1
|
Schmidt D, Endres C, Hoefflin R, Andrieux G, Zwick M, Karantzelis N, Staehle HF, Vinnakota JM, Duquesne S, Mozaffari M, Pfeifer D, Becker H, Blazar BR, Zähringer A, Duyster J, Brummer T, Boerries M, Baumeister J, Shoumariyeh K, Li J, Green AR, Heidel FH, Tirosh I, Pahl HL, Leimkühler N, Köhler N, de Toledo MAS, Koschmieder S, Zeiser R. Oncogenic Calreticulin Induces Immune Escape by Stimulating TGFβ Expression and Regulatory T-cell Expansion in the Bone Marrow Microenvironment. Cancer Res 2024; 84:2985-3003. [PMID: 38885318 PMCID: PMC11405138 DOI: 10.1158/0008-5472.can-23-3553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Increasing evidence supports the interplay between oncogenic mutations and immune escape mechanisms. Strategies to counteract the immune escape mediated by oncogenic signaling could provide improved therapeutic options for patients with various malignancies. As mutant calreticulin (CALR) is a common driver of myeloproliferative neoplasms (MPN), we analyzed the impact of oncogenic CALRdel52 on the bone marrow (BM) microenvironment in MPN. Single-cell RNA sequencing revealed that CALRdel52 led to the expansion of TGFβ1-producing erythroid progenitor cells and promoted the expansion of FoxP3+ regulatory T cells (Treg) in a murine MPN model. Treatment with an anti-TGFβ antibody improved mouse survival and increased the glycolytic activity in CD4+ and CD8+ T cells in vivo, whereas T-cell depletion abrogated the protective effects conferred by neutralizing TGFβ. TGFβ1 reduced perforin and TNFα production by T cells in vitro. TGFβ1 production by CALRdel52 cells was dependent on JAK1/2, PI3K, and ERK activity, which activated the transcription factor Sp1 to induce TGFβ1 expression. In four independent patient cohorts, TGFβ1 expression was increased in the BM of patients with MPN compared with healthy individuals, and the BM of patients with MPN contained a higher frequency of Treg compared with healthy individuals. Together, this study identified an ERK/Sp1/TGFβ1 axis in CALRdel52 MPNs as a mechanism of immunosuppression that can be targeted to elicit T-cell-mediated cytotoxicity. Significance: Targeting the mutant calreticulin/TGFβ1 axis increases T-cell activity and glycolytic capacity, providing the rationale for conducting clinical trials on TGFβ antagonists as an immunotherapeutic strategy in patients with myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Dominik Schmidt
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Cornelia Endres
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Rouven Hoefflin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melissa Zwick
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Nikolaos Karantzelis
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Hans F. Staehle
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Janaki Manoja Vinnakota
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Sandra Duquesne
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Miriam Mozaffari
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Heiko Becker
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Bruce R. Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alexander Zähringer
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Justus Duyster
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tilman Brummer
- IMMZ, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg
| | - Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, and Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Khalid Shoumariyeh
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Juan Li
- Department of Hematology, University of Cambridge, Cambridge, UK
| | - Anthony R. Green
- Department of Hematology, University of Cambridge, Cambridge, UK
| | - Florian H. Heidel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany
- Leibniz Institute on Aging, Fritz-Lipmann Institute, Jena, Germany
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Heike L. Pahl
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Nils Leimkühler
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Germany
| | - Natalie Köhler
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg
| | - Marcelo A. S. de Toledo
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, and Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, and Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Robert Zeiser
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg
- Signalling Research Centres BIOSS and CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg
| |
Collapse
|
2
|
Sobas M, Ianotto JC, Kiladjian JJ, Harrison C. Myeloproliferative neoplasms: young patients, current data and future considerations. Ann Hematol 2024; 103:3287-3291. [PMID: 39110200 DOI: 10.1007/s00277-024-05920-8] [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: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024]
Abstract
The Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell disorders predominantly occurring in elderly, whereas in children and young adults are quite infrequent. Therefore, less is known about clinical presentation, genetic abnormalities, prognosis and best management strategies for this groups of patients. Currently, more cases of younger MPN patients are diagnosed. Nevertheless, diagnosis of MPNs, especially in childhood, may be difficult due to lower incidence of JAK2V617F and CALR mutations and differences in peripheral blood counts between adults and children. Challenges for younger MPN patients are longer life expectances, specific psychosocial need, fertility and pregnancy need and a long term therapy side effect (including second cancers). The most severe MPNs complication is transformation to secondary myelofibrosis (MF) or acute myeloid leukemia (AML). Optimal management of young MPNs remains a challenge as the classical risk scores fail in young MPNs. Moreover, the main objective of young MPNs therapy should be the disease outcome modification. Therefore, international collaborative work between pediatricians and "adult hematologists" is required to measure outcomes and generate protocol of management of young MPNs.
Collapse
Affiliation(s)
- Marta Sobas
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland.
| | - Jean-Christophe Ianotto
- Department of Hematology, Institute of Oncology and Hematology, University Hospital, Brest, France
| | - Jean-Jacques Kiladjian
- Assitance Publique - Hôpitaux de Paris, Hôpital Saint-Louis, Université de Paris, Inserm, Paris, France
| | - Claire Harrison
- Department of Hematology, Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| |
Collapse
|
3
|
Liu Q, Ma H. Cancer biotherapy: review and prospect. Clin Exp Med 2024; 24:114. [PMID: 38801637 PMCID: PMC11130057 DOI: 10.1007/s10238-024-01376-2] [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: 02/02/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Malignant tumors pose a grave threat to the quality of human life. The prevalence of malignant tumors in China is steadily rising. Presently, clinical interventions encompass surgery, radiotherapy, and pharmaceutical therapy in isolation or combination. Nonetheless, these modalities fail to completely eradicate malignant tumor cells, frequently leading to metastasis and recurrence. Conversely, tumor biotherapy has emerged as an encouraging fourth approach in preventing and managing malignant tumors owing to its safety, efficacy, and minimal adverse effects. Currently, a range of tumor biotherapy techniques are employed, including gene therapy, tumor vaccines, monoclonal antibody therapy, cancer stem cell therapy, cytokine therapy, and adoptive cellular immunotherapy. This study aims to comprehensively review the latest developments in biological treatments for malignant tumors.
Collapse
Affiliation(s)
- Qi Liu
- Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Thoracic Oncology, The Second Affiliated Hospital of Zunyi Medical University, Guizhou, 56300, Zunyi, China
| | - Hu Ma
- Zunyi Medical University, Zunyi, Guizhou, 563000, China.
- Department of Thoracic Oncology, The Second Affiliated Hospital of Zunyi Medical University, Guizhou, 56300, Zunyi, China.
| |
Collapse
|
4
|
Francis S, King T, Zeidler MP. Case report: Effectiveness of low-dose methotrexate monotherapy in post-essential thrombocythemia myelofibrosis. Front Med (Lausanne) 2024; 11:1285772. [PMID: 38698784 PMCID: PMC11063320 DOI: 10.3389/fmed.2024.1285772] [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: 08/30/2023] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
JAK/STAT pathway signalling is associated with both chronic inflammatory conditions such as psoriasis and haematological malignancies such as the myeloproliferative neoplasms (MPNs). Here we describe a 73yo female patient with a history of chronic plaque psoriasis, post-essential thrombocythemia myelofibrosis (MF) and a quality of life substantially impacted by both conditions. We report that 15 mg oral Methotrexate (MTX) weekly as a monotherapy is well tolerated, provides a substantial clinical improvement for both conditions and significantly improves quality of life. We suggest that the recently identified mechanism of action of MTX as a JAK inhibitor is likely to explain this efficacy and suggest that repurposing MTX for MPNs may represent a clinical- and cost-effective therapeutic option.
