Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2021 Oct 7;196(5):1149–1158. doi: 10.1111/bjh.17850

The immune landscape in BCR‐ABL negative myeloproliferative neoplasms: inflammation, infections and opportunities for immunotherapy

Marie Strickland 1,2, Lynn Quek 3,4, Bethan Psaila 1,2,
PMCID: PMC9135025  PMID: 34618358

Summary

Breakpoint cluster region‐Abelson (BCR‐ABL) negative myeloproliferative neoplasms (MPNs) are chronic myeloid neoplasms initiated by the acquisition of gene mutation(s) in a haematopoietic stem cell, leading to clonal expansion and over‐production of blood cells and their progenitors. MPNs encompass a spectrum of disorders with overlapping but distinct molecular, laboratory and clinical features. This includes polycythaemia vera, essential thrombocythaemia and myelofibrosis. Dysregulation of the immune system is key to the pathology of MPNs, supporting clonal evolution, mediating symptoms and resulting in varying degrees of immunocompromise. Targeting immune dysfunction is an important treatment strategy. In the present review, we focus on the immune landscape in patients with MPNs – the role of inflammation in disease pathogenesis, susceptibility to infection and emerging strategies for therapeutic immune modulation. Further detailed work is required to delineate immune perturbation more precisely in MPNs to determine how and why vulnerability to infection differs between clinical subtypes and to better understand how inflammation results in a competitive advantage for the MPN clone. These studies may help shed light on new designs for disease‐modifying therapies.

Keywords: Chronic myeloid malignancies, immunity, immunocompromise, immuno‐oncology, JAK inhibitors

The role of innate and adaptive immunity in myeloproliferative neoplasms (MPNs)

The innate and adaptive immune systems work synergistically to protect against the emergence of cancer via a series of stepwise events referred to as the ‘cancer‐immunity cycle’. 1 This process depends on the release of aberrant proteins by malignant cells and their detection and capture by antigen‐presenting cells (APCs) of the innate immune system (e.g. dendritic cells and macrophages). APCs then prime and activate effector T cells, leading to target elimination. This process is carefully moderated by co‐stimulatory and co‐inhibitory signals and the ratio of effector versus regulatory T cells (Tregs). Cancer cells must evade this cycle in order to survive and proliferate. This is achieved via mechanisms that downregulate anti‐tumoral immunity, such as reduced expression of human leucocyte antigen (HLA) class I molecules preventing antigen presentation, or upregulation of molecules such as programmed death‐ligand 1 (PD‐L1) that inhibit T‐cell activity (reviewed in Sukari et al. 2 and Sánchez‐Paulete et al. 3 ).

In the present review, we first discuss data indicating how MPN driver mutations result in inflammatory signalling, and how this is permissive for clonal expansion and disease progression. The mechanisms by which the MPN clone evades anti‐tumoral immunity and the impact of immune perturbations on susceptibility to infection are then considered. Finally, emerging strategies for targeted immunomodulatory therapy for MPNs are discussed.

MPN gene mutations result in increased production of inflammatory cytokines from both clonal and non‐clonal cells

Myeloproliferative neoplasms arise following the acquisition of gene mutations in haematopoietic stem cells (HSCs) that result in cytokine‐independent activation of Janus kinase‐signal transducer and activator of transcription (JAK‐STAT) signalling. The most common mutations occur in the genes encoding JAK2 (JAK2 V617F), calreticulin (CALR) and the thrombopoietin receptor, MPL. MPN ‘driver’ mutations result in the activation of pro‐inflammatory signalling, in particular tumour necrosis factor (TNF)/nuclear factor κ‐light‐chain‐enhancer of activated B cells (NF‐κB) pathways, in mutated HSCs and their progeny. 4 , 5 , 6 Studies at the single cell level have shown that the increased production of inflammatory cytokines results from both an increase in the percentage of cytokine‐secreting cells, as well as augmented cytokine secretion per cell. 4 In addition, in patients with mutant (mut)CALR‐driven essential thrombocythaemia (ET) and myelofibrosis, there is a direct pro‐inflammatory effect of the mutant protein itself, as secreted mutCALR exaggerates cytokine production from normal monocytes. 7

In addition to production of inflammatory cytokines by the MPN clone, immune dysregulation also results from paracrine/endocrine effects on non‐clonal haematopoietic and stromal cells (Fig 1). For example, in patients with myelofibrosis, Wang et al. 8 observed that excessive soluble interleukin‐2 receptor α (IL‐2Rα) was produced by non‐clonal cluster of differentiation (CD)4+CD25+ forkhead box protein 3 (FoxP3)+ Tregs. This has also been carefully documented by studies at the single cell level in which accurate discrimination of mutated and wild‐type cells can be performed in parallel with transcriptomic analysis. For example, simultaneous interrogation of clonal and non‐clonal populations in individuals with BCR‐ABL‐negative MPNs, as well as chronic myeloid leukaemia (CML), showed significant enrichment of inflammatory pathways including IL‐6, transforming growth factor beta (TGFβ) and TNFα‐associated signalling in wild‐type HSCs in patients with CML 9 and in patients with JAK2 V617F+ myelofibrosis, 10 in comparison to HSCs in age‐matched, healthy donors. In pre‐clinical models, mice injected with JAK V617F+ cells demonstrated increased TNFα and IL‐6 production by host cells in response to proteins secreted by the malignant clone. 11

Fig 1.

Fig 1

Pro‐inflammatory interactions between clonal and non‐clonal cells. Inflammatory cytokines and growth factors such as TNFα, IL‐6, and IL‐8 confer a competitive advantage to the malignant MPN clone and are associated with increased symptom burden. JAK2 V617F mutations also alter the epigenetic regulation of TNF/NF‐κB inflammatory signalling pathways, further increasing inflammation. Extra‐cellular signals released by the MPN clone result in excess production of inflammatory cytokines by non‐clonal myeloid cells, lymphoid cells and bone marrow stroma. HSPCs, haematopoietic stem and progenitor cells; IL, interleukin; JAK2, Janus kinase 2; MPN, myeloproliferative neoplasm; NF‐κB, nuclear factor κ‐light‐chain‐enhancer of activated B cells; TGFβ, transforming growth factor beta; TNF, tumour necrosis factor.

