1
|
Brunmeir R, Ying L, Yan J, Hee YT, Lin B, Kaur H, Leong QZ, Teo WW, Choong G, Jen WY, Koh LP, Tan LK, Chan E, Ooi M, Yang H, Chng WJ. EZH2 modulates mRNA splicing and exerts part of its oncogenic function through repression of splicing factors in CML. Leukemia 2025; 39:650-662. [PMID: 39774797 PMCID: PMC11879851 DOI: 10.1038/s41375-024-02509-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 12/01/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
The polycomb protein EZH2 is up-regulated in Chronic Myeloid Leukaemia (CML) and associated with transcriptional reprogramming. Here we tested whether EZH2 might also act as a modulator of the mRNA splicing landscape to elicit its oncogenic function in CML. We treated CML cell lines with EZH2 inhibitors and detected differential splicing of several hundreds of events, potentially caused by the transcriptional regulation of splicing factors. Amongst those genes, CELF2 was identified as a candidate to mediate part of the EZH2 inhibitor induced phenotype. Upon over-expression, we observed (1) reduced cell growth, viability, and colony formation of CML cell lines, (2) a change in the splicing landscape, partially overlapping with EZH2 mediated changes, (3) the down-regulation of MYC signalling. Importantly, these findings were successfully validated in a cohort of CML patient samples, confirming the role of CELF2 as EZH2-regulated tumour-suppressor, contributing to the severe splicing de-regulation present in CML. Based on this we propose that EZH2 exerts part of its oncogenic function in CML through the transcriptional repression of splicing factors. Finally, analysis of publicly available datasets suggests that splicing modulation by EZH2 might not be restricted to CML.
Collapse
MESH Headings
- Humans
- Enhancer of Zeste Homolog 2 Protein/genetics
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- RNA Splicing/genetics
- RNA Splicing Factors/genetics
- RNA Splicing Factors/metabolism
- RNA, Messenger/genetics
- Gene Expression Regulation, Leukemic
- Cell Proliferation
- Cell Line, Tumor
- CELF Proteins
- Nerve Tissue Proteins
Collapse
Affiliation(s)
- Reinhard Brunmeir
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Li Ying
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Junli Yan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Yan Ting Hee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Baohong Lin
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Harvinder Kaur
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Qiao Zheng Leong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wei Wen Teo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Gerald Choong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wei-Ying Jen
- Division of Haematology, Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Liang Piu Koh
- Division of Haematology, Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Lip Kun Tan
- Division of Haematology, Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Esther Chan
- Division of Haematology, Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Melissa Ooi
- Division of Haematology, Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
2
|
Jia MX, Di DL, Liu ZZ, Wang HY, Chen L. Successful treatment of a chronic myeloid leukemia patient with extreme thrombocytosis by a combination of imatinib and interferon‑α: A case report. Exp Ther Med 2025; 29:50. [PMID: 39885917 PMCID: PMC11775754 DOI: 10.3892/etm.2025.12800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
Chronic myeloid leukemia with extreme thrombocytosis (CML-T), defined by a platelet count >1,000x109/l is a rare leukemia subtype. The present case report described a 66-year-old female CML-T patient presenting with a platelet count of 3,798x109/l, but a consistently normal spleen size. Following treatment with imatinib combined with interferon-α, the patient achieved hematological remission within 2 months, with a platelet count reduction to 311x109/l and complete cytogenetic remission after 10 months. The patient experienced myocardial infarction and liver injury during treatment, which was managed with supportive care. The present case suggested that imatinib combined with interferon-α may be a safe and effective treatment option for patients with CML-T and extreme thrombocytosis and suboptimal response to imatinib monotherapy.
Collapse
Affiliation(s)
- Meng-Xiao Jia
- Department of Hematology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, P.R. China
| | - Da-Lin Di
- Department of Immunology, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhen-Zhen Liu
- Department of Hematology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, P.R. China
| | - Hai-Ying Wang
- Department of Hematology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, P.R. China
| | - Lei Chen
- Department of Hematology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, P.R. China
| |
Collapse
|
3
|
Stagno F, Stella S, Leotta S, Caserta S, Vitale SR, Markovic U, Tomarchio C, Mirabile G, Russo S, Massimino M, Vigneri P, Manzella L, Di Raimondo F, Allegra A. No Benefit from Hydroxyurea Pre-Treatment in Frontline Chronic Myeloid Leukemia Therapy and Evidence of Quantitative Changes in the BCR::ABL1 Transcript Level. Int J Mol Sci 2025; 26:1840. [PMID: 40076467 PMCID: PMC11899297 DOI: 10.3390/ijms26051840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Hydroxyurea (HU) cytoreduction is usually administered to patients with chronic myeloid leukemia before starting any tyrosine kinase inhibitors (TKIs) therapy. However, up to date, there is no evidence of any benefit of hydroxyurea pre-treatment. Conversely, evidence has been provided on both the prognostic significance of the quantitative assessment of BCR::ABL1 expression at diagnosis and the individual decline of the BCR::ABL1 slope. In this view, we assumed that any kind of treatment administered before a confirmed diagnosis of chronic myeloid leukemia might change the amount of BCR::ABL1 transcript levels. To this purpose, we evaluated leukocyte counts and BCR::ABL1 quantitative expression either at diagnosis (baseline and no therapy) and on day 7 and day 14 of treatment in a cohort of 45 unselected patients with newly diagnosed chronic myeloid leukemia in the chronic phase. After informed consent, 21 of them received HU cytoreduction for 14 days before starting TKI treatment (HU group), whereas the other 24 patients received frontline TKI therapy without HU pre-treatment (TKI group). Our findings showed that: (i) there is no benefit from HU cytoreduction in patients affected with chronic myeloid leukemia before starting treatment with TKIs; (ii) any kind of therapy administered before a confirmed diagnosis of chronic myeloid leukemia might change the amount of BCR::ABL1 expression levels.
Collapse
Affiliation(s)
- Fabio Stagno
- Hematology Section, A.O.U. Policlinico “G. Martino”, University of Messina, 98100 Messina, Italy; (S.C.); (S.R.); (A.A.)
| | - Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (S.S.); (S.R.V.); (C.T.); (P.V.); (L.M.)
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “Rodolico-San Marco”, 95123 Catania, Italy;
| | - Salvatore Leotta
- Hematology and BMT Unit, A.O.U. Policlinico “Rodolico-San Marco”, University of Catania, 95123 Catania, Italy; (S.L.); (U.M.); (F.D.R.)
| | - Santino Caserta
- Hematology Section, A.O.U. Policlinico “G. Martino”, University of Messina, 98100 Messina, Italy; (S.C.); (S.R.); (A.A.)
| | - Silvia Rita Vitale
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (S.S.); (S.R.V.); (C.T.); (P.V.); (L.M.)
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “Rodolico-San Marco”, 95123 Catania, Italy;
| | - Uros Markovic
- Hematology and BMT Unit, A.O.U. Policlinico “Rodolico-San Marco”, University of Catania, 95123 Catania, Italy; (S.L.); (U.M.); (F.D.R.)
| | - Cristina Tomarchio
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (S.S.); (S.R.V.); (C.T.); (P.V.); (L.M.)
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “Rodolico-San Marco”, 95123 Catania, Italy;
| | - Giuseppe Mirabile
- Hematology Section, A.O.U. Policlinico “G. Martino”, University of Messina, 98100 Messina, Italy; (S.C.); (S.R.); (A.A.)
| | - Sabina Russo
- Hematology Section, A.O.U. Policlinico “G. Martino”, University of Messina, 98100 Messina, Italy; (S.C.); (S.R.); (A.A.)
| | - Michele Massimino
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “Rodolico-San Marco”, 95123 Catania, Italy;
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (S.S.); (S.R.V.); (C.T.); (P.V.); (L.M.)
- University Oncology Department, Humanitas Istituto Clinico Catanese, 95045 Catania, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (S.S.); (S.R.V.); (C.T.); (P.V.); (L.M.)
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “Rodolico-San Marco”, 95123 Catania, Italy;
| | - Francesco Di Raimondo
- Hematology and BMT Unit, A.O.U. Policlinico “Rodolico-San Marco”, University of Catania, 95123 Catania, Italy; (S.L.); (U.M.); (F.D.R.)
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - Alessandro Allegra
- Hematology Section, A.O.U. Policlinico “G. Martino”, University of Messina, 98100 Messina, Italy; (S.C.); (S.R.); (A.A.)
| |
Collapse
|
4
|
Rea D, Fodil S, Lengline E, Raffoux E, Cayuela JM. Tyrosine Kinase Inhibitor Discontinuation in Chronic Myeloid Leukemia: Strategies to Optimize Success and New Directions. Curr Hematol Malig Rep 2024; 19:104-110. [PMID: 38393431 DOI: 10.1007/s11899-024-00728-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE OF REVIEW The discovery that patients suffering from chronic myeloid leukemia who obtain deep and long-lasting molecular responses upon treatment with tyrosine kinase inhibitors may maintain their disease silent for many years after therapy discontinuation launched the era of treatment-free remission as a key management goal in clinical practice. The purpose of this review on treatment-free remission is to discuss clinical advances, highlight knowledge gaps, and describe areas of research. RECENT FINDINGS Patients in treatment-free remission are a minority, and it is believed that some may still retain a reservoir of leukemic stem cells; thus, whether they can be considered as truly cured is uncertain. Strengthening BCR::ABL1 inhibition increases deep molecular responses but is not sufficient to improve treatment-free remission, and we lack biomarkers to identify and specifically target residual cells with aggressive potential. Another level of complexity resides in the intra- and inter-patient clonal heterogeneity of minimal residual disease and characteristics of the bone marrow environment. Finding determinants of deep molecular responses achievement and elucidating varying biological mechanisms enabling either post-tyrosine kinase inhibitor chronic myeloid leukemia control or relapse may help develop innovative and safe therapies. In the light of the increasing prevalence of CML, targeting the residual leukemic stem cell pool is thought to be the key.
Collapse
Affiliation(s)
- Delphine Rea
- Service d'Hématologie Adulte, Hôpital Saint-Louis APHP, 75010, Paris, France.
