1
|
Lin C, Patel AA, Huo D, Karrison T, van Besien K, Godwin J, Sher D, Weiner H, Green M, Wade JL, Klisovic R, Baer MR, Larson RA, Stock W, Odenike O. A multicenter phase 2 clinical trial of low-dose subcutaneous decitabine in myelofibrosis. Blood Adv 2024; 8:5735-5743. [PMID: 39250708 DOI: 10.1182/bloodadvances.2024013215] [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: 03/22/2024] [Revised: 07/15/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
ABSTRACT Myelofibrosis (MF) in the chronic phase is a challenging disease to treat, and conventional treatment options are geared toward symptom palliation. In this prospective, multicenter, phase 2 trial, 21 patients with MF (18 chronic phase, 2 accelerated phase, and 1 blast phase) were treated with a 10-day schedule of subcutaneous decitabine at 0.3 mg/kg per day. The overall response rate was 33% (95% confidence interval, 15-57), primarily manifested as an improvement in cytopenias. The median duration of response was 7 months (range, 3-44). A high International Prognostic Scoring System risk score, high baseline fetal hemoglobin level, and sustained decrease in circulating CD34+ cell counts were associated with response to decitabine. All patients experienced at least 1 grade 3/4 cytopenia. Nonhematologic toxicities were less frequent, with fatigue, anorexia, and hypocalcemia being the most common. Given the lack of effective therapies in MF with severe cytopenias, this study supports further investigation into the use of hypomethylating agents as single agents or in combination therapies. This trial was registered at www.ClinicalTrials.gov as #NCT00095784.
Collapse
Affiliation(s)
- Chenyu Lin
- The University of Chicago Pritzker School of Medicine, Chicago, IL
| | - Anand A Patel
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Dezheng Huo
- Department of Public Health Sciences, The University of Chicago, Chicago, IL
| | - Theodore Karrison
- Department of Public Health Sciences, The University of Chicago, Chicago, IL
| | - Koen van Besien
- Division of Hematology and Cellular Therapy, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - John Godwin
- Earle A. Chiles Research Institute, Providence Portland Cancer Institute, Portland, OR
| | - Dorie Sher
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Howie Weiner
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Margaret Green
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - James L Wade
- Medical Oncology, Decatur Memorial Hospital, Decatur, IL
| | - Rebecca Klisovic
- Division of Hematology & Medical Oncology, Emory University School of Medicine, Atlanta, GA
| | - Maria R Baer
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Richard A Larson
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
- The University of Chicago Comprehensive Cancer Center, Chicago, IL
| | - Wendy Stock
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Olatoyosi Odenike
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| |
Collapse
|
2
|
Mangione R, Giallongo C, Duminuco A, La Spina E, Longhitano L, Giallongo S, Tibullo D, Lazzarino G, Saab MW, Sbriglione A, Palumbo GA, Graziani A, Alanazi AM, Di Pietro V, Tavazzi B, Amorini AM, Lazzarino G. Targeted Metabolomics Highlights Dramatic Antioxidant Depletion, Increased Oxidative/Nitrosative Stress and Altered Purine and Pyrimidine Concentrations in Serum of Primary Myelofibrosis Patients. Antioxidants (Basel) 2024; 13:490. [PMID: 38671937 PMCID: PMC11047794 DOI: 10.3390/antiox13040490] [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: 03/03/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
To date, little is known concerning the circulating levels of biochemically relevant metabolites (antioxidants, oxidative/nitrosative stress biomarkers, purines, and pyrimidines) in patients with primary myelofibrosis (PMF), a rare form of myeloproliferative tumor causing a dramatic decrease in erythropoiesis and angiogenesis. In this study, using a targeted metabolomic approach, serum samples of 22 PMF patients and of 22 control healthy donors were analyzed to quantify the circulating concentrations of hypoxanthine, xanthine, uric acid (as representative purines), uracil, β-pseudouridine, uridine (as representative pyrimidines), reduced glutathione (GSH), ascorbic acid (as two of the main water-soluble antioxidants), malondialdehyde, nitrite, nitrate (as oxidative/nitrosative stress biomarkers) and creatinine, using well-established HPLC method for their determination. Results showed that PMF patients have dramatic depletions of both ascorbic acid and GSH (37.3- and 3.81-times lower circulating concentrations, respectively, than those recorded in healthy controls, p < 0.0001), accompanied by significant increases in malondialdehyde (MDA) and nitrite + nitrate (4.73- and 1.66-times higher circulating concentrations, respectively, than those recorded in healthy controls, p < 0.0001). Additionally, PMF patients have remarkable alterations of circulating purines, pyrimidines, and creatinine, suggesting potential mitochondrial dysfunctions causing energy metabolism imbalance and consequent increases in these cell energy-related compounds. Overall, these results, besides evidencing previously unknown serum metabolic alterations in PMF patients, suggest that the determination of serum levels of the aforementioned compounds may be useful to evaluate PMF patients on hospital admission for adjunctive therapies aimed at recovering their correct antioxidant status, as well as to monitor patients' status and potential pharmacological treatments.
Collapse
Affiliation(s)
- Renata Mangione
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of the Sacred Heart of Rome, Largo F. Vito 1, 00168 Rome, Italy;
- Departmental Faculty of Medicine, UniCamillus—Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy; (A.G.); (G.L.)
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Division of Hematology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (C.G.); (S.G.); (G.A.P.)
| | - Andrea Duminuco
- Hematology Unit with BMT, A.O.U. Policlinico “G.Rodolico-San Marco”, Via S. Sofia 78, 95123 Catania, Italy;
| | - Enrico La Spina
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Lucia Longhitano
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Sebastiano Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Division of Hematology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (C.G.); (S.G.); (G.A.P.)
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Miriam Wissam Saab
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Arianna Sbriglione
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Giuseppe A. Palumbo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Division of Hematology, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (C.G.); (S.G.); (G.A.P.)
| | - Andrea Graziani
- Departmental Faculty of Medicine, UniCamillus—Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy; (A.G.); (G.L.)
| | - Amer M. Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Valentina Di Pietro
- Neurotrauma and Ophthalmology Research Group, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital, Edgbaston, Birmingham B15 2TH, UK
| | - Barbara Tavazzi
- Departmental Faculty of Medicine, UniCamillus—Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy; (A.G.); (G.L.)
| | - Angela Maria Amorini
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (E.L.S.); (L.L.); (D.T.); (G.L.); (M.W.S.); (A.S.)
| | - Giacomo Lazzarino
- Departmental Faculty of Medicine, UniCamillus—Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy; (A.G.); (G.L.)
| |
Collapse
|
3
|
Liu L, Song X, Dong W, Li Z, Guo D. Case report: Safety and efficacy of synergistic treatment using selinexor and azacitidine in patients with atypical chronic myeloid leukemia with resistance to decitabine. Front Oncol 2024; 14:1353818. [PMID: 38384813 PMCID: PMC10879427 DOI: 10.3389/fonc.2024.1353818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Background Atypical chronic myeloid leukemia (aCML) is a BCR::ABL1 negative myelodysplastic/myeloproliferative neoplasm with poor overall survival. Some patients can be treated by allogeneic hematopoietic stem cell transplantation (allo-HSCT) from suitable donors. The effectiveness of decitabine or azacitidine (AZA) has recently been reported; however, their combined efficacy with selinexor has not yet been reported. Case description In this study, we report the case of a patient with aCML who was successfully treated with selinexor combined with AZA. A 67-year-old man with a history of gastric mucosa-associated lymphoid tissue (MALT) lymphoma was admitted to the hospital with fatigue and emaciation. He was diagnosed with aCML and no longer responded to decitabine treatment after undergoing seven cycles. The patient was subsequently administered hydroxyurea (HU), selinexor, and AZA. After four courses of combination therapy, his blood cell counts improved; he no longer required transfusions and was able to discontinue HU. The patient continued receiving selinexor and AZA without severe complications. This case is the first to show that combinatorial selinexor and AZA therapy can effectively treat aCML. Conclusion Our case sheds light on the importance of selinexor and AZA combined therapy in the exploration of new treatment strategies for aCML. Moreover, this treatment approach offers the possibility of bridging with allo-HSCT.
Collapse
Affiliation(s)
- Lu Liu
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Xiaofeng Song
- Department of Hand and Foot Surgery, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Wenhao Dong
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Zhao Li
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Dongmei Guo
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| |
Collapse
|
4
|
Barosi G, Campanelli R, Catarsi P, Abbà C, Carolei A, Massa M, Gale RP, Rosti V. Type 1 CALR mutation allele frequency correlates with CD34/CXCR4 expression in myelofibrosis-type megakaryocyte dysplasia: A mechanism of disease progression? Blood Cancer J 2024; 14:18. [PMID: 38253566 PMCID: PMC10803778 DOI: 10.1038/s41408-024-00991-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Affiliation(s)
- Giovanni Barosi
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy.
| | - Rita Campanelli
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy
| | - Paolo Catarsi
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy
| | - Carlotta Abbà
- General Medicine 2, Center for Sistemic Amyloidosis and High Complexity Diseases, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo Foundation, Pavia, Italy
| | - Adriana Carolei
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy
| | - Margherita Massa
- General Medicine 2, Center for Sistemic Amyloidosis and High Complexity Diseases, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo Foundation, Pavia, Italy
| | - Robert Peter Gale
- Centre for Haematology Research, Imperial College London, London, UK
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy
| |
Collapse
|
5
|
Tremblay D, Mascarenhas J. Pharmacotherapeutic advances for splenomegaly in myelofibrosis. Expert Opin Pharmacother 2023; 24:577-585. [PMID: 36922391 DOI: 10.1080/14656566.2023.2192350] [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: 03/18/2023]
Abstract
INTRODUCTION Myelofibrosis is a hematologic malignancy with a variety of clinical manifestations including splenomegaly, which is present in approximately 80% of newly diagnosed patients. JAK inhibitors are the mainstay of pharmacologic treatment for splenomegaly in myelofibrosis, although spleen size reduction is not universal, and the duration of benefit is only moderately durable. AREAS COVERED We first discuss the pathobiology of splenomegaly in myelofibrosis before detailing approved and novel pharmacotherapies that can reduce spleen size while also highlighting non-pharmacologic approaches. In this review, efficacy of these treatments is measured solely by spleen volume reduction, acknowledging that other outcome measures such as symptom improvement and survival are also critical. EXPERT OPINION Currently, ruxolitinib can be administered to the majority of frontline patients although those with severe thrombocytopenia should receive pacritinib to address spleen burden. Momelotinib may be particularly well suited for patients with significant anemia and novel combination treatments in clinical development may improve the depth and duration of spleen responses. After frontline treatment failure, fedratinib, or pacritinib are commercial options for patients with persistent symptomatic splenomegaly. Novel agents given alone or in combination with a JAK inhibitor are being explored in trials, which may ameliorate splenomegaly and ultimately improve disease progression.
Collapse
Affiliation(s)
- Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
6
|
Chia YC, Siti Asmaa MJ, Ramli M, Woon PY, Johan MF, Hassan R, Islam MA. Molecular Genetics of Thrombotic Myeloproliferative Neoplasms: Implications in Precision Oncology. Diagnostics (Basel) 2023; 13:163. [PMID: 36611455 PMCID: PMC9818412 DOI: 10.3390/diagnostics13010163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 01/06/2023] Open
Abstract
Classical BCR-ABL-negative myeloproliferative neoplasms (MPN) include polycythaemia vera, essential thrombocythaemia, and primary myelofibrosis. Unlike monogenic disorders, a more complicated series of genetic mutations are believed to be responsible for MPN with various degrees of thromboembolic and bleeding complications. Thrombosis is one of the early manifestations in patients with MPN. To date, the driver genes responsible for MPN include JAK2, CALR, MPL, TET2, ASXL1, and MTHFR. Affords have been done to elucidate these mutations and the incidence of thromboembolic events. Several lines of evidence indicate that mutations in JAK2, MPL, TET2 and ASXL1 gene and polymorphisms in several clotting factors (GPIa, GPIIa, and GPIIIa) are associated with the occurrence and prevalence of thrombosis in MPN patients. Some polymorphisms within XRCC1, FBG, F2, F5, F7, F12, MMP9, HPA5, MTHFR, SDF-1, FAS, FASL, TERT, ACE, and TLR4 genes may also play a role in MPN manifestation. This review aims to provide an insightful overview on the genetic perspective of thrombotic complications in patients with MPN.
