1
|
Carturan A, Morè S, Poloni A, Rupoli S, Morsia E. Shaping the Future of Myeloproliferative Neoplasm Therapy: Immune-Based Strategies and Targeted Innovations. Cancers (Basel) 2024; 16:4113. [PMID: 39682299 DOI: 10.3390/cancers16234113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Numerous cutting-edge immunotherapy approaches have been developed for hematological malignancies, such as immune-checkpoint inhibitors for lymphomas, chimeric antigen receptor (CAR)-T-cell treatments for B-cell cancers, and monoclonal antibody therapies for acute myeloid leukemia (AML). However, achieving similar breakthroughs in MPNs has proven challenging. The key obstacles include the absence of universally expressed and MPN-specific surface markers, significant cellular and molecular variability among both individual patients and across different MPN subtypes, and the failure of treatments to stimulate an anti-tumor immune response due to the immune system disruptions caused by the myeloid neoplasm. Currently, there are several innovative therapies in clinical trials for MPNs. These include new JAK inhibitors with greater specificity for JAK2, as well as "add-on" medications designed to enhance the effectiveness of ruxolitinib, in both patients who are new to the drug and in those who have shown suboptimal responses. Additionally, there is ongoing exploration of novel therapeutic targets. In this review, we will explore the immunotherapy approaches that are currently used in clinical practice for MPNs, as well as emerging strategies that are likely to change the treatment of these diseases in the coming years.
Collapse
Affiliation(s)
- Alberto Carturan
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Hematology and Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sonia Morè
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60126 Ancona, Italy
- Hematology Clinic, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Antonella Poloni
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60126 Ancona, Italy
- Hematology Clinic, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Serena Rupoli
- Hematology Clinic, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Erika Morsia
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60126 Ancona, Italy
- Hematology Clinic, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| |
Collapse
|
2
|
Holmström MO, Ruders JH, Riley CH, Larsen MK, Grauslund JH, Kjær L, Skov V, Ellervik C, Guo BB, Linden M, Hasselbalch HC, Andersen MH. The CALR mutations enhance the expression of the immunosuppressive proteins GARP and LAP on peripheral blood lymphocytes through increased binding of activated platelets. Br J Haematol 2024; 205:1417-1429. [PMID: 39161981 DOI: 10.1111/bjh.19711] [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: 07/02/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
Recently, an antibody which inhibits the glycoprotein A repetitions predominant (GARP)-mediated release of active transforming growth factor beta (TGFβ) from the TGFβ propeptide latency-associated peptide (LAP) showed preclinical activity in a murine model of the chronic myeloproliferative neoplasms (MPN). Consequently, we investigated the expression of the immunosuppressive molecules LAP and GARP on peripheral blood lymphocytes from 56 MPN patients and 11 healthy donors (HD). We found that lymphocytes from patients with MPN express higher levels of LAP and GARP with no strong differences found between the different MPN diagnoses. The impact of clinical parameters on the expression of LAP and GARP by lymphocytes showed that patients with calreticulin (CALR)mut MPN have increased expression compared with HD and patients with the Januskinase2 (JAK2) mutation. The fraction of lymphocytes bound to activated platelets (aPLT) strongly correlate to LAP and GARP expression suggesting that it is not the lymphocytes themselves but aPLT, which confer the increased expression of GARP and LAP on MPN patient lymphocytes. Notably, no differences in neither platelet counts nor anti-thrombotic therapy was identified between patients with JAK2- and CALRmut patients. Analysis of platelet gene expression failed to identify differences in expression of relevant genes between JAK2- and CALRmut patients.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
| | - Josephine Hallundbæk Ruders
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
| | | | | | - Jacob Handlos Grauslund
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
| | - Lasse Kjær
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Department of Clinical Biochemistry, Zealand University Hospital, Koege, Denmark
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Belinda B Guo
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Matthew Linden
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | | | - Mads Hald Andersen
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Tavernari L, Rontauroli S, Norfo R, Mirabile M, Maccaferri M, Mora B, Genovese E, Parenti S, Carretta C, Bianchi E, Bertesi M, Pedrazzi F, Tenedini E, Martinelli S, Bochicchio MT, Guglielmelli P, Potenza L, Lucchesi A, Passamonti F, Tagliafico E, Luppi M, Vannucchi AM, Manfredini R. Targeting exhausted cytotoxic T cells through CTLA-4 inhibition promotes elimination of neoplastic cells in human myelofibrosis xenografts. Am J Hematol 2024; 99:1939-1950. [PMID: 38953347 DOI: 10.1002/ajh.27428] [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: 03/28/2024] [Revised: 05/30/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Myeloproliferative neoplasms represent a group of clonal hematopoietic disorders of which myelofibrosis (MF) is the most aggressive. In the context of myeloid neoplasms, there is a growing recognition of the dysregulation of immune response and T-cell function as significant contributors to disease progression and immune evasion. We investigated cytotoxic T-cell exhaustion in MF to restore immune response against malignant cells. Increased expression of inhibitory receptors like CTLA-4 was observed on cytotoxic T cells from MF patients together with a reduced secretion of IFNɣ and TNFɑ. CTLA-4 ligands CD80 and CD86 were increased on MF granulocytes and monocytes highlighting a possible role for myeloid cells in suppressing T-cell activation in MF patients. Unlike healthy donors, the activation of cytotoxic T cells from MF patients was attenuated in the presence of myeloid cells and restored when T cells were cultured alone or treated with anti-CTLA-4. Moreover, anti-CTLA-4 treatment promoted elimination of neoplastic monocytes and granulocytes in a co-culture system with cytotoxic T cells. To test CTLA-4 inhibition in vivo, patient-derived xenografts were generated by transplanting MF CD34+ cells and by infusing homologous T cells in NSGS mice. CTLA-4 blockade reduced human myeloid chimerism and led to T-cell expansion in spleen and bone marrow. Overall, these findings shed light on T-cell dysfunction in MF and suggest that CTLA-4 blockade can boost the cytotoxic T cell-mediated immune response against tumor cells.