- France Intergroupe Des Leucémies Myéloïdes Chroniques FiLMC, Paris, France.
| | - Sofiane Fodil
- Service d'Hématologie Adulte, Hôpital Saint-Louis APHP, 75010, Paris, France
| | - Etienne Lengline
- Service d'Hématologie Adulte, Hôpital Saint-Louis APHP, 75010, Paris, France
| | - Emmanuel Raffoux
- Service d'Hématologie Adulte, Hôpital Saint-Louis APHP, 75010, Paris, France
| | - Jean-Michel Cayuela
- France Intergroupe Des Leucémies Myéloïdes Chroniques FiLMC, Paris, France
- Laboratoire Central d'Hématologie, Hôpital Saint-Louis APHP, Paris, France
| |
Collapse
|
5
|
Hall T, Gurbuxani S, Crispino JD. Malignant progression of preleukemic disorders. Blood 2024; 143:2245-2255. [PMID: 38498034 PMCID: PMC11181356 DOI: 10.1182/blood.2023020817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024] Open
Abstract
ABSTRACT The spectrum of myeloid disorders ranges from aplastic bone marrow failure characterized by an empty bone marrow completely lacking in hematopoiesis to acute myeloid leukemia in which the marrow space is replaced by undifferentiated leukemic blasts. Recent advances in the capacity to sequence bulk tumor population as well as at a single-cell level has provided significant insight into the stepwise process of transformation to acute myeloid leukemia. Using models of progression in the context of germ line predisposition (trisomy 21, GATA2 deficiency, and SAMD9/9L syndrome), premalignant states (clonal hematopoiesis and clonal cytopenia of unknown significance), and myelodysplastic syndrome, we review the mechanisms of progression focusing on the hierarchy of clonal mutation and potential roles of transcription factor alterations, splicing factor mutations, and the bone marrow environment in progression to acute myeloid leukemia. Despite major advances in our understanding, preventing the progression of these disorders or treating them at the acute leukemia phase remains a major area of unmet medical need.
Collapse
Affiliation(s)
- Trent Hall
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Sandeep Gurbuxani
- Section of Hematopathology, Department of Pathology, University of Chicago, Chicago, IL
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
6
|
Chelysheva E, Apperley J, Turkina A, Yassin MA, Rea D, Nicolini FE, Barraco D, Kazakbaeva K, Saliev S, Abulafia AS, Al-Kindi S, Byrne J, Robertson HF, Cerrano M, Shmakov R, Polushkina E, de Fabritiis P, Trawinska MM, Abruzzese E. Chronic myeloid leukemia diagnosed in pregnancy: management and outcome of 87 patients reported to the European LeukemiaNet international registry. Leukemia 2024; 38:788-795. [PMID: 38388649 PMCID: PMC11408247 DOI: 10.1038/s41375-024-02183-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
The management of chronic myeloid leukemia (CML) diagnosed during pregnancy is a rare and challenging situation. We report the treatment and outcome of 87 cases diagnosed in chronic phase from 2001-2022 derived from the largest international observational registry, supported by the European LeukemiaNet (ELN), of 400 pregnancies in 299 CML women. Normal childbirth occurred in 76% without an increased rate of birth abnormalities or life-threatening events, including in patients untreated or treated with interferon-α and/or imatinib in 2nd-3rd trimester. The low birth weight rate of 12% was comparable to that seen in the normal population. Elective and spontaneous abortions occurred in 21% and 3%, respectively. The complete hematologic response rate before labor was 95% with imatinib and 47% with interferon only. No disease progression during pregnancy was observed, 28% of the patients switched their therapy at varying times after delivery. Treatment options balance the efficacy and safety for mother and infant: interferon-α can commence in the 1st trimester and continued throughout in cases of good disease control and tolerability. Because of limited placental crossing, selected tyrosine kinase inhibitors (imatinib and nilotinib) seem to be safe and effective options in 2nd and 3rd trimester while hydroxycarbamide offers few benefits.
Collapse
Affiliation(s)
| | - Jane Apperley
- Centre for Haematology, Imperial College London, London, UK
| | - Anna Turkina
- National Medical Research Center for Hematology, Moscow, Russian Federation
| | - Mohamed A Yassin
- Department of Medical Oncology/ Hematology Section, National Centre For Cancer Care & Research, Doha, Qatar
| | - Delphine Rea
- Service d'hématologie Adulte and FiLMC Hôpital Saint-Louis, Paris, France
| | - Franck E Nicolini
- Hematology department and INSERM 1052 CRCL, Centre Léon Bérard, Lyon, France
| | - Daniela Barraco
- Division of Hematology, University Hospital "Ospedale di Circolo e Fondazione Macchi - ASST Sette Laghi", Varese, Italy
| | - Khamida Kazakbaeva
- Republician Specilized Scientific and Practical Medical Centre of Hematology, Tashkent, Uzbekistan
| | - Sukhrob Saliev
- Republician Specilized Scientific and Practical Medical Centre of Hematology, Tashkent, Uzbekistan
| | - Adi Shacham Abulafia
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
| | - Salam Al-Kindi
- Department of Haematology, Sultan Qaboos University, Muscat, Oman
| | - Jennifer Byrne
- Centre for Clinical Haematology, Nottingham University Hospitals NHS Trust, Nottingham, England, UK
| | | | - Marco Cerrano
- Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| | - Roman Shmakov
- FSBI «National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov» Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| | - Evgenia Polushkina
- FSBI «National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov» Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| | - Paolo de Fabritiis
- Hematology, S. Eugenio Hospital, Tor Vergata University, ASL Roma2, Rome, Italy
| | | | | |
Collapse
|
7
|
Elsabagh AA, Benkhadra M, Elmakaty I, Elsayed A, Elsayed B, Elmarasi M, Abutineh M, Qasem NM, Ali E, Yassin M. Male Fertility and Fatherhood in Chronic Myeloid Leukemia: Current Understanding and Future Perspectives. Cancers (Basel) 2024; 16:791. [PMID: 38398181 PMCID: PMC10886940 DOI: 10.3390/cancers16040791] [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: 10/16/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 02/25/2024] Open
Abstract
Chronic myeloid leukemia (CML), while traditionally a disease of the elderly, has recently risen in incidence among younger patients. Hence, fertility concerns have emerged considering the disease process and treatments, especially with the current scarce and conflicting recommendations. This review explores the impact of CML treatments including the first-line tyrosine kinase inhibitors (TKIs) and other treatments on male fertility in chronic myeloid leukemia (CML) patients. The aim of this review was to compile the available evidence on male fertility to ultimately tailor treatment plans for male CML patients for whom fertility and future chances for conception pose a concern. The data available on the conventional and newer TKIs to address fertility concerns were reviewed, particularly the potential long- and short-term effects. Also, the possible side effects on subsequent generations were a crucial focus point of this review to reach a more comprehensive CML management approach. We found and compared the evidence on TKIs approved to treat CML. We also reported the effects of hydroxyurea, interferon, and transplantation, which are considered second-line treatments. Our findings suggest that these drugs might have an undiscovered effect on fertility. More research with larger sample sizes and longer follow-up periods is essential to solidify our understanding of these effects.
Collapse
Affiliation(s)
- Ahmed Adel Elsabagh
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (I.E.); (B.E.); (M.E.)
| | - Maria Benkhadra
- National Center for Cancer Care & Research, Hamad General Hospital, Doha P.O. Box 3050, Qatar;
| | - Ibrahim Elmakaty
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (I.E.); (B.E.); (M.E.)
| | - Abdelrahman Elsayed
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (I.E.); (B.E.); (M.E.)
| | - Basant Elsayed
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (I.E.); (B.E.); (M.E.)
| | - Mohamed Elmarasi
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (I.E.); (B.E.); (M.E.)
| | - Mohammad Abutineh
- Hematology Section, Medical Oncology, National Center for Cancer Care and Research (NCCCR), Hamad Medical Corporation, Doha P.O. Box 3050, Qatar (N.M.Q.)
| | - Nabeel Mohammad Qasem
- Hematology Section, Medical Oncology, National Center for Cancer Care and Research (NCCCR), Hamad Medical Corporation, Doha P.O. Box 3050, Qatar (N.M.Q.)
| | - Elrazi Ali
- One Brooklyn Health, Interfaith Medical Center, Brooklyn, NY 11213, USA;
| | - Mohamed Yassin
- Hematology Section, Medical Oncology, National Center for Cancer Care and Research (NCCCR), Hamad Medical Corporation, Doha P.O. Box 3050, Qatar (N.M.Q.)
| |
Collapse
|
8
|
Vukotić M, Kapor S, Simon F, Cokic V, Santibanez JF. Mesenchymal stromal cells in myeloid malignancies: Immunotherapeutic opportunities. Heliyon 2024; 10:e25081. [PMID: 38314300 PMCID: PMC10837636 DOI: 10.1016/j.heliyon.2024.e25081] [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: 10/05/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Myeloid malignancies are clonal disorders of the progenitor cells or hematopoietic stem cells, including acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid neoplastic cells affect the proliferation and differentiation of other hematopoietic lineages in the bone marrow and peripheral blood, leading to severe and life-threatening complications. Mesenchymal stromal cells (MSCs) residing in the bone marrow exert immunosuppressive functions by suppressing innate and adaptive immune systems, thus creating a supportive and tolerant microenvironment for myeloid malignancy progression. This review summarizes the significant features of MSCs in myeloid malignancies, including their role in regulating cell growth, cell death, and antineoplastic resistance, in addition to their immunosuppressive contributions. Understanding the implications of MSCs in myeloid malignancies could pave the path for potential use in immunotherapy.
