope to improve patient outcome in AML. However, the implementation of targeted agents in AML therapy has been unexpectedly cumbersome and remains a difficult task due to a variety of disease- and patient-specific factors. In this review, we describe current standard and investigational therapeutic strategies with a focus on targeted agents and highlight potential tools that might facilitate the development of targeted therapies for this fatal disease. The classes of agents described in this review include constitutively activated signalling pathway inhibitors, surface receptor targets, epigenetic modifiers, drugs targeting the interaction of the hematopoietic progenitor cell with the stroma and drugs that target the apoptotic machinery. The clinical context and outcome with these agents will be examined to gain insight about their optimal utilization.
Collapse
Affiliation(s)
- Bryan Brinda
- Indiana University Melvin and Bren Simon Cancer CenterIndianapolisINUSA
| | - Irum Khan
- Division of Hematology and OncologyCollege of Medicine at ChicagoUniversity of IllinoisChicagoILUSA
| | - Brian Parkin
- University of Michigan Comprehensive Cancer CenterAnn ArborMIUSA
| | - Heiko Konig
- Indiana University Melvin and Bren Simon Cancer CenterIndianapolisINUSA
| |
Collapse
|
8
|
Liu J, Qiu L, Xia J, Chen S, Yu X, Zhou Y. ZGDHu-1 for cancer therapy. Oncol Lett 2018; 14:6334-6340. [PMID: 29344112 PMCID: PMC5754890 DOI: 10.3892/ol.2017.7096] [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: 07/20/2016] [Accepted: 01/31/2017] [Indexed: 11/06/2022] Open
Abstract
N,N'-di-(m-methylphenyl)-3,6-dimethyl-1,4-dihydro-1,2,4,5-tetrazine-1,4-dicarboamide (ZGDHu-1) is a novel tetrazine derivative that was initially designed and produced by Professor W.X. Hu, and which has been reported by our group to exhibit antitumor activity. Accumulating evidence suggests that the anticancer mechanisms of ZGDHu-1 may be involved indifferent biological activities, particularly in acute myeloid leukemia (AML) cells. At a high concentration, ZGDHu-1 has been demonstrated to inhibit the proliferation of the leukemia cells by arresting the cell cycle at the G2/M phase, and by inducing cell apoptosis via inducing the accumulation of reactive oxygen species, the translocation of phosphatidylserine across the plasma membrane and the loss of mitochondrial membrane potential. Furthermore, at a low concentration, it was demonstrated to induce the differentiation and degrade the AML1-eight-twenty-one fusion protein in AML cells. Finally, results from a previous study indicate that ZGDHu-1 is a potential proteasome inhibitor. Overall, our preliminary research suggests that ZGDHu-1 may be a promising anticancer drug; however, further research is warranted to identify the exact drug target and potential clinical application in leukemia cells or solid tumors. In the present review, the application of ZGDHu-1 in cancer research, in addition to the specific underlying targets of ZGDHu-1, are discussed.
Collapse
Affiliation(s)
- Jinlin Liu
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Liannv Qiu
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Jun Xia
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Sufeng Chen
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiping Yu
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Yonglie Zhou
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
9
|
Gbolahan OB, Zeidan AM, Stahl M, Abu Zaid M, Farag S, Paczesny S, Konig H. Immunotherapeutic Concepts to Target Acute Myeloid Leukemia: Focusing on the Role of Monoclonal Antibodies, Hypomethylating Agents and the Leukemic Microenvironment. Int J Mol Sci 2017; 18:E1660. [PMID: 28758974 PMCID: PMC5578050 DOI: 10.3390/ijms18081660] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/22/2017] [Accepted: 07/24/2017] [Indexed: 12/20/2022] Open
Abstract
Intensive chemotherapeutic protocols and allogeneic stem cell transplantation continue to represent the mainstay of acute myeloid leukemia (AML) treatment. Although this approach leads to remissions in the majority of patients, long-term disease control remains unsatisfactory as mirrored by overall survival rates of approximately 30%. The reason for this poor outcome is, in part, due to various toxicities associated with traditional AML therapy and the limited ability of most patients to tolerate such treatment. More effective and less toxic therapies therefore represent an unmet need in the management of AML, a disease for which therapeutic progress has been traditionally slow when compared to other cancers. Several studies have shown that leukemic blasts elicit immune responses that could be exploited for the development of novel treatment concepts. To this end, early phase studies of immune-based therapies in AML have delivered encouraging results and demonstrated safety and feasibility. In this review, we discuss opportunities for immunotherapeutic interventions to enhance the potential to achieve a cure in AML, thereby focusing on the role of monoclonal antibodies, hypomethylating agents and the leukemic microenvironment.