Collapse
Affiliation(s)
- Yuh Cai Chia
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Mat Jusoh Siti Asmaa
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Marini Ramli
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Peng Yeong Woon
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan
| | - Muhammad Farid Johan
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Rosline Hassan
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Md Asiful Islam
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
7
|
Arumugam T, Ramphal U, Adimulam T, Chinniah R, Ramsuran V. Deciphering DNA Methylation in HIV Infection. Front Immunol 2021; 12:795121. [PMID: 34925380 PMCID: PMC8674454 DOI: 10.3389/fimmu.2021.795121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
With approximately 38 million people living with HIV/AIDS globally, and a further 1.5 million new global infections per year, it is imperative that we advance our understanding of all factors contributing to HIV infection. While most studies have focused on the influence of host genetic factors on HIV pathogenesis, epigenetic factors are gaining attention. Epigenetics involves alterations in gene expression without altering the DNA sequence. DNA methylation is a critical epigenetic mechanism that influences both viral and host factors. This review has five focal points, which examines (i) fluctuations in the expression of methylation modifying factors upon HIV infection (ii) the effect of DNA methylation on HIV viral genes and (iii) host genome (iv) inferences from other infectious and non-communicable diseases, we provide a list of HIV-associated host genes that are regulated by methylation in other disease models (v) the potential of DNA methylation as an epi-therapeutic strategy and biomarker. DNA methylation has also been shown to serve as a robust therapeutic strategy and precision medicine biomarker against diseases such as cancer and autoimmune conditions. Despite new drugs being discovered for HIV, drug resistance is a problem in high disease burden settings such as Sub-Saharan Africa. Furthermore, genetic therapies that are under investigation are irreversible and may have off target effects. Alternative therapies that are nongenetic are essential. In this review, we discuss the potential role of DNA methylation as a novel therapeutic intervention against HIV.
Collapse
Affiliation(s)
- Thilona Arumugam
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Upasana Ramphal
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Theolan Adimulam
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Romona Chinniah
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Veron Ramsuran
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
8
|
Campanelli R, Massa M, Rosti V, Barosi G. New Markers of Disease Progression in Myelofibrosis. Cancers (Basel) 2021; 13:5324. [PMID: 34771488 PMCID: PMC8582535 DOI: 10.3390/cancers13215324] [Citation(s) in RCA: 3] [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: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/30/2022] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm due to the clonal proliferation of a hematopoietic stem cell. The vast majority of patients harbor a somatic gain of function mutation either of JAK2 or MPL or CALR genes in their hematopoietic cells, resulting in the activation of the JAK/STAT pathway. Patients display variable clinical and laboratoristic features, including anemia, thrombocytopenia, splenomegaly, thrombotic complications, systemic symptoms, and curtailed survival due to infections, thrombo-hemorrhagic events, or progression to leukemic transformation. New drugs have been developed in the last decade for the treatment of PMF-associated symptoms; however, the only curative option is currently represented by allogeneic hematopoietic cell transplantation, which can only be offered to a small percentage of patients. Disease prognosis is based at diagnosis on the classical International Prognostic Scoring System (IPSS) and Dynamic-IPSS (during disease course), which comprehend clinical parameters; recently, new prognostic scoring systems, including genetic and molecular parameters, have been proposed as meaningful tools for a better patient stratification. Moreover, new biological markers predicting clinical evolution and patient survival have been associated with the disease. This review summarizes basic concepts of PMF pathogenesis, clinics, and therapy, focusing on classical prognostic scoring systems and new biological markers of the disease.
Collapse
Affiliation(s)
- Rita Campanelli
- Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (V.R.); (G.B.)
| | - Margherita Massa
- General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (V.R.); (G.B.)
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (V.R.); (G.B.)
| |
Collapse
|
9
|
Chia YC, Ramli M, Woon PY, Johan MF, Hassan R, Islam MA. WITHDRAWN: Molecular genetics of thrombotic myeloproliferative neoplasms: Implications in precision oncology. Genes Dis 2021. [DOI: 10.1016/j.gendis.2021.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
10
|
Rambaldi B, Diral E, Donsante S, Di Marzo N, Mottadelli F, Cardinale L, Dander E, Isimbaldi G, Pioltelli P, Biondi A, Riminucci M, D'Amico G, Elli EM, Pievani A, Serafini M. Heterogeneity of the bone marrow niche in patients with myeloproliferative neoplasms: ActivinA secretion by mesenchymal stromal cells correlates with the degree of marrow fibrosis. Ann Hematol 2020; 100:105-116. [PMID: 33089365 DOI: 10.1007/s00277-020-04306-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/15/2020] [Indexed: 01/19/2023]
Abstract
Mesenchymal stromal cells (MSCs) represent an essential component of the bone marrow (BM) niche and display disease-specific alterations in several myeloid malignancies. The aim of this work was to study possible MSC abnormalities in Philadelphia-negative myeloproliferative neoplasms (MPNs) in relationship to the degree of BM fibrosis. MSCs were isolated from BM of 6 healthy donors (HD) and of 23 MPN patients, classified in 3 groups according to the diagnosis and the grade of BM fibrosis: polycythemia vera and essential thrombocythemia (PV/ET), low fibrosis myelofibrosis (LF-MF), and high fibrosis MF (HF-MF). MSC cultures were established from 21 of 23 MPN patients. MPN-derived MSCs did not exhibit any functional impairment in their adipogenic/osteogenic/chondrogenic differentiation potential and displayed a phenotype similar to HD-derived MSCs but with a decreased expression of CD146. All MPN-MSC lines were negative for the patient-specific hematopoietic clone mutations (JAK2, MPL, CALR). MSCs derived from HF-MF patients displayed a reduced clonogenic potential and a lower growth kinetic compared to MSCs from HD, LF-MF, and PV/ET patients. mRNA levels of hematopoiesis regulatory molecules were unaffected in MSCs from HF-MF compared to HD. Finally, in vitro ActivinA secretion by MSCs was increased in HF-MF compared to LF-MF patients, in association with a lower hemoglobin value. Increased ActivinA immunolabeling on stromal cells and erythroid precursors was also observed in HF-MF BM biopsies. In conclusion, higher grade of BM fibrosis is associated with functional impairment of MSCs and the increased secretion of ActivinA may represent a suitable target for anemia treatment in MF patients.
Collapse
Affiliation(s)
- Benedetta Rambaldi
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy.,Department of Hematology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy.,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Elisa Diral
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy.,Department of Hematology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy.,Hematology Department, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Noemi Di Marzo
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Federica Mottadelli
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Lucia Cardinale
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Erica Dander
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Giuseppe Isimbaldi
- Department of Pathology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy.,Department of Pathology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Pietro Pioltelli
- Department of Hematology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Andrea Biondi
- Department of Pediatrics, Fondazione MBBM/San Gerardo Hospital, Monza, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Giovanna D'Amico
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Elena Maria Elli
- Department of Hematology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy.
| | - Alice Pievani
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Marta Serafini
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy.
| |
Collapse
|
11
|
Vigorelli V, Resta J, Bianchessi V, Lauri A, Bassetti B, Agrifoglio M, Pesce M, Polvani G, Bonalumi G, Cavallotti L, Alamanni F, Genovese S, Pompilio G, Vinci MC. Abnormal DNA Methylation Induced by Hyperglycemia Reduces CXCR 4 Gene Expression in CD 34 + Stem Cells. J Am Heart Assoc 2020; 8:e010012. [PMID: 31018749 PMCID: PMC6512087 DOI: 10.1161/jaha.118.010012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background CD 34+ stem/progenitor cells are involved in vascular homeostasis and in neovascularization of ischemic tissues. The number of circulating CD 34+ stem cells is a predictive biomarker of adverse cardiovascular outcomes in diabetic patients. Here, we provide evidence that hyperglycemia can be "memorized" by the stem cells through epigenetic changes that contribute to onset and maintenance of their dysfunction in diabetes mellitus. Methods and Results Cord-blood-derived CD 34+ stem cells exposed to high glucose displayed increased reactive oxygen species production, overexpression of p66shc gene, and downregulation of antioxidant genes catalase and manganese superoxide dismutase when compared with normoglycemic cells. This altered oxidative state was associated with impaired migration ability toward stromal-cell-derived factor 1 alpha and reduced protein and mRNA expression of the C-X-C chemokine receptor type 4 ( CXCR 4) receptor. The methylation analysis by bisulfite Sanger sequencing of the CXCR 4 promoter revealed a significant increase in DNA methylation density in high-glucose CD 34+ stem cells that negatively correlated with mRNA expression (Pearson r=-0.76; P=0.004). Consistently, we found, by chromatin immunoprecipitation assay, a more transcriptionally inactive chromatin conformation and reduced RNA polymerase II engagement on the CXCR 4 promoter. Notably, alteration of CXCR 4 DNA methylation, as well as transcriptional and functional defects, persisted in high-glucose CD 34+ stem cells despite recovery in normoglycemic conditions. Importantly, such an epigenetic modification was thoroughly confirmed in bone marrow CD 34+ stem cells isolated from sternal biopsies of diabetic patients undergoing coronary bypass surgery. Conclusions CD 34+ stem cells "memorize" the hyperglycemic environment in the form of epigenetic modifications that collude to alter CXCR 4 receptor expression and migration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Francesco Alamanni
- 1 IRCCS Centro Cardiologico Monzino Milan Italy.,3 Department of Clinical Sciences and Community Health Università degli Studi di Milano Milan Italy
| | | | - Giulio Pompilio
- 1 IRCCS Centro Cardiologico Monzino Milan Italy.,3 Department of Clinical Sciences and Community Health Università degli Studi di Milano Milan Italy
| | | |
Collapse
|
12
|
Longhitano L, Li Volti G, Giallongo C, Spampinato M, Barbagallo I, Di Rosa M, Romano A, Avola R, Tibullo D, Palumbo GA. The Role of Inflammation and Inflammasome in Myeloproliferative Disease. J Clin Med 2020; 9:E2334. [PMID: 32707883 PMCID: PMC7464195 DOI: 10.3390/jcm9082334] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/01/2020] [Accepted: 07/17/2020] [Indexed: 12/22/2022] Open
Abstract
Polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF) are rare hematological conditions known as myeloproliferative neoplasms (MPNs). They are characterized for being BCR-ABL negative malignancies and affected patients often present with symptoms which can significantly impact their quality of life. MPNs are characterized by a clonal proliferation of an abnormal hematopoietic stem/progenitor cell. In MPNs; cells of all myeloid lineages; including those involved in the immune and inflammatory response; may belong to the malignant clone thus leading to an altered immune response and an overexpression of cytokines and inflammatory receptors; further worsening chronic inflammation. Many of these cytokines; in particular, IL-1β and IL-18; are released in active form by activating the inflammasome complexes which in turn mediate the inflammatory process. Despite this; little is known about the functional effects of stem cell-driven inflammasome signaling in MPN pathogenesis. In this review we focused on the role of inflammatory pathway and inflammasome in MPN diseases. A better understanding of the inflammatory-state-driving MPNs and of the role of the inflammasome may provide new insights on possible therapeutic strategies.