Inflammation is permissive for clonal expansion and MPN disease progression

Recent data suggest that MPN mutations may be acquired in utero or during early childhood, although they typically do not manifest until adult life. 12 , 13 This suggests a model in which progression to clinically overt disease may be driven by gradual clonal expansion supported by ageing‐associated immune dysfunction and inflammatory bone marrow changes. 14 , 15 , 16 In support of this hypothesis, certain inflammatory cytokines that are increased in patients with MPN, including TNFα and interferon α (IFNα), have been shown to confer a selective growth advantage to JAK2 V617F‐mutant over wild‐type cells in vitro, 17 , 18 enabling clonal expansion (Fig 1). Recently published data indicates that JAK2V617F‐mutant HSCs are hyper‐responsive to IFN, and that IFN signalling promotes megakaryocyte‐biased haematopoiesis, which may be an important determinant of MPN phenotype and disease evolution. 19 , 20 Increased TNFα as well as hepatocyte growth factor (HGF), platelet‐derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and interleukins especially IL‐6 and IL‐8 have also been associated with increased JAK2 V617F mutation burden and poor outcomes. 17 , 21 , 22

Associations have also been noted between certain cytokines and disease stage and/or fibrotic progression. In a study of >120 patients with myelofibrosis, IL‐8 and the IL‐2R were highly predictive of disease stage, with increased levels of one or both associated with an increased Dynamic International Prognostic Scoring System (DIPSS) risk score. 22 Higher levels of soluble IL‐2Rα (sIL‐2Rα) have also been observed in patients with myelofibrosis relative to either patients with polycythaemia vera (PV)/ET or healthy controls 8 and increased IL‐6, IL‐2 and sIL‐2Rα have been demonstrated in patients following progression from ET and PV to myelofibrosis. 23 Further support for a key role of inflammation in MPN progression is that the development of fibrosis in myelofibrosis has been reported in some studies to correlate more strongly with the levels of certain cytokines including IL‐8 and TGF‐β than malignant clone burden (e.g. JAK2 V617F variant allele frequency). 24 , 25 In addition to supporting clonal expansion, inflammatory cytokines result in high symptom burden for some patients, including fatigue, itching, sweats and weight loss. 26

Breaking of the tumour‐immunity cycle and evasion of anti‐tumour immunosurveillance

Subversion of innate and adaptive immune surveillance and breaking of the tumour‐immunity cycle contribute to enabling the neoplastic myeloid clone to expand. APCs in patients with MPNs have a lower ability to process and present antigens, leading to suboptimal priming and activation of T cells. Both class I and II HLA genes are down‐regulated in PV, ET and myelofibrosis, with progressive downregulation of certain genes [BAT2L, HLA Complex Group 11 (HCG11) and major histocompatibility complex (MHC), Class I‐related (MR1)] in ET versus PV versus myelofibrosis. 27 Numbers of circulating dendritic cells are reduced in patients with myelofibrosis, most significantly in those with the JAK2 V617F mutation. 28 Alongside this, the differentiation of monocytes into dendritic cells in vitro has been shown to be impaired in a study of patients with myelofibrosis, with an associated defect in their capacity to prime T cells. 28 An increase in myeloid‐derived suppressor cells (MDSCs) has also been documented, specifically in myelofibrosis. 29 MDSCs lack expression of the MHC class II receptor HLA‐DR and have an overall immunosuppressive impact on T lymphocytes, aiding immune evasion. Loss or down‐regulation of HLA class I antigens results in escape from immune surveillance in several solid tumours and other haematological malignancies. 30

In addition to impaired antigen presentation and T‐cell priming, mutations in CALR also subvert cellular immunity via a specific mechanism, as the normal CALR protein has a key role in tumour immunosurveillance. CALR expressed on the cell surface promotes phagocytosis of malignant cells by engaging low‐density lipoprotein receptor‐related protein (LRP) on macrophages. 31 To evade phagocytosis, neoplastic cells upregulate expression of CD47, a cell surface molecule that binds signal‐regulatory protein α (SIRP‐α) on macrophages and delivers a ‘do not eat me’ signal. 32 An altered ratio of expression of pro‐phagocytic CALR and anti‐phagocytic CD47 mediates evasion from immunosurveillance and is the basis for CD47‐targeting molecules as novel anti‐cancer therapies. 33 In patients with mutCALR+ MPN, mutant CALR protein inhibits phagocytosis of apoptotic MPN cells by dendritic cells, preventing effective antigen presentation. 34

Alterations in immune effector cell number and function have been reported, including a reduced number of CD56+ CD3 natural killer (NK) cells in untreated patients with MPN compared to healthy controls, 35 a reduced number of CD3+ T cells, 36 and impaired IFNγ production by T cells. 22 Unlike in solid cancers where Tregs are generally increased, numbers of Tregs do not appear to be substantially increased in MPNs. 8 , 36 However, their ability to produce soluble IL‐2Rα is reduced in myelofibrosis relative to other MPN diagnoses and compared to healthy controls. 8

Myelofibrosis in particular is associated with pronounced changes to the bone marrow stroma and non‐cellular matrix. It is possible that these structural changes may prevent immune cell surveillance within the bone marrow microenvironment, coupled with immune ‘exhaustion’ induced by chronic and high‐level exposure to the MPN cancer antigens. 37 , 38 Together, these changes impact the efficiency of identification and targeting of the MPN clone by innate and adaptive immune mechanisms.

Disease‐mediated susceptibility to infection

The altered immune landscape in MPNs results in an increased incidence of bacterial, viral, and fungal infections. A recent population‐based cohort study including >8000 patients with MPNs and 32 000 controls in Sweden identified a hazard ratio (HR) of ~2·0 for both bacterial and viral infections across all MPN subgroups. 39 The highest risk of any infection was in those with myelofibrosis (HR 3·7) compared to those with PV or ET (HR 1·7), and there was no difference in infection rates in patients with ET or PV who were not receiving treatment compared to those who were receiving hydroxyurea, IFN or anagrelide. 39 This suggests that a large component of susceptibility to infection is mediated by the underlying disease and present to varying degrees in all three MPN subtypes, irrespective of immunocompromise resulting from particular treatments. As for many other haematological disorders, infection rates are particularly high in those who are transfusion dependent with iron overload. 40

Vulnerability to infection in patients with MPN was rapidly highlighted by the coronavirus disease 2019 (COVID‐19) pandemic. Several studies documented a substantially higher risk of severe infection and death from COVID‐19 in patients with haematological malignancy, including those with MPNs. 41 , 42 , 43 , 44 In a systematic review and meta‐analysis of 3377 patients with haematological malignancies and COVID‐19, risk of death in patients with MPNs was 34%, substantially higher than in the general population, although lower than for those with acquired bone marrow failure syndromes and acute leukaemias. 45 A UK‐wide national audit of COVID‐19 in MPN highlighted the strong correlation between advanced age and male sex with severe COVID‐19 within MPN patients, similar to that observed in the general population. 46 In this study, patients not receiving cytoreductive therapy were under‐represented in the cohort, suggesting a lower risk in untreated patients, while ruxolitinib‐treated patients were over‐represented, although this study was not able to assess the relative impact of a more advanced underlying disease or age and comorbidities versus a direct association with ruxolitinib treatment itself. A particularly poor outcome was observed in ruxolitinib‐treated patients aged >75 years. 46 One USA study suggested a substantially stronger effect of recently diagnosed ET on the odds of COVID‐19 infection as compared to PV. 43

As a result of increased susceptibility to infection in MPN and higher risk of developing serious complications, most guidelines recommend that patients with MPN receive the annual influenza vaccine and other inactivated vaccines but that live attenuated vaccines (e.g. shingles vaccine) are avoided. However, responses to vaccination in some patients with MPN may be suboptimal, with evidence, albeit in a small cohort, of delayed and impaired B cell and T cell memory responses 3‐weeks and 3‐months after Influenza A vaccination. 47 Evidence of significantly reduced seroconversion rates and lower antibody titres has also been documented in patients with MPNs after the first dose of the COVID‐19 vaccine, particularly in patients with ET and PV receiving hydroxycarbamide and patients receiving ruxolitinib, while responses in patients with ET and PV treated with pegylated IFN were relatively unimpaired. 48