Collapse
Affiliation(s)
- Sebastian Francis
- Department of Haematology, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Tom King
- Department of Dermatology, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Martin P. Zeidler
- The Bateson Centre and the School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
5
|
Masarova L, Chifotides HT. SOHO State of the Art Update and Next Questions: Novel Therapies for Polycythemia Vera. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:141-148. [PMID: 38135633 DOI: 10.1016/j.clml.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023]
Abstract
In the recent years, landmark advancements in the treatment of polycythemia vera (PV) have been achieved. We witnessed the regulatory approval of ropeginterferon and the advanced clinical development of other novel agents that may affect the underlying pathophysiological mechanisms of the disease. Agents with the potential of disease modification may soon overtake preceding treatment options that were based on the patient's age and history of thrombosis. Recent studies using ropeginterferon in low-risk PV patients earlier in the disease course challenge the current treatment paradigm and shift the focus on modifying the course of the disease. Hepcidin mimetics offer an excellent alternative to phlebotomy, providing better quality of life, and may lead to improved outcomes in PV by tight hematocrit control. Novel agents, such as histone deacetylase inhibitors, hold promise to complement the therapeutic landscape of PV and might be particularly promising in rationale combinations. Ruxolitinib is well established as an approved second-line treatment for PV. In the frontline setting, the precise role of ruxolitinib, which also represents an appealing agent in combination regimens, will be determined in ongoing research studies. Longer follow-up is necessary to assess whether novel agents/regimens elicit fewer thromboembolic/ hemorrhagic events and halt disease progression to myelofibrosis and acute myeloid leukemia. We aspire that disease-modifying approaches in PV are on the horizon, and that we will be empowered to ultimately change the natural course of the disease and profoundly impact the lives of PV patients in the near future.
Collapse
Affiliation(s)
- Lucia Masarova
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030.
| | - Helen T Chifotides
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| |
Collapse
|
6
|
Vachhani P, Mascarenhas J, Bose P, Hobbs G, Yacoub A, Palmer JM, Gerds AT, Masarova L, Kuykendall AT, Rampal RK, Mesa R, Verstovsek S. Interferons in the treatment of myeloproliferative neoplasms. Ther Adv Hematol 2024; 15:20406207241229588. [PMID: 38380373 PMCID: PMC10878223 DOI: 10.1177/20406207241229588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/14/2023] [Indexed: 02/22/2024] Open
Abstract
Interferons are cytokines with immunomodulatory properties and disease-modifying effects that have been used to treat myeloproliferative neoplasms (MPNs) for more than 35 years. The initial use of interferons was limited due to difficulties with administration and a significant toxicity profile. Many of these shortcomings were addressed by covalently binding polyethylene glycol to the interferon structure, which increases the stability, prolongs activity, and reduces immunogenicity of the molecule. In the current therapeutic landscape, pegylated interferons are recommended for use in the treatment of polycythemia vera, essential thrombocythemia, and primary myelofibrosis. We review recent efficacy, molecular response, and safety data for the two available pegylated interferons, peginterferon alfa-2a (Pegasys) and ropeginterferon alfa-2b-njft (BESREMi). The practical management of interferon-based therapies is discussed, along with our opinions on whether to and how to switch from hydroxyurea to one of these therapies. Key topics and questions related to use of interferons, such as their safety and tolerability, the significance of variant allele frequency, advantages of early treatment, and what the future of interferon therapy may look like, will be examined. Pegylated interferons represent an important therapeutic option for patients with MPNs; however, more research is still required to further refine interferon therapy.
Collapse
Affiliation(s)
- Pankit Vachhani
- Hematology Oncology at The Kirklin Clinic of UAB Hospital, North Pavilion, Room 2540C, 1720 2 Ave S, Birmingham, AL 35294-3300, USA
| | - John Mascarenhas
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Prithviraj Bose
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriela Hobbs
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Abdulraheem Yacoub
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Cancer Center, Westwood, KS, USA
| | | | - Aaron T. Gerds
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Lucia Masarova
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew T. Kuykendall
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Raajit K. Rampal
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ruben Mesa
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Srdan Verstovsek
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
7
|
Pasquer H, Daltro de Oliveira R, Vasseur L, Soret-Dulphy J, Maslah N, Zhao LP, Marcault C, Cazaux M, Gauthier N, Verger E, Parquet N, Vainchenker W, Raffoux E, Ugo V, Luque Paz D, Roy L, Lambert WC, Ianotto JC, Lippert E, Giraudier S, Cassinat B, Kiladjian JJ, Benajiba L. Distinct clinico-molecular arterial and venous thrombosis scores for myeloproliferative neoplasms risk stratification. Leukemia 2024; 38:326-339. [PMID: 38148396 DOI: 10.1038/s41375-023-02114-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 12/28/2023]
Abstract
Current recommended risk scores to predict thrombotic events associated with myeloproliferative neoplasms (MPN) do not discriminate between arterial and venous thrombosis despite their different physiopathology. To define novel stratification systems, we delineated a comprehensive landscape of MPN associated thrombosis across a large long-term follow-up MPN cohort. Prior arterial thrombosis, age >60 years, cardiovascular risk factors and presence of TET2 or DNMT3A mutations were independently associated with arterial thrombosis in multivariable analysis. ARTS, an ARterial Thrombosis Score, based on these four factors, defined low- (0.37% patients-year) and high-risk (1.19% patients-year) patients. ARTS performance was superior to the two-tiered conventional risk stratification in our training cohort, across all MPN subtypes, as well as in two external validation cohorts. Prior venous thrombosis and presence of a JAK2V617F mutation with a variant allelic frequency ≥50% were independently associated with venous thrombosis. The discrimination potential of VETS, a VEnous Thrombosis Score based on these two factors, was poor, similar to the two-tiered conventional risk stratification. Our study pinpoints arterial and venous thrombosis clinico-molecular differences and proposes an arterial risk score for more accurate patients' stratification. Further improvement of venous risk scores, accounting for additional factors and considering venous thrombosis as a heterogeneous entity is warranted.
Collapse
Affiliation(s)
- Hélène Pasquer
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
- INSERM UMR 944, Institut de Recherche Saint-Louis, Paris, France
| | - Rafael Daltro de Oliveira
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Loic Vasseur
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Juliette Soret-Dulphy
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Nabih Maslah
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Lin-Pierre Zhao
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Clémence Marcault
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Marine Cazaux
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Nicolas Gauthier
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Emmanuelle Verger
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Nathalie Parquet
- Université Paris Cité, APHP, Hôpital Saint-Louis, Département d'hématologie et d'Immunologie, Paris, France
| | - William Vainchenker
- APHP, Hôpital Saint-Louis, Département d'hématologie et d'Immunologie, Paris, France
| | - Emmanuel Raffoux
- Université Paris Cité, APHP, Hôpital Saint-Louis, Département d'hématologie et d'Immunologie, Paris, France
| | - Valérie Ugo
- Univ Angers, Nantes Université, CHU Angers, Inserm, CNRS, CRCI2NA, Angers, France
| | - Damien Luque Paz
- Univ Angers, Nantes Université, CHU Angers, Inserm, CNRS, CRCI2NA, Angers, France
| | - Lydia Roy
- Université Paris Est Créteil, APHP, Hôpital Henri Mondor, Service d'hématologie, Créteil, France
| | - Wayne-Corentin Lambert
- Université de Bretagne Occidentale, CHU de Brest, Service d'Hématologie Biologique, Brest, France
| | - Jean-Christophe Ianotto
- Université de Bretagne Occidentale, CHU de Brest, Service d'Hématologie et d'Hémostase Clinique, Brest, France
| | - Eric Lippert
- Université de Bretagne Occidentale, CHU de Brest, Service d'Hématologie Biologique, Brest, France
| | - Stéphane Giraudier
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Bruno Cassinat
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Jean-Jacques Kiladjian
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Lina Benajiba
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France.
- INSERM UMR 944, Institut de Recherche Saint-Louis, Paris, France.