Collapse
Affiliation(s)
- Lara Tavernari
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sebastiano Rontauroli
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Ruggiero Norfo
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Margherita Mirabile
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Monica Maccaferri
- Department Oncology and Hematology, Hematology Unit, Modena University Hospital, Modena, Italy
| | | | - Elena Genovese
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sandra Parenti
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Carretta
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Bianchi
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Bertesi
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Pedrazzi
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Tenedini
- Department of Laboratory Medicine, Diagnostic Hematology and Clinical Genomics Unit, Modena University Hospital, Modena, Italy
| | - Silvia Martinelli
- Department of Laboratory Medicine, Diagnostic Hematology and Clinical Genomics Unit, Modena University Hospital, Modena, Italy
| | - Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paola Guglielmelli
- CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, AOU Careggi, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Leonardo Potenza
- Department Oncology and Hematology, Hematology Unit, Modena University Hospital, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, Modena, Italy
| | - Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | | | - Enrico Tagliafico
- Department of Laboratory Medicine, Diagnostic Hematology and Clinical Genomics Unit, Modena University Hospital, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, Modena, Italy
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Luppi
- Department Oncology and Hematology, Hematology Unit, Modena University Hospital, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, Modena, Italy
| | - Alessandro Maria Vannucchi
- CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, AOU Careggi, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Rossella Manfredini
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
4
|
Ibrahim M, Murphree C, Patel K, Mastrodomenico M, Saba NS, Safah H, Schmid J, Socola F. Programmed death-ligand 1 expression in patients with primary or secondary myelofibrosis. Cancer Rep (Hoboken) 2024; 7:e2054. [PMID: 39233645 PMCID: PMC11375330 DOI: 10.1002/cnr2.2054] [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: 10/08/2023] [Revised: 03/06/2024] [Accepted: 03/09/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND It has been described in mice models that myeloproliferative neoplasm (MPN) with JAK2-V617F mutation has an increased expression of programmed death-ligand 1 (PD-L1) in megakaryocytes leading to cancer immune evasion by inhibiting the T-lymphocytes. AIMS To quantify and compare the PD-L1 expression on bone marrow (BM) of patients with MPN JAK2 positive, negative, and normal controls. METHODS We collected BM of patients with MPN JAK2 positive, negative and normal controls from 1990 to 2019. We also created a scoring system to quantify PD-L1 expression in megakaryocytes. RESULTS We obtained 14 BM with JAK2 positive PMF, 5 JAK2 negative PMF, and 10 patients with normal BM biopsies. PD-L1 expression was higher in the JAK2 positive group compared with the control group with a score of 212.6 versus 121.1 (t-value 2.05, p-value 0.025). In addition, the score was higher in the PMF group regardless of JAK2 mutational status when compared with the control group with score of 205.9 versus 121.1 (t-value 2.12, p-value 0.021). There was no difference in the PD-L1 score between the JAK2 negative versus the control group 187.2 versus 121.1 (t-value 1.02, p-value 0.162). CONCLUSION These findings suggest that PMF patients with a JAK2 mutation have a higher PD-L1 expression in megakaryocytes compared with the control group. We postulate that the combination of checkpoint and JAK2 inhibitors may be an active treatment option in JAK2 mutated PMF given the higher PD-L1 expression.
Collapse
Affiliation(s)
- Moayed Ibrahim
- Section of Hematology and Medical Oncology, Deming Department of MedicineTulane UniversityNew OrleansLouisianaUSA
| | - Catherine Murphree
- Section of Hematology and Medical Oncology, Deming Department of MedicineTulane UniversityNew OrleansLouisianaUSA
| | | | | | - Nakhle S. Saba
- Section of Hematology and Medical Oncology, Deming Department of MedicineTulane UniversityNew OrleansLouisianaUSA
| | - Hana Safah
- Section of Hematology and Medical Oncology, Deming Department of MedicineTulane UniversityNew OrleansLouisianaUSA
| | - Janet Schmid
- Pathology DepartmentTulane UniversityNew OrleansLouisianaUSA
| | - Francisco Socola
- Hematology and Bone Marrow TransplantUniversity of California DavisSacramentoCaliforniaUSA
| |
Collapse
|
5
|
Isfort S, von Bubnoff N, Al-Ali HK, Becker H, Götze T, le Coutre P, Griesshammer M, Moskwa C, Wohn L, Riedel J, Palandri F, Manz K, Hochhaus A, Döhner K, Heidel FH. FRACTION: protocol of a phase II study of Fedratinib and Nivolumab combination in patients with myelofibrosis and resistance or suboptimal response to JAK-inhibitor treatment of the German MPN study group (GSG-MPN). Ann Hematol 2024; 103:2775-2785. [PMID: 38967662 PMCID: PMC11283433 DOI: 10.1007/s00277-024-05867-w] [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: 05/24/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Development of Janus-kinase (JAK) inhibitors has revolutionized the therapeutic landscape for patients with myeloproliferative neoplasia (MPN). Following approval of the first JAK1/2-inhibitor Ruxolitinib, symptoms of this inflammatory disease, characterized by splenomegaly, release of inflammatory cytokines and appearance of thrombosis, could be effectively reduced for the first time. However, JAK-inhibitor treatment is limited in several aspects: 1) duration of response: 3 years after initiation of therapy more than 50% of patients have discontinued JAK-inhibitor treatment due to lack of efficacy or resistance; 2) reduction of disease burden: while effective in reducing inflammation and constitutional symptoms, JAK-inhibitors fail to reduce the malignant clone in the majority of patients and therefore lack long-term efficacy. Early clinical trials for patients with myelofibrosis (MF) have tried to address these issues for patients with suboptimal response to Ruxolitinib therapy while combination therapies with Fedratinib are rare. Recent reports provided first evidence on how the JAK2-V617F mutated myeloid cells may influence T-cell responses. JAK2-V617F promoted the synthesis of PD-L1 in MPN cells leading to limited anti-neoplastic T-cell responses, metabolic changes in T-cells and eventually JAK2-V617F-driven immune-escape of MPN cells. These findings may facilitate the use of immunotherapeutic approaches for JAK-mutated clones. Immune checkpoints refer to a variety of inhibitory pathways that are crucial for maintaining self-tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage. The FRACTION study is a single arm, open label Phase II trial investigating the combination of Fedratinib with the PD-1 inhibitor Nivolumab in patients with myelofibrosis and suboptimal or lack of response to JAK-inhibitor therapy. Over a 12 months period the trial assesses longer term outcomes, particularly the effects on clinical outcomes, such as induction of clinical remissions, quality of life and improvement of anemia. No prospective clinical trial data exist for combinations of JAK- and immune-checkpoint-inhibitors in the planned MF study population and this study will provide new findings that may contribute to advancing the treatment landscape for MF patients with suboptimal responses and limited alternatives.