Immunomodulatory impact of current cytoreductive MPN treatments

Susceptibility to infection is a well‐documented consequence of ruxolitinib, a JAK1/JAK2 inhibitor that is highly effective at reducing symptoms and splenomegaly patients with symptomatic myelofibrosis. A recent study of 948 patients with MPN in Germany and Italy found that the proportion of patients experiencing infections was significantly higher in patients receiving ruxolitinib or ruxolitinib combinations as compared to patients not receiving any treatment or hydroxycarbamide alone, and that ruxolitinib‐treated patients were more likely to require hospital outpatients visits or admission for infections. 49 This finding was observed for both upper respiratory tract and gastro‐intestinal infections. 49 Of note, there was no difference in infection rates between patients with different driver mutation status (JAK2 V617F, CALR, MPL, triple negative) but patients with myelofibrosis had significantly more infections than those with ET or PV. 49 One consideration in interpreting these data is that generally patients treated with ruxolitinib tend to be older and with more advanced disease, and non‐drug related factors may contribute to their vulnerability to infection. Certain specific infections such as herpes virus reactivation and urinary tract infections are more prevalent in patients with MPN treated with ruxolitinib, 50 and reactivation of rare opportunistic infections have also been reported, including progressive multifocal leukoencephalopathy (PML), 51 hepatitis B, 52 pneumonia and disseminated tuberculosis. 53 , 54 , 55 , 56 As a result, patients should be screened for latent and atypical infections such as hepatitis C and B and tuberculous, and prophylactic acyclovir considered to prevent shingles, prior to the commencement of ruxolitinib treatment.

The mechanism of ruxolitinib‐induced immunosuppression is multifactorial. The success of ruxolitinib in reducing symptoms and spleen sizes is largely due to its strong anti‐proliferative and anti‐inflammatory properties, with clear evidence of a reduction in circulating inflammatory cytokines including TNFα, IL‐1, IL‐6, IL‐8, IL‐1RA and IFNγ and reduced cytokine‐mediated symptoms. 57 , 58 Ruxolitinib treatment results in impaired effector T‐cell function, 59 as well as changes in Tregs, NK and dendritic cells, 60 , 61 , 62 reviewed in McLornan et al. 63 As the effects on T cells are largely due to the inhibition of JAK1, it is likely that JAK inhibitors that are more selective for JAK2, such as fedratinib and pacritinib, may be associated with a lower risk of infection. 64 However, whilst the trial data are promising, more time is needed to properly evaluate the impact of these ‘second‐generation’ JAK inhibitors on susceptibility to infection.

There does not appear to be a substantial increase in infections in patients with MPN receiving IFNα or hydroxycarbamide treatment as compared to patients not receiving cytoreductive treatment. 49 IFNα has been used for many decades as an immunomodulatory agent in MPN and remains the only licenced agent for which complete or major molecular remissions have been observed, supporting the hypothesis that modulating the immune system is important for clonal response. 18 Patients treated with IFNα have an altered balance of regulatory versus effector T cells, with studies reporting double the proportion of CD25+FoxP3+ Tregs in the CD4+ T‐cell compartment as compared to untreated patients or healthy donors, 65 and increased numbers of effector T cells. 66 However, it is unclear whether or how this mediates clonal responses.

Novel strategies for targeted immunotherapy in MPN

Many modern immunotherapy strategies have been pioneered in haematological cancers, such as immune checkpoint blockade in lymphomas, chimeric antigen receptor (CAR)‐T cell therapies in B‐cell neoplasms and antibody therapies in acute myeloid leukaemia (AML). Obstacles to similar successes in MPNs include a lack of ubiquitous and MPN clone‐specific cell surface targets, substantial cellular and/or molecular heterogeneity between patients and between disease subtypes, and an inability of the therapies to ignite anti‐tumour immunity due to the immune derangement resulting from the myeloid neoplasm.

There are many new therapies in clinical development for MPNs, including novel JAK inhibitors that are more selective for JAK2, ‘add‐on’ drugs that improve efficacy of ruxolitinib both in ruxolitinib‐naïve patients and in those who have a suboptimal response, and new targets (Table I and reviewed in Venugopal and Mascarenhas). 67 Many of these have anti‐inflammatory activity, such as the bromodomain and extra‐terminal motif (BET)‐inhibitor CPI‐0610· 68 In the next section, we outline some emerging targeted immunotherapy strategies that aim to ameliorate the deficiencies in anti‐tumoral immunity described above (Fig 2; also reviewed in Holmstrom et al. 69 ).

Table I.

Examples of immune modulating therapies employed in myeloproliferative neoplasms.

Strategy Disease References
Immunomodulatory agents currently in use Interferon‐α Myelofibrosis, PV and ET 91, 92, 93
Haematopoietic stem cell transplantation Myelofibrosis 94, 95
Corticosteroids Myelofibrosis 96
JAK inhibitors – ruxolitinib, fedratinib, pacritinib, momelotinib Myelofibrosis, PV and ET 97, 98, 99, 100
Immunomodulators – thalidomide, lenalidomide, pomalidomide Myelofibrosis 101, 102, 103
Targeted immunomodulatory agents in clinical‐stage development Anti‐CD123 – tagraxofusp Myelofibrosis 104
mutCALR vaccination Myelofibrosis and ET 88
Anti‐PD‐1 – pembrolizumab, nivolumab Myelofibrosis 89, 105

CD, cluster of differentiation; ET, essential thrombocythaemia; JAK, Janus kinase, mutCALR, mutant calreticulin; PD‐1, programmed cell death‐protein 1; PV, polycythaemia vera.

Fig 2.

Fig 2

Opportunities for immune‐based therapies in myeloproliferative neoplasm (MPN). (1) Identification of MPN neoantigens that elicit immune responses, such as JAK2 V617F and mutant CALR epitopes, and alternatively spliced proteins, such as eIF‐2α and protamine‐2 could be exploited for immune therapy; (2) Antibody therapies can be directed against cell surface proteins aberrantly expressed on the cell surface of MPN clone cells; (3) CAR‐T cells and adoptive T‐cell transfer confer T cells with the ability to target a specific protein, enhancing T‐cell recognition and destruction of mutant cells; (4) Blockade of the PD‐1 and CTLA‐4 inhibitory pathways on T cells enables better activation of naïve T cells by APCs such as dendritic cells, which present tumour antigens to the T cells for recognition. APC, antigen‐presenting cell; CALR, calreticulin; CAR, chimeric antigen receptor; CD, cluster of differentiation; CTLA‐4, cytotoxic T‐lymphocyte protein 4; eIF‐2α, eukaryotic initiation factor‐2α; IFN, interferon; JAK2, Janus kinase 2; MPN, myeloproliferative neoplasm; NF‐κB, nuclear factor κ‐light‐chain‐enhancer of activated B cells; PD‐1, programmed cell death‐protein 1; TNF, tumour necrosis factor.