| |
Collapse
|
8
|
Ligia S, Scalzulli E, Carmosino I, Palumbo G, Molinari MC, Poggiali R, Costa A, Bisegna ML, Martelli M, Breccia M. Ropeginterferon alfa-2b treatment in a young patient with multi-refractory polycythemia vera and double JAK2 gene mutation: a case report. Front Oncol 2024; 13:1338417. [PMID: 38264737 PMCID: PMC10803620 DOI: 10.3389/fonc.2023.1338417] [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: 11/14/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
This case report presents a 3-year-old female patient initially diagnosed with polycythemia vera (PV) in 2001. The patient exhibited elevated red blood cell (RBC) counts, high hemoglobin (Hb) levels, hyperleukocytosis, and moderate thrombocytosis, with sporadic abdominal pain and significant splenomegaly. Despite various treatments, including phlebotomies, hydroxyurea, and alpha-interferon, the patient struggled to maintain optimal hematocrit levels and experienced persistent symptoms. Subsequent genomic analysis revealed a rare JAK2 G301R mutation alongside the canonical JAK2 V617F mutation, potentially contributing to disease severity. In 2023, the patient started Ropeginterferon alfa-2b, leading to improved hematological parameters and symptom relief. The case underscores the challenges in managing PV, particularly in young patients, and highlights the potential clinical significance of additional JAK2 mutations/variants and the potential benefits of Ropeginterferon alfa-2b in achieving better disease control.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Massimo Breccia
- Hematology, Department of Translational and Precision Medicine, Azienda Policlinico Umberto I - Sapienza University, Rome, Italy
| |
Collapse
|
9
|
de Castro FA, Mehdipour P, Chakravarthy A, Ettayebi I, Loo Yau H, Medina TS, Marhon SA, de Almeida FC, Bianco TM, Arruda AGF, Devlin R, de Figueiredo-Pontes LL, Chahud F, da Costa Cacemiro M, Minden MD, Gupta V, De Carvalho DD. Ratio of stemness to interferon signalling as a biomarker and therapeutic target of myeloproliferative neoplasm progression to acute myeloid leukaemia. Br J Haematol 2024; 204:206-220. [PMID: 37726227 DOI: 10.1111/bjh.19107] [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: 05/02/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/21/2023]
Abstract
Progression to aggressive secondary acute myeloid leukaemia (sAML) poses a significant challenge in the management of myeloproliferative neoplasms (MPNs). Since the physiopathology of MPN is closely linked to the activation of interferon (IFN) signalling and that AML initiation and aggressiveness is driven by leukaemia stem cells (LSCs), we investigated these pathways in MPN to sAML progression. We found that high IFN signalling correlated with low LSC signalling in MPN and AML samples, while MPN progression and AML transformation were characterized by decreased IFN signalling and increased LSC signature. A high LSC to IFN expression ratio in MPN patients was associated with adverse clinical prognosis and higher colony forming potential. Moreover, treatment with hypomethylating agents (HMAs) activates the IFN signalling pathway in MPN cells by inducing a viral mimicry response. This response is characterized by double-stranded RNA (dsRNA) formation and MDA5/RIG-I activation. The HMA-induced IFN response leads to a reduction in LSC signature, resulting in decreased stemness. These findings reveal the frequent evasion of viral mimicry during MPN-to-sAML progression, establish the LSC-to-IFN expression ratio as a progression biomarker, and suggests that HMAs treatment can lead to haematological response in murine models by re-activating dsRNA-associated IFN signalling.
Collapse
Affiliation(s)
- Fabíola Attié de Castro
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Parinaz Mehdipour
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | - Ankur Chakravarthy
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ilias Ettayebi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Helen Loo Yau
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Tiago Silva Medina
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Translational Immuno-Oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Sajid A Marhon
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Felipe Campos de Almeida
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto de Investigação em Imunologia, Institutos Nacionais de Ciência e Tecnologia (INCT-iii), Salvador, Brazil
| | - Thiago Mantello Bianco
- Hematology Division, Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Andrea G F Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Rebecca Devlin
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Lorena Lobo de Figueiredo-Pontes
- Hematology Division, Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Chahud
- Department of Pathology and Forensic Medicine, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maira da Costa Cacemiro
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Vikas Gupta
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Gill H, Leung GMK, Ooi MGM, Teo WZY, Wong CL, Choi CW, Wong GC, Lao Z, Rojnuckarin P, Castillo MRID, Xiao Z, Hou HA, Kuo MC, Shih LY, Gan GG, Lin CC, Chng WJ, Kwong YL. Management of classical Philadelphia chromosome-negative myeloproliferative neoplasms in Asia: consensus of the Asian Myeloid Working Group. Clin Exp Med 2023; 23:4199-4217. [PMID: 37747591 DOI: 10.1007/s10238-023-01189-9] [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] [Received: 03/18/2023] [Accepted: 09/04/2023] [Indexed: 09/26/2023]
Abstract
Myeloproliferative neoplasms (MPN) are a heterogeneous group of clonal hematopoietic stem cell disorders characterized clinically by the proliferation of one or more hematopoietic lineage(s). The classical Philadelphia-chromosome (Ph)-negative MPNs include polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). The Asian Myeloid Working Group (AMWG) comprises representatives from fifteen Asian centers experienced in the management of MPN. This consensus from the AMWG aims to review the current evidence in the risk stratification and treatment of Ph-negative MPN, to identify management gaps for future improvement, and to offer pragmatic approaches for treatment commensurate with different levels of resources, drug availabilities and reimbursement policies in its constituent regions. The management of MPN should be patient-specific and based on accurate diagnostic and prognostic tools. In patients with PV, ET and early/prefibrotic PMF, symptoms and risk stratification will guide the need for early cytoreduction. In younger patients requiring cytoreduction and in those experiencing resistance or intolerance to hydroxyurea, recombinant interferon-α preparations (pegylated interferon-α 2A or ropeginterferon-α 2b) should be considered. In myelofibrosis, continuous risk assessment and symptom burden assessment are essential in guiding treatment selection. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) in MF should always be based on accurate risk stratification for disease-risk and post-HSCT outcome. Management of classical Ph-negative MPN entails accurate diagnosis, cytogenetic and molecular evaluation, risk stratification, and treatment strategies that are outcome-oriented (curative, disease modification, improvement of quality-of-life).
Collapse
Affiliation(s)
- Harinder Gill
- Department of Medicine, LKS Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China.
- Department of Medicine, Professorial Block, Queen Mary Hospital, Pokfulam Road, Pok Fu Lam, Hong Kong, China.
| | - Garret M K Leung
- Department of Medicine, LKS Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - Melissa G M Ooi
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University, Singapore, Singapore
| | - Winnie Z Y Teo
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Fast and Chronic Program, Alexandra Hospital, Singapore, Singapore
| | - Chieh-Lee Wong
- Department of Medicine, Sunway Medical Centre, Shah Alam, Selangor, Malaysia
| | - Chul Won Choi
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Gee-Chuan Wong
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Zhentang Lao
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Ponlapat Rojnuckarin
- King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | | | - Zhijian Xiao
- Blood Disease Hospital and Institute of Hematology, Chinese Academy of Medical Sciences Peking Union Medical College, Tianjin, China
| | - Hsin-An Hou
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Chung Kuo
- Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Lee-Yung Shih
- Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Gin-Gin Gan
- University of Malaya, Kuala Lumpur, Malaysia
| | - Chien-Chin Lin
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wee-Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University, Singapore, Singapore
| | - Yok-Lam Kwong
- Department of Medicine, LKS Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| |
Collapse
|
11
|
Krecak I, Skelin M, Verstovsek S. Evaluating ropeginterferon alfa-2b for the treatment of adults with polycythemia vera. Expert Rev Hematol 2023; 16:305-316. [PMID: 37002907 DOI: 10.1080/17474086.2023.2199151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
INTRODUCTION Interferons (IFNs) have been used for decades to treat polycythemia vera (PV). Single-arm clinical trials assessing IFN in PV patients demonstrated high hematological and molecular response rates, indicating potential disease-modifying activity of IFN. However, discontinuation rates of IFNs have been rather high due to frequent treatment-related side-effects. AREAS COVERED Ropeginterferon alfa-2b (ROPEG) is a monopegylated IFN consisting of a single isoform, which differentiates it from previous IFNs with respect to tolerability and dosing frequency. ROPEG has improved pharmacokinetic and pharmacodynamic properties, which allow extended dosing every 2 weeks and monthly administration during maintenance phase. This review covers ROPEG's pharmacokinetic and pharmacodynamic properties, presents results of randomized clinical trials (RCT) that evaluated ROPEG in the treatment of PV patients, and discusses contemporary findings regarding the potential disease-modifying activity of ROPEG. EXPERT OPINION RCT have demonstrated high rates of hematological and molecular responses in PV patients treated with ROPEG, irrespective of thrombotic risk. Drug discontinuation rates were generally low. However, even though RCT captured the most important surrogate endpoints of thrombotic risk and disease progression in PV, they were not statistically powered to fully determine whether therapeutic intervention with ROPEG indeed has a direct positive effect on these important clinical outcomes.