Collapse
Affiliation(s)
- Milica Vukotić
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Suncica Kapor
- Department of Hematology, Clinical Hospital Center “Dr. Dragisa Misovic-Dedinje,” University of Belgrade, Serbia
| | - Felipe Simon
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases, Universidad de Chile, Santiago, Chile
| | - Vladan Cokic
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Juan F. Santibanez
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| |
Collapse
|
9
|
Bishehsari F, Drees M, Adnan D, Sharma D, Green S, Koshy J, Giron LB, Goldman A, Abdel-Mohsen M, Rasmussen HE, Miller GE, Keshavarzian A. Multi-omics approach to socioeconomic disparity in metabolic syndrome reveals roles of diet and microbiome. Proteomics 2023; 23:e2300023. [PMID: 37525324 DOI: 10.1002/pmic.202300023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/02/2023]
Abstract
The epidemy of metabolic syndrome (MetS) is typically preceded by adoption of a "risky" lifestyle (e.g., dietary habit) among populations. Evidence shows that those with low socioeconomic status (SES) are at an increased risk for MetS. To investigate this, we recruited 123 obese subjects (body mass index [BMI] ≥ 30) from Chicago. Multi-omic data were collected to interrogate fecal microbiota, systemic markers of inflammation and immune activation, plasma metabolites, and plasma glycans. Intestinal permeability was measured using the sugar permeability testing. Our results suggest a heterogenous metabolic dysregulation among obese populations who are at risk of MetS. Systemic inflammation, linked to poor diet, intestinal microbiome dysbiosis, and gut barrier dysfunction may explain the development of MetS in these individuals. Our analysis revealed 37 key features associated with increased numbers of MetS features. These features were used to construct a composite metabolic-inflammatory (MI) score that was able to predict progression of MetS among at-risk individuals. The MI score was correlated with several markers of poor diet quality as well as lower levels of gut microbial diversity and abnormalities in several species of bacteria. This study reveals novel targets to reduce the burden of MetS and suggests access to healthy food options as a practical intervention.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Michael Drees
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Darbaz Adnan
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Deepak Sharma
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Stefan Green
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Jane Koshy
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Leila B Giron
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Aaron Goldman
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | | | - Gregory E Miller
- Institute for Policy Research and Dept of Psychology, Northwestern Univ, Evanston, Illinois, USA
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
10
|
Ansari S, Verma M. Control of Ph + and additional chromosomal abnormalities in chronic myeloid leukemia by tyrosine kinase inhibitors. Med Oncol 2023; 40:237. [PMID: 37439908 DOI: 10.1007/s12032-023-02116-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Chronic myeloid leukemia (CML) is a type of blood cancer that is known to affect hematopoietic stem cells. The presence of the Philadelphia chromosome (Ph+) is the major characteristic of CML. A protein expressed by the Philadelphia chromosome shows elevated tyrosine kinase activity and is considered a tumorigenic factor. The first line of therapy that had been established for CML was "imatinib," a potent tyrosine kinase inhibitor. Various other second- and third-generation TKIs are taken into account in cases of imatinib failure/resistance. With the subsequent rise in the development of tyrosine kinase inhibitors, optimization in the treatment of CML and amplified total survival were observed throughout TKI dosage. As the disease progresses, additional chromosomal abnormalities (ACAs) have been reported, but their prognostic effect and impact on the response to treatment are still unknown. However, some substantial understandings have been achieved into the disease transformation mechanisms, including the role of somatic mutations, ACAs, and several different genomic mutations that occur during diagnosis or have evolved during treatment. The acquisition of ACAs impedes CML treatment. Due to additional chromosomal lesions, there are greater chances of future disease progression at the time of CML diagnosis beyond the Ph+ translocation. The synchronous appearance of two or more ACAs leads to lower survival and is classified as a poor prognostic group. The key objective of this review is to provide detailed insights into TKIs and their role in controlling Ph+ and ACAs, along with their response, treatment, overall persistence, and survival rate.
Collapse
Affiliation(s)
- Sana Ansari
- School of Biotechnology, Banaras Hindu University, Varanasi, U.P., 221005, India
| | - Malkhey Verma
- School of Biotechnology, Banaras Hindu University, Varanasi, U.P., 221005, India.
| |
Collapse
|
11
|
Hiroki H, Ishii Y, Piao J, Namikawa Y, Masutani M, Honda H, Akahane K, Inukai T, Morio T, Takagi M. Targeting Poly(ADP)ribose polymerase in BCR/ABL1-positive cells. Sci Rep 2023; 13:7588. [PMID: 37165001 PMCID: PMC10172294 DOI: 10.1038/s41598-023-33852-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/20/2023] [Indexed: 05/12/2023] Open
Abstract
BCR/ABL1 causes dysregulated cell proliferation and is responsible for chronic myelogenous leukemia (CML) and Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph1-ALL). In addition to the deregulatory effects of its kinase activity on cell proliferation, BCR/ABL1 induces genomic instability by downregulating BRCA1. PARP inhibitors (PARPi) effectively induce cell death in BRCA-defective cells. Therefore, PARPi are expected to inhibit growth of CML and Ph1-ALL cells showing downregulated expression of BRCA1. Here, we show that PARPi effectively induced cell death in BCR/ABL1 positive cells and suppressed colony forming activity. Prevention of BCR/ABL1-mediated leukemogenesis by PARP inhibition was tested in two in vivo models: wild-type mice that had undergone hematopoietic cell transplantation with BCR/ABL1-transduced cells, and a genetic model constructed by crossing Parp1 knockout mice with BCR/ABL1 transgenic mice. The results showed that a PARPi, olaparib, attenuates BCR/ABL1-mediated leukemogenesis. One possible mechanism underlying PARPi-dependent inhibition of leukemogenesis is increased interferon signaling via activation of the cGAS/STING pathway. This is compatible with the use of interferon as a first-line therapy for CML. Because tyrosine kinase inhibitor (TKI) monotherapy does not completely eradicate leukemic cells in all patients, combined use of PARPi and a TKI is an attractive option that may eradicate CML stem cells.
Collapse
Affiliation(s)
- Haruka Hiroki
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Yuko Ishii
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Jinhua Piao
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Yui Namikawa
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Mitsuko Masutani
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, 852-8523, Nagasaki, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Koshi Akahane
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Takeshi Inukai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
12
|
Patterson SD, Copland M. The Bone Marrow Immune Microenvironment in CML: Treatment Responses, Treatment-Free Remission, and Therapeutic Vulnerabilities. Curr Hematol Malig Rep 2023; 18:19-32. [PMID: 36780103 PMCID: PMC9995533 DOI: 10.1007/s11899-023-00688-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 02/14/2023]
Abstract
PURPOSE OF REVIEW Tyrosine kinase inhibitors (TKIs) are very successful for the treatment of chronic myeloid leukaemia (CML) but are not curative in most patients due to persistence of TKI-resistant leukaemia stem cells (LSCs). The bone marrow immune microenvironment (BME) provides protection to the LSC through multidimensional interactions, driving therapy resistance, and highlighting the need to circumvent these protective niches therapeutically. This review updates the evidence for interactions between CML cells and the immune microenvironment with a view to identifying targetable therapeutic vulnerabilities and describes what is known about the role of immune regulation in treatment-free remission (TFR). RECENT FINDINGS Intracellular signalling downstream of the chemotactic CXCL12-CXCR4 axis, responsible for disrupted homing in CML, has been elucidated in LSCs, highlighting novel therapeutic opportunities. In addition, LSCs expressing CXCL12-cleaving surface protein CD26 were highly correlated with CML burden, building on existing evidence. Newer findings implicate the adhesion molecule CD44 in TKI resistance, while JAK/STAT-mediated resistance to TKIs may occur downstream of extrinsic signalling in the BME. Exosomal BME-LSC cross-communication has also been explored. Finally, further detail on the phenotypes of natural killer (NK) cells putatively involved in maintaining successful TFR has been published, and NK-based immunotherapies are discussed. Recent studies highlight and build on our understanding of the BME in CML persistence and TKI resistance, pinpointing therapeutically vulnerable interactions. Repurposing existing drugs and/or the development of novel inhibitors targeting these relationships may help to overcome these issues in TKI-resistant CML and be used as adjuvant therapy for sustained TFR.
Collapse
Affiliation(s)
- Shaun David Patterson
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Paul O'Gorman Leukaemia Research Centre, University of Glasgow, 21 Shelley Road, Glasgow, G12 0ZD, UK.
| | - Mhairi Copland
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Paul O'Gorman Leukaemia Research Centre, University of Glasgow, 21 Shelley Road, Glasgow, G12 0ZD, UK.
| |
Collapse
|
13
|
Holicek P, Truxova I, Rakova J, Salek C, Hensler M, Kovar M, Reinis M, Mikyskova R, Pasulka J, Vosahlikova S, Remesova H, Valentova I, Lysak D, Holubova M, Kaspar P, Prochazka J, Kasikova L, Spisek R, Galluzzi L, Fucikova J. Type I interferon signaling in malignant blasts contributes to treatment efficacy in AML patients. Cell Death Dis 2023; 14:209. [PMID: 36964168 PMCID: PMC10039058 DOI: 10.1038/s41419-023-05728-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 03/26/2023]
Abstract
While type I interferon (IFN) is best known for its key role against viral infection, accumulating preclinical and clinical data indicate that robust type I IFN production in the tumor microenvironment promotes cancer immunosurveillance and contributes to the efficacy of various antineoplastic agents, notably immunogenic cell death inducers. Here, we report that malignant blasts from patients with acute myeloid leukemia (AML) release type I IFN via a Toll-like receptor 3 (TLR3)-dependent mechanism that is not driven by treatment. While in these patients the ability of type I IFN to stimulate anticancer immune responses was abolished by immunosuppressive mechanisms elicited by malignant blasts, type I IFN turned out to exert direct cytostatic, cytotoxic and chemosensitizing activity in primary AML blasts, leukemic stem cells from AML patients and AML xenograft models. Finally, a genetic signature of type I IFN signaling was found to have independent prognostic value on relapse-free survival and overall survival in a cohort of 132 AML patients. These findings delineate a clinically relevant, therapeutically actionable and prognostically informative mechanism through which type I IFN mediates beneficial effects in patients with AML.
Collapse
Affiliation(s)
- Peter Holicek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | | | | | - Cyril Salek
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
- Institute of Clinical and Experimental Hematology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Marek Kovar
- Laboratory of Tumor Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Milan Reinis
- Laboratory of Immunological and Tumour Models, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Romana Mikyskova
- Laboratory of Immunological and Tumour Models, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | | | | | - Hana Remesova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Iva Valentova
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Daniel Lysak
- Department of Hematology and Oncology, Faculty Hospital in Pilsen, Pilsen, Czech Republic
| | - Monika Holubova
- Biomedical Center, Medical Faculty in Pilsen, Charles University, Pilsen, Czech Republic
| | - Petr Kaspar
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | | | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic.