Identifying neoantigens in MPNs

Tumour neoantigens are cancer cell‐specific molecules that activate T cells through MHC class I and II presentation by APCs. Neoantigens can result from gene mutations, fusions, alternative splicing and post‐translational protein modifications. Selective CD8+ cytotoxic T‐cell reactivity against epitopes within JAK2 V617F have been documented. 70 In this study, dendritic cell‐mediated stimulation of CD8+ T cells induced a significant release of IFNγ and TNFα, two markers of T‐cell activation, although this release required co‐stimulation with IL‐2, IL‐7 and IL‐12. 70 In contrast to the JAK2 V617F mutation, which is a single amino acid change, mutant CALR protein represents a large neoantigen that is expressed on the cell surface, and therefore is an attractive target for tumour‐specific immunotherapy in patients with mutCALR‐driven MPN. Immunoreactivity to mutant CALR has been observed in a subset of patients with MPN. 36 , 71 Responses against two neoantigens, CALRlong1 and CALRlong2 were reported in 50% and 42% of patients respectively. The response rate was found to be higher in ET than for myelofibrosis, with 80% of the patients with ET studied showing a response compared to only 36% of the patients with myelofibrosis. 71 Notably, memory T‐cell responses to CALR mutant peptides have also been detected in healthy donors without MPN. 36 This suggests that CALR may be a mutation that is frequently generated but held in check by the immune system, and that an inability to invoke responses against CALR mutant peptides may mediate immune evasion in patients who develop overt disease.

In MPNs, tumour neoantigens have also been identified by screening expression complementary DNA (cDNA) libraries with sera from patients. This method identified that peptides normally expressed only in the human testis, an immune‐privileged site, are over‐expressed in both CML and PV. 72 , 73 In one study, two proteins, eukaryotic translation initiation factor‐2A (eIF‐2A) and protamine 2, were shown to elicit immunoglobulin G (IgG) antibody reactions, in particular in those patients treated with IFNα, although expression was not specific to the mutant clone and there was no difference in expression levels of these antigens between clonal and wild‐type cells. 73

A recent study examining the granulocyte transcriptome profile in >100 patients with MPNs using RNA‐sequencing identified 13 gene fusions, 231 non‐synonymous single nucleotide variants and 21 insertions and deletions in 106 of 113 patients. 74 Using a ‘virtual peptide library’, the authors predicted that both CALR and JAK2 mutations resulted in a large number of neoantigens and 35 of these peptides were predicted to be strong MHC class I binders. The frequency of neoantigens was higher in patients with splicing factor 3B subunit 1 (SF3B1) mutations. This suggests a role for transcriptomics in designing personalised vaccine or adoptive cell‐based therapies. 74

Targeting the MPN clone with therapeutic antibodies

Antibody‐based therapies may be used to deliver cytotoxic drugs (e.g. antibody drug conjugates), induce antibody‐dependent cellular cytotoxicity (ADCC), or augment complement‐mediated cytotoxicity or the innate immune response. CD123, the receptor for IL‐3, has been identified as a therapeutic target in several myeloid malignancies including MPNs. Tagraxofusp is a targeted therapy directed to CD123 that comprises recombinant IL‐3 fused to a truncated diphtheria toxin. Following United States Food and Drug Administration (FDA) approval in blastic plasmacytoid dendritic neoplasm, a Phase I/II trial of tagraxofusp in MPN demonstrated some clinical efficacy, highlighting a potential role for CD123 targeting in this setting. 75

Single cell technologies are also increasingly being employed to uncover cell type‐specific targets. ‘Multi‐omic’ methods that enable simultaneous detection of gene mutations, transcriptome analysis and immunophenotyping of individual cells 10 , 76 , 77 , 78 offer the necessary resolution to identify cancer cell‐specific targets. For example, expression of the megakaryocyte gene G6B was found to be dramatically increased in stem/progenitor cells in myelofibrosis, specifically in JAK2 V617F‐mutant versus wild‐type stem cells within the same patients. 79 G6B was validated in vitro as a potential immunotherapy target, capable of supporting receptor‐mediated internalisation of a tool antibody.

As discussed previously, CD47 represents an attractive target for activating the innate immune system. Targeting CD47 blocks the ‘do not eat me’ signal from leukaemic cells resulting in their elimination by macrophages. 80 The use of targeting antibodies against CD47 are currently under development for the treatment of AML and MDS, 81 , 82 with demonstrated efficacy in combination with azacitidine including in patients with TP53‐mutant AML who are generally refractory to standard therapies. CD47 is also overexpressed in patients with MPNs and TP53 is a common driver of leukaemic transformation in patients with MPNs, suggesting potential utility here too. 83 , 84

Vaccines and adoptive cell therapy

To date, the development of vaccination and CAR‐T cell therapies in myeloid disease have focussed on AML and not extensively implemented in MPNs. Wilms tumour 1 (WT1) is frequently over‐expressed in AML, and has been targeted by several vaccine trials with modest success. 85 ‘Personalised’ vaccine approaches have also been tested, e.g. where a patient’s leukaemic blasts are fused with autologous dendritic cells, generating a hybridoma that is then reinfused into the patient. 86 There are several CAR‐T cell trials targeting putative AML antigens including C‐type lectin‐like molecule‐1 (CLL‐1), CD33 and CD123, 87 which if successful could also have activity in MPN, especially in patients with accelerated phase disease or post‐MPN AML.

The safety and efficacy of vaccination with a peptide derived from the CALR exon 9 mutation was recently tested in a phase I clinical trial in 10 patients with mutCALR MPN. 88 Although a decline in platelet count was observed in the initial 100‐day period after vaccination, unfortunately none of the patients had either a clinical or molecular response. In this study, IFNγ enzyme‐linked immunospot (ELISPOT) responses were observed in four of 10 patients before vaccination, and in four additional patients after vaccination, confirming an increase in in vitro anti‐cancer immune activity and a potential role for therapeutic vaccines combined with other modalities to enhance the efficacy of anti‐tumour T‐cell activity. 88

Immune checkpoint blockade

Increased expression of the immune checkpoint mediators programmed cell death‐protein 1 (PD‐1) and PD‐L1 dampens the ability of T cells to react to tumour neoantigens. Both JAK2 V617F and CALR mutations enhance PD‐L1 expression. 36 , 89 , 90 Increased expression of the immune checkpoint receptors PD‐1 and cytotoxic T‐lymphocyte protein 4 (CTLA‐4) on CD4+ and CD8+ T cells in MPN has been shown, 88 and blockade of PD‐1 and CTLA‐4 can recover T‐cell reactivity against mutCALR ex vivo, and result in the production of IFNγ and TNFα. 87 Early trials with pembrolizumab have shown a reduction in the number of PD‐1‐expressing cells and increased expansion of the CALR‐specific CD4+ and CD8+ T cells. 89 Trials with other checkpoint inhibitors including nivolumab and ipilimumab are currently underway.