Collapse
Affiliation(s)
- Susanne Isfort
- Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital Lübeck, Lübeck, Germany
| | - Haifa Kathrin Al-Ali
- University Hospital Halle (Saale), Krukenberg Cancer Center Halle, Halle, Germany
| | - Heiko Becker
- Department of Medicine I - Medical Center - University of Freiburg, Faculty of Medicine, University of , Freiburg, Freiburg, Germany
| | - Thorsten Götze
- Krankenhaus Nordwest, University Cancer Center (UCT), Frankfurt, Germany
- Institut für Klinische Krebsforschung IKF Am Krankenhaus Nordwest, Frankfurt, Germany
| | - Philipp le Coutre
- Department of Hematology, Oncology and Stem Cell Transplantation, Charite Berlin, Germany
| | | | - Claudia Moskwa
- Internal Medicine C, Hematology, Oncology, Stem Cell Transplantation and Palliative Care, University Medicine Greifswald, Greifswald, Germany
| | - Luisa Wohn
- Institut für Klinische Krebsforschung IKF Am Krankenhaus Nordwest, Frankfurt, Germany
| | - Johanna Riedel
- Institut für Klinische Krebsforschung IKF Am Krankenhaus Nordwest, Frankfurt, Germany
| | - Francesca Palandri
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Istituto Di Ematologia "Seràgnoli", Bologna, Italy
| | - Kirsi Manz
- Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, Hannover, 30625, Germany
- Institute for Community Medicine - SHIP-KEF, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Andreas Hochhaus
- Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Jena, Germany
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Florian H Heidel
- Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, Hannover, 30625, Germany.
- Internal Medicine C, Hematology, Oncology, Stem Cell Transplantation and Palliative Care, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
6
|
Pandey G, Mazzacurati L, Rowsell TM, Horvat NP, Amin NE, Zhang G, Akuffo AA, Colin-Leitzinger CM, Haura EB, Kuykendall AT, Zhang L, Epling-Burnette PK, Reuther GW. SHP2 inhibition displays efficacy as a monotherapy and in combination with JAK2 inhibition in preclinical models of myeloproliferative neoplasms. Am J Hematol 2024; 99:1040-1055. [PMID: 38440831 PMCID: PMC11096011 DOI: 10.1002/ajh.27282] [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: 09/26/2023] [Revised: 01/30/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024]
Abstract
Myeloproliferative neoplasms (MPNs), including polycythemia vera, essential thrombocytosis, and primary myelofibrosis, are clonal hematopoietic neoplasms driven by mutationally activated signaling by the JAK2 tyrosine kinase. Although JAK2 inhibitors can improve MPN patients' quality of life, they do not induce complete remission as disease-driving cells persistently survive therapy. ERK activation has been highlighted as contributing to JAK2 inhibitor persistent cell survival. As ERK is a component of signaling by activated RAS proteins and by JAK2 activation, we sought to inhibit RAS activation to enhance responses to JAK2 inhibition in preclinical MPN models. We found the SHP2 inhibitor RMC-4550 significantly enhanced growth inhibition of MPN cell lines in combination with the JAK2 inhibitor ruxolitinib, effectively preventing ruxolitinib persistent growth, and the growth and viability of established ruxolitinib persistent cells remained sensitive to SHP2 inhibition. Both SHP2 and JAK2 inhibition diminished cellular RAS-GTP levels, and their concomitant inhibition enhanced ERK inactivation and increased apoptosis. Inhibition of SHP2 inhibited the neoplastic growth of MPN patient hematopoietic progenitor cells and exhibited synergy with ruxolitinib. RMC-4550 antagonized MPN phenotypes and increased survival of an MPN mouse model driven by MPL-W515L. The combination of RMC-4550 and ruxolitinib, which was safe and tolerated in healthy mice, further inhibited disease compared to ruxolitinib monotherapy, including extending survival. Given SHP2 inhibitors are undergoing clinical evaluation in patients with solid tumors, our preclinical findings suggest that SHP2 is a candidate therapeutic target with potential for rapid translation to clinical assessment to improve current targeted therapies for MPN patients.