Collapse
Affiliation(s)
- Lucia Longhitano
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Giovanni Li Volti
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Cesarina Giallongo
- Department of Scienze Mediche Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | - Mariarita Spampinato
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Ignazio Barbagallo
- Section of Biochemistry, Department of Drug Sciences, University of Catania, 95123 Catania, Italy;
| | - Michelino Di Rosa
- Section of Human Anatomy, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Alessandra Romano
- Division of Hematology, Department of General Surgery and Medical-Surgical Specialties, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy;
| | - Roberto Avola
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Daniele Tibullo
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Giuseppe Alberto Palumbo
- Department of Scienze Mediche Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
13
|
Yang X, Chen D, Long H, Zhu B. The mechanisms of pathological extramedullary hematopoiesis in diseases. Cell Mol Life Sci 2020; 77:2723-2738. [PMID: 31974657 PMCID: PMC11104806 DOI: 10.1007/s00018-020-03450-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/24/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
Extramedullary hematopoiesis (EMH) is the expansion and differentiation of hematopoietic stem and progenitor cells outside of the bone marrow. In postnatal life, as a compensatory mechanism for ineffective hematopoiesis of the bone marrow, pathological EMH is triggered by hematopoietic disorders, insufficient hematopoietic compensation, and other pathological stress conditions, such as infection, advanced tumors, anemia, and metabolic stress. Pathological EMH has been reported in many organs, and the sites of pathological EMH may be related to reactivation of the embryonic hematopoietic structure in these organs. As a double-edged sword (blood and immune cell supplementation as well as clinical complications), pathological EMH has been widely studied in recent years. In particular, pathological EMH induced by late-stage tumors contributes to tumor immunosuppression. Thus, a deeper understanding of the mechanism of pathological EMH may be conducive to the development of therapies against the pathological processes that induce EMH. This article reviews the recent progress of research on the cellular and molecular mechanisms of pathological EMH in specific diseases.
Collapse
Affiliation(s)
- Xinxin Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
14
|
Reduced CXCR4-expression on CD34-positive blood cells predicts outcomes of persons with primary myelofibrosis. Leukemia 2020; 35:468-475. [PMID: 32536689 DOI: 10.1038/s41375-020-0926-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 01/20/2023]
Abstract
The expression of the CXCR4 chemokine receptor on CD34-positive blood cells is reduced in persons with primary myelofibrosis (PMF). We analyzed the relevance of cytofluorimetric assessment of the percentage of CD34-positive blood cells that had a positive CXCR4 surface expression (CD34/CXCR4-se) in a large cohort of subjects with myeloproliferative neoplasms. Mean CD34/CXCR4-se was lower in subjects with PMF compared with those with essential thrombocythemia (ET) or polycythemia vera (PV). A cutoff value of 39% was associated with a diagnosis of pre-fibrotic PMF vs. ET with a positive predictive value of 97%. In PMF male sex, older age, and MPL mutation were independent correlates of reduced CD34/CXCR4-se and associated with a briefer interval to development of severe anemia, large splenomegaly, thrombocytopenia, leukopenia, elevated CD34-positive blood cells, blast transformation and death. We constructed a prognostic model including age >65 years, hemoglobin < 100 g/L, CD34-positive blood cells > 50 × 106/L, and CD34/CXCR4-se <39% at diagnosis. The model identified three risk cohorts with greater accuracy compared with the International Prognostic Scoring System. In conclusion, CD34/CXCR4-se is a highly sensitive marker of disease activity and a new potential diagnostic and prognostic biomarker in PMF.
Collapse
|
15
|
Zingariello M, Martelli F, Verachi P, Bardelli C, Gobbo F, Mazzarini M, Migliaccio AR. Novel targets to cure primary myelofibrosis from studies on Gata1 low mice. IUBMB Life 2019; 72:131-141. [PMID: 31749302 DOI: 10.1002/iub.2198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/24/2019] [Indexed: 01/06/2023]
Abstract
In 2002, we discovered that mice carrying the hypomorphic Gata1low mutation that reduces expression of the transcription factor GATA1 in megakaryocytes (Gata1low mice) develop myelofibrosis, a phenotype that recapitulates the features of primary myelofibrosis (PMF), the most severe of the Philadelphia-negative myeloproliferative neoplasms (MPNs). At that time, this discovery had a great impact on the field because mutations driving the development of PMF had yet to be discovered. Later studies identified that PMF, as the others MPNs, is associated with mutations activating the thrombopoietin/JAK2 axis raising great hope that JAK inhibitors may be effective to treat the disease. Unfortunately, ruxolitinib, the JAK1/2 inhibitor approved by FDA and EMEA for PMF, ameliorates symptoms but does not improve the natural course of the disease, and the cure of PMF is still an unmet clinical need. Although GATA1 is not mutated in PMF, reduced GATA1 content in megakaryocytes as a consequence of ribosomal deficiency is a hallmark of myelofibrosis (both in humans and mouse models) and, in fact, a driving event in the disease. Conversely, mice carrying the hypomorphic Gata1low mutation express an activated TPO/JAK2 pathway and partially respond to JAK inhibitors in a fashion similar to PMF patients (reduction of spleen size but limited improvement of the natural history of the disease). These observations cross-validated Gata1low mice as a bona fide animal model for PMF and prompted the use of this model to identify abnormalities that might be targeted to cure the disease. We will summarize here data generated in Gata1low mice indicating that the TGF-β/P-selectin axis is abnormal in PMF and represents a novel target for its treatment.
Collapse
Affiliation(s)
- Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | | | - Paola Verachi
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Claudio Bardelli
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Francesca Gobbo
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Maria Mazzarini
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy.,Myeloproliferative Neoplasms Research Consortium, New York, New York
| |
Collapse
|
16
|
Kang N, Choi SY, Kim BN, Yeo CD, Park CK, Kim YK, Kim TJ, Lee SB, Lee SH, Park JY, Park MS, Yim HW, Kim SJ. Hypoxia-induced cancer stemness acquisition is associated with CXCR4 activation by its aberrant promoter demethylation. BMC Cancer 2019; 19:148. [PMID: 30760238 PMCID: PMC6375212 DOI: 10.1186/s12885-019-5360-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 02/07/2019] [Indexed: 01/17/2023] Open
Abstract
Background A hypoxic microenvironment leads to an increase in the invasiveness and the metastatic potential of cancer cells within tumors via the epithelial-mesenchymal transition (EMT) and cancer stemness acquisition. However, hypoxia-induced changes in the expression and function of candidate stem cell markers and their possible molecular mechanism is still not understood. Methods Lung cell lines were analyzed in normoxic or hypoxic conditions. For screening among the stem cell markers, a transcriptome analysis using next-generation sequencing was performed. For validation, the EMT and stem cell characteristics were analyzed. To determine whether an epigenetic mechanism was involved, the cell lines were treated with a DNA methyltransferase inhibitor (AZA), and methylation-specific PCR and bisulfite sequencing were performed. Results Next-generation sequencing revealed that the CXCR4 expression was significantly higher after the hypoxic condition, which functionally resulted in the EMT and cancer stemness acquisition. The acquisition of the EMT and stemness properties was inhibited by treatment with CXCR4 siRNA. The CXCR4 was activated by either the hypoxic condition or treatment with AZA. The methylation-specific PCR and bisulfite sequencing displayed a decreased CXCR4 promoter methylation in the hypoxic condition. Conclusions These results suggest that hypoxia-induced acquisition of cancer stem cell characteristics was associated with CXCR4 activation by its aberrant promoter demethylation.
Collapse
Affiliation(s)
- Nahyeon Kang
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Su Yeon Choi
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Bit Na Kim
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Chan Kwon Park
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Young Kyoon Kim
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Tae-Jung Kim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seong-Beom Lee
- Department of Pathology, Institute of Hansen's Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sug Hyung Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Y Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mi Sun Park
- Department of Biostatistics, Clinical Research Coordinating Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyeon Woo Yim
- Department of Biostatistics, Clinical Research Coordinating Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Joon Kim
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
17
|
Selicean SE, Tomuleasa C, Grewal R, Almeida-Porada G, Berindan-Neagoe I. Mesenchymal stem cells in myeloproliferative disorders - focus on primary myelofibrosis. Leuk Lymphoma 2018; 60:876-885. [PMID: 30277128 DOI: 10.1080/10428194.2018.1516881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Primary myelofibrosis (PMF) is the most aggressive Philadelphia-negative (Ph-) myeloproliferative neoplasm (MPN), characterized by bone marrow (BM) insufficiency, myelofibrosis (MF), osteosclerosis, neoangiogenesis, and extramedullary hematopoiesis (EMH) in spleen and liver. Presently, there is no curative treatment for this disease and therapy consists primarily of symptom relief and, in selected cases, allogeneic hematopoietic stem cell transplant (alloHSCT). PMF's major defining characteristics, as well as several recently described aspects of its cellular and molecular pathophysiology all support a critical role for dysregulated cell-cell/cell-extracellular matrix interactions and cytokine/chemokine signaling within the BM niche in the natural history of this disease. This review will highlight current data concerning the involvement of the BM niche, particularly of mesenchymal stem cells (MSC), in PMF, and will then discuss the rationale for a stroma-directed treatment, and the advantages such an approach would offer over the current treatments focused on targeting the malignant clone.
Collapse
Affiliation(s)
- Sonia Emilia Selicean
- a Research Center for Functional Genomics and Translational Medicine , Iuliu Haţieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania.,b Department of Hematology , Iuliu Haţieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Ciprian Tomuleasa
- a Research Center for Functional Genomics and Translational Medicine , Iuliu Haţieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania.,b Department of Hematology , Iuliu Haţieganu University of Medicine and Pharmacy , Cluj Napoca , Romania.,c Department of Hematology , Ion Chiricuta Clinical Research Center , Cluj Napoca , Romania
| | - Ravnit Grewal
- d Department of Pathology , South African National Bioinformatics Institute , Cape Town , South Africa
| | - Graca Almeida-Porada
- e Wake Forest Institute for Regenerative Medicine , Wake Forest University School of Medicine , Winston-Salem , NC , USA
| | - Ioana Berindan-Neagoe
- a Research Center for Functional Genomics and Translational Medicine , Iuliu Haţieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| |
Collapse
|
18
|
Understanding Splenomegaly in Myelofibrosis: Association with Molecular Pathogenesis. Int J Mol Sci 2018; 19:ijms19030898. [PMID: 29562644 PMCID: PMC5877759 DOI: 10.3390/ijms19030898] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 12/31/2022] Open
Abstract
Myelofibrosis (MF) is a clinical manifestation of chronic BCR-ABL1-negative chronic myeloproliferative neoplasms. Splenomegaly is one of the major clinical manifestations of MF and is directly linked to splenic extramedullary hematopoiesis (EMH). EMH is associated with abnormal trafficking patterns of clonal hematopoietic cells due to the dysregulated bone marrow (BM) microenvironment leading to progressive splenomegaly. Several recent data have emphasized the role of several cytokines for splenic EMH. Alteration of CXCL12/CXCR4 pathway could also lead to splenic EMH by migrated clonal hematopoietic cells from BM to the spleen. Moreover, low Gata1 expression was found to be significantly associated with the EMH. Several gene mutations were found to be associated with significant splenomegaly in MF. In recent data, JAK2V617F homozygous mutation was associated with a larger spleen size. In other data, CALR mutations in MF were signigicantly associated with longer larger splenomegaly-free survivals than others. In addition, MF patients with ≥1 mutations in AZXL1, EZH1 or IDH1/2 had significantly low spleen reduction response in ruxolitinib treatment. Developments of JAK inhibitors, such as ruxolitinib, pacritinib, momelotinib, and febratinib enabled the effective management in MF patients. Especially, significant spleen reduction responses of the drugs were demonstrated in several randomized clinical studies, although those could not eradicate allele burdens of MF.