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic.
| |
Collapse
|
14
|
Hasselbalch H, Skov V, Kjær L, Larsen MK, Knudsen TA, Lucijanić M, Kusec R. Recombinant Interferon-β in the Treatment of Polycythemia Vera and Related Neoplasms: Rationales and Perspectives. Cancers (Basel) 2022; 14:5495. [PMID: 36428587 PMCID: PMC9688061 DOI: 10.3390/cancers14225495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
About 30 years ago, the first clinical trials of the safety and efficacy of recombinant interferon-α2 (rIFN-α2) were performed. Since then, several single-arm studies have shown rIFN-α2 to be a highly potent anticancer agent against several cancer types. Unfortunately, however, a high toxicity profile in early studies with rIFN-α2 -among other reasons likely due to the high dosages being used-disqualified rIFN-α2, which was accordingly replaced with competitive drugs that might at first glance look more attractive to clinicians. Later, pegylated IFN-α2a (Pegasys) and pegylated IFN-α2b (PegIntron) were introduced, which have since been reported to be better tolerated due to reduced toxicity. Today, treatment with rIFN-α2 is virtually outdated in non-hematological cancers, where other immunotherapies-e.g., immune-checkpoint inhibitors-are routinely used in several cancer types and are being intensively investigated in others, either as monotherapy or in combination with immunomodulatory agents, although only rarely in combination with rIFN-α2. Within the hematological malignancies, rIFN-α2 has been used off-label for decades in patients with Philadelphia-negative chronic myeloproliferative neoplasms (MPNs)-i.e., essential thrombocythemia, polycythemia vera, and myelofibrosis-and in recent years rIFN-α2 has been revived with the marketing of ropeginterferon-α2b (Besremi) for the treatment of polycythemia vera patients. Additionally, rIFN-α2 has been revived for the treatment of chronic myelogenous leukemia in combination with tyrosine kinase inhibitors. Another rIFN formulation-recombinant interferon-β (rIFN-β)-has been used for decades in the treatment of multiple sclerosis but has never been studied as a potential agent to be used in patients with MPNs, although several studies and reviews have repeatedly described rIFN-β as an effective anticancer agent as well. In this paper, we describe the rationales and perspectives for launching studies on the safety and efficacy of rIFN-β in patients with MPNs.
Collapse
Affiliation(s)
- Hans Hasselbalch
- Department of Hematology, Zealand University, 4000 Roskilde, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University, 4000 Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University, 4000 Roskilde, Denmark
| | | | - Trine A. Knudsen
- Department of Hematology, Zealand University, 4000 Roskilde, Denmark
| | - Marko Lucijanić
- Department of Hematology, University Hospital Dubrava, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Rajko Kusec
- Department of Hematology, University Hospital Dubrava, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
15
|
Puzzolo MC, Breccia M, Mariglia P, Colafigli G, Pepe S, Scalzulli E, Mariggiò E, Latagliata R, Guarini A, Foà R. Immunomodulatory Effects of IFNα on T and NK Cells in Chronic Myeloid Leukemia Patients in Deep Molecular Response Preparing for Treatment Discontinuation. J Clin Med 2022; 11:jcm11195594. [PMID: 36233461 PMCID: PMC9570842 DOI: 10.3390/jcm11195594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/06/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
A deep and stable molecular response (DMR) is a prerequisite for a successful treatment-free remission (TFR) in chronic myeloid leukemia (CML). In order to better identify and analyze potential candidates of successful TFR, we examined the phenotypic and functional host immune compartment in DMR patients who had received TKI treatment only (TKI-only) or had been previously treated with interferon-alpha (IFNα + TKI) or had received IFNα treatment only (IFNα-only). The T/NK-cell subset distribution, NK- and T-cell cytokine production, activation and maturation markers were measured in 44 patients in DMR treated with IFNα only (9), with IFNα + TKI (11) and with TKI-only (24). IFNα + TKI and TKI-only groups were eligible to TKI discontinuation according to the NCCN and ESMO guidelines (stable MR4 for more than two years). In IFNα-treated patients, we documented an increased number of lymphocytes capable of producing IFNγ and TNFα compared to the TKI-only group. In INFα + TKI patients, the percentage of NKG2C expression and its mean fluorescence intensity were significantly higher compared to the TKI-only group and to the INFα-only group in the CD56dim/CD16+ NK cell subsets (INFα + TKI vs. TKI-only p = 0.041, p = 0.037; INFα + TKI vs. INFα-only p = 0.03, p = 0.033, respectively). Furthermore, in INFα-only treated patients, we observed an increase of NKp46 MFI in the CD56bright/CD16- NK cell subset that becomes significant compared to the INFα + TKI group (p = 0.008). Our data indicate that a previous exposure to IFNα substantially and persistently modified the immune system of CML patients in memory T lymphocytes, differentiated NKG2C+ “long-lived” NK cells responses, even years after the last IFNα contact.
Collapse
Affiliation(s)
- Maria Cristina Puzzolo
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto 1, ‘Sapienza’ University, 00161 Rome, Italy
| | - Massimo Breccia
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto 1, ‘Sapienza’ University, 00161 Rome, Italy
- Correspondence: ; Tel.: +39-06-857-951; Fax: +39-06-4424-1984
| | - Paola Mariglia
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto 1, ‘Sapienza’ University, 00161 Rome, Italy
| | - Gioia Colafigli
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto 1, ‘Sapienza’ University, 00161 Rome, Italy
| | - Sara Pepe
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto 1, ‘Sapienza’ University, 00161 Rome, Italy
| | - Emilia Scalzulli
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto 1, ‘Sapienza’ University, 00161 Rome, Italy
| | - Elena Mariggiò
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto 1, ‘Sapienza’ University, 00161 Rome, Italy
| | - Roberto Latagliata
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto 1, ‘Sapienza’ University, 00161 Rome, Italy
| | - Anna Guarini
- Hematology, Department of Molecular Medicine, ‘Sapienza’ University, 00161 Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto 1, ‘Sapienza’ University, 00161 Rome, Italy
| |
Collapse
|
16
|
Abruzzese E, Aureli S, Bondanini F, Ciccarone M, Cortis E, Di Paolo A, Fabiani C, Galimberti S, Malagola M, Malato A, Martino B, Trawinska MM, Russo D, de Fabritiis P. Chronic Myeloid Leukemia and Pregnancy: When Dreams Meet Reality. State of the Art, Management and Outcome of 41 Cases, Nilotinib Placental Transfer. J Clin Med 2022; 11:1801. [PMID: 35407407 PMCID: PMC8999799 DOI: 10.3390/jcm11071801] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/26/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023] Open
Abstract
The overwhelming success of tyrosine kinase inhibitor (TKI) therapy in chronic myeloid leukemia (CML) patients has opened a discussion among medical practitioners and the lay public on the real possibility of pregnancy and conception in females and males with CML. In the past 10 years this subject has acquired growing interest in the scientific community and specific knowledge has been obtained "from bench to bedside". Embryological, pharmacological, and pathophysiological studies have merged with worldwide patient databases to provide a roadmap to a successful pregnancy and birth in CML patients. Male conception does not seem to be affected by TKI therapy, since this class of drugs is neither genotoxic nor mutagenic, however, caution should be used specially with newer drugs for which little or no data are available. In contrast, female patients should avoid TKI therapy specifically during the embryonic stage of organogenesis (5-12 weeks) because TKIs can be teratogenic. In the last 15 years, 41 pregnancies have been followed in our center. A total of 11 male conceptions and 30 female pregnancies are described. TKI treatment was generally terminated as soon as the pregnancy was discovered (3-5 weeks), to avoid exposure during embryonic period and to reduce the risk of needing treatment in the first trimester. Eleven pregnancies were treated with interferon, imatinib or nilotinib during gestation. Nilotinib plasma levels in cord blood and maternal blood at delivery were studied in 2 patients and reduced or absent placental crossing of nilotinib was observed. All of the patients were managed by a multidisciplinary team of physicians with obligatory hematological and obgyn consultations. This work provides an update on the state of the art and detailed description of pregnancy management and outcomes in CML patients.
Collapse
Affiliation(s)
- Elisabetta Abruzzese
- Hematology, Sant’Eugenio Hospital, ASL Roma 2, Tor Vergata University, 00144 Rome, Italy; (M.M.T.); (P.d.F.)
| | - Stefano Aureli
- Obstetrics and Gynecology, Sant’Eugenio Hospital, ASL Roma 2, 00144 Rome, Italy;
| | | | - Mariavita Ciccarone
- “Dormant Buds” Association, Obstetrics and Gynecology, San Carlo di Nancy Hospital, 00165 Rome, Italy;
| | | | - Antonello Di Paolo
- Section of Pharmacology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Cristina Fabiani
- Reproduction Pathophisiology and Andrology, Sandro Pertini Hospital, ASL Roma 2, 00157 Rome, Italy;
| | - Sara Galimberti
- Hematology Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Michele Malagola
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Sciences, ASST Spedali Civili di Brescia, University of Brescia, 25123 Brescia, Italy; (M.M.); (D.R.)
| | - Alessandra Malato
- Division of Hematology, Ospedali Riuniti Villa Sofia-Cervello, 90146 Palermo, Italy;
| | - Bruno Martino
- Hematology Unit, Grande Ospedale Metropolitano “Bianchi-Melacrino-Morelli”, 89124 Reggio Calabria, Italy;
| | - Malgorzata Monika Trawinska
- Hematology, Sant’Eugenio Hospital, ASL Roma 2, Tor Vergata University, 00144 Rome, Italy; (M.M.T.); (P.d.F.)
| | - Domenico Russo
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Sciences, ASST Spedali Civili di Brescia, University of Brescia, 25123 Brescia, Italy; (M.M.); (D.R.)
| | - Paolo de Fabritiis
- Hematology, Sant’Eugenio Hospital, ASL Roma 2, Tor Vergata University, 00144 Rome, Italy; (M.M.T.); (P.d.F.)
| |
Collapse
|
17
|
Innate Immune Mechanisms and Immunotherapy of Myeloid Malignancies. Biomedicines 2021; 9:biomedicines9111631. [PMID: 34829860 PMCID: PMC8615731 DOI: 10.3390/biomedicines9111631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Similar to other cancers, myeloid malignancies are thought to subvert the immune system during their development. This subversion occurs via both malignant cell-autonomous and non-autonomous mechanisms and involves manipulation of the innate and adaptive immune systems. Multiple strategies are being studied to rejuvenate, redirect, or re-enforce the immune system in order to fight off myeloid malignancies. So far, the most successful strategies include interferon treatment and antibody-based therapies, though chimeric antigen receptor (CAR) cells and immune checkpoint inhibitors are also promising therapies. In this review, we discuss the inherent immune mechanisms of defense against myeloid malignancies, currently-approved agents, and agents under investigation. Overall, we evaluate the efficacy and potential of immuno-oncology in the treatment of myeloid malignancies.