Collapse
Affiliation(s)
- Garima Pandey
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
| | - Lucia Mazzacurati
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
| | - Tegan M. Rowsell
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
| | | | - Narmin E. Amin
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
| | - Guolin Zhang
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL USA
| | - Afua A. Akuffo
- Department of Immunology, Moffitt Cancer Center, Tampa, FL USA
| | | | - Eric B. Haura
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL USA
| | | | - Ling Zhang
- Department of Pathology, Moffitt Cancer Center, Tampa, FL USA
| | | | - Gary W. Reuther
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL USA
| |
Collapse
|
7
|
Bilgihan MT, Eryigit AN, Ciftciler R. Efficacy and Safety of Immune Checkpoint Inhibitors in Hematologic Malignancies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:23-31. [PMID: 37863681 DOI: 10.1016/j.clml.2023.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 10/22/2023]
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has led to a dramatic paradigm shift within the landscape of cancer treatment, igniting significant interest in their potential application in treating hematologic malignancies. This comprehensive review critically has examined the existing body of literature to shed light on the evolving understanding of the efficacy and safety of ICIs, both as a single agent and in combination regimens in hematologic malignancies. Across distinct lymphoma subtypes, the observed treatment responses exhibit diversity, and conflicts. Notably, Hodgkin lymphoma and certain non-Hodgkin lymphomas such as primary mediastinal B-cell lymphoma, emerge as remarkable cases, showing encouraging response rates and outcomes. However, the efficacy of ICIs reveals variations among subtypes such as chronic lymphocytic leukemia and multiple myeloma. Combination therapies consistently demonstrated superior outcomes compared to monotherapy in several malignancies. While the potential benefits of ICIs in hematologic malignancies are evident, the safety profile warrants careful consideration. Immune-related and other adverse events, though generally tolerable and manageable, highlight the necessity of meticulous monitoring and appropriate intervention. The discussions prompted by these findings underscore the need for tailored treatment approaches, driven by disease subtype, patient characteristics, and potential biomarkers. Moreover, the emerging realm of combination therapies involving immune checkpoint inhibitors holds promise for enhanced treatment outcomes, and ongoing research endeavors aim to unravel the optimal strategies.
Collapse
Affiliation(s)
| | | | - Rafiye Ciftciler
- Department of Hematology, Selcuk University Faculty of Medicine, Konya, Turkey.
| |
Collapse
|
8
|
Mahdi D, Spiers J, Rampotas A, Polverelli N, McLornan DP. Updates on accelerated and blast phase myeloproliferative neoplasms: Are we making progress? Br J Haematol 2023; 203:169-181. [PMID: 37527977 DOI: 10.1111/bjh.19010] [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: 04/30/2023] [Revised: 07/13/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023]
Abstract
Management approaches for accelerated and blast phase myeloproliferative neoplasms remain challenging for clinicians and patients alike. Despite many therapeutic advances, outcomes for those patients who are not allogeneic haematopoietic cell transplant eligible remain, in general, very poor. Estimated survival rates for such blast phase patients is frequently reported as less than 6 months. No specific immunological, genomic or clinicopathological signature currently exists that accurately predicts the risk and timing of transformation, which frequently induces a high degree of anxiety among patients and clinicians alike. Within this review article, we provide an up-to-date summary of current understanding of the underlying pathogenesis of accelerated and blast phase disease and discuss current therapeutic approaches and realistic outcomes. Finally, we discuss how the horizon may look with the introduction of more novel agents into the clinical arena.
Collapse
Affiliation(s)
- Dina Mahdi
- Department of Haematology, University College Hospital, London, UK
| | - Jessica Spiers
- Department of Haematology, University College Hospital, London, UK
| | | | - Nicola Polverelli
- Unit of Blood Diseases and Stem Cell Transplantation, University of Brescia, Brescia, Italy
| | - Donal P McLornan
- Department of Haematology, University College Hospital, London, UK
| |
Collapse
|
9
|
Vainchenker W, Yahmi N, Havelange V, Marty C, Plo I, Constantinescu SN. Recent advances in therapies for primary myelofibrosis. Fac Rev 2023; 12:23. [PMID: 37771602 PMCID: PMC10523375 DOI: 10.12703/r/12-23] [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] [Indexed: 09/30/2023] Open
Abstract
Primary myelofibrosis (PMF), polycythemia vera (PV) and essential thrombocythemia (ET) form the classical BCR-ABL1-negative myeloproliferative neoplasms (MPNs) that are driven by a constitutive activation of JAK2 signaling. PMF as well as secondary MF (post-ET and post-PV MF) are the most aggressive MPNs. Presently, there is no curative treatment, except allogenic hematopoietic stem cell transplantation. JAK inhibitors, essentially ruxolitinib, are the therapy of reference for intermediate and high-risk MF. However, presently the current JAK inhibitors behave mainly as anti-inflammatory drugs, improving general symptoms and spleen size without major impact on disease progression. A better understanding of the genetics of MF, the biology of its leukemic stem cells (LSCs), the mechanisms of fibrosis and of cytopenia and the role of inflammatory cytokines has led to new approaches with the development of numerous therapeutic agents that target epigenetic regulation, telomerase, apoptosis, cell cycle, cytokines and signaling. Furthermore, the use of a new less toxic form of interferon-α has been revived, as it is presently one of the only molecules that targets the mutated clone. These new approaches have different aims: (a) to provide alternative therapy to JAK inhibition; (b) to correct cytopenia; and (c) to inhibit fibrosis development. However, the main important goal is to find new disease modifier treatments, which will profoundly modify the progression of the disease without major toxicity. Presently the most promising approaches consist of the inhibition of telomerase and the combination of JAK2 inhibitors (ruxolitinib) with either a BCL2/BCL-xL or BET inhibitor. Yet, the most straightforward future approaches can be considered to be the development of and/or selective inhibition of JAK2V617F and the targeting MPL and calreticulin mutants by immunotherapy. It can be expected that the therapy of MF will be significantly improved in the coming years.