Collapse
|
19
|
Yeboah A, Maguire T, Schloss R, Berthiaume F, Yarmush ML. Stromal Cell-Derived Growth Factor-1 Alpha-Elastin Like Peptide Fusion Protein Promotes Cell Migration and Revascularization of Experimental Wounds in Diabetic Mice. Adv Wound Care (New Rochelle) 2017; 6:10-22. [PMID: 28116224 DOI: 10.1089/wound.2016.0694] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/28/2016] [Indexed: 12/12/2022] Open
Abstract
Objective: In previous work, we demonstrated the development of a novel fusion protein containing stromal cell-derived growth factor-1 alpha juxtaposed to an elastin-like peptide (SDF1-ELP), which has similar bioactivity, but is more stable in elastase than SDF1. Herein, we compare the ability of a single topical application of SDF1-ELP to that of SDF1 in healing 1 × 1 cm excisional wounds in diabetic mice. Approach: Human Leukemia-60 cells were used to demonstrate the chemotactic potential of SDF1-ELP versus SDF1 in vitro. Human umbilical vascular endothelial cells were used to demonstrate the angiogenic potential of SDF1-ELP versus SDF1 in vitro. The bioactivity of SDF1-ELP versus SDF1 after incubation in ex-vivo diabetic wound fluid was compared. The in-vivo effectiveness of SDF1-ELP versus SDF1 was compared in diabetic mice wound model by monitoring for the number of CD31+ cells in harvested wound tissues. Results: SDF1-ELP promotes the migration of cells and induces vascularization similar to SDF1 in vitro. SDF1-ELP is more stable in wound fluids compared to SDF1. In vivo, SDF1-ELP induced a higher number of vascular endothelial cells (CD31+ cells) compared to SDF1 and other controls, suggesting increased vascularization. Innovation: While growth factors have been shown to improve wound healing, this strategy is largely ineffective in chronic wounds. In this work, we show that SDF1-ELP is a promising agent for the treatment of chronic skin wounds. Conclusion: The superior in vivo performance and stability of SDF1-ELP makes it a promising agent for the treatment of chronic skin wounds.
Collapse
Affiliation(s)
- Agnes Yeboah
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, New Jersey
| | - Tim Maguire
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Rene Schloss
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Martin L. Yarmush
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Burns Hospital, Boston, Massachusetts
| |
Collapse
|
20
|
Grinfeld J, Nangalia J, Green AR. Molecular determinants of pathogenesis and clinical phenotype in myeloproliferative neoplasms. Haematologica 2017; 102:7-17. [PMID: 27909216 PMCID: PMC5210228 DOI: 10.3324/haematol.2014.113845] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022] Open
Abstract
The myeloproliferative neoplasms are a heterogeneous group of clonal disorders characterized by the overproduction of mature cells in the peripheral blood, together with an increased risk of thrombosis and progression to acute myeloid leukemia. The majority of patients with Philadelphia-chromosome negative myeloproliferative neoplasms harbor somatic mutations in Janus kinase 2, leading to constitutive activation. Acquired mutations in calreticulin or myeloproliferative leukemia virus oncogene are found in a significant number of patients with essential thrombocythemia or myelofibrosis, and mutations in numerous epigenetic regulators and spliceosome components are also seen. Although the cellular and molecular consequences of many of these mutations remain unclear, it seems likely that they interact with germline and microenvironmental factors to influence disease pathogenesis. This review will focus on the determinants of specific myeloproliferative neoplasm phenotypes as well as on how an improved understanding of molecular mechanisms can inform our understanding of the disease entities themselves.
Collapse
Affiliation(s)
- Jacob Grinfeld
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| | - Jyoti Nangalia
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| | - Anthony R Green
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
21
|
Bianchessi V, Lauri A, Vigorelli V, Toia M, Vinci MC. Evaluating the methylation status of CXCR4 promoter: A cost-effective and sensitive two-step PCR method. Anal Biochem 2016; 519:84-91. [PMID: 28007399 DOI: 10.1016/j.ab.2016.12.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/12/2016] [Accepted: 12/18/2016] [Indexed: 12/22/2022]
Abstract
The chemokine receptor CXCR4 plays a key role in the bone marrow microenvironment maintenance and in the hematopoietic stem and progenitor cells migration. In addition, CXCR4 is expressed in a broad spectrum of solid tumors where its methylation state has been recently proposed as a biomarker for cancer prognosis. To evaluate methylation status of CXCR4 promoter we developed a sensitive, accurate, specific and cost-effective two-step PCR method that does not require any specific equipment other than a conventional real-time PCR instrument. The principle of the technique relies on a novel normalization strategy which allows the detection and quantification of small methylation differences among pre-amplified DNA samples deriving from low amount of starting material. In addition, the analysis of melting curve profiles of PCR products provides additional information about the methylation status of CpG sites in between the primers. Finally, the principle of this technique can potentially be adapted for the investigation of the methylation status of any other DNA region.
Collapse
Affiliation(s)
| | - Andrea Lauri
- Axxam SpA, Molecular Biology Unit, via Meucci 3 - 20091 Bresso, Milano, Italy
| | | | - Martina Toia
- Centro Cardiologico Monzino, IRCCS, Milano, Italy
| | | |
Collapse
|
22
|
Olekson MP, Faulknor RA, Hsia HC, Schmidt AM, Berthiaume F. Soluble Receptor for Advanced Glycation End Products Improves Stromal Cell-Derived Factor-1 Activity in Model Diabetic Environments. Adv Wound Care (New Rochelle) 2016; 5:527-538. [PMID: 28078186 DOI: 10.1089/wound.2015.0674] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/06/2015] [Indexed: 12/30/2022] Open
Abstract
Objective: In diabetes, hyperglycemia causes the accumulation of advanced glycation end products (AGEs) that trigger reactive oxygen species (ROS) generation through binding the receptor for AGEs (RAGE). Because exogenous growth factors have had little success in enhancing chronic wound healing, we investigated whether hyperglycemia-induced AGEs interfere with cellular responses to extracellular signals. We used stromal cell-derived factor-1 (SDF-1), an angiogenic chemokine also known to promote stem cell recruitment in skin wounds. Approach: Human leukemia-60 (HL-60) cells and mouse peripheral blood mononuclear cells (PBMCs), which express the SDF-1 receptor CXCR-4, were incubated for 24 h in medium supplemented with 25 mM d-glucose. Soluble RAGE (sRAGE) was used to block RAGE activation. Response to SDF-1 was measured in cellular migration and ROS assays. A diabetic murine excisional wound model measured SDF-1 liposome and sRAGE activity in vivo. Results: Hyperglycemia led to significant accumulation of AGEs, decreased SDF-1-directed migration, and elevated baseline ROS levels; it suppressed the ROS spike normally triggered by SDF-1. sRAGE decreased the ROS baseline and restored both the SDF-1-mediated spike and cell migration. Topically applied sRAGE alone promoted healing and enhanced the effect of exogenous SDF-1 on diabetic murine wounds. Innovation: While there is interest in using growth factors to improve wound healing, this strategy is largely ineffective in diabetic wounds. We show that sRAGE may restore signaling, thus potentiating the effect of exogenously applied growth factors. Conclusion: Blocking RAGE with sRAGE restores SDF-1-mediated cellular responses in hyperglycemic environments and may potentiate the effectiveness of SDF-1 applied in vivo.
Collapse
Affiliation(s)
| | - Renea A. Faulknor
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Henry C. Hsia
- Department of Surgery, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Ann Marie Schmidt
- Departments of Medicine, Pharmacology, and Pathology, New York University Medical Center, New York
| | - François Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
23
|
Abstract
INTRODUCTION Primary myelofibrosis (PMF) is the least common but the most aggressive of the classic Philadelphia chromosome-negative myeloproliferative neoplasms. Survival is much shorter in PMF than in polycythemia vera (PV) or essential thrombocythemia (ET). Post-PV/ET myelofibrosis (MF) is clinically indistinguishable from PMF and approached similarly. Areas covered: Current pharmacologic therapy of MF revolves around the Janus kinase 1/2 (JAK1/2) inhibitor ruxolitinib, which dramatically improves constitutional symptoms and splenomegaly in the majority of patients, and improves overall survival (OS). However, allogeneic stem cell transplantation remains the only potential cure. Other JAK inhibitors continue to be developed for MF, and momelotinib and pacritinib are in phase III clinical trials. Anemia is common in MF, and initially worsened by ruxolitinib. Momelotinib and pacritinib may prove advantageous in this regard. Current strategies for managing anemia of MF include danazol, immunomodulatory drugs and erythroid stimulating agents, either alone or in combination with ruxolitinib. Expert opinion: A number of other agents, representing diverse drug classes, are in various stages of development for MF. These include newer JAK inhibitors, other signaling inhibitors, epigenetic modifiers, anti-fibrotic agents, telomerase inhibitors, and activin receptor ligand traps (for anemia). Hopefully, these novel therapies will further extend the clinical benefits of ruxolitinib.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Srdan Verstovsek
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
24
|
Bose P, Verstovsek S. Investigational histone deacetylase inhibitors (HDACi) in myeloproliferative neoplasms. Expert Opin Investig Drugs 2016; 25:1393-1403. [PMID: 27756180 DOI: 10.1080/13543784.2016.1250882] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The Philadelphia chromosome negative myeloproliferative neoplasms (MPN) mainly comprise polycythemia vera (PV), essential thrombocythemia (ET) and myelofibrosis (MF, primary or post-PV/ET). Therapy in PV and ET focuses on minimizing thrombosis and bleeding risk, while in MF, prolongation of survival is an important goal. Different cytoreductive agents are employed in high risk PV and ET, while the JAK inhibtior ruxolitinib is the cornerstone of therapy in MF. Histone deacetylase inhibitors (HDACi) are pleiotropic agents with diverse epigenetic and non-epigenetic actions, selectively in transformed cells. A number of HDACi have been or are being investigated in MPN. Areas covered: The mechanisms of action of HDACI in neoplastic cells are summarized, and the preclinical rationale and data supporting their development in MPN specifically examined, particularly their synergism with JAK inhibitors. Major findings of clinical trials of HDACi, both alone and in combination with ruxolitinib, in MPN are then discussed, with particular attention to their toxicities and disease-modifying effects. Expert opinion: HDACi are clearly active in MPN, and there is good preclinical rationale for this. Their combination with ruxolitinib in MF is promising, but the long-term tolerability of these agents is an important concern. Further development in PV or ET appears unlikely.
Collapse
Affiliation(s)
- Prithviraj Bose
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Srdan Verstovsek
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
25
|
Abdelouahab H, Zhang Y, Wittner M, Oishi S, Fujii N, Besancenot R, Plo I, Ribrag V, Solary E, Vainchenker W, Barosi G, Louache F. CXCL12/CXCR4 pathway is activated by oncogenic JAK2 in a PI3K-dependent manner. Oncotarget 2016; 8:54082-54095. [PMID: 28903325 PMCID: PMC5589564 DOI: 10.18632/oncotarget.10789] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/17/2016] [Indexed: 12/26/2022] Open
Abstract
JAK2 activation is the driver mechanism in BCR-ABL-negative myeloproliferative neoplasms (MPN). These diseases are characterized by an abnormal retention of hematopoietic stem cells within the bone marrow microenvironment and their increased trafficking to extramedullary sites. The CXCL12/CXCR4 axis plays a central role in hematopoietic stem cell/ progenitor trafficking and retention in hematopoietic sites. The present study explores the crosstalk between JAK2 and CXCL12/CXCR4 signaling pathways in MPN. We show that JAK2, activated by either MPL-W515L expression or cytokine stimulation, cooperates with CXCL12/CXCR4 signaling to increase the chemotactic response of human cell lines and primary CD34+ cells through an increased phosphatidylinositol-3-kinase (PI3K) signaling. Accordingly, primary myelofibrosis (MF) patient cells demonstrate an increased CXCL12-induced chemotaxis when compared to controls. JAK2 inhibition by knock down or chemical inhibitors decreases this effect in MPL-W515L expressing cell lines and reduces the CXCL12/CXCR4 signaling in some patient primary cells. Taken together, these data indicate that CXCL12/CXCR4 pathway is overactivated in MF patients by oncogenic JAK2 that maintains high PI3K signaling over the threshold required for CXCR4 activation. These results suggest that inhibition of this crosstalk may contribute to the therapeutic effects of JAK2 inhibitors.