Collapse
Affiliation(s)
- Ivan Krecak
- Department of Internal Medicine, General Hospital of Sibenik-Knin County, Sibenik, Croatia
- Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Marko Skelin
- Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Pharmacy Department, General Hospital of Šibenik-Knin County, Šibenik, Croatia
| | - Srdan Verstovsek
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
12
|
Puglianini OC, Peker D, Zhang L, Papadantonakis N. Essential Thrombocythemia and Post-Essential Thrombocythemia Myelofibrosis: Updates on Diagnosis, Clinical Aspects, and Management. Lab Med 2023; 54:13-22. [PMID: 35960786 DOI: 10.1093/labmed/lmac074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Although several decades have passed since the description of myeloproliferative neoplasms (MPN), many aspects of their pathophysiology have not been elucidated. In this review, we discuss the mutational landscape of patients with essential thrombocythemia (ET), prognostic scores and salient pathology, and clinical points. We discuss also the diagnostic challenges of differentiating ET from prefibrotic MF. We then focus on post-essential thrombocythemia myelofibrosis (post-ET MF), a rare subset of MPN that is usually studied in conjunction with post-polycythemia vera MF. The transition of ET to post-ET MF is not well studied on a molecular level, and we present available data. Patients with secondary MF could benefit from allogenic hematopoietic stem cell transplantation, and we present available data focusing on post-ET MF.
Collapse
Affiliation(s)
- Omar Castaneda Puglianini
- H. Lee Moffitt Cancer Center & Research Institute, Department of Blood & Marrow Transplant & Cellular Immunotherapy, Tampa, FL, USA
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Deniz Peker
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Linsheng Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Nikolaos Papadantonakis
- Winship Cancer Institute of Emory University, Department of Hematology and Medical Oncology, Atlanta, GA, USA
| |
Collapse
|
13
|
Real world experience with ropeginterferon alpha-2b (Besremi) in essential thrombocythaemia and polycythaemia vera following exposure to pegylated interferon alfa-2a (Pegasys). Leuk Res Rep 2022; 19:100360. [PMID: 36590864 PMCID: PMC9801096 DOI: 10.1016/j.lrr.2022.100360] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 11/25/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Despite widespread use of Pegylated forms of Inteferon in the management of Myeloproliferative Neoplasms (MPN), most clinicians have experience predominantly with peginterferon alfa-2a (Pegasys). Third generation pegylated IFNα, ropeginterferon alfa-2b (ropegIFN; Besremi), was recommended by the European Medicine Authority (EMA) for treatment of Polycythaemia Vera (PV) following a Phase III trial (PROUD-PV / CONTINUATION-PV). FDA approval for PV, regardless of treatment history, was subsequently granted in November 2021. We hereby demonstrate the safety and tolerability of ropegIFN in a series of MPN patients at variable doses. It corroborates reports of efficacy of ropegIFN in patients with PV and use in pregnancy.
Collapse
|
14
|
Jiang YZ, Wei ZL, Wang NN, Huang C, Huang J, Yan JW, Wang R, Yu ZZ, Huang DP. Clinical characteristics of a patient with de novo acute promyelocytic leukemia with JAK2 v617f mutation. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2022; 27:1290-1293. [PMID: 36476114 DOI: 10.1080/16078454.2022.2153200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The V617F mutation of Janus-associated kinase 2 (JAK2) is common in myeloproliferative neoplasms (MPN). JAK2 V617F mutation can be detected in patients with de novo acute myeloid leukemia (AML), but de novo acute promyelocytic leukemia (APL) with JAK2 V617F mutation is rare. CASE PRESENTATION We report a case of APL with both the t(15;17) translocation as well as the JAK2 V617F mutation that transformed into MPN (PV/ET). CONCLUSIONS A de novo APL patient presented initially with JAK2 V617F. After ATRA and ATO dual induction and chemotherapy consolidation, the patient achieved complete remission (CR) with undetectable PML/RARα. However, the JAK2 V617F remained positive, and the patient developed MPN (PV/ET) 22 months later, which responded well to interferon therapy.AML, acute myeloid leukemia; APL, acute promyelocytic leukemia; ATRA, all-trans retinoic acid; ATO, arsenic trioxide; BM, bone marrow; CR, complete remission; ET, essential thrombocythemia; Hb, hemoglobin; JAK2, Janus-associated kinase 2; MPN, myeloproliferative neoplasms; PLT, platelets; PMF, primary myelofibrosis; PML/RARα; PV, polycythemia vera; WBC, white blood cells.
Collapse
Affiliation(s)
- Yi-Zhi Jiang
- Department of Hematology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, People's Republic of China
| | - Zhong-Ling Wei
- Department of Hematology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, People's Republic of China
| | - Na-Na Wang
- Department of Hematology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, People's Republic of China
| | - Chen Huang
- Department of Hematology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, People's Republic of China
| | - Jun Huang
- Department of Hematology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, People's Republic of China
| | - Jia-Wei Yan
- Department of Hematology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, People's Republic of China
| | - Ran Wang
- Department of Hematology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, People's Republic of China
| | - Zheng-Zhi Yu
- Department of Hematology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, People's Republic of China
| | - Dong-Ping Huang
- Department of Hematology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, People's Republic of China
| |
Collapse
|
15
|
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: 2.0] [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
|
16
|
Mughal TI, Pemmaraju N, Bejar R, Gale RP, Bose P, Kiladjian JJ, Prchal J, Royston D, Pollyea D, Valent P, Brümmendorf TH, Skorski T, Patnaik M, Santini V, Fenaux P, Kucine N, Verstovsek S, Mesa R, Barbui T, Saglio G, Van Etten RA. Perspective: Pivotal translational hematology and therapeutic insights in chronic myeloid hematopoietic stem cell malignancies. Hematol Oncol 2022; 40:491-504. [PMID: 35368098 DOI: 10.1002/hon.2987] [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: 12/28/2021] [Revised: 02/21/2022] [Accepted: 03/03/2022] [Indexed: 11/10/2022]
Abstract
Despite much of the past 2 years being engulfed by the devastating consequences of the SAR-CoV-2 pandemic, significant progress, even breathtaking, occurred in the field of chronic myeloid malignancies. Some of this was show-cased at the 15th Post-American Society of Hematology (ASH) and the 25th John Goldman workshops on myeloproliferative neoplasms (MPN) held on 9th-10th December 2020 and 7th-10th October 2021, respectively. The inaugural Post-ASH MPN workshop was set out in 2006 by John Goldman (deceased) and Tariq Mughal to answer emerging translational hematology and therapeutics of patients with these malignancies. Rather than present a resume of the discussions, this perspective focuses on some of the pivotal translational hematology and therapeutic insights in these diseases.
Collapse
Affiliation(s)
- Tariq I Mughal
- Tufts University School of Medicine, Boston, Massachusetts, USA
- University of Buckingham, Buckingham, UK
| | - Naveen Pemmaraju
- MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Rafael Bejar
- University of California San Diego, La Jolla, California, USA
| | | | - Prithviraj Bose
- MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | | | - Josef Prchal
- Huntsman Cancer Center, Salt Lake City, Utah, USA
| | - Daniel Royston
- John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Daniel Pollyea
- University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Peter Valent
- Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Tomasz Skorski
- Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Valeria Santini
- Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Pierre Fenaux
- Hospital St Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | - Srdan Verstovsek
- MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Ruben Mesa
- Mays Cancer Center, UT Health San Antonio MD Anderson Cancer Center, San Antonio, Texas, USA
| | - Tiziano Barbui
- Fondazione per la Ricerca Ospedale Maggiore di Bergamo, Bergamo, Italy
| | | | - Richard A Van Etten
- Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| |
Collapse
|
17
|
Current and emerging therapies for neuroendocrine prostate cancer. Pharmacol Ther 2022; 238:108255. [DOI: 10.1016/j.pharmthera.2022.108255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022]
|
18
|
Stutz H. Advances and applications of electromigration methods in the analysis of therapeutic and diagnostic recombinant proteins – A Review. J Pharm Biomed Anal 2022; 222:115089. [DOI: 10.1016/j.jpba.2022.115089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022]
|
19
|
Verstovsek S, Komatsu N, Gill H, Jin J, Lee SE, Hou HA, Sato T, Qin A, Urbanski R, Shih W, Zagrijtschuk O, Zimmerman C, Mesa RA. SURPASS-ET: phase III study of ropeginterferon alfa-2b versus anagrelide as second-line therapy in essential thrombocythemia. Future Oncol 2022; 18:2999-3009. [PMID: 35924546 DOI: 10.2217/fon-2022-0596] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Patients diagnosed with high-risk essential thrombocythemia (ET) have limited treatment options to reduce the risk of thrombosis and lessen the progression of the disease by targeting the molecular source. Hydroxyurea is the recommended treatment, but many patients experience resistance or intolerance. Anagrelide is an approved second-line option for ET, but concerns of a higher frequency of disease transformation may affect its role as a suitable long-term option. Interferons have been evaluated in myeloproliferative neoplasms for over 30 years, but early formulations had safety and tolerability issues. SURPASS-ET (NCT04285086) is a phase III, open-label, multicenter, global, randomized, active-controlled trial that will evaluate the safety, efficacy, tolerability and pharmacokinetics of ropeginterferon alfa-2b compared with anagrelide as second-line therapy in high-risk ET.