Collapse
Affiliation(s)
- William Vainchenker
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Nasrine Yahmi
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Violaine Havelange
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Cliniques universitaires Saint Luc, Department of Hematology, Université Catholique de Louvain, Brussels, Belgium
| | - Caroline Marty
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Stefan N Constantinescu
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
- WEL Research Institute, WELBIO Department, Wavre, Belgium
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| |
Collapse
|
10
|
Holmström MO, Andersen M, Traynor S, Ahmad SM, Lisle TL, Handlos Grauslund J, Skov V, Kjær L, Ottesen JT, Gjerstorff MF, Hasselbalch HC, Andersen MH. Therapeutic cancer vaccination against mutant calreticulin in myeloproliferative neoplasms induces expansion of specific T cells in the periphery but specific T cells fail to enrich in the bone marrow. Front Immunol 2023; 14:1240678. [PMID: 37662956 PMCID: PMC10470021 DOI: 10.3389/fimmu.2023.1240678] [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: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 09/05/2023] Open
Abstract
Background Therapeutic cancer vaccination against mutant calreticulin (CALR) in patients with CALR-mutant (CALRmut) myeloproliferative neoplasms (MPN) induces strong T-cell responses against mutant CALR yet fails to demonstrate clinical activity. Infiltration of tumor specific T cells into the tumor microenvironment is needed to attain a clinical response to therapeutic cancer vaccination. Aim Determine if CALRmut specific T cells isolated from vaccinated patients enrich in the bone marrow upon completion of vaccination and explore possible explanations for the lack of enrichment. Methods CALRmut specific T cells from four of ten vaccinated patients were expanded, enriched, and analyzed by T-cell receptor sequencing (TCRSeq). The TCRs identified were used as fingerprints of CALRmut specific T cells. Bone marrow aspirations from the four patients were acquired at baseline and at the end of trial. T cells were enriched from the bone marrow aspirations and analyzed by TCRSeq to identify the presence and fraction of CALRmut specific T cells at the two different time points. In silico calculations were performed to calculate the ratio between transformed cells and effector cells in patients with CALRmut MPN. Results The fraction of CALRmut specific T cells in the bone marrow did not increase upon completion of the vaccination trial. In general, the T cell repertoire in the bone marrow remains relatively constant through the vaccination trial. The enriched and expanded CALRmut specific T cells recognize peripheral blood autologous CALRmut cells. In silico analyses demonstrate a high imbalance in the fraction of CALRmut cells and CALRmut specific effector T-cells in peripheral blood. Conclusion CALRmut specific T cells do not enrich in the bone marrow after therapeutic cancer peptide vaccination against mutant CALR. The specific T cells recognize autologous peripheral blood derived CALRmut cells. In silico analyses demonstrate a high imbalance between the number of transformed cells and CALRmut specific effector T-cells in the periphery. We suggest that the high burden of transformed cells in the periphery compared to the number of effector cells could impact the ability of specific T cells to enrich in the bone marrow.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Morten Andersen
- Centre for Mathematical Modeling – Human Health and Disease, IMFUFA, Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Sofie Traynor
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Shamaila Munir Ahmad
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Thomas Landkildehus Lisle
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Jacob Handlos Grauslund
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Johnny T. Ottesen
- Centre for Mathematical Modeling – Human Health and Disease, IMFUFA, Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Morten Frier Gjerstorff
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Yadav R, Hakobyan N, Wang JC. Role of Next Generation Immune Checkpoint Inhibitor (ICI) Therapy in Philadelphia Negative Classic Myeloproliferative Neoplasm (MPN): Review of the Literature. Int J Mol Sci 2023; 24:12502. [PMID: 37569880 PMCID: PMC10420159 DOI: 10.3390/ijms241512502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/17/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The Philadelphia chromosome-negative (Ph-) myeloproliferative neoplasms (MPNs), which include essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF), are enduring and well-known conditions. These disorders are characterized by the abnormal growth of one or more hematopoietic cell lineages in the body's stem cells, leading to the enlargement of organs and the manifestation of constitutional symptoms. Numerous studies have provided evidence indicating that the pathogenesis of these diseases involves the dysregulation of the immune system and the presence of chronic inflammation, both of which are significant factors. Lately, the treatment of cancer including hematological malignancy has progressed on the agents aiming for the immune system, cytokine environment, immunotherapy agents, and targeted immune therapy. Immune checkpoints are the molecules that regulate T cell function in the tumor microenvironment (TME). The first line of primary immune checkpoints are programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte antigen-4 (CTLA-4). Immune checkpoint inhibitor therapy (ICIT) exerts its anti-tumor actions by blocking the inhibitory pathways in T cells and has reformed cancer treatment. Despite the impressive clinical success of ICIT, tumor internal resistance poses a challenge for oncologists leading to a low response rate in solid tumors and hematological malignancies. A Phase II trial on nivolumab for patients with post-essential thrombocythemia myelofibrosis, primary myelofibrosis, or post-polycythemia myelofibrosis was performed (Identifier: NCT02421354). This trial tested the efficacy of a PD-1 blockade agent, namely nivolumab, but was terminated prematurely due to adverse events and lack of efficacy. A multicenter, Phase II, single-arm open-label study was conducted including pembrolizumab in patients with primary thrombocythemia, post-essential thrombocythemia or post-polycythemia vera myelofibrosis that were ineligible for or were previously treated with ruxolitinib. This study showed that pembrolizumab treatment did not have many adverse events, but there were no pertinent clinical responses hence it was terminated after the first stage was completed. To avail the benefits from immunotherapy, the paradigm has shifted to new immune checkpoints in the TME such as lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin and mucin domain 3 (TIM-3), T cell immunoglobulin and ITIM domain (TIGIT), V-domain immunoglobulin-containing suppressor of T cell activation (VISTA), and human endogenous retrovirus-H long terminal repeat-associating protein 2 (HHLA2) forming the basis of next-generation ICIT. The primary aim of this article is to underscore and elucidate the significance of next-generation ICIT in the context of MPN. Specifically, we aim to explore the potential of monoclonal antibodies as targeted immunotherapy and the development of vaccines targeting specific MPN epitopes, with the intent of augmenting tumor-related immune responses. It is anticipated that these therapeutic modalities rooted in immunotherapy will not only expand but also enhance the existing treatment regimens for patients afflicted with MPN. Preliminary studies from our laboratory showed over-expressed MDSC and over-expressed VISTA in MDSC, and in progenitor and immune cells directing the need for more clinical trials using next-generation ICI in the treatment of MPN.