Collapse
Affiliation(s)
- Hadjer Abdelouahab
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris Diderot, Paris, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Yanyan Zhang
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Monika Wittner
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Shinya Oishi
- Kyoto University, Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Nobutaka Fujii
- Kyoto University, Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Rodolphe Besancenot
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France.,Equipe labellisée Ligue Nationale contre le Cancer, UMR 1170, Institut Gustave Roussy, Villejuif, France.,Grex, Laboratoire d'Excellence, Paris, France
| | - Vincent Ribrag
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Eric Solary
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - William Vainchenker
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France.,Grex, Laboratoire d'Excellence, Paris, France
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Fawzia Louache
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris Diderot, Paris, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| |
Collapse
|
26
|
Moens L, Frans G, Bosch B, Bossuyt X, Verbinnen B, Poppe W, Boeckx N, Slatter M, Brusselmans C, Diaz G, Tousseyn T, Flipts H, Corveleyn A, Dierickx D, Meyts I. Successful hematopoietic stem cell transplantation for myelofibrosis in an adult with warts-hypogammaglobulinemia-immunodeficiency-myelokathexis syndrome. J Allergy Clin Immunol 2016; 138:1485-1489.e2. [PMID: 27484033 DOI: 10.1016/j.jaci.2016.04.057] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/04/2016] [Accepted: 04/29/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Leen Moens
- Department of Microbiology and Immunology, Experimental Laboratory Immunology, University Hospitals Leuven, Leuven, Belgium
| | - Glynis Frans
- Department of Microbiology and Immunology, Experimental Laboratory Immunology, University Hospitals Leuven, Leuven, Belgium
| | - Barbara Bosch
- Department of Pediatric Pulmonology, University Hospitals Leuven, Leuven, Belgium
| | - Xavier Bossuyt
- Department of Microbiology and Immunology, Experimental Laboratory Immunology, University Hospitals Leuven, Leuven, Belgium; Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Bert Verbinnen
- Department of Microbiology and Immunology, Experimental Laboratory Immunology, University Hospitals Leuven, Leuven, Belgium; Biomedical Laboratory Technology, Life Sciences & Chemistry, Thomas More Kempen, Geel, Belgium
| | - Willy Poppe
- Department of Obstetrics-Gynaecology, UZ Gasthuisberg Herestraat, Leuven, Belgium
| | - Nancy Boeckx
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Mary Slatter
- Department of Paediatric Immunology, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | | | - George Diaz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Thomas Tousseyn
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium; Translational Cell and Tissue Research, Catholic University Leuven, Leuven, Belgium
| | - Helena Flipts
- Department of Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Anniek Corveleyn
- Department of Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Daan Dierickx
- Laboratory for Experimental Hematology, KU Leuven, Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Isabelle Meyts
- Department of Microbiology and Immunology, Childhood Immunology, KU Leuven, Leuven, Belgium; Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
27
|
Leukocytosis and presence of CALR mutation is associated with non-hepatosplenic extramedullary hematopoiesis in primary myelofibrosis. Blood Cancer J 2016; 6:e436. [PMID: 27315113 PMCID: PMC5141359 DOI: 10.1038/bcj.2016.44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
28
|
The development and characterization of SDF1α-elastin-like-peptide nanoparticles for wound healing. J Control Release 2016; 232:238-47. [PMID: 27094603 DOI: 10.1016/j.jconrel.2016.04.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 12/18/2022]
Abstract
Chronic skin wounds are characterized by poor re-epithelialization, angiogenesis and granulation. Previous work has demonstrated that topical stromal cell-derived growth factor-1 (SDF1) promotes neovascularization, resulting in faster re-epithelialization of skin wounds in diabetic mice. However, the clinical usefulness of such bioactive peptides is limited because they are rapidly degraded in the wound environment due to high levels of proteases. Here, we describe the development of a recombinant fusion protein comprised of SDF1 and an elastin-like peptide that confers the ability to self-assemble into nanoparticles. The fusion protein and recombinant human SDF1 showed similar binding characteristics, as indicated by the measured equilibrium dissociation constant (Kd) for the binding of free SDF1 or the fusion protein to the CXCR4 receptor. The biological activity of SDF1-ELP, as measured by intracellular calcium release in HL60 cells was dose dependent, and also very similar to that of free SDF1. In contrast, the biological activity of SDF1-ELP in vivo was significantly superior to that of free SDF1. When applied to full thickness skin wounds in diabetic mice, wounds treated with SDF1-ELP nanoparticles were 95% closed by day 21, and fully closed by day 28, while wounds treated with free SDF1, ELP alone, or vehicle were only 80% closed by day 21, and took 42days to fully close. In addition, the SDF1-ELP nanoparticles significantly increased the epidermal and dermal layer of the healed wound, as compared to the other groups. These results indicate that SDF1-ELP fusion protein nanoparticles are promising agents for the treatment of chronic skin wounds.
Collapse
|
29
|
Yadav SS, Prasad SB, Prasad CB, Pandey LK, Pradhan S, Singh S, Narayan G. CXCL12 is a key regulator in tumor microenvironment of cervical cancer: an in vitro study. Clin Exp Metastasis 2016; 33:431-9. [PMID: 26970955 DOI: 10.1007/s10585-016-9787-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 02/25/2016] [Indexed: 12/21/2022]
Abstract
CXCL12 is a small pro-inflammatory chemo-attractant cytokine which signals through chemokine receptor CXCR4. The importance of CXCL12/CXCR4 axis is coming to the fore in several divergent signaling pathway-initiating signals related to cell survival and/or proliferation and cancer metastasis. In the present study we have investigated whether deregulation in CXCR4 signaling (as a consequence of deregulated expression of CXCL12) modulate the metastatic potential of cervical carcinoma cells. We demonstrate that CXCL12 is frequently down regulated and its promoter is hypermethylated in cervical cancer cell lines and primary tumor biopsies. Exogenous treatment of cervical cancer cell lines (HeLa, SiHa and C-33A) with recombinant CXCL12 inhibited the metastasis promoting cell migration, cell invasion and anchorage independent cell growth events. Although this study will need further in vivo validation, our observations suggest that (a) silencing of CXCL12 in cervical cancer cells may be critical in migration and invasion, the key events in cancer cell metastases; (b) cervical cancer cells having down regulated CXCL12 are more prone to being attracted to CXCL12 expressed at secondary sites of metastases; and (c) CXCL12 inhibits anchorage independent cell growth via anoikis. These findings suggest the tumor suppressor functions of CXCL12 in cervical cancer.
Collapse
Affiliation(s)
- Suresh Singh Yadav
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi, 221005, India
| | - Shyam Babu Prasad
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi, 221005, India
| | - Chandra Bhushan Prasad
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi, 221005, India
| | - Lakshmi Kant Pandey
- Department of Obstetrics and Gynecology, Banaras Hindu University, Varanasi, 221005, India
| | - Satyajit Pradhan
- Department of Radiotherapy & Radiation Medicine, Banaras Hindu University, Varanasi, 221005, India
| | - Sunita Singh
- Department of Zoology, MahilaMahavidyalaya, Banaras Hindu University, Varanasi, 221005, India.
| | - Gopeshwar Narayan
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
30
|
Agarwal A, Morrone K, Bartenstein M, Zhao ZJ, Verma A, Goel S. Bone marrow fibrosis in primary myelofibrosis: pathogenic mechanisms and the role of TGF-β. Stem Cell Investig 2016; 3:5. [PMID: 27358897 DOI: 10.3978/j.issn.2306-9759.2016.02.03] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/15/2016] [Indexed: 12/21/2022]
Abstract
Primary myelofibrosis (PMF) is a Philadelphia chromosome negative myeloproliferative neoplasm (MPN) with adverse prognosis and is associated with bone marrow fibrosis and extramedullary hematopoiesis. Even though the discovery of the Janus kinase 2 (JAK2), thrombopoietin receptor (MPL) and calreticulin (CALR) mutations have brought new insights into the complex pathogenesis of MPNs, the etiology of fibrosis is not well understood. Furthermore, since JAK2 inhibitors do not lead to reversal of fibrosis further understanding of the biology of fibrotic process is needed for future therapeutic discovery. Transforming growth factor beta (TGF-β) is implicated as an important cytokine in pathogenesis of bone marrow fibrosis. Various mouse models have been developed and have established the role of TGF-β in the pathogenesis of fibrosis. Understanding the molecular alterations that lead to TGF-β mediated effects on bone marrow microenvironment can uncover newer therapeutic targets against myelofibrosis. Inhibition of the TGF-β pathway in conjunction with other therapies might prove useful in the reversal of bone marrow fibrosis in PMF.
Collapse
Affiliation(s)
- Archana Agarwal
- 1 Steward Carney Hospital, 2100 Dorchester Avenue, Dorchester, MA, USA ; 2 Albert Einstein College of Medicine, Bronx, NY, USA ; 3 University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kerry Morrone
- 1 Steward Carney Hospital, 2100 Dorchester Avenue, Dorchester, MA, USA ; 2 Albert Einstein College of Medicine, Bronx, NY, USA ; 3 University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matthias Bartenstein
- 1 Steward Carney Hospital, 2100 Dorchester Avenue, Dorchester, MA, USA ; 2 Albert Einstein College of Medicine, Bronx, NY, USA ; 3 University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zhizhuang Joe Zhao
- 1 Steward Carney Hospital, 2100 Dorchester Avenue, Dorchester, MA, USA ; 2 Albert Einstein College of Medicine, Bronx, NY, USA ; 3 University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Amit Verma
- 1 Steward Carney Hospital, 2100 Dorchester Avenue, Dorchester, MA, USA ; 2 Albert Einstein College of Medicine, Bronx, NY, USA ; 3 University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Swati Goel
- 1 Steward Carney Hospital, 2100 Dorchester Avenue, Dorchester, MA, USA ; 2 Albert Einstein College of Medicine, Bronx, NY, USA ; 3 University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
31
|
Tedjaseputra A, Galli S, Ibrahim M, Harrison CN, McLornan DP. Histone deacetylase inhibitors in myeloproliferative neoplasms: current roles and future prospects. Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1149467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
32
|
Horton JD, Arbini AA, Perle MA, Raphael BG. Rapid and robust reversion to essential thrombocythemia on treatment with Decitabine in a case of hydroxyurea-induced t-MDS/AML. Clin Case Rep 2016; 4:46-50. [PMID: 26783435 PMCID: PMC4706408 DOI: 10.1002/ccr3.431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 09/15/2015] [Accepted: 10/01/2015] [Indexed: 01/06/2023] Open
Abstract
Rapid remission of MDS/AML may be induced with Decitabine; however, significant megakaryocyte expansion and subsequent thrombocytosis may occur. Decitabine‐mediated reversion of the MDS to benign ET via hypomethylation of JAK/STAT pathway repressors is one potential mechanism to explain this observed phenomenon.
Collapse
Affiliation(s)
- Joshua D Horton
- New York University School of Medicine 550 First Avenue New York New York 10010
| | - Arnaldo A Arbini
- Department of Pathology New York University Medical Center 530 First Avenue New York New York 10010
| | - Mary Ann Perle
- Department of Pathology New York University Medical Center 530 First Avenue New York New York 10010
| | - Bruce G Raphael
- Hematology Division Department of Medicine New York University Medical Center 550 First Avenue New York New York 10010
| |
Collapse
|
33
|
Hussein K, Stucki-Koch A, Alchalby H, Triviai I, Kröger N, Kreipe H. Cytokine Expression Pattern in Bone Marrow Microenvironment after Allogeneic Stem Cell Transplantation in Primary Myelofibrosis. Biol Blood Marrow Transplant 2015; 22:644-650. [PMID: 26708839 DOI: 10.1016/j.bbmt.2015.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/07/2015] [Indexed: 02/03/2023]
Abstract
The only curative therapy for primary myelofibrosis (PMF) is allogeneic stem cell transplantation (ASCT). However, although we know that patients can benefit from ASCT, we do not know the extent of the changes of the expression profile of cytokines and matrix modulation factors. In this first systematic analysis, we evaluated the expression profile of 103 factors before and after transplantation to identify potential biomarkers. The expression of fibrosis-, inflammation-, and angiogenesis-associated genes was analyzed in a total of 52 bone marrow biopsies: PMF patients (n = 14) before and after ASCT and, for control purposes, post-ASCT multiple myeloma patients (n = 14) and non-neoplastic hematopoiesis (n = 10). In post-ASCT PMF cases, decreased expression of tissue inhibitor of metalloproteinases (TIMP) and platelet-derived growth factor alpha (PDGFA) correlated with bone marrow remodeling and hematological remission. Expression of several other matrix factors remained at high levels and may contribute to post-ASCT remodeling. This is the first systematic analysis of cytokine expression in post-ASCT PMF bone marrow that shows that normalization of bone marrow microenvironment is paralleled by decreased expression of TIMP and PDGFA.