Collapse
Affiliation(s)
- Olumide Babajide Gbolahan
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Amer M Zeidan
- Department of Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Maximilian Stahl
- Department of Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Mohammad Abu Zaid
- Department of Medicine, Bone Marrow and Stem Cell Transplantation, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Sherif Farag
- Department of Medicine, Bone Marrow and Stem Cell Transplantation, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Sophie Paczesny
- Wells Center for Pediatric Research, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Heiko Konig
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
10
|
Abstract
Midostaurin was recently approved by the US Food and Drug Administration for the treatment of FLT3-mutant acute myeloid leukemia (AML). This is the first drug to receive regulatory approval for AML in the United States since the year 2000. Midostaurin is a small-molecule kinase inhibitor with activity against the receptor tyrosine kinase FLT3, and its approval will hopefully mark the beginning of an era of targeted agents for the treatment of molecularly defined subtypes of AML.
Collapse
|
11
|
Sak K, Everaus H. Established Human Cell Lines as Models to Study Anti-leukemic Effects of Flavonoids. Curr Genomics 2017; 18:3-26. [PMID: 28503087 PMCID: PMC5321770 DOI: 10.2174/1389202917666160803165447] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 11/20/2015] [Accepted: 11/27/2015] [Indexed: 12/19/2022] Open
Abstract
Despite the extensive work on pathological mechanisms and some recent advances in the treatment of different hematological malignancies, leukemia continues to present a significant challenge being frequently considered as incurable disease. Therefore, the development of novel therapeutic agents with high efficacy and low toxicity is urgently needed to improve the overall survival rate of patients. In this comprehensive review article, the current knowledge about the anticancer activities of flavonoids as plant secondary polyphenolic metabolites in the most commonly used human established leukemia cell lines (HL-60, NB4, KG1a, U937, THP-1, K562, Jurkat, CCRF- CEM, MOLT-3, and MOLT-4) is compiled, revealing clear anti-proliferative, pro-apoptotic, cell cycle arresting, and differentiation inducing effects for certain compounds. Considering the low toxicity of these substances in normal blood cells, the presented data show a great potential of flavonoids to be developed into novel anti-leukemia agents applicable also in the malignant cells resistant to the current conventional chemotherapeutic drugs.
Collapse
Affiliation(s)
- Katrin Sak
- Department of Hematology and Oncology, University of Tartu, Tartu, Estonia
| | - Hele Everaus
- Department of Hematology and Oncology, University of Tartu, Tartu, Estonia
| |
Collapse
|
12
|
Ronson A, Tvito A, Rowe JM. Liposomal cytarabine and daunorubicin (CPX-351) for treatment of acute myeloid leukemia. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2016.1256768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Aaron Ronson
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Ariella Tvito
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Jacob M. Rowe
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
- Department of Hematology and Bone Marrow Transplantation, Haifa, Israel
- Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
13
|
Khan M, Mansoor AER, Kadia TM. Future prospects of therapeutic clinical trials in acute myeloid leukemia. Future Oncol 2016; 13:523-535. [PMID: 27771959 DOI: 10.2217/fon-2016-0262] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is a markedly heterogeneous hematological malignancy that is most commonly seen in elderly adults. The response to current therapies to AML is quite variable, and very few new drugs have been recently approved for use in AML. This review aims to discuss the issues with current trial design for AML therapies, including trial end points, patient enrollment, cost of drug discovery and patient heterogeneity. We also discuss the future directions in AML therapeutics, including intensification of conventional therapy and new drug delivery mechanisms; targeted agents, including epigenetic therapies, cell cycle regulators, hypomethylating agents and chimeric antigen receptor T-cell therapy; and detail of the possible agents that may be incorporated into the treatment of AML in the future.
Collapse
Affiliation(s)
- Maliha Khan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
14
|
[Clinical and biological prognostic factors in relapsed acute myeloid leukemia patients]. Med Clin (Barc) 2016; 147:185-191. [PMID: 27374030 DOI: 10.1016/j.medcli.2016.05.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 05/17/2016] [Accepted: 05/19/2016] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND OBJECTIVE Acute myeloid leukemia (AML) is the most frequent type of acute leukemia in adults. Despite recent advances in the characterization of pathogenesis of AML, the cure rates are under 40%, being leukemia relapse the most common cause of treatment failure. Leukaemia relapse occurs due to clonal evolution or clonal escape. In this study, we aimed to analyze the clinical and biological factors influencing outcomes in patients with AML relapse. PATIENTS AND METHODS We included a total of 75 AML patients who experienced leukaemia relapse after achieving complete remission. We performed complete immunophenotyping and conventional karyotyping in bone marrow aspirates obtained at diagnosis and at leukemia relapse. RESULTS Overall survival (OS) of the series was 3.7%±2.3, leukaemia progression being the most common cause of death. Patients relapsing before 12 months and those with adverse cytogenetic-molecular risk had statistically significant worse outcomes. A percentage of 52.5 of patients showed phenotypic changes and 50% cytogenetic changes at relapse. We did not find significant clinical factors predicting clonal evolution. The presence of clonal evolution at relapse did not have a significant impact on outcome. CONCLUSIONS Patients with relapsed AML have a dismal prognosis, especially those with early relapse and adverse cytogenetic-molecular risk. Clonal evolution with phenotypic and cytogenetic changes occurred in half of the patients without predictive clinical factors or impact on outcome.