Collapse
Affiliation(s)
- Ruchi Yadav
- Department of Internal Medicine, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA; (R.Y.); (N.H.)
| | - Narek Hakobyan
- Department of Internal Medicine, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA; (R.Y.); (N.H.)
| | - Jen-Chin Wang
- Department of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA
| |
Collapse
|
12
|
Ajufo HO, Waksal JA, Mascarenhas JO, Rampal RK. Treating accelerated and blast phase myeloproliferative neoplasms: progress and challenges. Ther Adv Hematol 2023; 14:20406207231177282. [PMID: 37564898 PMCID: PMC10410182 DOI: 10.1177/20406207231177282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/03/2023] [Indexed: 08/12/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are a group of clonal hematologic malignancies that include polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF). MPNs are characterized by activating mutations in the JAK/STAT pathway and an increased risk of transformation to an aggressive form of acute leukemia, termed MPN-blast phase (MPN-BP). MPN-BP is characterized by the presence of ⩾20% blasts in the blood or bone marrow and is almost always preceded by an accelerated phase (MPN-AP) defined as ⩾10-19% blasts in the blood or bone marrow. These advanced forms of disease are associated with poor prognosis with a median overall survival (mOS) of 3-5 months in MPN-BP and 13 months in MPN-AP. MPN-AP/BP has a unique molecular landscape characterized by increased intratumoral complexity. Standard therapies used in de novo acute myeloid leukemia (AML) have not demonstrated improvement in OS. Allogeneic hematopoietic stem cell transplant (HSCT) remains the only curative therapy but is associated with significant morbidity and mortality and infrequently utilized in clinical practice. Therefore, an urgent unmet need persists for effective therapies in this advanced phase patient population. Here, we review the current management and future directions of therapy in MPN-AP/BP.
Collapse
Affiliation(s)
- Helen O. Ajufo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julian A. Waksal
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John O. Mascarenhas
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1079, New York, NY 10029, USA
| | | |
Collapse
|
13
|
Gagelmann N, Badbaran A, Salit RB, Schroeder T, Gurnari C, Pagliuca S, Panagiota V, Rautenberg C, Cassinat B, Thol F, Wolschke C, Robin M, Heuser M, Rubio MT, Maciejewski JP, Reinhardt HC, Scott BL, Kröger N. Impact of TP53 on outcome of patients with myelofibrosis undergoing hematopoietic stem cell transplantation. Blood 2023; 141:2901-2911. [PMID: 36940410 PMCID: PMC10933704 DOI: 10.1182/blood.2023019630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/22/2023] Open
Abstract
TP53 mutations (TP53MTs) have been associated with poor outcomes in various hematologic malignancies, but no data exist regarding its role in patients with myelofibrosis undergoing hematopoietic stem cell transplantation (HSCT). Here, we took advantage of a large international multicenter cohort to evaluate the role of TP53MT in this setting. Among 349 included patients, 49 (13%) had detectable TP53MT, of whom 30 showed a multihit configuration. Median variant allele frequency was 20.3%. Cytogenetic risk was favorable (71%), unfavorable (23%), and very high (6%), with complex karyotype present in 36 patients (10%). Median survival of patients with TP53MT was 1.5 vs 13.5 years for those with wild-type TP53 (TP53WT; P < .001). Outcome was driven by multihit TP53MT constellation (P < .001), showing 6-year survival of 56% for individuals with single-hit vs 25% for those with multihit TP53MT vs 64% for those with TP53WT. Outcome was independent of current transplantation-specific risk factors and conditioning intensity. Similarly, cumulative incidence of relapse was 17% for single-hit vs 52% for multihit vs 21% for TP53WT. Ten patients with TP53MT (20%) presented as leukemic transformation vs only 7 (2%) in the TP53WT group (P < .001). Out of the 10 patients with TP53MT, 8 showed multihit constellation. Median time to leukemic transformation was shorter for multihit and single-hit TP53MT (0.7 and 0.5 years, respectively) vs 2.5 years for TP53WT. In summary, multihit TP53MT represents a very high-risk group in patients with myelofibrosis who are undergoing HSCT, whereas single-hit TP53MT alone showed similar outcome to patients with nonmutated TP53, informing prognostication for survival and relapse together with current transplantation-specific tools.
Collapse
Affiliation(s)
- Nico Gagelmann
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Badbaran
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rachel B. Salit
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Thomas Schroeder
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital of Essen, Essen, Germany
| | - Carmelo Gurnari
- Translational Hematology and Oncology Research Department, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy
| | - Simona Pagliuca
- Department of Hematology, Brabois Hospital, Centre Hospitalier Régional Universitaire, Nancy, France
| | - Victoria Panagiota
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Christina Rautenberg
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital of Essen, Essen, Germany
| | - Bruno Cassinat
- Laboratoire de Biologie Cellulaire, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Felicitas Thol
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Christine Wolschke
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marie Robin
- Service d'Hématologie-Greffe, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Marie-Thérèse Rubio
- Department of Hematology, Brabois Hospital, Centre Hospitalier Régional Universitaire, Nancy, France
| | - Jaroslaw P. Maciejewski
- Translational Hematology and Oncology Research Department, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH
- Leukemia Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital of Essen, Essen, Germany
| | - Bart L. Scott
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
How J, Garcia JS, Mullally A. Biology and therapeutic targeting of molecular mechanisms in MPNs. Blood 2023; 141:1922-1933. [PMID: 36534936 PMCID: PMC10163317 DOI: 10.1182/blood.2022017416] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell disorders characterized by activated Janus kinase (JAK)-signal transducer and activator of transcription signaling. As a result, JAK inhibitors have been the standard therapy for treatment of patients with myelofibrosis (MF). Although currently approved JAK inhibitors successfully ameliorate MPN-related symptoms, they are not known to substantially alter the MF disease course. Similarly, in essential thrombocythemia and polycythemia vera, treatments are primarily aimed at reducing the risk of cardiovascular and thromboembolic complications, with a watchful waiting approach often used in patients who are considered to be at a lower risk for thrombosis. However, better understanding of MPN biology has led to the development of rationally designed therapies, with the goal of not only addressing disease complications but also potentially modifying disease course. We review the most recent data elucidating mechanisms of disease pathogenesis and highlight emerging therapies that target MPN on several biologic levels, including JAK2-mutant MPN stem cells, JAK and non-JAK signaling pathways, mutant calreticulin, and the inflammatory bone marrow microenvironment.