Collapse
Affiliation(s)
- Kais Hussein
- Institute of Pathology, Hannover Medical School, Hannover, Germany.
| | | | - Haefaa Alchalby
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ioanna Triviai
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans Kreipe
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
34
|
Inflammation as a Keystone of Bone Marrow Stroma Alterations in Primary Myelofibrosis. Mediators Inflamm 2015; 2015:415024. [PMID: 26640324 PMCID: PMC4660030 DOI: 10.1155/2015/415024] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/08/2015] [Accepted: 10/15/2015] [Indexed: 01/11/2023] Open
Abstract
Primary myelofibrosis (PMF) is a clonal myeloproliferative neoplasm where severity as well as treatment complexity is mainly attributed to a long lasting disease and presence of bone marrow stroma alterations as evidenced by myelofibrosis, neoangiogenesis, and osteosclerosis. While recent understanding of mutations role in hematopoietic cells provides an explanation for pathological myeloproliferation, functional involvement of stromal cells in the disease pathogenesis remains poorly understood. The current dogma is that stromal changes are secondary to the cytokine “storm” produced by the hematopoietic clone cells. However, despite therapies targeting the myeloproliferation-sustaining clones, PMF is still regarded as an incurable disease except for patients, who are successful recipients of allogeneic stem cell transplantation. Although the clinical benefits of these inhibitors have been correlated with a marked reduction in serum proinflammatory cytokines produced by the hematopoietic clones, further demonstrating the importance of inflammation in the pathological process, these treatments do not address the role of the altered bone marrow stroma in the pathological process. In this review, we propose hypotheses suggesting that the stroma is inflammatory-imprinted by clonal hematopoietic cells up to a point where it becomes “independent” of hematopoietic cell stimulation, resulting in an inflammatory vicious circle requiring combined stroma targeted therapies.
Collapse
|
35
|
Cytokine Regulation of Microenvironmental Cells in Myeloproliferative Neoplasms. Mediators Inflamm 2015; 2015:869242. [PMID: 26543328 PMCID: PMC4620237 DOI: 10.1155/2015/869242] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/13/2015] [Indexed: 12/13/2022] Open
Abstract
The term myeloproliferative neoplasms (MPN) refers to a heterogeneous group of diseases including not only polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), but also chronic myeloid leukemia (CML), and systemic mastocytosis (SM). Despite the clinical and biological differences between these diseases, common pathophysiological mechanisms have been identified in MPN. First, aberrant tyrosine kinase signaling due to somatic mutations in certain driver genes is common to these MPN. Second, alterations of the bone marrow microenvironment are found in all MPN types and have been implicated in the pathogenesis of the diseases. Finally, elevated levels of proinflammatory and microenvironment-regulating cytokines are commonly found in all MPN-variants. In this paper, we review the effects of MPN-related oncogenes on cytokine expression and release and describe common as well as distinct pathogenetic mechanisms underlying microenvironmental changes in various MPN. Furthermore, targeting of the microenvironment in MPN is discussed. Such novel therapies may enhance the efficacy and may overcome resistance to established tyrosine kinase inhibitor treatment in these patients. Nevertheless, additional basic studies on the complex interplay of neoplastic and stromal cells are required in order to optimize targeting strategies and to translate these concepts into clinical application.
Collapse
|
36
|
Tamari R, Mughal TI, Rondelli D, Hasserjian R, Gupta V, Odenike O, Fauble V, Finazzi G, Pane F, Mascarenhas J, Prchal J, Giralt S, Hoffman R. Allo-SCT for myelofibrosis: reversing the chronic phase in the JAK inhibitor era? Bone Marrow Transplant 2015; 50:628-36. [PMID: 25665047 PMCID: PMC6394215 DOI: 10.1038/bmt.2014.323] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/16/2014] [Accepted: 11/17/2014] [Indexed: 01/30/2023]
Abstract
At present, allo-SCT is the only curative treatment for patients with myelofibrosis (MF). Unfortunately, a significant proportion of candidate patients are considered transplant ineligible due to their poor general condition and advanced age at the time of diagnosis. The approval of the first JAK inhibitor, ruxolitinib, for patients with advanced MF in 2011 has had a qualified impact on the treatment algorithm. The drug affords substantial improvement in MF-associated symptoms and splenomegaly but no major effect on the natural history. There has, therefore, been considerable support for assessing the drug's candidacy in the peritransplant period. The drug's precise impact on clinical outcome following allo-SCT is currently not known; nor are the drug's long-term efficacy and safety known. Considering the rarity of MF and the small proportion of patients who undergo allo-SCT, well designed collaborative efforts are required. In order to address some of the principal challenges, an expert panel of laboratory and clinical experts in this field was established, and an independent workshop held during the 54th American Society of Hematology Annual Meeting in New Orleans, USA on 6 December 2013, and the European Hematology Association's Annual Meeting in Milan, Italy on 13 June 2014. This document summarizes the results of these efforts.
Collapse
Affiliation(s)
- R Tamari
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T I Mughal
- Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| | - D Rondelli
- University of Illinois, Chicago, IL, USA
| | | | - V Gupta
- Princess Margaret Cancer Center, Toronto, Canada
| | - O Odenike
- University of Chicago, Chicago, IL, USA
| | - V Fauble
- Mayo Clinic Cancer Center, Scottsdale, AZ, USA
| | - G Finazzi
- Papa Giovani XXIII Hospital and Research Center, Bergamo, Italy
| | - F Pane
- Federico II University, Naples, Italy
| | - J Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Prchal
- Huntsman Cancer Center, Salt Lake City, UT, USA
| | - S Giralt
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - R Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
37
|
Desterke C, Martinaud C, Guerton B, Pieri L, Bogani C, Clay D, Torossian F, Lataillade JJ, Hasselbach HC, Gisslinger H, Demory JL, Dupriez B, Boucheix C, Rubinstein E, Amsellem S, Vannucchi AM, Le Bousse-Kerdilès MC. Tetraspanin CD9 participates in dysmegakaryopoiesis and stromal interactions in primary myelofibrosis. Haematologica 2015; 100:757-67. [PMID: 25840601 DOI: 10.3324/haematol.2014.118497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 03/23/2015] [Indexed: 12/11/2022] Open
Abstract
Primary myelofibrosis is characterized by clonal myeloproliferation, dysmegakaryopoiesis, extramedullary hematopoiesis associated with myelofibrosis and altered stroma in the bone marrow and spleen. The expression of CD9, a tetraspanin known to participate in megakaryopoiesis, platelet formation, cell migration and interaction with stroma, is deregulated in patients with primary myelofibrosis and is correlated with stage of myelofibrosis. We investigated whether CD9 participates in the dysmegakaryopoiesis observed in patients and whether it is involved in the altered interplay between megakaryocytes and stromal cells. We found that CD9 expression was modulated during megakaryocyte differentiation in primary myelofibrosis and that cell surface CD9 engagement by antibody ligation improved the dysmegakaryopoiesis by restoring the balance of MAPK and PI3K signaling. When co-cultured on bone marrow mesenchymal stromal cells from patients, megakaryocytes from patients with primary myelofibrosis displayed modified behaviors in terms of adhesion, cell survival and proliferation as compared to megakaryocytes from healthy donors. These modifications were reversed after antibody ligation of cell surface CD9, suggesting the participation of CD9 in the abnormal interplay between primary myelofibrosis megakaryocytes and stroma. Furthermore, silencing of CD9 reduced CXCL12 and CXCR4 expression in primary myelofibrosis megakaryocytes as well as their CXCL12-dependent migration. Collectively, our results indicate that CD9 plays a role in the dysmegakaryopoiesis that occurs in primary myelofibrosis and affects interactions between megakaryocytes and bone marrow stromal cells. These results strengthen the "bad seed in bad soil" hypothesis that we have previously proposed, in which alterations of reciprocal interactions between hematopoietic and stromal cells participate in the pathogenesis of primary myelofibrosis.
Collapse
Affiliation(s)
- Christophe Desterke
- INSERM UMR-S1197, Paul Brousse Hospital, Paris-Sud University, Villejuif, France INSERM UMS33, Paul Brousse Hospital, Paris-Sud University, Villejuif, France
| | - Christophe Martinaud
- INSERM UMR-S1197, Paul Brousse Hospital, Paris-Sud University, Villejuif, France CTS of Army, Percy Hospital, Clamart, France
| | - Bernadette Guerton
- INSERM UMR-S1197, Paul Brousse Hospital, Paris-Sud University, Villejuif, France
| | - Lisa Pieri
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Costanza Bogani
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Denis Clay
- INSERM UMR-S1197, Paul Brousse Hospital, Paris-Sud University, Villejuif, France INSERM UMS33, Paul Brousse Hospital, Paris-Sud University, Villejuif, France
| | - Frederic Torossian
- INSERM UMR-S1197, Paul Brousse Hospital, Paris-Sud University, Villejuif, France INSERM UMS33, Paul Brousse Hospital, Paris-Sud University, Villejuif, France
| | - Jean-Jacques Lataillade
- INSERM UMR-S1197, Paul Brousse Hospital, Paris-Sud University, Villejuif, France Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Hans C Hasselbach
- Department of Hematology, Herlev University Hospital, Copenhagen, Denmark
| | - Heinz Gisslinger
- Department of Hematology, University Klinik Fur Innere Medizin, Vienna, Austria
| | - Jean-Loup Demory
- Université Catholique de Lille, France French Intergroup on Myeloproliferative Neoplasms (FIM), France
| | - Brigitte Dupriez
- French Intergroup on Myeloproliferative Neoplasms (FIM), France Department of Hematology, Dr Schaffner Hospital, Lens, France
| | - Claude Boucheix
- INSERM UMS33, Paul Brousse Hospital, Paris-Sud University, Villejuif, France Inserm U935, Paul Brousse Hospital, Paris-Sud University, Villejuif, France
| | - Eric Rubinstein
- INSERM UMS33, Paul Brousse Hospital, Paris-Sud University, Villejuif, France Inserm U935, Paul Brousse Hospital, Paris-Sud University, Villejuif, France
| | - Sophie Amsellem
- Department of Hematology, Gustave Roussy Institute, Villejuif, France
| | | | - Marie-Caroline Le Bousse-Kerdilès
- INSERM UMR-S1197, Paul Brousse Hospital, Paris-Sud University, Villejuif, France INSERM UMS33, Paul Brousse Hospital, Paris-Sud University, Villejuif, France French Intergroup on Myeloproliferative Neoplasms (FIM), France
| |
Collapse
|
38
|
Triviai I, Stübig T, Niebuhr B, Hussein K, Tsiftsoglou A, Fehse B, Stocking C, Kröger N. CD133 marks a stem cell population that drives human primary myelofibrosis. Haematologica 2015; 100:768-79. [PMID: 25724578 DOI: 10.3324/haematol.2014.118463] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 02/26/2015] [Indexed: 01/01/2023] Open
Abstract
Primary myelofibrosis is a myeloproliferative neoplasm characterized by bone marrow fibrosis, megakaryocyte atypia, extramedullary hematopoiesis, and transformation to acute myeloid leukemia. To date the stem cell that undergoes the spatial and temporal chain of events during the development of this disease has not been identified. Here we describe a CD133(+) stem cell population that drives the pathogenesis of primary myelofibrosis. Patient-derived circulating CD133(+) but not CD34(+)CD133(-) cells, with a variable burden for JAK2 (V617F) mutation, had multipotent cloning capacity in vitro. CD133(+) cells engrafted for up to 10 months in immunocompromised mice and differentiated into JAK2-V617F(+) myeloid but not lymphoid progenitors. We observed the persistence of human, atypical JAK2-V617F(+) megakaryocytes, the initiation of a prefibrotic state, bone marrow/splenic fibrosis and transition to acute myeloid leukemia. Leukemic cells arose from a subset of CD133(+) cells harboring EZH2 (D265H) but lacking a secondary JAK2 (V617F) mutation, consistent with the hypothesis that deregulation of EZH2 activity drives clonal growth and increases the risk of acute myeloid leukemia. This is the first characterization of a patient-derived stem cell population that drives disease resembling both chronic and acute phases of primary myelofibrosis in mice. These results reveal the importance of the CD133 antigen in deciphering the neoplastic clone in primary myelofibrosis and indicate a new therapeutic target for myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Ioanna Triviai
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Germany Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Greece
| | - Thomas Stübig
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Germany
| | - Birte Niebuhr
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Kais Hussein
- Institute of Pathology, Hannover Medical School, Germany
| | - Asterios Tsiftsoglou
- Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Greece
| | - Boris Fehse
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Germany
| | - Carol Stocking
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
39
|
Schulenburg A, Blatt K, Cerny-Reiterer S, Sadovnik I, Herrmann H, Marian B, Grunt TW, Zielinski CC, Valent P. Cancer stem cells in basic science and in translational oncology: can we translate into clinical application? J Hematol Oncol 2015; 8:16. [PMID: 25886184 PMCID: PMC4345016 DOI: 10.1186/s13045-015-0113-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/14/2015] [Indexed: 02/08/2023] Open
Abstract
Since their description and identification in leukemias and solid tumors, cancer stem cells (CSC) have been the subject of intensive research in translational oncology. Indeed, recent advances have led to the identification of CSC markers, CSC targets, and the preclinical and clinical evaluation of the CSC-eradicating (curative) potential of various drugs. However, although diverse CSC markers and targets have been identified, several questions remain, such as the origin and evolution of CSC, mechanisms underlying resistance of CSC against various targeted drugs, and the biochemical basis and function of stroma cell-CSC interactions in the so-called ‘stem cell niche.’ Additional aspects that have to be taken into account when considering CSC elimination as primary treatment-goal are the genomic plasticity and extensive subclone formation of CSC. Notably, various cell fractions with different combinations of molecular aberrations and varying proliferative potential may display CSC function in a given neoplasm, and the related molecular complexity of the genome in CSC subsets is considered to contribute essentially to disease evolution and acquired drug resistance. In the current article, we discuss new developments in the field of CSC research and whether these new concepts can be exploited in clinical practice in the future.