Collapse
Affiliation(s)
- Srdan Verstovsek
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Norio Komatsu
- Department of Hematology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
- PharmaEssentia Japan KK, Tokyo, 107-0051, Japan
| | - Harinder Gill
- Department of Medicine, School of Clinical Medicine, the University of Hong Kong, Hong Kong SAR, China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Sung-Eun Lee
- Department of Hematology, Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, TX, 06591, Korea, Republic of (South) Korea
| | - Hsin-An Hou
- Department of Internal Medicine, Division of Hematology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | | | - Albert Qin
- PharmaEssentia Corp., Taipei, 115, Taiwan
| | | | - Weichung Shih
- Biostatistics School of Public Health, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | | | | | - Ruben A Mesa
- UT Health San Antonio Cancer Center, San Antonio, TX 78229, USA
| |
Collapse
|
20
|
Interferon-alpha2 treatment of patients with polycythemia vera and related neoplasms favorably impacts deregulation of oxidative stress genes and antioxidative defense mechanisms. PLoS One 2022; 17:e0270669. [PMID: 35771847 PMCID: PMC9246201 DOI: 10.1371/journal.pone.0270669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/14/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammation is considered a major driving force for clonal expansion and evolution in the Philadelphia-negative myeloproliferative neoplasms, which include essential thrombocythemia, polycythemia vera and primary myelofibrosis (MPNs). One of the key mutation drivers is the JAK2V617F mutation, which has been shown to induce the generation of reactive oxygen species (ROS). Using whole blood gene expression profiling, deregulation of several oxidative stress and anti-oxidative defense genes has been identified in MPNs, including significant downregulation of TP53, the NFE2L2 or NRF2 genes. These genes have a major role for maintaining genomic stability, regulation of the oxidative stress response and in modulating migration or retention of hematopoietic stem cells. Therefore, their deregulation might give rise to increasing genomic instability, increased chronic inflammation and disease progression with egress of hematopoietic stem cells from the bone marrow to seed in the spleen, liver and elsewhere. Interferon-alpha2 (rIFNα) is increasingly being recognized as the drug of choice for the treatment of patients with MPNs. Herein, we report the first gene expression profiling study on the impact of rIFNα upon oxidative stress and antioxidative defense genes in patients with MPNs (n = 33), showing that rIFNα downregulates several upregulated oxidative stress genes and upregulates downregulated antioxidative defense genes. Treatment with rIFNα induced upregulation of 19 genes in ET and 29 genes in PV including CXCR4 and TP53. In conclusion, this rIFNα- mediated dampening of genotoxic damage to hematopoietic cells may ultimately diminish the risk of additional mutations and accordingly clonal evolution and disease progression towards myelofibrotic and leukemic transformation.
Collapse
|
21
|
Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol 2022; 19:237-253. [PMID: 34997230 DOI: 10.1038/s41571-021-00588-9] [Citation(s) in RCA: 379] [Impact Index Per Article: 189.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 12/14/2022]
Abstract
During the past 40 years, cytokines and cytokine receptors have been extensively investigated as either cancer targets or cancer treatments. A strong preclinical rationale supports therapeutic strategies to enhance the growth inhibitory and immunostimulatory effects of interferons and interleukins, including IL-2, IL-7, IL-12 and IL-15, or to inhibit the inflammatory and tumour-promoting actions of cytokines such as TNF, IL-1β and IL-6. This rationale is underscored by the discovery of altered and dysregulated cytokine expression in all human cancers. These findings prompted clinical trials of several cytokines or cytokine antagonists, revealing relevant biological activity but limited therapeutic efficacy. However, most trials involved patients with advanced-stage disease, which might not be the optimal setting for cytokine-based therapy. The advent of more effective immunotherapies and an increased understanding of the tumour microenvironment have presented new approaches to harnessing cytokine networks in the treatment of cancer, which include using cytokine-based therapies to enhance the activity or alleviate the immune-related toxicities of other treatments as well as to target early stage cancers. Many challenges remain, especially concerning delivery methods, context dependencies, and the pleiotropic, redundant and often conflicting actions of many cytokines. Herein, we discuss the lessons learnt from the initial trials of single-agent cytokine-based therapies and subsequent efforts to better exploit such agents for the treatment of solid tumours.
Collapse
Affiliation(s)
- David J Propper
- Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Frances R Balkwill
- Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
22
|
Dholakia J, Cohen AC, Leath CA, Evans ET, Alvarez RD, Thaker PH. Development of Delivery Systems for Local Administration of Cytokines/Cytokine Gene-Directed Therapeutics: Modern Oncologic Implications. Curr Oncol Rep 2022; 24:389-397. [PMID: 35141857 PMCID: PMC10466172 DOI: 10.1007/s11912-022-01221-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW In this review, we discuss modern cytokine delivery systems in oncologic care, focusing on modalities being developed in the clinical trials or currently in use. These include pegylation, immune-cytokine drug conjugates, cytokine-expressing plasmid nanoparticles, nonviral cytokine nanoparticles, viral systems, and AcTakines. RECENT FINDINGS Cytokine therapy has the potential to contribute to cancer treatment options by modulating the immune system towards an improved antitumor response and has shown promise both independently and in combination with other immunotherapy agents. Despite promising preliminary studies, systemic toxicities and challenges with administration have limited the impact of unmodified cytokine therapy. In the last decade, novel delivery systems have been developed to address these challenges and facilitate cytokine-based oncologic treatments. Novel delivery systems provide potential solutions to decrease dose-limiting side effects, facilitate administration, and increase the therapeutic activity of cytokine treatments in oncology care. The expanding clinical and translational research in these systems provides an opportunity to augment the armamentarium of immune oncology and may represent the next frontier of cytokine-based immuno-oncology.
Collapse
Affiliation(s)
- Jhalak Dholakia
- Department of Obstetrics & Gynecology, University of Alabama Division of Gynecologic Oncology, 1700 6th Avenue South, Room 10250, Birmingham, AL, 35249-7333, USA.
| | - Alexander C Cohen
- Department of Obstetrics & Gynecology, Washington University in St. Louis Division of Gynecologic Oncology, St. Louis, MO, USA
| | - Charles A Leath
- Department of Obstetrics & Gynecology, University of Alabama Division of Gynecologic Oncology, 1700 6th Avenue South, Room 10250, Birmingham, AL, 35249-7333, USA
| | - Elizabeth T Evans
- Department of Obstetrics & Gynecology, University of Alabama Division of Gynecologic Oncology, 1700 6th Avenue South, Room 10250, Birmingham, AL, 35249-7333, USA
| | - Ronald D Alvarez
- Department of Obstetrics & Gynecology, Vanderbilt University Division of Gynecologic Oncology, Nashville, TN, USA
| | - Premal H Thaker
- Department of Obstetrics & Gynecology, Washington University in St. Louis Division of Gynecologic Oncology, St. Louis, MO, USA
| |
Collapse
|
23
|
Dias E, Liberal R, Costa-Moreira P, Príncipe F, Fonseca E, Macedo G. Primary Myelofibrosis in the Prefibrotic Stage Presenting as Portal, Splenic, and Superior Mesenteric Vein Thrombosis: A Case Report and Review of the Literature. GE PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2022; 29:125-131. [PMID: 35497670 PMCID: PMC8995663 DOI: 10.1159/000514658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/29/2020] [Indexed: 08/30/2023]
Abstract
INTRODUCTION Myeloproliferative neoplasms are the most common cause of splanchnic vein thrombosis in the absence of cirrhosis or nearby malignancy. CASE PRESENTATION A 31-year-old male presented to the emergency department with epigastric pain associated with mild thrombocytosis and elevated levels of aminotransferases, lactate dehydrogenase, and C-reactive protein. Contrast-enhanced abdominal computed tomography revealed splanchnic venous thrombosis that involved the portal, splenic, and superior mesenteric veins, without signs of chronic liver disease. Anticoagulation with warfarin was immediately started. Diagnostic work-up was remarkable for the presence of the JAK2 V617T mutation and hypercellular bone marrow, with increased myeloid cells and atypical megakaryocytes, consistent with primary myelofibrosis in a prefibrotic stage. No other hypercoagulable conditions were identified. DISCUSSION We present a rare case of primary myelofibrosis in the prefibrotic stage presenting as portal-splenic-superior mesenteric vein thrombosis. This demonstrates that extensive splanchnic vein thrombosis may be the onset manifestation of myeloproliferative neoplasms, even in early stages and in the absence of concomitant hypercoagulable conditions. The presence of the JAK2 mutation is an important prothrombotic risk factor that can, per se, contribute to large venous thrombosis.