Conclusions

The ‘holy grail’ in MPN therapy is a targeted approach able to selectively ablate mutant clone stem cells or provide a dramatic competitive advantage to wild‐type stem cells, enabling normal haematopoiesis to recover. There is a clear rationale for immunomodulatory therapy, strongly backed by decades of experience with agents such as steroids, IFN and JAK‐inhibitors. More comprehensive profiling of the immune cell and protein repertoire in MPNs will be an important step in facilitating design of effective immunotherapies, as well as providing better insights into mechanisms and extent of immunocompromise and vulnerability to infection. Future treatments that harness the immune system may be highly effective; however, a personalised approach may be necessary given the vast heterogeneity between patients.

Author contributions

All three authors agreed the scope of the article, reviewed the relevant literature, wrote the manuscript and designed the figures.

Acknowledgements

Bethan Psaila and Marie Strickland are supported by a Cancer Research UK Advanced Clinician Scientist Fellowship to Bethan Psaila, and Bethan Psaila also received support from the UK National Institute for Health Research (NIHR) Biomedical Research Centre Haematology Subtheme, an NIHR BRC Senior Fellowship and a Blood Cancer UK Project Grant. Lynn Quek is supported by a UK Medical Research Council (MRC) Clinician Scientist Fellowship. Figures created with BioRender.com.