Collapse
Affiliation(s)
- Axel Schulenburg
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, A-1090, Wien, Austria. .,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Stem Cell Transplantation Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Wien, Austria.
| | - Katharina Blatt
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Sabine Cerny-Reiterer
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Irina Sadovnik
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Harald Herrmann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Radiation Therapy, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria.
| | - Brigitte Marian
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Institute for Cancer Research, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Thomas W Grunt
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Clinical Oncology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Christoph C Zielinski
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Clinical Oncology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Peter Valent
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| |
Collapse
|
40
|
Wang X, Cho SY, Hu CS, Chen D, Roboz J, Hoffman R. C-X-C motif chemokine 12 influences the development of extramedullary hematopoiesis in the spleens of myelofibrosis patients. Exp Hematol 2014; 43:100-9.e1. [PMID: 25461253 DOI: 10.1016/j.exphem.2014.10.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/19/2014] [Indexed: 12/11/2022]
Abstract
Myelofibrosis (MF) is characterized by the constitutive mobilization of hematopoietic stem cells (HSC) and hematopoietic progenitor cells (HPC) and the establishment of extramedullary hematopoiesis. The mechanisms underlying this abnormal HSC/HPC trafficking pattern remain poorly understood. We demonstrated that both splenic and peripheral blood (PB) MF CD34(+) cells equally share a defective ability to home to the marrow, but not to the spleens, of NOD/LtSz-Prkdc(scid) mice. This trafficking pattern could not be attributed to discordant expression of integrins or chemokine receptors other than the downregulation of C-X-C chemokine receptor type 4 by both PB and splenic MF CD34(+) cells. The number of both splenic MF CD34(+) cells and HPCs that migrated toward splenic MF plasma was, however, significantly greater than the number that migrated toward PB MF plasma. The concentration of the intact HSC/HPC chemoattractant C-X-C motif chemokine 12 (CXCL12) was greater in splenic MF plasma than PB MF plasma, as quantified using mass spectrometry. Functionally inactive truncated products of CXCL12, which are the product of proteolytic degradation by serine proteases, were detected at similar levels in both splenic and PB MF plasma. Treatment with an anti-CXCL12 neutralizing antibody resulted in a reduction in the degree of migration of splenic MF CD34(+) cells toward both PB and splenic MF plasma, validating the role of CXCL12 as a functional chemoattractant. Our data indicate that the MF splenic microenvironment is characterized by increased levels of intact, functional CXCL12, which contributes to the localization of MF CD34(+) cells to the spleen and the establishment of extramedullary hematopoiesis.
Collapse
Affiliation(s)
- Xiaoli Wang
- Division of Hematology/Oncology, The Tisch Cancer Institute, Department of Medicine, Myeloproliferative Disorders Research Consortium, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sool Yeon Cho
- Division of Hematology/Oncology, The Tisch Cancer Institute, Department of Medicine, Myeloproliferative Disorders Research Consortium, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cing Siang Hu
- Division of Hematology/Oncology, The Tisch Cancer Institute, Department of Medicine, Myeloproliferative Disorders Research Consortium, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Chen
- Division of Hematology/Oncology, The Tisch Cancer Institute, Department of Medicine, Myeloproliferative Disorders Research Consortium, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Roboz
- Division of Hematology/Oncology, The Tisch Cancer Institute, Department of Medicine, Myeloproliferative Disorders Research Consortium, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Hoffman
- Division of Hematology/Oncology, The Tisch Cancer Institute, Department of Medicine, Myeloproliferative Disorders Research Consortium, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
41
|
Wang J, Xu J, Gale RP, Xu Z, Li B, Qin T, Zhang Y, Fang L, Zhang H, Pan L, Qu S, Zhang P, Xiao Z. Prognostic impact of splenomegaly on survival of Chinese with primary myelofibrosis. Leuk Res 2014; 38:1207-11. [PMID: 25182689 DOI: 10.1016/j.leukres.2014.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/16/2014] [Accepted: 08/13/2014] [Indexed: 11/25/2022]
Abstract
Predicting survival in persons with primary myelofibrosis (PMF) is typically based on the International Prognostic Scoring System (IPSS), the Dynamic IPSS (DIPSS) or the DIPSS-Plus. These scoring systems use clinical and laboratory data developed predominately in persons of European descent. Splenomegaly is not a prognostic variable in any of these scoring systems. Recently, we reported differences in clinical and laboratory features between Chinese vs. persons of European descent with PMF. Based on this we developed a modified prognostic model to predict survival of Chinese subjects in which splenomegaly is an independent favorable prognostic factor. In the current study, we analyzed data from 874 Chinese with PMF including 495 with splenomegaly. Subjects with splenomegaly had significantly higher hemoglobin concentrations (P<0.001), higher levels of WBCs (P<0.001), platelets (P<0.001), excess blood blasts (≥ 1%; P=0.012), less RBC-transfusion-dependence (P<0.001) and lower DIPSS risk distribution (P=0.024). Frequency of JAK2(V617F) (62% vs. 50%; P=0.003) was also different. In univariate analyses subjects without splenomegaly had briefer survival (median, 64 mo [95% CI, 43-85] vs. 110 mo [95% CI, 67-153]; P<0.001). In multivariate analyses, splenomegaly was a favorable prognostic correlate of survival independent of DIPSS risk-cohort (hazard ratio [HR]=1.445; [95% CI, 1.101-1.895]; P=0.008). Our data suggest including splenomegaly improves the predictive accuracy of the prognostic model to estimate survival of Chinese with PMF.
Collapse
Affiliation(s)
- Jingya Wang
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Junqing Xu
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Robert Peter Gale
- Haematology Research Center, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Zefeng Xu
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Bing Li
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Tiejun Qin
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yue Zhang
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Liwei Fang
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Hongli Zhang
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Lijuan Pan
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Shiqiang Qu
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Peihong Zhang
- Department of Pathology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zhijian Xiao
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| |
Collapse
|
42
|
Mascarenhas J. Rationale for combination therapy in myelofibrosis. Best Pract Res Clin Haematol 2014; 27:197-208. [PMID: 25189730 DOI: 10.1016/j.beha.2014.07.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 07/11/2014] [Indexed: 12/18/2022]
Abstract
Agents targeting the JAK-STAT pathway have dominated the investigational therapeutic portfolio over the last five years resulting in the first and only approved agent for the treatment of patients with myelofibrosis (MF). However, chromatin modifying agents, anti-fibrosing agents, and other signaling pathway inhibitors have also demonstrated activity and offer the potential to improve upon the clinical success of JAK2 inhibition. Due to the complex pathobiological mechanisms underlying MF, it is likely that a combination of biologically active therapies will be required to target the MF hematopoietic stem cell in order to achieve significant disease course modification. Ruxolitinib in partnership with panobinostat, decitabine, and LDE225 are being evaluated in current combination therapy trials based on pre-clinical studies that provide strong scientific rationale. The rationale of combination of danazol or lenalidomide with ruxolitinib is mainly based on mitigation of anti-JAK2-mediated myelosuppression. Combination trials of ruxolitinib and novel anti-fibrosing agents such as PRM-151 represent an attempt to address therapeutic limitations of JAK2 inhibitors such as reversal of bone marrow fibrosis. Ruxolitinib is also being incorporated in novel treatment strategies in the setting of hematopoietic stem cell transplantation for MF. As the pathogenetic mechanisms are better understood, potential drug combinations in MF will increase dramatically and demonstration of biologic activity in effective preclinical models will be required to efficiently evaluate the most active combinations with least toxicity in future trials. This manuscript will address the proposed goals of combination therapy approach and review the state of the art in combination experimental therapy for MF.
Collapse
Affiliation(s)
- John Mascarenhas
- Myeloproliferative Disorder Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY, USA.
| |
Collapse
|
43
|
Epigenetic silencing of CXCR4 promotes loss of cell adhesion in cervical cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:581403. [PMID: 25114911 PMCID: PMC4119908 DOI: 10.1155/2014/581403] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/31/2014] [Accepted: 05/31/2014] [Indexed: 12/15/2022]
Abstract
In the network of chemokine signaling pathways, recent reports have described the SDF-1α/CXCR4 axis and its role in cancer progression and metastasis. Interestingly, we found downregulation of CXCR4 at both transcript and protein level in cervical cancer cell lines and primary tumors. We also found CXCR4 promoter hypermethylation in cervical cancer cell lines and primary biopsy samples. DNA hypomethylating drug 5-AZA-2′-deoxycytidine and histone deacetylase inhibitor Trichostatin A treatments in cell lines reactivate both CXCR4 transcription and protein expression. Cell adhesion assay demonstrated that autocrine SDF-1α promotes the loss of cell adhesion while paracrine SDF-1α predominantly protects the normal cervical cells from loss of cell adhesion. Cervical cancer cell line C-33A having increased expression of CXCR4 after TSA treatment showed increased cell adhesion by paracrine source of SDF-1α in comparison to untreated C-33A. These findings demonstrate the first evidence that epigenetic silencing of CXCR4 makes the cells inefficient to respond to the paracrine source of SDF-1α leading to loss of cell adhesion, one of the key events in metastases and progression of the disease. Our results provide novel insight of SDF-1α/CXCR4 signaling in tumor microenvironment which may be promising to further delineate molecular mechanism of cervical carcinogenesis.