Collapse
|
18
|
Matsushita M. Novel Treatment Strategies Utilizing Immune Reactions against Chronic Myelogenous Leukemia Stem Cells. Cancers (Basel) 2021; 13:cancers13215435. [PMID: 34771599 PMCID: PMC8582551 DOI: 10.3390/cancers13215435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 01/21/2023] Open
Abstract
Simple Summary Although tyrosine kinase inhibitors (TKIs) are highly effective in the treatment of patients with chronic myelogenous leukemia (CML), leukemic stem cells (LSCs) are known to be resistant to TKIs. As a result, the application of immunotherapies against LSCs may cure CML. Abstract Introduction of tyrosine kinase inhibitors (TKIs) has improved the prognosis of patients with chronic myelogenous leukemia (CML), and treatment-free remission (TFR) is now a treatment goal. However, about half of the patients experience molecular relapse after cessation of TKIs, suggesting that leukemic stem cells (LSCs) are resistant to TKIs. Eradication of the remaining LSCs using immunotherapies including interferon-alpha, vaccinations, CAR-T cells, and other drugs would be a key strategy to achieve TFR.
Collapse
Affiliation(s)
- Maiko Matsushita
- Division of Clinical Physiology and Therapeutics, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| |
Collapse
|
19
|
Saifullah HH, Lucas CM. Treatment-Free Remission in Chronic Myeloid Leukemia: Can We Identify Prognostic Factors? Cancers (Basel) 2021; 13:cancers13164175. [PMID: 34439327 PMCID: PMC8392063 DOI: 10.3390/cancers13164175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/04/2021] [Accepted: 08/08/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Chronic myeloid leukemia (CML) is a blood cancer. Unlike other cancers CML treatment is lifelong and many patients experience side effects. For those patients who respond well to treatment and achieve deep molecular remission, quality of life is impacted because of continuous treatment. In this review, we look at emerging clinical trials which aim to investigate which patients can safely stop treatment. Treatment-free remission is the ultimate goal for CML patients, but there is still a gap in our knowledge as to why some patients can achieve treatment-free remission, while others relapse when treatment is stopped. Here we discuss if there are any prognostic factors that can predict the best candidates who qualify for treatment discontinuation, with a view to keeping them in remission. Abstract Following the development of tyrosine kinase inhibitors (TKI), the survival of patients with chronic myeloid leukaemia (CML) drastically improved. With the introduction of these agents, CML is now considered a chronic disease for some patients. Taking into consideration the side effects, toxicity, and high cost, discontinuing TKI became a goal for patients with chronic phase CML. Patients who achieved deep molecular response (DMR) and discontinued TKI, remained in treatment-free remission (TFR). Currently, the data from the published literature demonstrate that 40–60% of patients achieve TFR, with relapses occurring within the first six months. In addition, almost all patients who relapsed regained a molecular response upon retreatment, indicating TKI discontinuation is safe. However, there is still a gap in understanding the mechanisms behind TFR, and whether there are prognostic factors that can predict the best candidates who qualify for TKI discontinuation with a view to keeping them in TFR. Furthermore, the information about a second TFR attempt and the role of gradual de-escalation of TKI before complete cessation is limited. This review highlights the factors predicting success or failure of TFR. In addition, it examines the feasibility of a second TFR attempt after the failure of the first one, and the current guidelines concerning TFR in clinical practice.
Collapse
Affiliation(s)
- Hilbeen Hisham Saifullah
- Chester Medical School, University of Chester, Bache Hall, Chester CH2 1BR, UK
- Correspondence: (H.H.S.); (C.M.L.)
| | - Claire Marie Lucas
- Chester Medical School, University of Chester, Bache Hall, Chester CH2 1BR, UK
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GA, UK
- Correspondence: (H.H.S.); (C.M.L.)
| |
Collapse
|
20
|
Yung Y, Lee E, Chu HT, Yip PK, Gill H. Targeting Abnormal Hematopoietic Stem Cells in Chronic Myeloid Leukemia and Philadelphia Chromosome-Negative Classical Myeloproliferative Neoplasms. Int J Mol Sci 2021; 22:ijms22020659. [PMID: 33440869 PMCID: PMC7827471 DOI: 10.3390/ijms22020659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 02/02/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are unique hematopoietic stem cell disorders sharing mutations that constitutively activate the signal-transduction pathways involved in haematopoiesis. They are characterized by stem cell-derived clonal myeloproliferation. The key MPNs comprise chronic myeloid leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). CML is defined by the presence of the Philadelphia (Ph) chromosome and BCR-ABL1 fusion gene. Despite effective cytoreductive agents and targeted therapy, complete CML/MPN stem cell eradication is rarely achieved. In this review article, we discuss the novel agents and combination therapy that can potentially abnormal hematopoietic stem cells in CML and MPNs and the CML/MPN stem cell-sustaining bone marrow microenvironment.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Autophagy
- Biomarkers, Tumor
- Cell Survival/drug effects
- Cell Transformation, Neoplastic/genetics
- Combined Modality Therapy
- Disease Susceptibility
- Genetic Predisposition to Disease
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Molecular Targeted Therapy
- Myeloproliferative Disorders/etiology
- Myeloproliferative Disorders/pathology
- Myeloproliferative Disorders/therapy
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Philadelphia Chromosome
- Signal Transduction/drug effects
- Stem Cell Niche
- Tumor Microenvironment
Collapse
Affiliation(s)
| | | | | | | | - Harinder Gill
- Correspondence: ; Tel.: +852-2255-4542; Fax: +852-2816-2863
| |
Collapse
|
21
|
Abstract
New insights have emerged from maturing long-term academic and commercial clinical trials regarding optimum management of chronic myeloid leukemia (CML). Velocity of response has unexpectedly proved less important than hitherto thought, does not predict survival, and is of unclear relevance for treatment-free remission (TFR). Serious and cumulative toxicity has been observed with tyrosine kinase inhibitors that had been expected to replace imatinib. Generic imatinib has become cost-effective first-line treatment in chronic phase despite chronic low-grade side-effects in many patients. Earlier recognition of end-phase by genetic assessment might improve prospects for blast crisis (BC). TFR has become an important new treatment goal of CML. To reflect this new situation ELN has recently revised and updated its recommendations for treating CML. After a brief review of 175 years of CML history this review will focus on recent developments and on current evidence for treating CML in 2020.
Collapse
Affiliation(s)
- Rüdiger Hehlmann
- ELN Foundation, Weinheim; Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Germany
| |
Collapse
|
22
|
Galimberti S, Petrini M, Baratè C, Ricci F, Balducci S, Grassi S, Guerrini F, Ciabatti E, Mechelli S, Di Paolo A, Baldini C, Baglietto L, Macera L, Spezia PG, Maggi F. Tyrosine Kinase Inhibitors Play an Antiviral Action in Patients Affected by Chronic Myeloid Leukemia: A Possible Model Supporting Their Use in the Fight Against SARS-CoV-2. Front Oncol 2020; 10:1428. [PMID: 33014780 PMCID: PMC7493657 DOI: 10.3389/fonc.2020.01428] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
SARS-CoV-2 is the viral agent responsible for the pandemic that in the first months of 2020 caused about 400,000 deaths. Among compounds proposed to fight the SARS-CoV-2-related disease (COVID-19), tyrosine kinase inhibitors (TKIs), already effective in Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL) and chronic myeloid leukemia (CML), have been proposed on the basis of their antiviral action already demonstrated against SARS-CoV-1. Very few cases of COVID-19 have been reported in Ph+ ALL and in CML Italian cohorts; authors suggested that this low rate of infections might depend on the use of TKIs, but the biological causes of this phenomenon remain unknown. In this study, the CML model was used to test if TKIs would sustain or not the viral replication and if they could damage patient immunity. Firstly, the infection and replication rate of torquetenovirus (TTV), whose load is inversely proportional to the host immunological control, have been measured in CML patients receiving nilotinib. A very low percentage of subjects were infected at baseline, and TTV did not replicate or at least showed a low replication rate during the follow-up, with a mean load comparable to the measured one in healthy subjects. Then, after gene expression profiling experiments, we found that several “antiviral” genes, such as CD28 and IFN gamma, were upregulated, while genes with “proviral” action, such as ARG-1, CEACAM1, and FUT4, were less expressed during treatment with imatinib, thus demonstrating that TKIs are not detrimental from the immunological point of view. To sum up, our data could offer some biological explanations to the low COVID-19 occurrence in Ph+ ALL and CML patients and sustain the use of TKIs in COVID-19, as already proposed by several international ongoing studies.
Collapse
Affiliation(s)
- Sara Galimberti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Mario Petrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Federica Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Serena Balducci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Susanna Grassi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesca Guerrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Ciabatti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Antonello Di Paolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Baldini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Laura Baglietto
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lisa Macera
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Pietro Giorgio Spezia
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Fabrizio Maggi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
23
|
Pegylated interferon-2α invokes graft-versus-leukemia effects in patients relapsing after allogeneic stem cell transplantation. Blood Adv 2020; 3:3013-3019. [PMID: 31648324 DOI: 10.1182/bloodadvances.2019000453] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/08/2019] [Indexed: 01/01/2023] Open
Abstract
Allogeneic stem cell transplantation (SCT) is a curative therapy for patients with hematological malignancies related largely to an immunological graft-versus-leukemia (GVL) effect mediated by donor T cells and natural killer cells. Relapse of disease after SCT represents failure of GVL and is now the major cause of treatment failure. We sought to augment GVL effects in patients (n = 29) relapsing after SCT in a prospective phase I/II clinical trial of dose-escalated pegylated interferon-2α (peg-IFNα). The administration of peg-IFNα after reinduction chemotherapy, with or without subsequent donor lymphocyte infusion (DLI), resulted in a 2-year overall survival (OS) of 31% (95% confidence interval, 17.3%-49.2%), which rejects the null hypothesis of 7% generated by observations in an institutional historical cohort. As expected, peg-IFNα was associated with graft-versus-host disease (GVHD) and hematological toxicity, which was manageable with scheduled dose modifications. Progression-free survival (PFS) was greatest in patients who experienced GVHD, although the majority of those patients still eventually progressed. Higher PFS and OS were associated with pretreatment proportions of immune cell populations with regulatory function, including mucosal invariant T cells, regulatory T cells, and plasmacytoid dendritic cells, independent of any association with GVHD. Peg-IFNα administration after relapse thus constitutes a logical strategy to invoke GVL effects and should be studied in a larger, multicenter cohort. This trial was registered at www.anzctr.org.au as #ACTRN12612000728831.