REFERENCES

  • 1. Chen DS, Mellman I. Oncology meets immunology: the cancer‐immunity cycle. Immunity. 2013;39:1–10. [DOI] [PubMed] [Google Scholar]
  • 2. Sukari A, Nagasaka M, Al‐Hadidi A, Lum LG. Cancer immunology and immunotherapy. Anticancer Res. 2016;36:5593–606. [DOI] [PubMed] [Google Scholar]
  • 3. Sánchez‐Paulete AR, Teijeira A, Cueto FJ, Garasa S, Pérez‐Gracia JL, Sánchez‐Arráez A, et al. Antigen cross‐presentation and T‐cell cross‐priming in cancer immunology and immunotherapy. Ann Oncol. 2017;28:xii44–55. [DOI] [PubMed] [Google Scholar]
  • 4. Kleppe M, Kwak M, Koppikar P, Riester M, Keller M, Bastian L, et al. JAK‐STAT pathway activation in malignant and nonmalignant cells contributes to MPN pathogenesis and therapeutic response. Cancer Discov. 2015;5:316–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Kleppe M, Koche R, Zou L, van Galen P, Hill CE, Dong L, et al. Dual targeting of oncogenic activation and inflammatory signaling increases therapeutic efficacy in myeloproliferative neoplasms. Cancer Cell. 2018;33:785–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Jutzi JS, Mullally A. Remodeling the bone marrow microenvironment – a proposal for targeting pro‐inflammatory contributors in MPN. Front Immunol. 2020;11:2093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Garbati MR, Welgan CA, Landefeld SH, Newell LF, Agarwal A, Dunlap JB, et al. Mutant calreticulin‐expressing cells induce monocyte hyperreactivity through a paracrine mechanism. Am J Hematol. 2016;91:211–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Wang JC, Sindhu H, Chen C, Kundra A, Kafeel MI, Wong C, et al. Immune derangements in patients with myelofibrosis: the role of Treg, Th17, and sIL2Ralpha. PLoS One. 2015;10:e0116723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Giustacchini A, Thongjuea S, Barkas N, Woll PS, Povinelli BJ, Booth CA, et al. Single‐cell transcriptomics uncovers distinct molecular signatures of stem cells in chronic myeloid leukemia. Nat Med. 2017;23:692–702. [DOI] [PubMed] [Google Scholar]
  • 10. Rodriguez‐Meira A, Buck G, Clark SA, Povinelli BJ, Alcolea V, Louka E, et al. Unravelling intratumoral heterogeneity through high‐sensitivity single‐cell mutational analysis and parallel RNA sequencing. Mol Cell. 2019;73:1292–305.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Hoermann G, Cerny‐Reiterer S, Herrmann H, Blatt K, Bilban M, Gisslinger H, et al. Identification of oncostatin M as a JAK2 V617F‐dependent amplifier of cytokine production and bone marrow remodeling in myeloproliferative neoplasms. FASEB J. 2012;26:894–906. [DOI] [PubMed] [Google Scholar]
  • 12. Van Egeren D, Escabi J, Nguyen M, Liu S, Reilly CR, Patel S, et al. Reconstructing the lineage histories and differentiation trajectories of individual cancer cells in myeloproliferative neoplasms. Cell Stem Cell. 2021;28:514–23.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Wiliams N, Lee J, Moore L, Baxter EJ, Hewison J, Dawson KJ, et al. Phylogenetic reconstruction of myeloproliferative neoplasm reveals very early origins and lifelong evolution. BioRxiv. 2020; 10.1101/2020.11.09.374710 [DOI] [Google Scholar]
  • 14. Pang WW, Price EA, Sahoo D, Beerman I, Maloney WJ, Rossi DJ, et al. Human bone marrow hematopoietic stem cells are increased in frequency and myeloid‐biased with age. Proc Natl Acad Sci. 2011;108:20012–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Pierre RV, Hoagland HC. Age‐associated aneuploidy: loss of Y chromosome from human bone marrow cells with aging. Cancer. 1972;30:889–94. [DOI] [PubMed] [Google Scholar]
  • 16. Valletta S, Thomas A, Meng Y, Ren X, Drissen R, Sengul H, et al. Micro‐environmental sensing by bone marrow stroma identifies IL‐6 and TGFbeta1 as regulators of hematopoietic ageing. Nat Commun. 2020;11:4075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Fleischman AG, Aichberger KJ, Luty SB, Bumm TG, Petersen CL, Doratotaj S, et al. TNFalpha facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. Blood. 2011;118:6392–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Tong J, Sun T, Ma S, Zhao Y, Ju M, Gao Y, et al. Hematopoietic stem cell heterogeneity is linked to the initiation and therapeutic response of myeloproliferative neoplasms. Cell Stem Cell. 2021;28:502–13.e6. [DOI] [PubMed] [Google Scholar]
  • 19. Tong J, Sun T, Ma S, Zhao Y, Ju M, Gao Y. Hematopoietic stem cell heterogeneity is linked to the initiation and therapeutic response of myeloproliferative neoplasms. Cell Stem Cell. 2021;28(3):502–513.e6. 10.1016/j.stem.2021.01.018 [DOI] [PubMed] [Google Scholar]
  • 20. Rao TN, Hansen N, Stetka J, Luque Paz D, Kalmer M, Hilfiker J. JAK2‐V617F and interferon‐α induce megakaryocyte‐biased stem cells characterized by decreased long‐term functionality. Blood. 2021;137(16):2139–2151. 10.1182/blood.2020005563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Pourcelot E, Trocme C, Mondet J, Bailly S, Toussaint B, Mossuz P. Cytokine profiles in polycythemia vera and essential thrombocythemia patients: clinical implications. Exp Hematol. 2014;42:360–8. [DOI] [PubMed] [Google Scholar]
  • 22. Tefferi A, Vaidya R, Caramazza D, Finke C, Lasho T, Pardanani A. Circulating interleukin (IL)‐8, IL‐2R, IL‐12, and IL‐15 levels are independently prognostic in primary myelofibrosis: a comprehensive cytokine profiling study. J Clin Oncol. 2011;29:1356–63. [DOI] [PubMed] [Google Scholar]
  • 23. Panteli KE, Hatzimichael EC, Bouranta PK, Katsaraki A, Seferiadis K, Stebbing J, et al. Serum interleukin (IL)‐1, IL‐2, sIL‐2Ra, IL‐6 and thrombopoietin levels in patients with chronic myeloproliferative diseases. Br J Haematol. 2005;130:709–15. [DOI] [PubMed] [Google Scholar]
  • 24. Tefferi A. Pathogenesis of myelofibrosis with myeloid metaplasia. J Clin Oncol. 2005;23(33):8520–30. [DOI] [PubMed] [Google Scholar]
  • 25. Bock O, Höftmann J, Theophile K, Hussein K, Wiese B, Schlué J, et al. Bone morphogenetic proteins are overexpressed in the bone marrow of primary myelofibrosis and are apparently induced by fibrogenic cytokines. American J Pathol. 2008;172:951–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Geyer HL, Dueck AC, Scherber RM, Mesa RA. Impact of inflammation on myeloproliferative neoplasm symptom development. Mediators Inflamm. 2015;2015:284706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Skov V, Riley CH, Thomassen M, Larsen TS, Jensen MK, Bjerrum OW, et al. Whole blood transcriptional profiling reveals significant down‐regulation of human leukocyte antigen class I and II genes in essential thrombocythemia, polycythemia vera and myelofibrosis. Leuk Lymphoma. 2013;54:2269–73. [DOI] [PubMed] [Google Scholar]
  • 28. Romano M, Sollazzo D, Trabanelli S, Barone M, Polverelli N, Perricone M, et al. Mutations in JAK2 and Calreticulin genes are associated with specific alterations of the immune system in myelofibrosis. Oncoimmunology. 2017;6:e1345402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Wang JC, Kundra A, Andrei M, Baptiste S, Chen C, Wong C, et al. Myeloid‐derived suppressor cells in patients with myeloproliferative neoplasm. Leuk Res. 2016;43:39–43. [DOI] [PubMed] [Google Scholar]
  • 30. Majumder D, Bandyopadhyay D, Chandra S, Mukhopadhayay A, Mukherjee N, Bandyopadhyay SK, et al. Analysis of HLA class Ia transcripts in human leukaemias. Immunogenetics. 2005;57:579–89. [DOI] [PubMed] [Google Scholar]
  • 31. Chao MP, Jaiswal S, Weissman‐Tsukamoto R, Alizadeh AA, Gentles AJ, Volkmer J, et al. Calreticulin is the dominant pro‐phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci Transl Med. 2010;2:63ra94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Chao MP, Weissman IL, Majeti R. The CD47‐SIRPalpha pathway in cancer immune evasion and potential therapeutic implications. Curr Opin Immunol. 2012;24:225–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Chao MP, Jaiswal S, Weissman‐Tsukamoto R, Alizadeh AA, Gentles AJ, Volkmer J, et al. Calreticulin is the dominant pro‐phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci Transl Med. 2010;2:3001375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Liu P, Zhao L, Loos F, Marty C, Xie W, Martins I, et al. Immunosuppression by mutated calreticulin released from malignant cells. Mol Cell. 2020;77:748–60.e9. [DOI] [PubMed] [Google Scholar]