Collapse
|
44
|
Bartalucci N, Bogani C, Vannucchi AM. Preclinical models for drug selection in myeloproliferative neoplasms. Curr Hematol Malig Rep 2014; 8:317-24. [PMID: 24146202 DOI: 10.1007/s11899-013-0182-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The discovery that an abnormally activated JAK-STAT signaling pathway is central to the pathogenesis of myeloproliferative neoplasms has promoted the clinical development of small-molecule JAK2 inhibitors. These agents have shown remarkable efficacy in disease control, but do not induce molecular remission; on the other hand, interferon holds the promise to target the putative hematopoietic progenitor cell initiating the disease. The presence of additional molecular abnormalities indicates a high molecular complexity of myeloproliferative neoplasms, and the need for simultaneously targeting different targets. Several drugs are currently under study as single agents and in combination. This review briefly describes the several in vitro and in vivo models of myeloproliferative neoplasms that are being used as preclinical models for drug development.
Collapse
Affiliation(s)
- Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | | |
Collapse
|
45
|
Abstract
Besides 5-azacytidine (azacitidine, Vidaza®), 5-aza-2'-deoxycytidine (decitabine, Dacogen®) is the most widely used inhibitor of DNA methylation, which triggers demethylation leading to consecutive reactivation of epigenetically silenced tumor suppressor genes in vitro and in vivo. Although antileukemic activity of decitabine is known for almost 40 years, its therapeutic potential in hematologic malignancies has only recently led to its approval in higher-risk MDS patients and as first-line treatment in AML patients>65 years who are not candidates for intensive chemotherapy. Several clinical trials showed promising activity of low-dose decitabine also in CML and hemoglobinopathies, whereas its efficacy in solid tumors is very limited. Clinical responses appear to be exerted both by epigenetic alterations and by induction of cell-cycle arrest and/or apoptosis. Recent and ongoing clinical trials investigate new dosing schedules, routes of administration, and combination of decitabine with other agents, including histone deacetylase inhibitors.
Collapse
|
46
|
Abstract
Abstract
Myelofibrosis (MF), including primary MF, postpolycythemia vera MF, and postessential thrombocythemia MF, is a clonal stem cell disorder characterized by BM fibrosis, extramedullary hematopoiesis, and a variable propensity to transform into acute leukemia. Allogeneic stem cell transplantation is the only known cure for MF, but its applicability is limited by the advanced age of most patients and by comorbid conditions. In the past decade, there has been an explosion of information on the molecular-genetic features associated with these diseases, fueled recently by the discovery of the JAK2V617F mutation. The development of JAK inhibitors has represented a significant therapeutic advance for these diseases; however, their use in MF has not yet been associated with eradication or a significant suppression of the malignant clone. In this era, much remains to be understood about MF, but it is likely that the identification of key pathogenetic drivers of the disease, coupled with the availability of novel molecularly targeted agents, will result in the discovery of new agents that significantly alter the natural history of the disease. This review focuses on recent and ongoing efforts in the development of novel agents in MF that go beyond the field of JAK inhibitors.
Collapse
|
47
|
Mascarenhas JO, Orazi A, Bhalla KN, Champlin RE, Harrison C, Hoffman R. Advances in myelofibrosis: a clinical case approach. Haematologica 2013; 98:1499-509. [PMID: 24091929 PMCID: PMC3789453 DOI: 10.3324/haematol.2013.086348] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 07/02/2013] [Indexed: 12/11/2022] Open
Abstract
Primary myelofibrosis is a member of the myeloproliferative neoplasms, a diverse group of bone marrow malignancies. Symptoms of myelofibrosis, particularly those associated with splenomegaly (abdominal distention and pain, early satiety, dyspnea, and diarrhea) and constitutional symptoms, represent a substantial burden to patients. Most patients eventually die from the disease, with a median survival ranging from approximately 5-7 years. Mutations in Janus kinase 2 (JAK2), a kinase that is essential for the normal development of erythrocytes, granulocytes, and platelets, notably the V617F mutation, have been identified in approximately 50% of patients with myelofibrosis. The approval of a JAK2 inhibitor in 2011 has improved the outlook of many patients with myelofibrosis and has changed the treatment landscape. This article focuses on some of the important issues in current myelofibrosis treatment management, including differentiation of myelofibrosis from essential thrombocythemia and polycythemia vera, up-dated data on the results of JAK2 inhibitor therapy, the role of epigenetic mechanisms in myelofibrosis pathogenesis, investigational therapies for myelofibrosis, and advances in hematopoietic stem cell transplant. Three myelofibrosis cases are included to underscore the issues in diagnosing and treating this complex disease.
Collapse
|
48
|
Characterization of the TGF-β1 signaling abnormalities in the Gata1low mouse model of myelofibrosis. Blood 2013; 121:3345-63. [PMID: 23462118 DOI: 10.1182/blood-2012-06-439661] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Primary myelofibrosis (PMF) is characterized by fibrosis, ineffective hematopoiesis in marrow, and hematopoiesis in extramedullary sites and is associated with abnormal megakaryocyte (MK) development and increased transforming growth factor (TGF)-β1 release. To clarify the role of TGF-β1 in the pathogenesis of this disease, the TGF-β1 signaling pathway of marrow and spleen of the Gata1(low) mouse model of myelofibrosis (MF) was profiled and the consequences of inhibition of TGF-β1 signaling on disease manifestations determined. The expression of 20 genes in marrow and 36 genes in spleen of Gata1(low) mice was altered. David-pathway analyses identified alterations of TGF-β1, Hedgehog, and p53 signaling in marrow and spleen and of mammalian target of rapamycin (mTOR) in spleen only and predicted that these alterations would induce consequences consistent with the Gata1(low) phenotype (increased apoptosis and G1 arrest both in marrow and spleen and increased osteoblast differentiation and reduced ubiquitin-mediated proteolysis in marrow only). Inhibition of TGF-β1 signaling normalized the expression of p53-related genes, restoring hematopoiesis and MK development and reducing fibrosis, neovascularization, and osteogenesis in marrow. It also normalized p53/mTOR/Hedgehog-related genes in spleen, reducing extramedullary hematopoiesis. These data identify altered expression signatures of TGF-β1 signaling that may be responsible for MF in Gata1(low) mice and may represent additional targets for therapeutic intervention in PMF.
Collapse
|
49
|
Nischal S, Bhattacharyya S, Christopeit M, Yu Y, Zhou L, Bhagat T, Sohal D, Will B, Mo Y, Suzuki M, Pardanani A, McDevitt M, Maciejewski JP, Melnick AM, Greally J, Steidl U, Moliterno A, Verma A. Methylome profiling reveals distinct alterations in phenotypic and mutational subgroups of myeloproliferative neoplasms. Cancer Res 2013; 73:1076-85. [PMID: 23066032 PMCID: PMC5500294 DOI: 10.1158/0008-5472.can-12-0735] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Even though mutations in epigenetic regulators frequently occur in myeloproliferative neoplasms, their effects on the epigenome have not been well studied. Furthermore, even though primary myelofibrosis (PMF) has a markedly worse prognosis than essential thrombocytosis or polycythemia vera, the molecular distinctions between these subgroups are not well elucidated. We conducted the HELP (HpaII tiny fragment enriched by LM-PCR) assay to study genome-wide methylation in polycythemia vera, essential thrombocytosis, and PMF samples compared with healthy controls. We determined that polycythemia vera and essential thrombocytosis are characterized by aberrant promoter hypermethylation, whereas PMF is an epigenetically distinct subgroup characterized by both aberrant hyper- and hypomethylation. Aberrant hypomethylation in PMF was seen to occur in non-CpG island loci, showing further qualitative differences between the disease subgroups. The differentially methylated genes in polycythemia vera and essential thrombocytosis were involved predominantly in cell signaling pathways and were enriched for binding sites of GATA1 and other transcription factors. In contrast, aberrantly methylated genes in PMF were involved in inflammatory pathways and were enriched for NF1, LEF1, and other transcription factors. Within the PMF subgroup, cases with ASXL1 disruptions formed an epigenetically distinct subgroup with relatively increased methylation. Cases of myeloproliferative neoplasms (MPN) with TET2 mutations showed decreased levels of hydroxymethylation and distinct set of hypermethylated genes. In contrast, the JAK2V617F mutation did not drive epigenetic clustering within MPNs. Finally, the significance of aberrant methylation was shown by sensitivity of MPN-derived cell lines to decitabine. These results show epigenetic differences between PMF and polycythemia vera/essential thrombocytosis and reveal methylomic signatures of ASXL1 and TET2 mutations.
Collapse
Affiliation(s)
| | | | | | - Yiting Yu
- Albert Einstein College of Medicine, Bronx, NY
| | - Li Zhou
- Albert Einstein College of Medicine, Bronx, NY
| | | | | | - Britta Will
- Albert Einstein College of Medicine, Bronx, NY
| | - Yongkai Mo
- Albert Einstein College of Medicine, Bronx, NY
| | | | | | | | | | | | | | | | | | - Amit Verma
- Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
50
|
Li H, Niederkorn JY, Sadegh L, Mellon J, Chen PW. Epigenetic regulation of CXCR4 expression by the ocular microenvironment. Invest Ophthalmol Vis Sci 2013; 54:234-43. [PMID: 23188729 PMCID: PMC3544529 DOI: 10.1167/iovs.12-10643] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 11/12/2012] [Accepted: 11/17/2012] [Indexed: 01/25/2023] Open
Abstract
PURPOSE Expression of the chemokine receptor CXCR4 by tumors is associated with metastatic migration and invasion of tumor cells. The importance of CXCR4 expression by uveal melanomas in metastasis to the liver was recently demonstrated when injection of CXCR4-negative uveal melanoma cells into mice resulted in reduced liver metastasis compared with CXCR4-positive uveal melanoma cells. Factors in the eye can induce downregulation of genes by epigenetic mechanisms. This study examined whether epigenetic regulation by the ocular environment induced downregulation of CXCR4 expression. METHODS LS174T colon cancer cells were injected in the anterior chamber (AC), subcutaneously (SC), or in the spleen capsule to induce liver metastasis in immune-deficient mice. CXCR4 gene transcription was analyzed by RT-PCR, and protein expression was determined by flow cytometry. Methyltransferase and histone deacetylase activities were determined by ELISA. Treatment with either 5-Aza-2-deoxycytidine (5-Aza) or trichostatin A (TSA) was used to induce demethylation or inhibit histone deacetylases, respectively. RESULTS AC-derived LS174T cells showed lower CXCR4 gene expression compared with SC-, liver-derived, or wild-type tumor cells. AC-derived LS174T tumor cells expressed methyltransferase activity compared with SC-, liver-derived, and wild-type tumor cells. Deacetylase activity was elevated in AC-derived LS174T tumor cells compared with SC-derived, liver-derived, and wild-type tumor cells. Treatment of AC-derived LS174T tumor cells with 5-Aza upregulated CXCR4 expression. TSA treatment did not restore CXCR4 expression. CONCLUSIONS These studies demonstrate that ocular microenvironment factors induce methylation and downregulation of tumor CXCR4 expression.
Collapse
MESH Headings
- Animals
- Anterior Chamber/metabolism
- Anterior Chamber/pathology
- Antimetabolites, Antineoplastic/pharmacology
- Azacitidine/analogs & derivatives
- Azacitidine/pharmacology
- Cecum/metabolism
- Cecum/pathology
- Cell Line, Tumor
- Decitabine
- Down-Regulation
- Epigenesis, Genetic
- Flow Cytometry
- Gene Expression
- Gene Expression Regulation, Neoplastic/physiology
- Histone Deacetylase Inhibitors/administration & dosage
- Histone Deacetylases/metabolism
- Histones/metabolism
- Hydroxamic Acids/administration & dosage
- Injections
- Injections, Subcutaneous
- Lysine/metabolism
- Methylation
- Mice
- Mice, Inbred BALB C
- Neoplasm Transplantation
- Promoter Regions, Genetic
- Receptors, CXCR4/drug effects
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Spleen/metabolism
- Spleen/pathology
- Up-Regulation
Collapse
Affiliation(s)
- Haochuan Li
- Department of Ophthalmology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-9057, USA
| | | | | | | | | |
Collapse
|