Collapse
|
24
|
Bezerra ED, Flowers ME, Onstad LE, Chielens D, Radich J, Higano CS. A phase 2 study of alpha interferon for molecularly measurable residual disease in chronic myeloid leukemia after allogeneic hematopoietic cell transplantation. Leuk Lymphoma 2019; 60:2754-2761. [PMID: 31014151 DOI: 10.1080/10428194.2019.1605508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
CML therapy has improved dramatically with the development of tyrosine kinase inhibitors (TKIs). Prior to the TKI era, we conducted two trials of alpha-interferon (IFN) for post-transplant hematologic and cytogenetic relapse. The complete cytogenetic response rate was 33% and 57% respectively. This report describes a third trial in which 40 patients with molecular relapse between 6 and 12 months post-transplant were treated with IFN. The projected cytogenetic relapse at 4.5 years was 12.6% compared with 42% in the historical control group. Although this data may not apply to most patients with CML today due to the availability of multiple TKIs, the effectiveness of short term IFN in post-transplant molecular relapse is supported by long-term treatment-free-survival in 75% of patients after a median follow-up of 15.6 years. This report suggests that alpha-interferon is potentially useful in the rare patient who has post-transplant molecular relapse who does not tolerate, or is resistant to TKIs.
Collapse
Affiliation(s)
- Evandro D Bezerra
- Department of Medicine Division of Oncology, University of Washington, Seattle, WA, USA
| | - Mary E Flowers
- Department of Medicine Division of Oncology, University of Washington, Seattle, WA, USA.,Clinical Research Division Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Lynn E Onstad
- Clinical Research Division Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Jerald Radich
- Department of Medicine Division of Oncology, University of Washington, Seattle, WA, USA.,Clinical Research Division Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Celestia S Higano
- Department of Medicine Division of Oncology, University of Washington, Seattle, WA, USA.,Clinical Research Division Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Seattle Cancer Care Alliance, Seattle, WA, USA
| |
Collapse
|
25
|
Suprunenko T, Hofer MJ. Complexities of Type I Interferon Biology: Lessons from LCMV. Viruses 2019; 11:v11020172. [PMID: 30791575 PMCID: PMC6409748 DOI: 10.3390/v11020172] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Abstract
Over the past decades, infection of mice with lymphocytic choriomeningitis virus (LCMV) has provided an invaluable insight into our understanding of immune responses to viruses. In particular, this model has clarified the central roles that type I interferons play in initiating and regulating host responses. The use of different strains of LCMV and routes of infection has allowed us to understand how type I interferons are critical in controlling virus replication and fostering effective antiviral immunity, but also how they promote virus persistence and functional exhaustion of the immune response. Accordingly, these discoveries have formed the foundation for the development of novel treatments for acute and chronic viral infections and even extend into the management of malignant tumors. Here we review the fundamental insights into type I interferon biology gained using LCMV as a model and how the diversity of LCMV strains, dose, and route of administration have been used to dissect the molecular mechanisms underpinning acute versus persistent infection. We also identify gaps in the knowledge regarding LCMV regulation of antiviral immunity. Due to its unique properties, LCMV will continue to remain a vital part of the immunologists' toolbox.
Collapse
Affiliation(s)
- Tamara Suprunenko
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Markus J Hofer
- School of Life and Environmental Sciences, the Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, and the Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
26
|
Hasselbalch HC, Holmström MO. Perspectives on interferon-alpha in the treatment of polycythemia vera and related myeloproliferative neoplasms: minimal residual disease and cure? Semin Immunopathol 2019; 41:5-19. [PMID: 30203226 PMCID: PMC6323070 DOI: 10.1007/s00281-018-0700-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 08/06/2018] [Indexed: 12/19/2022]
Abstract
The first clinical trials of the safety and efficacy of interferon-alpha2 (IFN-alpha2) were performed about 30 years ago. Since then, several single-arm studies have convincingly demonstrated that IFN-alpha2 is a highly potent anti-cancer agent in several cancer types but unfortunately not being explored sufficiently due to a high toxicity profile when using non-pegylated IFN-alpha2 or high dosages or due to competitive drugs, that for clinicians at first glance might look more attractive. Within the hematological malignancies, IFN-alpha2 has only recently been revived in patients with the Philadelphia-negative myeloproliferative neoplasms-essential thrombocytosis, polycythemia vera, and myelofibrosis (MPNs)-and in patients with chronic myelogenous leukemia (CML) in combination with tyrosine kinase inhibitors. In this review, we tell the IFN story in MPNs from the very beginning in the 1980s up to 2018 and describe the perspectives for IFN-alpha2 treatment of MPNs in the future. The mechanisms of actions are discussed and the impact of chronic inflammation as the driving force for clonal expansion and disease progression in MPNs is discussed in the context of combination therapies with potent anti-inflammatory agents, such as the JAK1-2 inhibitors (licensed only ruxolitinib) and statins as well. Interferon-alpha2 being the cornerstone treatment in MPNs and having the potential of inducing minimal residual disease (MRD) with normalization of the bone marrow and low-JAK2V617F allele burden, we believe that combination therapy with ruxolitinib may be even more efficacious and hopefully revert disease progression in many more patients to enter the path towards MRD. In patients with advanced and transforming disease towards leukemic transformation or having transformed to acute myeloid leukemia, "triple therapy" is proposed as a novel treatment modality to be tested in clinical trials combining IFN-alpha2, DNA-hypomethylator, and ruxolitinib. The rationale for this "triple therapy" is given, including the fact that even in AML, IFN-alpha2 as monotherapy may revert disease progression. We envisage a new and bright future with many more patients with MPNs obtaining MRD on the above therapies. From this stage-and even before-vaccination strategies may open a new horizon with cure being the goal for some patients.
Collapse
Affiliation(s)
- Hans Carl Hasselbalch
- Department of Hematology, Zealand University Hospital, Sygehusvej 10, 4000, Roskilde, Denmark.
| | - Morten Orebo Holmström
- Department of Hematology, Zealand University Hospital, Sygehusvej 10, 4000, Roskilde, Denmark
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, Herlev, Denmark
| |
Collapse
|
27
|
Zhang H, Liang Z, Hu Y, Wang X, Wang B, Huang H. The effectiveness of interferon-α combined with imatinib in patient with chronic myeloid leukemia harboring T315I BCR-ABL1 mutation. Leuk Lymphoma 2018; 59:3018-3019. [PMID: 29616854 DOI: 10.1080/10428194.2018.1443329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Hao Zhang
- a Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , China
| | - Zuyu Liang
- a Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , China
| | - Yongxian Hu
- a Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , China
| | - Xiujian Wang
- a Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , China
| | - Binsheng Wang
- a Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , China
| | - He Huang
- a Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , China
| |
Collapse
|
28
|
Bloch J, Spertini O, Stucki A, Solly F, Blum S. Over 20 years of treatment-free remission after interferon-alpha monotherapy for chronic myeloid leukemia. Leuk Res 2018; 73:103-104. [DOI: 10.1016/j.leukres.2018.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/07/2018] [Accepted: 09/10/2018] [Indexed: 11/16/2022]
|
29
|
Interferon-α-induced cytoplasmic MxA structures in hepatoma Huh7 and primary endothelial cells. Contemp Oncol (Pozn) 2018; 22:86-94. [PMID: 30150884 PMCID: PMC6103230 DOI: 10.5114/wo.2018.76149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 02/07/2023] Open
Abstract
Aim of the study Interferon (IFN)-α is now established as a treatment modality in various human cancers. The IFN-α-inducible human “myxovirus resistance protein A” (MxA) is a cytoplasmic dynamin-family large GTPase primarily characterized for its broad-spectrum antiviral activity and, more recently, for its anti-tumor and anti-metastasis effects. We characterized the association of IFN-α-induced MxA with cytoplasmic structures in human Huh7 cancer cells and in primary endothelial cells. Material and methods We re-evaluated the long-standing inference that MxA associated with the smooth ER using double-label immunofluorescence techniques and the ER structural protein RTN4 as a marker for smooth ER in IFN-α-treated cells. We also evaluated the relationship of exogenously expressed HA-MxA and GFP-MxA with mitochondria, and characterized cytoplasmic GFP-MxA structures using correlated light and electron microscopy (CLEM). Results and conclusions We discovered that IFN-α-induced endogenous MxA associated with variably-sized endosome-like and reticular cytoplasmic structures which were distinct from the ER. Thin-section EM studies of GFP-MxA expressing Huh7 cells showed that GFP-MxA formed variably-sized clusters of vesiculotubular elements to form endosome-like “MxA bodies”. Many of these clusters stretched out alongside cytoskeletal elements to give the appearance of a cytoplasmic “MxA reticulum”. This MxA meshwork was distinct from but adjacent to mitochondria. GFP-MxA expressing Huh7 cells showed reduced MitoTracker uptake and swollen mitochondria by thin-section EM. The new data identify cytoplasmic MxA structures as novel organelles, and suggest cross-talk between MxA structures and mitochondria that might account for the increased anti-tumoral efficacy of IFN-α combined with ligands that activate other pattern-sensing receptor pathways.