Collapse
Affiliation(s)
- Emanuel Dias
- Gastroenterology Department, Centro Hospitalar de São João, Porto, Portugal
| | - Rodrigo Liberal
- Gastroenterology Department, Centro Hospitalar de São João, Porto, Portugal
| | | | - Fernando Príncipe
- Hematology Department, Centro Hospitalar de São João, Porto, Portugal
| | - Elsa Fonseca
- Pathology Department, Centro Hospitalar de São João, Porto, Portugal
| | - Guilherme Macedo
- Gastroenterology Department, Centro Hospitalar de São João, Porto, Portugal
| |
Collapse
|
24
|
Barbui T, Carobbio A, De Stefano V. Thrombosis in myeloproliferative neoplasms during cytoreductive and antithrombotic drug treatment. Res Pract Thromb Haemost 2022; 6:e12657. [PMID: 35155976 PMCID: PMC8822262 DOI: 10.1002/rth2.12657] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/31/2021] [Accepted: 01/12/2022] [Indexed: 12/16/2022] Open
Abstract
A state-of-the-art lecture titled "Myeloproliferative Neoplasm-associated Thrombosis" was presented at the ISTH congress in 2021. We summarize here the main points of the lecture with two purposes: to report the incidence rates of major thrombosis in polycythemia vera and essential thrombocythemia and to discuss to what extent cytoreductive therapy and antithrombotic drugs have reduced the incidence of these events. Unfortunately, the incidence rate of thrombosis remains high, ranging between 2 and 5/100 person-years. It is likely that new drugs such as interferon and ruxolitinib can be more efficacious given their cytoreductive and anti-inflammatory activities. Despite prophylaxis with vitamin K antagonists and direct oral anticoagulants after venous thrombosis in either common sites or splanchnic or cerebral sites, the incidence rate is still elevated, as high as 4 to 5/100 person-years. Future studies with new drugs or new strategies should consider thrombosis as the primary endpoint or surrogate biomarkers only if previously validated.
Collapse
Affiliation(s)
- Tiziano Barbui
- FROM Research FoundationPapa Giovanni XXIII HospitalBergamoItaly
| | | | - Valerio De Stefano
- Section of HematologyDepartment of Radiological and Hematological SciencesCatholic UniversityFondazione Policlinico A. Gemelli IRCCSRomeItaly
| |
Collapse
|
25
|
Vitiello GAF, Ferreira WAS, Cordeiro de Lima VC, Medina TDS. Antiviral Responses in Cancer: Boosting Antitumor Immunity Through Activation of Interferon Pathway in the Tumor Microenvironment. Front Immunol 2021; 12:782852. [PMID: 34925363 PMCID: PMC8674309 DOI: 10.3389/fimmu.2021.782852] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022] Open
Abstract
In recent years, it became apparent that cancers either associated with viral infections or aberrantly expressing endogenous retroviral elements (EREs) are more immunogenic, exhibiting an intense intra-tumor immune cell infiltration characterized by a robust cytolytic apparatus. On the other hand, epigenetic regulation of EREs is crucial to maintain steady-state conditions and cell homeostasis. In line with this, epigenetic disruptions within steady-state cells can lead to cancer development and trigger the release of EREs into the cytoplasmic compartment. As such, detection of viral molecules by intracellular innate immune sensors leads to the production of type I and type III interferons that act to induce an antiviral state, thus restraining viral replication. This knowledge has recently gained momentum due to the possibility of triggering intratumoral activation of interferon responses, which could be used as an adjuvant to elicit strong anti-tumor immune responses that ultimately lead to a cascade of cytokine production. Accordingly, several therapeutic approaches are currently being tested using this rationale to improve responses to cancer immunotherapies. In this review, we discuss the immune mechanisms operating in viral infections, show evidence that exogenous viruses and endogenous retroviruses in cancer may enhance tumor immunogenicity, dissect the epigenetic control of EREs, and point to interferon pathway activation in the tumor milieu as a promising molecular predictive marker and immunotherapy target. Finally, we briefly discuss current strategies to modulate these responses within tumor tissues, including the clinical use of innate immune receptor agonists and DNA demethylating agents.
Collapse
Affiliation(s)
| | - Wallax Augusto Silva Ferreira
- Translational Immuno-Oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SAMAM), Evandro Chagas Institute, Ananindeua, Brazil
| | | | - Tiago da Silva Medina
- Translational Immuno-Oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
- National Institute of Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo, Brazil
| |
Collapse
|
26
|
Hasselbalch HC, Silver RT. New Perspectives of Interferon-alpha2 and Inflammation in Treating Philadelphia-negative Chronic Myeloproliferative Neoplasms. Hemasphere 2021; 5:e645. [PMID: 34805764 PMCID: PMC8601345 DOI: 10.1097/hs9.0000000000000645] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
- Hans C Hasselbalch
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Richard T Silver
- Myeloproliferative Neoplasms Center, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
27
|
AIDS Related Kaposi's Sarcoma: A 20-Year Experience in a Clinic from the South-East of Romania. J Clin Med 2021; 10:jcm10225346. [PMID: 34830628 PMCID: PMC8620409 DOI: 10.3390/jcm10225346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Kaposi’s sarcoma (KS) was peculiarly described in the first notified cases of the acquired immunodeficiency syndrome as an opportunistic condition. However, the medical progress and the development of active antiretroviral therapy allowed the control of the HIV/AIDS epidemic, although the features of KS have changed throughout the past decades. The purpose of our study is to assess the epidemiological and clinical features of AIDS related KS in Romanian patients. A retrospective follow-up study was achieved in a single infectious diseases’ clinic from Galati—Romania, between 2001 and 2021. Referring to 290 new HIV diagnosed cases from our clinic retained in care, the prevalence of KS was 3.4%. The main characteristics of patients with KS are a median age of 33, a predominance of males, prevalent severe systemic forms of diseases, frequent association of past or concomitant tuberculosis, and context of immune reconstruction syndrome. The mortality rate was 70%. KS has occurred in patients with delayed HIV diagnoses and inadequate adherence to therapy. Early recognition of both infections, the close monitoring of latent or symptomatic tuberculosis, improving the antiretroviral adherence and raising the access to oncologic procedures in Romanian HIV patients could improve their prognosis related to KS.
Collapse
|
28
|
How J, Hobbs G. Management Issues and Controversies in Low-Risk Patients with Essential Thrombocythemia and Polycythemia Vera. Curr Hematol Malig Rep 2021; 16:473-482. [PMID: 34478054 DOI: 10.1007/s11899-021-00649-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Essential thrombocythemia (ET) and polycythemia vera (PV) are the most common myeloproliferative neoplasms (MPNs). Treatment of ET and PV is based on the risk for subsequent thrombosis. High-risk patients, defined as older than 60, JAK2 V617F-positive patients, or patients with a history of prior thrombosis, merit cytoreduction to control blood counts, whereas a watchful waiting paradigm is utilized in low-risk patients. However, low-risk patients have a host of other specific management issues that arise during their disease course. This review will discuss the most common management issues specific to the care of low-risk patients, including anti-platelet therapy dosing, pregnancy, and indications for early cytoreduction. RECENT FINDINGS Although low-dose aspirin is well established in PV, its indications and dosing regimens are less clear in ET. Recent evidence has supported twice daily low-dose aspirin in ET and observation alone in very low-risk ET patients. Pregnancy is not contraindicated in MPNs, and we recommend aspirin throughout pregnancy with consideration for prophylactic postpartum anticoagulation. High phlebotomy needs, symptom burden, and extreme thrombocytosis are common reasons for initiation of cytoreduction in low-risk patients, although we typically do not start cytoreduction for an isolated high platelet count alone. Recent data has also demonstrated a potential disease-modifying effect of interferons in MPNs, with some experts now advocating the early use of interferon in low-risk patients, although more mature data is needed before practice guidelines change. We evaluate the literature to inform clinical decision-making regarding these controversies, including most recent data that has challenged the "watchful waiting" paradigm. Our discussion provides guidance on common clinical scenarios seen in low-risk ET and PV patients, who face a myriad of complex management decisions in their care.