Collapse
Affiliation(s)
- Joan How
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Jacqueline S. Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute, Cambridge, MA
| |
Collapse
|
15
|
Grauslund JH, Holmström MO, Martinenaite E, Lisle TL, Glöckner HJ, El Fassi D, Klausen U, Mortensen REJ, Jørgensen N, Kjær L, Skov V, Svane IM, Hasselbalch HC, Andersen MH. An arginase1- and PD-L1-derived peptide-based vaccine for myeloproliferative neoplasms: A first-in-man clinical trial. Front Immunol 2023; 14:1117466. [PMID: 36911725 PMCID: PMC9996128 DOI: 10.3389/fimmu.2023.1117466] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Arginase-1 (ARG1) and Programed death ligand-1 (PD-L1) play a vital role in immunosuppression in myeloproliferative neoplasms (MPNs) and directly inhibit T-cell activation and proliferation. We previously identified spontaneous T-cell responses towards PD-L1 and ARG1 derived peptide epitopes in patients with MPNs. In the present First-in-Man study we tested dual vaccinations of ARG1- derived and PD-L1-derived peptides, combined with Montanide ISA-51 as adjuvant, in patients with Janus Kinase 2 (JAK2) V617F-mutated MPN. Methods Safety and efficacy of vaccination with ARG1- derived and PD-L1-derived peptides with montanide as an adjuvant was tested in 9 patients with MPN The primary end point was safety and toxicity evaluation. The secondary end point was assessment of the immune response to the vaccination epitope (www.clinicaltrials.gov identifier NCT04051307). Results The study included 9 patients with JAK2-mutant MPN of which 8 received all 24 planned vaccines within a 9-month treatment period. Patients reported only grade 1 and 2 vaccine related adverse events. No alterations in peripheral blood counts were identified, and serial measurements of the JAK2V617F allelic burden showed that none of the patients achieved a molecular response during the treatment period. The vaccines induced strong immune responses against both ARG1 and PD-L1- derived epitopes in the peripheral blood of all patients, and vaccine-specific skin-infiltrating lymphocytes from 5/6 patients could be expanded in vitro after a delayed-type hypersensitivity test. In two patients we also detected both ARG1- and PD-L1-specific T cells in bone marrow samples at the end of trial. Intracellular cytokine staining revealed IFNγ and TNFγ producing CD4+- and CD8+- T cells specific against both vaccine epitopes. Throughout the study, the peripheral CD8/CD4 ratio increased significantly, and the CD8+ TEMRA subpopulation was enlarged. We also identified a significant decrease in PD-L1 mRNA expression in CD14+ myeloid cells in the peripheral blood in all treated patients and a decrease in ARG1 mRNA expression in bone marrow of 6 out of 7 evaluated patients. Conclusion Overall, the ARG1- and PD-L1-derived vaccines were safe and tolerable and induced strong T-cell responses in all patients. These results warrant further studies of the vaccine in other settings or in combination with additional immune-activating treatments.
Collapse
Affiliation(s)
- Jacob Handlos Grauslund
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
- Institute for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Evelina Martinenaite
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
- Research and Development, IO Biotech ApS, Copenhagen, Denmark
| | - Thomas Landkildehus Lisle
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Hannah Jorinde Glöckner
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Daniel El Fassi
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Uffe Klausen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Rasmus E. J. Mortensen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Nicolai Jørgensen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
- Institute for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Lecomte S, Devreux J, de Streel G, van Baren N, Havelange V, Schröder D, Vaherto N, Vanhaver C, Vanderaa C, Dupuis N, Pecquet C, Coulie PG, Constantinescu SN, Lucas S. Therapeutic activity of GARP:TGF-β1 blockade in murine primary myelofibrosis. Blood 2023; 141:490-502. [PMID: 36322928 PMCID: PMC10651781 DOI: 10.1182/blood.2022017097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/23/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by the clonal expansion of myeloid cells, notably megakaryocytes (MKs), and an aberrant cytokine production leading to bone marrow (BM) fibrosis and insufficiency. Current treatment options are limited. TGF-β1, a profibrotic and immunosuppressive cytokine, is involved in PMF pathogenesis. While all cell types secrete inactive, latent TGF-β1, only a few activate the cytokine via cell type-specific mechanisms. The cellular source of the active TGF-β1 implicated in PMF is not known. Transmembrane protein GARP binds and activates latent TGF-β1 on the surface of regulatory T lymphocytes (Tregs) and MKs or platelets. Here, we found an increased expression of GARP in the BM and spleen of mice with PMF and tested the therapeutic potential of a monoclonal antibody (mAb) that blocks TGF-β1 activation by GARP-expressing cells. GARP:TGF-β1 blockade reduced not only fibrosis but also the clonal expansion of transformed cells. Using mice carrying a genetic deletion of Garp in either Tregs or MKs, we found that the therapeutic effects of GARP:TGF-β1 blockade in PMF imply targeting GARP on Tregs. These therapeutic effects, accompanied by increased IFN-γ signals in the spleen, were lost upon CD8 T-cell depletion. Our results suggest that the selective blockade of TGF-β1 activation by GARP-expressing Tregs increases a CD8 T-cell-mediated immune reaction that limits transformed cell expansion, providing a novel approach that could be tested to treat patients with myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Sara Lecomte
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Julien Devreux
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | - Nicolas van Baren
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Violaine Havelange
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Department of Hematology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - David Schröder
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Noora Vaherto
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | | | - Noémie Dupuis
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Christian Pecquet
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium
| | - Pierre G. Coulie
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology, Wavre, Belgium
| | - Stefan N. Constantinescu
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology, Wavre, Belgium
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research Oxford, University of Oxford, Oxford, United Kingdom
| | - Sophie Lucas
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology, Wavre, Belgium
| |
Collapse
|
17
|
Ghit A. Myelofibrosis treatment history and future prospects. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2022. [DOI: 10.1186/s43162-022-00169-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractMyelofibrosis (MF) is a haematopoietic stem cell tumour caused by the lack of BCR-ABL translocation due to point mutations in Janus kinases (JAKs). In previous years, dealing with MF included several protocols such as traditional drugs that control general symptoms, splenectomy, blood transfusion, and allogeneic haematopoietic stem-cell transplantation (HSCT). Allogeneic HSCT is remaining the only treatment that has the potential to alter MF’s progression. However, clinical trials of JAK inhibitors and non-JAK targeted therapies have been increasingly carried out in earlier years. The most prominent JAK inhibitors for the treatment of MF are ruxolitinib, fedratinib, momelotinib, pacritinib, gandotinib, ilginatinib, itacitinib, and lestaurtinib. On the other hand, the non-JAK targeted therapies that showed strong efficacy and safety are alisertib, imetelstat, pembrolizumab, nivolumab, and sotatercept. In this review, we summarized the recent clinical trials carried out on these drugs to understand their efficacy and safety. Also, we talked briefly about allogeneic HSCT as powerful therapy until the present for patients suffering from MF.