Collapse
|
30
|
Khouri R, Silva-Santos G, Dierckx T, Menezes SM, Decanine D, Theys K, Silva AC, Farré L, Bittencourt A, Mangino M, Roederer M, Vandamme AM, Van Weyenbergh J. A genetic IFN/STAT1/FAS axis determines CD4 T stem cell memory levels and apoptosis in healthy controls and Adult T-cell Leukemia patients. Oncoimmunology 2018; 7:e1426423. [PMID: 29721391 PMCID: PMC5927537 DOI: 10.1080/2162402x.2018.1426423] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 01/04/2018] [Accepted: 01/07/2018] [Indexed: 11/22/2022] Open
Abstract
Adult T-cell leukemia (ATL) is an aggressive, chemotherapy-resistant CD4+CD25+ leukemia caused by HTLV-1 infection, which usually develops in a minority of patients several decades after infection. IFN + AZT combination therapy has shown clinical benefit in ATL, although its mechanism of action remains unclear. We have previously shown that an IFN-responsive FAS promoter polymorphism in a STAT1 binding site (rs1800682) is associated to ATL susceptibility and survival. Recently, CD4 T stem cell memory (TSCM) Fashi cells have been identified as the hierarchical cellular apex of ATL, but a possible link between FAS, apoptosis, proliferation and IFN response in ATL has not been studied. In this study, we found significant ex vivo antiproliferative, antiviral and immunomodulatory effects of IFN-α treatment in short-term culture of primary mononuclear cells from ATL patients (n = 25). Bayesian Network analysis allowed us to integrate ex vivo IFN-α response with clinical, genetic and immunological data from ATL patients, thereby revealing a central role for FAS -670 polymorphism and apoptosis in the coordinated mechanism of action of IFN-α. FAS genotype-dependence of IFN-induced apoptosis was experimentally validated in an independent cohort of healthy controls (n = 20). The same FAS -670 polymorphism also determined CD4 TSCM levels in a genome-wide twin study (p = 7 × 10-11, n = 460), confirming a genetic link between apoptosis and TSCM levels. Transcriptomic analysis and cell type deconvolution confirmed the FAS genotype/TSCM link and IFN-α-induced downregulation of CD4 TSCM-specific genes in ATL patient cells. In conclusion, ex vivo IFN-α treatment exerts a pleiotropic effect on primary ATL cells, with a genetic IFN/STAT1/Fas axis determining apoptosis vs. proliferation and underscoring the CD4 TSCM model of ATL leukemogenesis.
Collapse
Affiliation(s)
- Ricardo Khouri
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
- Instituto Gonçalo Moniz (IGM) - Fundação Oswaldo Cruz (FIOCRUZ), Salvador-Bahia, Brazil
| | | | - Tim Dierckx
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | - Soraya Maria Menezes
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | - Daniele Decanine
- Instituto Gonçalo Moniz (IGM) - Fundação Oswaldo Cruz (FIOCRUZ), Salvador-Bahia, Brazil
| | - Kristof Theys
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | - Aline Clara Silva
- Instituto Gonçalo Moniz (IGM) - Fundação Oswaldo Cruz (FIOCRUZ), Salvador-Bahia, Brazil
| | - Lourdes Farré
- Instituto Gonçalo Moniz (IGM) - Fundação Oswaldo Cruz (FIOCRUZ), Salvador-Bahia, Brazil
| | - Achiléa Bittencourt
- Department of Pathology, Complexo Hospitalar Universitário Prof Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador-Bahia, Brazil
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King's College, London, UK
| | - Mario Roederer
- Immunotechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda-MD, USA
| | - Anne-Mieke Vandamme
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
- Center for Global Health and Tropical Medicine, Unidade de Microbiologia, Instituto de Highne e Medicina Tropical, Universidade, Nova de Lisboa, Lisbon, Portugal
| | - Johan Van Weyenbergh
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| |
Collapse
|
31
|
Atallah E, Ritchie E. Will tyrosine kinase inhibitors be part of the treatment armamentarium for CML in the future? Expert Opin Pharmacother 2018; 19:79-81. [PMID: 29308683 DOI: 10.1080/14656566.2017.1421633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Ehab Atallah
- a Division of Hematology/Oncology , Medical College of Wisconsin and Froedtert Hospital , Milwaukee , WI , USA
| | - Ellen Ritchie
- b Leukemia program , Weill Medical College of Cornell University and The New York Presbyterian Hospital , New York , NY , USA
| |
Collapse
|
32
|
Ren X, Xie W, Wang Y, Xu M, Liu F, Tang M, Li C, Wang M, Zhang J. VEGFR2-targeted fusion antibody improved NK cell-mediated immunosurveillance against K562 cells. Immunol Res 2017; 64:1060-70. [PMID: 27154226 DOI: 10.1007/s12026-016-8800-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
MHC class I polypeptide-related sequence A (MICA), which is normally expressed on cancer cells, activates NK cells via NK group 2-member D pathway. However, some cancer cells escape NK-mediated immune surveillance by shedding membrane MICA causing immune suppression. To address this issue, we designed an antibody-MICA fusion targeting tumor-specific antigen (vascular endothelial growth factor receptor 2, VEGFR2) based on our patented antibody (mAb04) against VEGFR2. In vitro results demonstrate that the fusion antibody retains both the antineoplastic and the immunomodulatory activity of mAb04. Further, we revealed that it enhanced NK-mediated immunosurveillance against K562 cells through increasing degranulation and cytokine production of NK cells. The overall data suggest our new fusion protein provides a promising approach for cancer-targeted immunotherapy and has prospects for potential application of chronic myeloid leukemia.
Collapse
Affiliation(s)
- Xueyan Ren
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Wei Xie
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Youfu Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Menghuai Xu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Fang Liu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Mingying Tang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Chenchen Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Min Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China.
| | - Juan Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
33
|
Gomez-de-León A, Gómez-Almaguer D, Ruiz-Delgado GJ, Ruiz-Arguelles GJ. Insights into the management of chronic myeloid leukemia in resource-poor settings: a Mexican perspective. Expert Rev Hematol 2017; 10:809-819. [PMID: 28742419 DOI: 10.1080/17474086.2017.1360180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The arrival of targeted therapy for chronic myeloid leukemia (CML) was revolutionary. However, due to the high cost of tyrosine kinase inhibitors, access to this highly effective therapy with strict monitoring strategies is limited in low to middle-income countries. In this context, following standard recommendations proposed by experts in developed countries is difficult. Areas covered: This review aims to provide an insight into the management of patients with CML living in a resource-limited setting. It addresses several issues: diagnosis, initial treatment, disease monitoring, and additional treatment alternatives including allogeneic hematopoietic stem cell transplantation. Expert commentary: Imatinib is probably the most cost-effective TKI for initial treatment in developing and underdeveloped countries. Generic imatinib preparations should be evaluated before considering their widespread use. Adherence to treatment should be emphasized. Adequate monitoring can be performed through several methods successfully and is important for predicting outcomes, particularly early in the first year, and if treatment suspension is being considered. Access to further therapeutic alternatives should define our actions after failure or intolerance to imatinib, preferring additional TKIs if possible. Allogeneic transplantation in chronic phase is a viable option in this context.
Collapse
Affiliation(s)
- Andrés Gomez-de-León
- a Universidad Autónoma de Nuevo León , Facultad de Medicina y Hospital Universitario "Dr.José Eleuterio González". Hematology Service, Monterrey , Nuevo León , México
| | - David Gómez-Almaguer
- a Universidad Autónoma de Nuevo León , Facultad de Medicina y Hospital Universitario "Dr.José Eleuterio González". Hematology Service, Monterrey , Nuevo León , México
| | | | | |
Collapse
|
34
|
Zhao B, Bhattacharya S, Yu Q, Fuchs SY. Expression of the IFNAR1 chain of type 1 interferon receptor in benign cells protects against progression of acute leukemia. Leuk Lymphoma 2017; 59:171-177. [PMID: 28503979 DOI: 10.1080/10428194.2017.1319053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Type I interferons (IFN) were widely used for leukemia treatment. These cytokines act on cell surface receptor consisting of the IFNAR1/2 chains to induce anti-tumorigenic effects. Given that levels of IFNAR1 can be regulated by phosphorylation-driven ubiquitination and degradation that undermines IFN signaling and anti-tumorigenic effects, we sought to determine the importance of IFNAR1 downregulation in progression of acute leukemia. Using knock-in mice deficient in downregulation of IFNAR1, we uncovered that IFNAR1 expression in stromal benign cells functions to protect against progression of leukemia. We discuss putative mechanisms of this regulation and potential of therapeutic targeting of IFNAR1 downregulation to treat leukemia.
Collapse
Affiliation(s)
- Bin Zhao
- a Department of Biomedical Sciences , Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Sabyasachi Bhattacharya
- a Department of Biomedical Sciences , Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Qiujing Yu
- a Department of Biomedical Sciences , Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Serge Y Fuchs
- a Department of Biomedical Sciences , Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
35
|
Manley PW, Stiefl NJ. Progress in the Discovery of BCR-ABL Kinase Inhibitors for the Treatment of Leukemia. TOPICS IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1007/7355_2017_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
36
|
da Cunha Vasconcelos F, Mauricio Scheiner MA, Moellman-Coelho A, Mencalha AL, Renault IZ, Rumjanek VM, Maia RC. Low ABCB1 and high OCT1 levels play a favorable role in the molecular response to imatinib in CML patients in the community clinical practice. Leuk Res 2016; 51:3-10. [DOI: 10.1016/j.leukres.2016.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/22/2016] [Accepted: 10/09/2016] [Indexed: 12/23/2022]
|
37
|
Saußele S, Richter J, Hochhaus A, Mahon FX. The concept of treatment-free remission in chronic myeloid leukemia. Leukemia 2016; 30:1638-47. [PMID: 27133824 PMCID: PMC4980559 DOI: 10.1038/leu.2016.115] [Citation(s) in RCA: 199] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/22/2016] [Accepted: 03/30/2016] [Indexed: 02/08/2023]
Abstract
The advent of tyrosine kinase inhibitors (TKI) into the management of patients with chronic myeloid leukemia (CML) has profoundly improved prognosis. Survival of responders is approaching that of the general population but lifelong treatment is still recommended. In several trials, TKI treatment has been stopped successfully in approximately half of the patients with deep molecular response. This has prompted the development of a new concept in the evaluation of CML patients known as 'treatment-free remission'. The future in CML treatment will be to define criteria for the safe and most promising discontinuation of TKI on one hand, and, on the other, to increase the number of patients available for such an attempt. Until safe criteria have been defined, discontinuation of therapy is still experimental and should be restricted to clinical trials or registries. This review will provide an overview of current knowledge as well as an outlook on future challenges.