Collapse
Affiliation(s)
- Joan How
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Zero Emerson, Office 138, Boston, MA, 02114, USA.,Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Gabriela Hobbs
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Zero Emerson, Office 138, Boston, MA, 02114, USA.
| |
Collapse
|
29
|
Kiem D, Wagner S, Magnes T, Egle A, Greil R, Melchardt T. The Role of Neutrophilic Granulocytes in Philadelphia Chromosome Negative Myeloproliferative Neoplasms. Int J Mol Sci 2021; 22:ijms22179555. [PMID: 34502471 PMCID: PMC8431305 DOI: 10.3390/ijms22179555] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 11/25/2022] Open
Abstract
Philadelphia chromosome negative myeloproliferative neoplasms (MPN) are composed of polycythemia vera (PV), essential thrombocytosis (ET), and primary myelofibrosis (PMF). The clinical picture is determined by constitutional symptoms and complications, including arterial and venous thromboembolic or hemorrhagic events. MPNs are characterized by mutations in JAK2, MPL, or CALR, with additional mutations leading to an expansion of myeloid cell lineages and, in PMF, to marrow fibrosis and cytopenias. Chronic inflammation impacting the initiation and expansion of disease in a major way has been described. Neutrophilic granulocytes play a major role in the pathogenesis of thromboembolic events via the secretion of inflammatory markers, as well as via interaction with thrombocytes and the endothelium. In this review, we discuss the molecular biology underlying myeloproliferative neoplasms and point out the central role of leukocytosis and, specifically, neutrophilic granulocytes in this group of disorders.
Collapse
Affiliation(s)
- Dominik Kiem
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Sandro Wagner
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Teresa Magnes
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Alexander Egle
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria
| | - Richard Greil
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria
| | - Thomas Melchardt
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria
- Correspondence: ; Tel.: +43-57255-25801
| |
Collapse
|
30
|
Zhang M, Fu S, Ren D, Wu Y, Yao N, Ni T, Feng Y, Chen Y, Chen T, Zhao Y, Liu J. Maternal and Fetal Outcomes After Interferon Exposure During Pregnancy: A Systematic Review With Meta-Analysis. FRONTIERS IN REPRODUCTIVE HEALTH 2021; 3:702929. [PMID: 36303990 PMCID: PMC9580814 DOI: 10.3389/frph.2021.702929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/12/2021] [Indexed: 12/30/2022] Open
Abstract
Interferon (IFN) treatment is widely applied in viral hepatitis and multiple myeloproliferative diseases. However, there is considerable controversy on how to deal with unintended pregnancy during IFN treatment, even selective termination is suggested by hepatologists. To settle this clinical dilemma, we conducted a systematic review to retrieve all published articles involving IFN exposure during pregnancy up until March 31, 2021. Only 8 case reports that were relevant with outcomes of pregnant women with viral hepatitis exposed to IFN-α were retrieved, and 17 studies reporting pregnancy outcomes after exposure to type I IFNs involving 3,543 pregnancies were eligible for meta-analysis. No birth defect was reported in the case reports of pregnant women with viral hepatitis. The meta-analysis showed that risks of pregnancy outcomes and birth defects were not increased after exposure to IFN-α. Further comprehensive meta-analysis concerning the IFN-α and IFN-β exposure demonstrated that the risks of live birth (OR 0.89, 95% CI: 0.62-1.27), spontaneous abortion (OR 1.09, 95% CI: 0.73-1.63), stillbirth (OR 1.38, 95% CI: 0.51-3.72), preterm delivery (OR 1.24, 95% CI: 0.85-1.81), and maternal complications (OR 0.72, 95% CI: 0.38-1.38) were not increased in patients exposed to IFNs. The pooled estimates of live birth, spontaneous abortion, stillbirth, preterm delivery, and maternal complications were 85.2, 9.4, 0, 7.5, and 6.5%, respectively. Importantly, the risk of birth defects was not increased (OR 0.68, 95% CI: 0.39-1.20) after IFN exposure, with a pooled rate of 0.51%. Therefore, IFN exposure does not increase the prevalence of spontaneous abortion, stillbirth, preterm delivery, and birth defects. Clinical decision should be made after weighing up all the evidence.
Collapse
Affiliation(s)
- Mengmeng Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| | - Shan Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| | - Danfeng Ren
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| | - Yuchao Wu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| | - Naijuan Yao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| | - Tianzhi Ni
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| | - YaLi Feng
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| | - Yaolong Chen
- Evidence-Based Medicine Center, Basic Medical Sciences, Lanzhou University, Lanzhou, China
- WHO Collaborating Center for Guideline Implementation and Knowledge Translation, Lanzhou, China
| | - Tianyan Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| | - Yingren Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| | - Jinfeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
- Department of Infectious Diseases, Xi'an, China
| |
Collapse
|
31
|
Lee SE. Disease modifying agents of myeloproliferative neoplasms: a review. Blood Res 2021; 56:S26-S33. [PMID: 33935032 PMCID: PMC8093995 DOI: 10.5045/br.2021.2020325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 01/14/2023] Open
Abstract
The identification of driver mutations in Janus kinase (JAK) 2, calreticulin (CALR), and myeloproliferative leukemia (MPL) has contributed to a better understanding of disease pathogenesis by highlighting the importance of JAK signal transducer and activator of transcription (STAT) signaling in classical myeloproliferative neoplasms (MPNs). This has led to the therapeutic use of novel targeted treatments, such as JAK2 inhibitors. More recently, with the development of next-generation sequencing, additional somatic mutations, which are not restricted to MPNs, have been elucidated. Treatment decisions for MPN patients are influenced by the MPN subtype, symptom burden, and risk classification. Although prevention of vascular events is the main objective of therapy for essential thrombocythemia (ET) and polycythemia vera (PV) patients, disease-modifying drugs are needed to eradicate clonal hematopoiesis and prevent progression to more aggressive myeloid neoplasms. JAK inhibitors are a valuable therapeutic strategy for patients with myelofibrosis (MF) who have splenomegaly and/or disease-related symptoms, but intolerance, refractory, resistance, and disease progression still present challenges. Currently, allogeneic stem cell transplantation remains the only curative treatment for MF, but it is typically limited by age-related comorbidities and high treatment-related mortality. Therefore, a better understanding of the molecular pathogenesis and potential new therapies with the aim of modifying the natural history of the disease is important. In this article, I review the current understanding of the molecular basis of MPNs and clinical studies on potential disease-modifying agents.
Collapse
Affiliation(s)
- Sung-Eun Lee
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
32
|
Yoon SY, Won JH. The clinical role of interferon alpha in Philadelphia-negative myeloproliferative neoplasms. Blood Res 2021; 56:S44-S50. [PMID: 33935035 PMCID: PMC8093996 DOI: 10.5045/br.2021.2020334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell malignancies. Chronic inflammation and a dysregulated immune system are central to the pathogenesis and progression of MPNs. Interferon alpha (IFNα) was first used for the treatment of MPNs approximately 40 years ago. It has significant antiviral effects and plays a role in anti-proliferative, pro-apoptotic, and immunomodulatory responses. IFNα is an effective drug that can simultaneously induce significant rates of clinical, hematological, molecular, and histopathological responses, suggesting that the disease may be cured in some patients. However, its frequent dosage and toxicity profile are major barriers to its widespread use. Pegylated IFNα (peg-IFNα), and more recently, ropeginterferon alpha-2b (ropeg-IFNα-2b), are expected to overcome these drawbacks. The objective of this article is to discuss the clinical role of IFNα in Philadelphia-negative MPNs through a review of recent studies. In particular, it is expected that new IFNs, such as peg-IFNα and ropeg-IFNα-2b, with lower rates of discontinuation due to fewer adverse effects, will play important clinical roles.