Collapse
|
15
|
Chisini M, Stefanizzi C, Ceglie T, Raponi S, Vozella F, Colafigli G, Salaroli A, D'Angiò M, Mancini M, Diverio D, Breccia M, Mancini F, Minotti C, Trisolini S, Capria S, Testi AM, Guarini A, Latagliata R, De Propris MS, Foà R. Independent prognostic impact of CD15 on complete remission achievement in patients with acute myeloid leukemia. Hematol Oncol 2016; 35:804-809. [PMID: 27400753 DOI: 10.1002/hon.2331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 02/06/2023]
Abstract
The prognostic role of CD15 in acute myeloid leukemia (AML) has been tested in different studies with conflicting results. To address this issue, we retrospectively evaluated a cohort of 460 AML patients of all ages with the exclusion of acute promyelocytic leukemia (M/F 243/217, median age 50.6 years [range 0.9-81.2]) intensively treated at our institute between January 1999 and December 2010. CD15 positivity was found in 171 of 406 evaluable patients (42.1%). Complete remission (CR) was achieved by 334 patients (72.6%), while 82 (17.8%) were resistant and 44 (9.6%) died during induction: the median CR duration was 15.5 months (range 0.6-176.0), with 2-year disease-free survival rate of 45.1% (95% confidence interval 39.6-50.6). The median overall survival was 14.4 months (range 0.3-177.0), with 2-year overall survival rate of 42.2% (95% confidence interval 37.5-46.9). At univariate analysis for CR achievement, age < 60 years (P < .001), World Health Organization classification (P = .045), low-risk karyotype (P < .001), no high-risk karyotype (P = .006), positivity for AML-ETO (P = .004)/CBFβ-MYH11 (P = .003)/CD15 (P = .006)/CD11b (P = .013), negativity for FLT3-ITD (P = .001), Hb > 8 g/dL (P = .020), and white blood cell < 50 × 109 /L (P = .034) had a favorable impact. At a multivariate logistic regression model, CD15 positivity (P = .002), age < 60 years (P = .008), white blood cell < 50 × 109 /L (P = .017), and low-risk/no high-risk karyotype (P = .026/P = .025) retained an independent prognostic role on CR achievement. The baseline assessment of CD15 positivity appears to have a role in the risk evaluation for CR achievement in AML patients undergoing intensive chemotherapy and should be assessed in prospective studies together with other clinical and biologic features already reported.
Collapse
Affiliation(s)
- Marta Chisini
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Caterina Stefanizzi
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Teresa Ceglie
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Sara Raponi
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Federico Vozella
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Gioia Colafigli
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Adriano Salaroli
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Mariella D'Angiò
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Marco Mancini
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Daniela Diverio
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Massimo Breccia
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Francesca Mancini
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Clara Minotti
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Silvia Trisolini
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Saveria Capria
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Anna Maria Testi
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Anna Guarini
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Roberto Latagliata
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | | | - Robin Foà
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
16
|
Islam A. Induction treatment of acute myeloid leukemia in an elderly patient with intramarrow injection/administration of cytarabine: first report of a case. Clin Case Rep 2015; 3:1026-9. [PMID: 26732947 PMCID: PMC4693691 DOI: 10.1002/ccr3.429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 09/28/2015] [Accepted: 10/01/2015] [Indexed: 11/08/2022] Open
Abstract
We have used intramarrow injection/administration of cytarabine (Ara-C) instead of conventional intravenous approach to induce remission in an elderly patient with acute myelogenous leukemia. We show for the first time that the intramarrow injection of chemotherapeutic agents such as Ara-C can be used safely and effectively.
Collapse
Affiliation(s)
- Anwarul Islam
- Division of Hematology/Oncology Department of Medicine Buffalo General Hospital Room E 318 Buffalo New York 14203
| |
Collapse
|
17
|
Abstract
The goal of precision medicine is to personalize therapy based on individual patient variation, to correctly select the right treatment, for the right patient, at the right time. Acute myeloid leukemia (AML) is a heterogeneous collection of myeloid malignancies with diverse genetic etiology and the potential for intra-patient clonal evolution over time. We discuss here how the precision medicine paradigm might be applied to the care of AML patients by focusing on the potential roles of targeting therapy by patient-specific somatic mutations and aberrant pathways, ex-vivo drug sensitivity and resistance testing, high sensitivity measurements of residual disease burden and biology along with potential clinical trial and regulatory constraints.
Collapse
Affiliation(s)
- Catherine Lai
- a Myeloid Malignancies Section, Hematology Branch, National Heart , Lung and Blood Institute, National Institutes of Health , Bethesda , MD , USA
| | - Judith E Karp
- a Myeloid Malignancies Section, Hematology Branch, National Heart , Lung and Blood Institute, National Institutes of Health , Bethesda , MD , USA.,b Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center , Johns Hopkins School of Medicine , Baltimore , MD , USA
| | - Christopher S Hourigan
- a Myeloid Malignancies Section, Hematology Branch, National Heart , Lung and Blood Institute, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|