Collapse
Affiliation(s)
- S Saußele
- III. Medizinische Klinik, Universitätsmedizin, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany
| | - J Richter
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - A Hochhaus
- Klinik für Innere Medizin II, Hämatologie/Onkologie, Universitätsklinikum Jena, Jena, Germany
| | - F-X Mahon
- Bergonié Cancer Institute, INSERM Unit 916, University of Bordeaux, Bordeaux, France
| |
Collapse
|
38
|
Takeishi S, Nakayama KI. To wake up cancer stem cells, or to let them sleep, that is the question. Cancer Sci 2016; 107:875-81. [PMID: 27116333 PMCID: PMC4946711 DOI: 10.1111/cas.12958] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 04/22/2016] [Indexed: 12/18/2022] Open
Abstract
Cancer stem cells (CSCs) generate transient-amplifying cells and thereby contribute to cancer propagation. A fuller understanding of the biological features of CSCs is expected to lead to the development of new anticancer therapies capable of eradicating this life-threatening disease. Cancer stem cells are known to maintain a non-proliferative state and to enter the cell cycle only infrequently. Given that conventional anticancer therapies preferentially target dividing cells, CSCs are resistant to such treatments, with those remaining after elimination of bulk cancer cells potentially giving rise to disease relapse and metastasis as they re-enter the cell cycle after a period of latency. Targeting of the switch between quiescence and proliferation in CSCs is therefore a potential strategy for preventing the reinitiation of malignancy, underscoring the importance of elucidation of the mechanisms by which these cells are maintained in the quiescent state. The fundamental properties of CSCs are thought to be governed cooperatively by internal molecules and cues from the external microenvironment (stem cell niche). Several such intrinsic and extrinsic regulators are responsible for the control of cell cycle progression in CSCs. In this review, we address two opposite approaches to the therapeutic targeting of CSCs - wake-up and hibernation therapies - that either promote or prevent the entry of CSCs into the cell cycle, respectively, and we discuss the potential advantages and risks of each strategy.
Collapse
Affiliation(s)
- Shoichiro Takeishi
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
39
|
Hackstein H, Tschipakow I, Bein G, Nold P, Brendel C, Baal N. Contact-dependent abrogation of bone marrow-derived plasmacytoid dendritic cell differentiation by murine mesenchymal stem cells. Biochem Biophys Res Commun 2016; 476:15-20. [PMID: 27233615 DOI: 10.1016/j.bbrc.2016.05.108] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 05/21/2016] [Indexed: 01/14/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) are rare central regulators of antiviral immunity and unsurpassed producers of interferon-α (IFN-α). Despite their crucial role as a link between innate and adaptive immunity, little is known about the modulation of pDC differentiation by other bone marrow (BM) cells. In this study, we investigated the modulation of pDC differentiation in Flt-3 ligand (Flt3L)-supplemented BM cultures, using highly purified mesenchymal stem cells (MSCs) that were FACS-isolated from murine BM based on surface marker expression and used after in vitro expansion. Initial analysis revealed an almost complete inhibition of BM-derived pDC expansion in the presence of >2% MSC. This inhibition was cell contact-dependent and soluble factor-independent, as indicated by trans-well experiments. The abrogation of functional pDC development by MSCs was confirmed after TLR9 stimulation, revealing a complete, contact-dependent suppression of the IFN-a producing capacity of pDCs in Flt3L MSC BM co-cultures. MSC selectively inhibited pDC development in contrast to myeloid DC development, as indicated by the significantly increased numbers of myeloid DC in Flt3L-supplemented BM cultures. The absence of significant MSC-mediated inhibitory effects on myeloid DC differentiation was confirmed by additional experiments in GM-CSF/IL-4-supplemented BM cultures. In summary, we describe a novel contact-dependent immunomodulatory mechanism of MSC that targets the BM-derived expansion of functional pDCs.
Collapse
Affiliation(s)
- Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Germany.
| | - Inna Tschipakow
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Germany
| | - Gregor Bein
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Germany
| | - Philipp Nold
- Department of Hematology, Oncology and Immunology, Philipps University Marburg, University Hospital Giessen und Marburg, Germany
| | - Cornelia Brendel
- Department of Hematology, Oncology and Immunology, Philipps University Marburg, University Hospital Giessen und Marburg, Germany
| | - Nelli Baal
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Germany
| |
Collapse
|
40
|
Rivero-Juarez A, Gonzalez R, Frias M, Manzanares-Martín B, Rodriguez-Cano D, Perez-Camacho I, Gordon A, Cuenca F, Camacho A, Pineda JA, Peña J, Rivero A. KIR2DS2 as predictor of thrombocytopenia secondary to pegylated interferon-alpha therapy. THE PHARMACOGENOMICS JOURNAL 2016; 17:360-365. [PMID: 26975229 DOI: 10.1038/tpj.2016.19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/22/2016] [Accepted: 01/27/2016] [Indexed: 12/28/2022]
Abstract
Our aim was to evaluate the killer cell immunoglobulin-like receptors (KIRs) as a marker for the development of thrombocytopenia secondary to Peg-interferon (IFN) therapy in a cohort of human immunodeficiency virus (HIV)/hepatitis C virus (HCV) co-infected patients. Patients were naive to HCV treatment, receiving a first course of Peg-IFN/Ribavirin combination therapy. Total platelet count (cells ml-1) was determined at each visit, determining platelet decline from baseline to weeks 1, 2, 4, 8 and 12 after starting therapy. The end point of the study was development of thrombocytopenia, defined as a platelet count of <1 50 000 cells ml-1. Fifty-eight HIV/HCV co-infected patients were included in the study, of whom 20 (34.4%) developed thrombocytopenia. The absence of KIR2DS2 was associated with higher and faster rate of thrombocytopenia (54.2% vs 22.5%; P=0.012; 6.6 vs 10.3 weeks; P=0.008). The absence of KIR2DS2 was associated with a greater decline in platelet count and development of thrombocytopenia during Peg-IFN treatment in HIV/HCV co-infected patients.
Collapse
Affiliation(s)
- A Rivero-Juarez
- Infectious Diseases Unit, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía de Córdoba, Universidad de Córdoba, Córdoba, Spain
| | - R Gonzalez
- Immunology Unit, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain
| | - M Frias
- Infectious Diseases Unit, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía de Córdoba, Universidad de Córdoba, Córdoba, Spain
| | - B Manzanares-Martín
- Immunology Unit, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain
| | - D Rodriguez-Cano
- Infectious Diseases Unit, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía de Córdoba, Universidad de Córdoba, Córdoba, Spain
| | - I Perez-Camacho
- Service de Medicina Tropical, Hospital de Poniente, El Ejido, Almería, Spain
| | - A Gordon
- Infectious Diseases Unit, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía de Córdoba, Universidad de Córdoba, Córdoba, Spain
| | - F Cuenca
- Infectious Diseases Unit, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía de Córdoba, Universidad de Córdoba, Córdoba, Spain
| | - A Camacho
- Infectious Diseases Unit, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía de Córdoba, Universidad de Córdoba, Córdoba, Spain
| | - J A Pineda
- Infectious Diseases and Clinical Microbiology Unit, Hospital Universitario de Valme, Seville, Spain
| | - J Peña
- Cellular Biology, Physiology and Immunology Department, Universidad de Córdoba, Córdoba, Spain
| | - A Rivero
- Infectious Diseases Unit, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía de Córdoba, Universidad de Córdoba, Córdoba, Spain
| |
Collapse
|
41
|
Stagno F, Stella S, Spitaleri A, Pennisi MS, Di Raimondo F, Vigneri P. Imatinib mesylate in chronic myeloid leukemia: frontline treatment and long-term outcomes. Expert Rev Anticancer Ther 2016; 16:273-8. [DOI: 10.1586/14737140.2016.1151356] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
42
|
Pediatric chronic myeloid leukemia is a unique disease that requires a different approach. Blood 2015; 127:392-9. [PMID: 26511135 DOI: 10.1182/blood-2015-06-648667] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/23/2015] [Indexed: 12/16/2022] Open
Abstract
Chronic myelogenous leukemia (CML) in children is relatively rare. Because of a lack of robust clinical study evidence, management of CML in children is not standardized and often follows guidelines developed for adults. Children and young adults tend to have a more aggressive clinical presentation than older adults, and prognostic scores for adult CML do not apply to children. CML in children has been considered to have the same biology as in adults, but recent data indicate that some genetic differences exist in pediatric and adult CML. Because children with CML may receive tyrosine kinase inhibitor (TKI) therapy for many decades, and are exposed to TKIs during a period of active growth, morbidities in children with CML may be distinct from those in adults and require careful monitoring. Aggressive strategies, such as eradication of CML stem cells with limited duration and intensive regimens of chemotherapy and TKIs, may be more advantageous in children as a way to avoid lifelong exposure to TKIs and their associated adverse effects. Blood and marrow transplantation in pediatric CML is currently indicated only for recurrent progressive disease, and the acute and long-term toxicities of this option should be carefully evaluated against the complications associated with lifelong use of TKIs.
Collapse
|
43
|
Effects of a Particular Heptapeptide on the IFN-α-Sensitive CML Cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:325026. [PMID: 26421285 PMCID: PMC4569753 DOI: 10.1155/2015/325026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 07/30/2015] [Accepted: 08/09/2015] [Indexed: 12/03/2022]
Abstract
Using the phage display biopanning technique, we have previously identified a heptapeptide KLWVIPQ which specifically binds to the surface of the IFN-α-sensitive but not the IFN-α-resistant CML cells. The effects of this heptapeptide on the IFN-α-sensitive CML cells were investigated in the present study. IFN-α-sensitive KT-1/A3 and IFN-α-resistant KT-1/A3R CML cells were transfected by pEGFP-KLWVIPQ expression vector and/or induced by IFN-α. WST-1 cell proliferation assay, flow cytometry, and western blotting were performed to determine the effects of this heptapeptide and/or IFN-α on CML cells. The viability of the KT-1/A3 cells was inhibited and apoptosis was induced by either expression of the heptapeptide KLWVIPQ or IFN-α treatment with concurrent upregulation of P53 and downregulation of P210bcr/abl. However, these effects were not observed in the IFN-α-resistant KT-1/A3R cells. These results suggest that the heptapeptide KLWVIPQ shares a similar mechanism with IFN-α in the regulation of CML cell growth and apoptosis, implying that the heptapeptide KLWVIPQ could be a novel target to go further into mechanisms of IFN-α sensitivity and/or resistance in CML.
Collapse
|