Collapse
|
18
|
Liu R, Xie Y, Xu JR, Luo Q, Ren YX, Chen M, Duan JL, Bao CJ, Liu YX, Li PS, Li JW, Wang GL, Lu WL. Engineered stem cell biomimetic liposomes carrying levamisole for macrophage immunity reconstruction in leukemia therapy. CHEMICAL ENGINEERING JOURNAL 2022; 447:137582. [DOI: 10.1016/j.cej.2022.137582] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
19
|
Varricchio L, Hoffman R. Megakaryocytes Are Regulators of the Tumor Microenvironment and Malignant Hematopoietic Progenitor Cells in Myelofibrosis. Front Oncol 2022; 12:906698. [PMID: 35646681 PMCID: PMC9130548 DOI: 10.3389/fonc.2022.906698] [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: 03/28/2022] [Accepted: 04/15/2022] [Indexed: 12/15/2022] Open
Abstract
Megakaryocytes (MKs) are multifunctional hematopoietic cells that produce platelets, serve as components of bone marrow (BM) niches that support the development of hematopoietic stem and progenitor cell (HSPC) and provide inflammatory signals. MKs can dynamically change their activities during homeostasis and following stress, thereby regulating hematopoietic stem cell (HSC) function. Myelofibrosis (MF) is a progressive chronic myeloproliferative neoplasm (MPN) characterized by hyperactivation of JAK/STAT signaling and MK hyperplasia, which is associated with an aberrant inflammatory signature. Since JAK1/2 inhibitor alone is incapable of depleting the malignant HSC clones or reversing BM fibrosis, the identification of mechanisms that cooperate with MF JAK/STAT signaling to promote disease progression might help in developing combination therapies to modify disease outcomes. Chronic inflammation and MK hyperplasia result in an abnormal release of TGFβ1, which plays a critical role in the pathobiology of MF by contributing to the development of BM fibrosis. Dysregulated TGFβ signaling can also alter the hematopoietic microenvironment supporting the predominance of MF-HSCs and enhance the quiescence of the reservoir of wild-type HSCs. Upregulation of TGFβ1 levels is a relatively late event in MF, while during the early pre-fibrotic stage of MF the alarmin S100A8/S100A9 heterocomplex promotes pro-inflammatory responses and sustains the progression of MF-HSCs. In this review, we will discuss the recent advances in our understanding of the roles of abnormal megakaryopoiesis, and the altered microenvironment in MF progression and the development of novel combined targeted therapies to disrupt the aberrant interplay between MKs, the BM microenvironment and malignant HSCs which would potentially limit the expansion of MF-HSC clones.
Collapse
Affiliation(s)
- Lilian Varricchio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
20
|
Chifotides HT, Bose P, Masarova L, Pemmaraju N, Verstovsek S. SOHO State of the Art Updates and Next Questions: Novel Therapies in Development for Myelofibrosis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:210-223. [PMID: 34840087 DOI: 10.1016/j.clml.2021.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 06/13/2023]
Abstract
Myeloproliferative neoplasms research has entered a dynamic and exciting era as we witness exponential growth of novel agents in advanced/early phase clinical trials for myelofibrosis (MF). Building on the success and pivotal role of ruxolitinib, many novel agents, spanning a wide range of mechanisms/targets (epigenetic regulation, apoptotic/intracellular signaling pathways, telomerase, bone marrow fibrosis) are in clinical development; several are studied in registrational trials and hold great potential to expand the therapeutic arsenal/shift the treatment paradigm if regulatory approval is granted. Insight into MF pathogenesis and its molecular underpinnings, preclinical studies demonstrating synergism of ruxolitinib with investigational agents, urgent unmet clinical needs (cytopenias, loss of response to JAK inhibitors); and progressive disease fueled the rapid rise of innovative therapeutics. New strategies include pairing ruxolitinib with erythroid maturation agents to manage anemia (luspatercept), designing rational combinations with ruxolitinib to boost responses in both the frontline and suboptimal response settings (pelabresib, navitoclax, parsaclisib), treatment with non-JAK inhibitor monotherapy in the second-line setting (navtemadlin, imetelstat), novel JAK inhibitors tailored to subgroups with challenging unmet needs (momelotinib and pacritinib for anemia and thrombocytopenia, respectively); and agents potentially enhancing longevity (imetelstat). Beyond typical endpoints evaluated in MF clinical trials (spleen volume reduction ≥ 35%, total symptom score reduction ≥ 50%) thus far, emerging endpoints include overall survival, progression-free survival, transfusion independence, anemia benefits, bone marrow fibrosis and driver mutation allele burden reduction. Novel biomarkers and additional clinical features are being sought to assess new agents and tailor emerging therapies to appropriate patients. New strategies are needed to optimize the design of clinical trials comparing novel combinations to standard agent monotherapy.
Collapse
Affiliation(s)
- Helen T Chifotides
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Lucia Masarova
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|