  • 35. Riley CH, Hansen M, Brimnes MK, Hasselbalch HC, Bjerrum OW, Straten PT, et al. Expansion of circulating CD56bright natural killer cells in patients with JAK2‐positive chronic myeloproliferative neoplasms during treatment with interferon‐alpha. Eur J Haematol. 2015;94:227–34. [DOI] [PubMed] [Google Scholar]
  • 36. Cimen Bozkus C, Roudko V, Finnigan JP, Mascarenhas J, Hoffman R, Iancu‐Rubin C, et al. Immune checkpoint blockade enhances shared neoantigen‐induced T‐cell immunity directed against mutated calreticulin in myeloproliferative neoplasms. Cancer Discov. 2019;9:1192–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492–9. [DOI] [PubMed] [Google Scholar]
  • 38. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15:486–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Landtblom AR, Andersson TL, Dickman PW, Smedby KE, Eloranta S, Batyrbekova N, et al. Risk of infections in patients with myeloproliferative neoplasms – a population‐based cohort study of 8363 patients. Leukemia. 2021;35:476–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Caocci G, Simula MP, Ghiani S, Mulas O, Mainas G, Atzeni S, et al. Increased incidence of infection in patients with myelofibrosis and transfusion‐associated iron overload in the clinical setting. Int J Hematol. 2020;111:614–8. [DOI] [PubMed] [Google Scholar]
  • 41. Lee LY, Cazier JB, Starkey T, Briggs SE, Arnold R, Bisht V, et al. COVID‐19 prevalence and mortality in patients with cancer and the effect of primary tumour subtype and patient demographics: a prospective cohort study. Lancet Oncol. 2020;21:1309–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. He W, Chen L, Chen LI, Yuan G, Fang Y, Chen W, et al. COVID‐19 in persons with haematological cancers. Leukemia. 2020;34:1637–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Wang Q, Berger NA, Xu R. When hematologic malignancies meet COVID‐19 in the United States: infections, death and disparities. Blood Rev. 2020;47:100775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Fattizzo B, Giannotta JA, Barcellini W, Barbanti MC, Bucelli C, Cassin R, et al. Ensuring continuity of care of hematologic patients during COVID‐19 pandemic in a tertiary hospital in Lombardy (Italy). Blood Adv. 2020;4:2996–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Vijenthira A, Gong IY, Fox TA, Booth S, Cook G, Fattizzo B, et al. Outcomes of patients with hematologic malignancies and COVID‐19: A systematic review and meta‐analysis of 3377 patients. Blood. 2020;136:2881–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Salisbury RA, Curto‐Garvia N, Chen F, Polzella P, Godfrey A, Russell J, et al. Results of a national UK audit of COVID‐19 infection in patients with myeloproliferative neoplasms. Leukaemia. 2021;35:2424–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Alimam S, Ann Timms J, Harrison CN, Dillon R, Mare T, DeLavallade H, et al. Altered immune response to the annual influenza A vaccine in patients with myeloproliferative neoplasms. Br J Haematol. 2020;193:150–4. [DOI] [PubMed] [Google Scholar]
  • 48. Chowdhury O, Bruguier H, Mallett G, Sousos N, Crozier K, Allman C, et al. Impaired antibody response to COVID‐19 vaccination in patients with chronic myeloid neoplasms. Br J Haematol. 2021;194:1010–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Crodel CC, Jentsch‐Ullrich K, Koschmieder S, Kämpfe D, Griesshammer M, Döhner K, et al. Frequency of infections in 948 MPN patients: a prospective multicenter patient‐reported pilot study. Leukemia. 2020;34:1949–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, et al. A double‐blind, placebo‐controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366:799–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Wathes R, Moule S, Milojkovic D. Progressive multifocal leukoencephalopathy associated with ruxolitinib. N Engl J Med. 2013;369:197–8. [DOI] [PubMed] [Google Scholar]
  • 52. Caocci G, Murgia F, Podda L, Solinas A, Atzeni S, La Nasa G. Reactivation of hepatitis B virus infection following ruxolitinib treatment in a patient with myelofibrosis. Leukemia. 2014;28:225–7. [DOI] [PubMed] [Google Scholar]
  • 53. Hopman RK, Lawrence SJ, Oh ST. Disseminated tuberculosis associated with ruxolitinib. Leukemia. 2014;28:1750–1. [DOI] [PubMed] [Google Scholar]
  • 54. Arana Yi C, Tam CS, Verstovsek S. Efficacy and safety of ruxolitinib in the treatment of patients with myelofibrosis. Future Oncol. 2015;11:719–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Lussana F, Cattaneo M, Rambaldi A, Squizzato A. Ruxolitinib‐associated infections: a systematic review and meta‐analysis. Am J Hematol. 2018;93:339–47. [DOI] [PubMed] [Google Scholar]
  • 56. Shamil E, Cunningham D, Wong BL, Jani P. Ruxolitinib associated tuberculosis presenting as a neck lump. Case Rep Infect Dis. 2015;2015:284168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Verstovsek S, Kantarjian H, Mesa RA, Pardanani AD, Cortes‐Franco J, Thomas DA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med. 2010;363:1117–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Yeleswaram S, Smith P, Burn T, Covington M, Juvekar A, Li Y, et al. Inhibition of cytokine signaling by ruxolitinib and implications for COVID‐19 treatment. Clin Immunol. 2020;218:108517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Parampalli Yajnanarayana S, Stubig T, Cornez I, Alchalby H, Schonberg K, Rudolph J, et al. JAK1/2 inhibition impairs T cell function in vitro and in patients with myeloproliferative neoplasms. Br J Haematol. 2015;169:824–33. [DOI] [PubMed] [Google Scholar]
  • 60. Heine A, Held SAE, Daecke SN, Wallner S, Yajnanarayana SP, Kurts C, et al. The JAK‐inhibitor ruxolitinib impairs dendritic cell function in vitro and in vivo. Blood. 2013;122:1192–202. [DOI] [PubMed] [Google Scholar]
  • 61. Massa M, Rosti V, Campanelli R, Fois G, Barosi G. Rapid and long‐lasting decrease of T‐regulatory cells in patients with myelofibrosis treated with ruxolitinib. Leukemia. 2014;28:449–51. [DOI] [PubMed] [Google Scholar]
  • 62. Keohane C, Kordasti S, Seidl T, Perez Abellan P, Thomas NS, Harrison CN, et al. JAK inhibition induces silencing of T Helper cytokine secretion and a profound reduction in T regulatory cells. Br J Haematol. 2015;171:60–73. [DOI] [PubMed] [Google Scholar]
  • 63. McLornan DP, Khan AA, Harrison CN. Immunological consequences of JAK inhibition: friend or foe? Curr Hematol Malig Rep. 2015;10:370–9. [DOI] [PubMed] [Google Scholar]
  • 64. Talpaz M, Kiladjian JJ. Fedratinib, a newly approved treatment for patients with myeloproliferative neoplasm‐associated myelofibrosis. Leukemia. 2021;35:1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Riley CH, Jensen MK, Brimnes MK, Hasselbalch HC, Bjerrum OW, Straten PT, et al. Increase in circulating CD4(+)CD25(+)Foxp3(+) T cells in patients with Philadelphia‐negative chronic myeloproliferative neoplasms during treatment with IFN‐alpha. Blood. 2011;118:2170–3. [DOI] [PubMed] [Google Scholar]
  • 66. Kovacsovics‐Bankowski M, Kelley TW, Efimova O, Kim SJ, Wilson A, Swierczek S, et al. Changes in peripheral blood lymphocytes in polycythemia vera and essential thrombocythemia patients treated with pegylated‐interferon alpha and correlation with JAK2(V617F) allelic burden. Exp Hematol Oncol. 2015;5:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Venugopal S, Mascarenhas J. Novel therapeutics in myeloproliferative neoplasms. J Hematol Oncol. 2020;13:162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Kremyanskaya M, Hoffman R, Mascarenhas J, Verstovsek S, Mertz J, Garner F, et al. A phase 2 study of CPI‐0610, a Bromodomain and Extraterminal (BET) inhibitor, in patients with Myelofibrosis (MF). Blood. 2018;132(Suppl 1):5481. [Google Scholar]
  • 69. Holmström MO, Hasselbalch HC, Andersen MH. Cancer immune therapy for philadelphia chromosome‐negative chronic myeloproliferative neoplasms. Cancers (Basel). 2020;12:1763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Holmström MO, Hjortsø MD, Ahmad SM, Met Ö, Martinenaite E, Riley C, et al. The JAK2V617F mutation is a target for specific T cells in the JAK2V617F‐positive myeloproliferative neoplasms. Leukemia. 2017;31:495–8. [DOI] [PubMed] [Google Scholar]
  • 71. Holmström MO, Riley CH, Svane IM, Hasselbalch HC, Andersen MH. The CALR exon 9 mutations are shared neoantigens in patients with CALR mutant chronic myeloproliferative neoplasms. Leukemia. 2016;30:2413–6. [DOI] [PubMed] [Google Scholar]