Collapse
Affiliation(s)
- Seug Yun Yoon
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Jong-Ho Won
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| |
Collapse
|
33
|
Arya Y, Syal A, Gupta M, Gaba S. Advances in the Treatment of Polycythemia Vera: Trends in Disease Management. Cureus 2021; 13:e14193. [PMID: 33936902 PMCID: PMC8084584 DOI: 10.7759/cureus.14193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 01/10/2023] Open
Abstract
Treatment modalities for polycythemia vera (PV) have evolved over time. Phlebotomy and low-dose aspirin suffice in low-risk patients, but cytoreductive therapies are indicated in all high-risk patients (age ≥ 65 years or those with a history of PV-related thrombotic event) and may be considered for low-risk patients with progressively increasing splenomegaly, progressively increasing leucocyte and platelet counts, and for those who do not tolerate phlebotomy. Hydroxyurea/hydroxycarbamide or interferons can be used as first-line drugs. Hydroxyurea may not be tolerated by some patients, and it also carries risk of myelosuppression. Interferon alfa is especially useful for PV symptoms, and the newer preparation, ropeginterferon alfa-2b, has lesser incidence of flu-like reactions. Ruxolitinib reduces the JAK2V617F mutation burden and is used as a second-line drug. Anagrelide reduces platelet production and can be used in conjunction with hydroxyurea in patients with excessive thrombocytosis. The alkylating agent, busulfan, can also be used as a last resort in patients with a limited life expectancy. Prospective future treatments include givinostat, a histone deacetylase inhibitor, and idasanutlin, a murine double minute 2 antagonist.
Collapse
Affiliation(s)
- Yajur Arya
- Internal Medicine, Government Medical College and Hospital, Chandigarh, Chandigarh, IND
| | - Arshi Syal
- Internal Medicine, Government Medical College and Hospital, Chandigarh, Chandigarh, IND
| | - Monica Gupta
- Internal Medicine, Government Medical College and Hospital, Chandigarh, Chandigarh, IND
| | - Saurabh Gaba
- Internal Medicine, Government Medical College and Hospital, Chandigarh, Chandigarh, IND
| |
Collapse
|
34
|
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: 3] [Impact Index Per Article: 1.0] [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
|
35
|
Sharma V, Wright KL, Epling-Burnette PK, Reuther GW. Metabolic Vulnerabilities and Epigenetic Dysregulation in Myeloproliferative Neoplasms. Front Immunol 2020; 11:604142. [PMID: 33329600 PMCID: PMC7734315 DOI: 10.3389/fimmu.2020.604142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/02/2020] [Indexed: 01/14/2023] Open
Abstract
The Janus kinase 2 (JAK2)-driven myeloproliferative neoplasms (MPNs) are associated with clonal myelopoiesis, elevated risk of death due to thrombotic complications, and transformation to acute myeloid leukemia (AML). JAK2 inhibitors improve the quality of life for MPN patients, but these approved therapeutics do not readily reduce the natural course of disease or antagonize the neoplastic clone. An understanding of the molecular and cellular changes requisite for MPN development and progression are needed to develop improved therapies. Recently, murine MPN models were demonstrated to exhibit metabolic vulnerabilities due to a high dependence on glucose. Neoplastic hematopoietic progenitor cells in these mice express elevated levels of glycolytic enzymes and exhibit enhanced levels of glycolysis and oxidative phosphorylation, and the disease phenotype of these MPN model mice is antagonized by glycolytic inhibition. While all MPN-driving mutations lead to aberrant JAK2 activation, these mutations often co-exist with mutations in genes that encode epigenetic regulators, including loss of function mutations known to enhance MPN progression. In this perspective we discuss how altered activity of epigenetic regulators (e.g., methylation and acetylation) in MPN-driving stem and progenitor cells may alter cellular metabolism and contribute to the MPN phenotype and progression of disease. Specific metabolic changes associated with epigenetic deregulation may identify patient populations that exhibit specific metabolic vulnerabilities that are absent in normal hematopoietic cells, and thus provide a potential basis for the development of more effective personalized therapeutic approaches.
Collapse
Affiliation(s)
- Vasundhara Sharma
- Department of Leukemia, Princess Margaret Cancer Center-University Health Network, Toronto, ON, Canada
| | - Kenneth L Wright
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | | | - Gary W Reuther
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
36
|
Schrickel L, Heidel FH, Sadjadian P, Becker T, Kolatzki V, Hochhaus A, Griesshammer M, Wille K. Interferon alpha for essential thrombocythemia during 34 high-risk pregnancies: outcome and safety. J Cancer Res Clin Oncol 2020; 147:1481-1491. [PMID: 33140210 DOI: 10.1007/s00432-020-03430-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/08/2020] [Indexed: 11/27/2022]
Abstract
PURPOSE Pregnancies in women with essential thrombocythemia (ET) are at a higher risk for obstetrical complications. Acetylsalicylic acid (ASA) and low-molecular weight heparin (LMWH) are common options to prevent miscarriages and maternal complications, whereas interferon alpha (IFN) seems to be the cytoreductive therapy of choice. This retrospective study analyzes the largest number of IFN pregnancies to date in terms of outcome and safety. METHODS Data of 34 high-risk pregnancies in 23 women presenting at the University hospitals of Minden and Jena from 01-Jun-2007 to 01-Jun-2020 were collected. Reasons defining high-risk ET pregnancy in all 23 patients were: Thrombosis (n = 9) or severe hemorrhage (n = 2) in history, platelet count ≥ 1500 × 103/µl (n = 8) or severe microcirculatory disturbances not completely responding to ASA (n = 4). RESULTS Without the use of IFN, live birth rate was 60% (6/10), however, after the use of IFN live birth rate increased to 73.5% (25/34 pregnancies). Nine pregnancies ended in miscarriages (9/34; 26.5%); all of them spontaneous abortions. Live birth rate significantly improved with ASA (90% versus 50%, p = 0.0168), however, if ASA and LMWH was added (n = 14), live birth rate was 100%. IFN compound (PEGylated versus standard IFN) and JAK2-driver mutation had no impact on pregnancy outcome. One major maternal complication occurred as a major peripartal bleeding after abortion curettage. CONCLUSION IFN was associated with an encouraging live birth rate of 73.5% with no fatal maternal events and manageable side effects.
Collapse
Affiliation(s)
- Lukas Schrickel
- University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care, Johannes Wesling Medical Center Minden, UKRUB, Ruhr-University Bochum, Minden, Germany.
| | - Florian H Heidel
- Department of Internal Medicine 2, Haematology and Oncology, University Hospital Jena, Jena, Germany.,Internal Medicine C, University Medicine Greifswald, Greifswald, Germany
| | - Parvis Sadjadian
- University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care, Johannes Wesling Medical Center Minden, UKRUB, Ruhr-University Bochum, Minden, Germany
| | - Tatjana Becker
- University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care, Johannes Wesling Medical Center Minden, UKRUB, Ruhr-University Bochum, Minden, Germany
| | - Vera Kolatzki
- University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care, Johannes Wesling Medical Center Minden, UKRUB, Ruhr-University Bochum, Minden, Germany
| | - Andreas Hochhaus
- Department of Internal Medicine 2, Haematology and Oncology, University Hospital Jena, Jena, Germany
| | - Martin Griesshammer
- University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care, Johannes Wesling Medical Center Minden, UKRUB, Ruhr-University Bochum, Minden, Germany
| | - Kai Wille
- University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care, Johannes Wesling Medical Center Minden, UKRUB, Ruhr-University Bochum, Minden, Germany
| | | |
Collapse
|
37
|
Bewersdorf JP, Zeidan AM. Novel and combination therapies for polycythemia vera and essential thrombocythemia: the dawn of a new era. Expert Rev Hematol 2020; 13:1189-1199. [PMID: 33076714 DOI: 10.1080/17474086.2020.1839887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Essential thrombocythemia (ET) and polycythemia vera (PV) belong to the BCR-ABL1-negative myeloproliferative neoplasms and are characterized by the clonal proliferation of hematopoietic stem and progenitor cells. The contribution of aberrant immune regulation within the bone marrow microenvironment to ET and PV pathogenesis as well as the underlying molecular landscape is becoming increasingly understood. AREAS COVERED Authors searched PubMed and conference abstracts in August 2020 for preclinical and clinical studies to provide an overview of the immune pathobiology in ET and PV and the rationale for several novel agents. A discussion of recent clinical trials on interferon and ruxolitinib in ET and PV patients is provided followed by an outline of the future challenges in the field particularly for novel therapeutics and an increasingly individualized, molecularly driven approach to treatment selection. Several novel agents are currently being actively evaluated and are reviewed herein as well. EXPERT OPINION While hydroxyurea remains the first-line treatment for cytoreduction in most high-risk ET and PV patients, the disease-modifying potential of IFN is promising and could make it a preferred option for selected patients. Advances in molecular testing will enable a more individualized approach to prognostication and treatment selection.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Yale School of Medicine and Yale Cancer Center , New Haven, CT, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Yale School of Medicine and Yale Cancer Center , New Haven, CT, USA.,Cancer Outcomes, Public Policy and Effectiveness Research (COPPER) Center, Yale Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center , New Haven, CT, USA
| |
Collapse
|