  • 72. Yan Y, Phan L, Yang F, Talpaz M, Yang YU, Xiong Z, et al. A novel mechanism of alternative promoter and splicing regulates the epitope generation of tumor antigen CML66‐L. J Immunol. 2004;172:651–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Xiong Z, Yan Y, Liu E, Silver RT, Verstovsek S, Yang F, et al. Novel tumor antigens elicit anti‐tumor humoral immune reactions in a subset of patients with polycythemia vera. Clin Immunol. 2007;122:279–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Schischlik F, Jäger R, Rosebrock F, Hug E, Schuster M, Holly R, et al. Mutational landscape of the transcriptome offers putative targets for immunotherapy of myeloproliferative neoplasms. Blood. 2019;134:199–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Krishnan A, Pagane M, Roshal M, McGovern E, Stone‐Molloy Z, Chen J, et al. Evaluation of tagraxofusp (SL‐401) alone and in combination with ruxolitinib for the treatment of myeloproliferative neoplasms. Blood. 2019;134(Suppl 1):2967. [Google Scholar]
  • 76. Nam AS, Kim KT, Chaligne R, Izzo F, Ang C, Taylor J, et al. Somatic mutations and cell identity linked by Genotyping of Transcriptomes. Nature. 2019;571:355–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Van Egeren D, Escabi J, Nguyen M, Liu S, Reilly CR, Patel S, et al. Reconstructing the lineage histories and differentiation trajectories of individual cancer cells in JAK2‐mutant myeloproliferative neoplasms. Cell Stem Cell. 2021;28:514–523.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Miles LA, Bowman RL, Merlinsky TR, Csete IS, Ooi AT, Durruthy‐Durruthy R, et al. Single‐cell mutational profiling delineates clonal trajectories in myeloid malignancies. Nature. 2020;587:477–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Psaila B, Wang G, Rodriguez‐Meira A, Li R, Heuston EF, Murphy L, et al. Single‐cell analyses reveal megakaryocyte‐biased hematopoiesis in myelofibrosis and identify mutant clone‐specific targets. Mol Cell. 2020;78:477–92.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Chao MP, Alizadeh AA, Tang C, Jan M, Weissman‐Tsukamoto R, Zhao F, et al. Therapeutic antibody targeting of CD47 eliminates human acute lymphoblastic leukemia. Cancer Res. 2011;71:1374–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Liu J, Wang L, Zhao F, Tseng S, Narayanan C, Shura L, et al. Pre‐clinical development of a humanized anti‐CD47 antibody with anti‐cancer therapeutic potential. PLoS One. 2015;10:e0137345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Zeidan AM, DeAngelo DJ, Palmer JM, Seet CS, Tallman MS, Wei X, et al. A phase I study of CC‐90002, a monoclonal antibody targeting CD47, in patients with relapsed and/or refractory (R/R) acute myeloid leukemia (AML) and high‐risk myelodysplastic syndromes (MDS): final results. Blood. 2019;134(Suppl 1):1320. [Google Scholar]
  • 83. Boasman K, Simmonds MJ, Rinaldi CR. CD47 but not calreticulin is over expressed in patients with myeloproliferative neoplasms. Blood. 2018;132(Suppl 1):5492. [Google Scholar]
  • 84. Boasman K, Simmonds MJ, Rinaldi CR. Expression of CD47 and CALR in myeloproliferative neoplasms and myelodysplastic syndrome: potential new therapeutical targets. Blood. 2019;134(Suppl 1):5031. [Google Scholar]
  • 85. Oka Y, Tsuboi A, Elisseeva OA, Udaka K, Sugiyama H. WT1 as a novel target antigen for cancer immunotherapy. Curr Cancer Drug Targets. 2002;2:45–54. [DOI] [PubMed] [Google Scholar]
  • 86. Rosenblatt J, Stone RM, Uhl L, Neuberg D, Joyce R, Levine JD, et al. Individualized vaccination of AML patients in remission is associated with induction of antileukemia immunity and prolonged remissions. Sci Transl Med. 2016;8:368ra171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Mardiana S, Gill S. CAR T cells for acute myeloid leukemia: state of the art and future directions. Front Oncol. 2020;10:697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Handlos Grauslund J, Holmström MO, Jørgensen NG, Klausen U, Weis‐Banke SE, El Fassi D, et al. Therapeutic cancer vaccination with a peptide derived from the calreticulin exon 9 mutations induces strong cellular immune responses in patients with CALR‐mutant chronic myeloproliferative neoplasms. Front Oncol. 2021;11:637420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Holmström MO, Hasselbalch Riley C, Skov V, Svane IM, Hasselbalch HC, Andersen MH. Spontaneous T‐cell responses against the immune check point programmed‐death‐ligand 1 (PD‐L1) in patients with chronic myeloproliferative neoplasms correlate with disease stage and clinical response. Oncoimmunology. 2018;7:e1433521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Prestipino A, Emhardt AJ, Aumann K, O'Sullivan D, Gorantla SP, Duquesne S, et al. Oncogenic JAK2(V617F) causes PD‐L1 expression, mediating immune escape in myeloproliferative neoplasms. Sci Transl Med. 2018;10:eaam7729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Pizzi M, Silver RT, Barel A, Orazi A. Recombinant interferon‐α in myelofibrosis reduces bone marrow fibrosis, improves its morphology and is associated with clinical response. Mod Pathol. 2015;28:1315–23. [DOI] [PubMed] [Google Scholar]
  • 92. Abu‐Zeinah G, Krichevsky S, Cruz T, Hoberman G, Jaber D, Savage N, et al. Interferon‐alpha for treating polycythemia vera yields improved myelofibrosis‐free and overall survival. Leukemia. 2021;35:2592–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Bewersdorf JP, Giri S, Wang R, Podoltsev N, Williams RT, Tallman MS, et al. Interferon alpha therapy in essential thrombocythemia and polycythemia vera – a systematic review and meta‐analysis. Leukemia. 2021;35:1643–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Savani M, Dulery R, Bazarbachi AH, Mohty R, Brissot E, Malard F, et al. Allogeneic haematopoietic cell transplantation for myelofibrosis: a real‐life perspective. Br J Haematol. 2021. 10.1111/bjh.17469 [DOI] [PubMed] [Google Scholar]
  • 95. Davidson MB, Gupta V. Application of stem cell therapy in myelofibrosis. Hematol Oncol Clin North Am. 2021;35:391–407. [DOI] [PubMed] [Google Scholar]
  • 96. Quintás‐Cardama A, Kantarjian HM, Manshouri T, Thomas D, Cortes J, Ravandi F, et al. Lenalidomide plus prednisone results in durable clinical, histopathologic, and molecular responses in patients with myelofibrosis. J Clin Oncol. 2009;27:4760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Tefferi A, Begna K, Hogan WJ, Litzow MR, Hanson CA, Pardanani A. Long‐term outcome of treatment with ruxolitinib in myelofibrosis. Blood. 2011;118(21):1752. [DOI] [PubMed] [Google Scholar]
  • 98. Vannucchi AM, Kiladjian JJ, Griesshammer M, Masszi T, Durrant S, Passamonti F, et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med. 2015;372:426–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Verstovsek S, Passamonti F, Rambaldi A, Barosi G, Rumi E, Gattoni E, et al. Ruxolitinib for essential thrombocythemia refractory to or intolerant of hydroxyurea: long‐term phase 2 study results. Blood. 2017;130:1768–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Asher S, McLornan DP, Harrison CN. Current and future therapies for myelofibrosis. Blood Rev. 2020;42:100715. [DOI] [PubMed] [Google Scholar]
  • 101. Tefferi A, Cortes J, Verstovsek S, Mesa RA, Thomas D, Lasho TL, et al. Lenalidomide therapy in myelofibrosis with myeloid metaplasia. Blood. 2006;108:1158–64. [DOI] [PubMed] [Google Scholar]
  • 102. Thomas DA, Giles FJ, Albitar M, Cortes JE, Verstovsek S, Faderl S, et al. Thalidomide therapy for myelofibrosis with myeloid metaplasia. Cancer. 2006;106:1974–84. [DOI] [PubMed] [Google Scholar]
  • 103. Tefferi A, Al‐Ali HK, Barosi G, Devos T, Gisslinger H, Jiang Q, et al. A randomized study of pomalidomide vs placebo in persons with myeloproliferative neoplasm‐associated myelofibrosis and RBC‐transfusion dependence. Leukemia. 2017;31:896–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Pemmaraju N, Gupta V, Ali H, Yacoub A, Wang E, Lee S, et al. MPN‐377: updated results from a phase 1/2 clinical trial of tagraxofusp, a CD123‐targeted therapy, in patients with poor‐risk myelofibrosis. Clin Lymphoma Myeloma Leuk. 2020;20(Suppl 1):S338–9. [Google Scholar]
  • 105. Abou Dalle I, Kantarjian H, Daver N, Masarova L, Pemmaraju N, Bose P, et al. Phase II study of single‐agent nivolumab in patients with myelofibrosis. Ann Hematol. 2021. 10.1007/s00277-021-04618-5 [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Haematology are provided here courtesy of Wiley

RESOURCES